1
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
2
|
Bostancı Ö, Karaduman E, Yılmaz AK, Kabadayı M, Bilgiç S. Midterm Effects of SARS-CoV-2 on Respiratory Function in Judokas With and Without Exercise-Induced Bronchoconstriction: A Retrospective Study. Clin J Sport Med 2025; 35:162-168. [PMID: 39626059 DOI: 10.1097/jsm.0000000000001312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/07/2023] [Indexed: 01/04/2025]
Abstract
OBJECTIVES The clinical consequences of coronavirus infection in elite judokas with exercise-induced bronchoconstriction (EIB) are unclear. We aimed to determine potential respiratory function abnormalities and recovery in athletes with and without EIB after severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. DESIGN Retrospective cohort study. SETTING Türkiye Olympic Preparation Centre. PARTICIPANTS This retrospective study analyzed data collected from 25 consecutive elite judokas diagnosed with and without EIB and SARS-CoV-2 infection, routinely followed at an Olympic Sports Center between September 2020 and 2021. INDEPENDENT VARIABLES Respiratory muscle strength and pulmonary function data were collected before and up to 90 days after SARS-CoV-2 infection. MAIN OUTCOME MEASURES Measurements included maximal inspiratory pressure (MIP), maximal expiratory pressure (MEP), forced expiratory volume in 1 second (FEV 1 ), forced vital capacity (FVC), FEV 1 /FVC ratio, and peak expiratory flow (PEF). RESULTS Infected athletes with EIB had more markedly reduced respiratory muscle strength and pulmonary function than those without EIB. Maximal inspiratory pressure was decreased by 14% and MEP by 8% from baseline in infected athletes with EIB during follow-up. Likewise, FEV 1 and FVC decreased by 4%. Maximal inspiratory pressure, MEP, FEV 1 , and FVC remained abnormal after 90 days of SARS-CoV-2 infection in EIB athletes but normalized rapidly in non-EIB athletes. Peak expiratory flow seemed unaffected during follow-up. Exercise-induced bronchoconstriction severity was moderately correlated with the maximum fall in MEP during follow-up. CONCLUSIONS Severe acute respiratory syndrome coronavirus-2 infection notably decreases respiratory muscle strength and pulmonary function in judokas, especially those with pre-existing EIB, thereby prolonging spontaneous recovery time.
Collapse
Affiliation(s)
- Özgür Bostancı
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Emre Karaduman
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Ali Kerim Yılmaz
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Menderes Kabadayı
- Faculty of Sport Sciences, University of Ondokuz Mayıs, Samsun, Türkiye ; and
| | - Sait Bilgiç
- Faculty of Medicine, University of Ondokuz Mayıs, Samsun, Türkiye
| |
Collapse
|
3
|
Richards A, Khalil AS, Friesen M, Whitfield TW, Gao X, Lungjangwa T, Kamm RD, Wan Z, Gehrke L, Mooney D, Jaenisch R. SARS-CoV-2 infection of human pluripotent stem cell-derived vascular cells reveals smooth muscle cells as key mediators of vascular pathology during infection. Nat Commun 2024; 15:10754. [PMID: 39737992 PMCID: PMC11685814 DOI: 10.1038/s41467-024-54917-4] [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: 08/06/2023] [Accepted: 11/22/2024] [Indexed: 01/01/2025] Open
Abstract
Although respiratory symptoms are the most prevalent disease manifestation of infection by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), nearly 20% of hospitalized patients are at risk for thromboembolic events. This prothrombotic state is considered a key factor in the increased risk of stroke, which is observed clinically during both acute infection and long after symptoms clear. Here, we develop a model of SARS-CoV-2 infection using human-induced pluripotent stem cell-derived endothelial cells (ECs), pericytes (PCs), and smooth muscle cells (SMCs) to recapitulate the vascular pathology associated with SARS-CoV-2 exposure. Our results demonstrate that perivascular cells, particularly SMCs, are a susceptible vascular target for SARS-CoV-2 infection. Utilizing RNA sequencing, we characterize the transcriptomic changes accompanying SARS-CoV-2 infection of SMCs, PCs, and ECs. We observe that infected SMCs shift to a pro-inflammatory state and increase the expression of key mediators of the coagulation cascade. Further, we show human ECs exposed to the secretome of infected SMCs produce hemostatic factors that contribute to vascular dysfunction despite not being susceptible to direct infection. The findings here recapitulate observations from patient sera in human COVID-19 patients and provide mechanistic insight into the unique vascular implications of SARS-CoV-2 infection at a cellular level.
Collapse
Affiliation(s)
- Alexsia Richards
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Andrew S Khalil
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Max Friesen
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Troy W Whitfield
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Xinlei Gao
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Tenzin Lungjangwa
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Lee Gehrke
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - David Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
4
|
Belete MA, Anley DT, Tsega SS, Moges N, Anteneh RM, Zemene MA, Gebeyehu AA, Dessie AM, Kebede N, Chanie ES, Alemayehu E. The potential of circulating microRNAs as novel diagnostic biomarkers of COVID-19: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:1011. [PMID: 39300343 PMCID: PMC11414062 DOI: 10.1186/s12879-024-09915-8] [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: 06/11/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
INTRODUCTION The COVID-19 pandemic has caused an unprecedented health threat globally, necessitating innovative and efficient diagnostic approaches for timely identification of infected individuals. Despite few emerging reports, the clinical utility of circulating microRNAs (miRNAs) in early and accurate diagnosis of COVID-19 is not well-evidenced. Hence, this meta-analysis aimed to explore the diagnostic potential of circulating miRNAs for COVID-19. The protocol for this study was officially recorded on PROSPERO under registration number CRD42023494959. METHODS Electronic databases including Embase, PubMed, Scopus, and other sources were exhaustively searched to recover studies published until 16th January, 2024. Pooled specificity, sensitivity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic ratio (DOR), positive predictive value (PPV), negative predictive value (NPV), and area under the curve (AUC) were computed from the metadata using Stata 14.0 software. Risk of bias appraisal of included articles was carried out using Review Manager (Rev-Man) 5.3 package through the modified QUADAS-2 tool. Subgroup, heterogeneity, meta-regression and sensitivity analyses were undertaken. Publication bias and clinical applicability were also evaluated via Deeks' funnel plot and Fagan nomogram (scattergram), respectively. RESULT A total of 43 studies from 13 eligible articles, involving 5175 participants (3281 COVID-19 patients and 1894 healthy controls), were analyzed. Our results depicted that miRNAs exhibit enhanced pooled specificity 0.91 (95% CI: 0.88-0.94), sensitivity 0.94 (95% CI: 0.91-0.96), DOR of 159 (95% CI: 87-288), and AUC values of 0.97 (95% CI: 0.95-0.98) with high pooled PPV 96% (95% CI: 94-97%) and NPV 88% (95% CI: 86-90%) values. Additionally, highest diagnostic capacity was observed in studies involving larger sample size (greater than 100) and those involving the African population, demonstrating consistent diagnostic effectiveness across various specimen types. Notably, a total of 12 distinct miRNAs were identified as suitable for both exclusion and confirmation of COVID-19 cases, denoting their potential clinical applicability. CONCLUSION Our study depicted that miRNAs show significantly high diagnostic accuracy in differentiating COVID-19 patients from healthy counterparts, suggesting their possible use as viable biomarkers. Nonetheless, thorough and wide-ranging longitudinal researches are necessary to confirm the clinical applicability of miRNAs in diagnosing COVID-19.
Collapse
Affiliation(s)
- Melaku Ashagrie Belete
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia.
| | - Denekew Tenaw Anley
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Sintayehu Simie Tsega
- Department of Medical Nursing, School of Nursing, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Natnael Moges
- Department of Pediatrics and Child Health Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Rahel Mulatie Anteneh
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Melkamu Aderajew Zemene
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Asaye Alamneh Gebeyehu
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Anteneh Mengist Dessie
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Natnael Kebede
- Department of Health Promotion, School of Public Health College of Medicine Health Sciences, Wollo University, Dessie, Ethiopia
| | - Ermias Sisay Chanie
- Department of Pediatrics and Child Health Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Ermiyas Alemayehu
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| |
Collapse
|
5
|
Skeeters S, Bagale K, Stepanyuk G, Thieker D, Aguhob A, Chan KK, Dutzar B, Shalygin S, Shajahan A, Yang X, DaRosa PA, Frazier E, Sauer MM, Bogatzki L, Byrnes-Blake KA, Song Y, Azadi P, Tarcha E, Zhang L, Procko E. Modulation of the pharmacokinetics of soluble ACE2 decoy receptors through glycosylation. Mol Ther Methods Clin Dev 2024; 32:101301. [PMID: 39185275 PMCID: PMC11342882 DOI: 10.1016/j.omtm.2024.101301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
The Spike of SARS-CoV-2 recognizes a transmembrane protease, angiotensin-converting enzyme 2 (ACE2), on host cells to initiate infection. Soluble derivatives of ACE2, in which Spike affinity is enhanced and the protein is fused to Fc of an immunoglobulin, are potent decoy receptors that reduce disease in animal models of COVID-19. Mutations were introduced into an ACE2 decoy receptor, including adding custom N-glycosylation sites and a cavity-filling substitution together with Fc modifications, which increased the decoy's catalytic activity and provided small to moderate enhancements of pharmacokinetics following intravenous and subcutaneous administration in humanized FcRn mice. Most prominently, sialylation of native glycans increases exposures by orders of magnitude, and the optimized decoy is therapeutically efficacious in a mouse COVID-19 model. Ultimately, an engineered and highly sialylated decoy receptor produced using methods suitable for manufacture of representative drug substance has high exposure with a 5- to 9-day half-life. Finally, peptide epitopes at mutated sites in the decoys generally have low binding to common HLA class II alleles and the predicted immunogenicity risk is low. Overall, glycosylation is a critical molecular attribute of ACE2 decoy receptors and modifications that combine tighter blocking of Spike with enhanced pharmacokinetics elevate this class of molecules as viable drug candidates.
Collapse
Affiliation(s)
| | - Kamal Bagale
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | - Sergei Shalygin
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Xu Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | | - Yifan Song
- Cyrus Biotechnology, Seattle, WA 98121, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Erik Procko
- Cyrus Biotechnology, Seattle, WA 98121, USA
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
6
|
Turner DL, Amoozadeh S, Baric H, Stanley E, Werder RB. Building a human lung from pluripotent stem cells to model respiratory viral infections. Respir Res 2024; 25:277. [PMID: 39010108 PMCID: PMC11251358 DOI: 10.1186/s12931-024-02912-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
To protect against the constant threat of inhaled pathogens, the lung is equipped with cellular defenders. In coordination with resident and recruited immune cells, this defence is initiated by the airway and alveolar epithelium following their infection with respiratory viruses. Further support for viral clearance and infection resolution is provided by adjacent endothelial and stromal cells. However, even with these defence mechanisms, respiratory viral infections are a significant global health concern, causing substantial morbidity, socioeconomic losses, and mortality, underlining the need to develop effective vaccines and antiviral medications. In turn, the identification of new treatment options for respiratory infections is critically dependent on the availability of tractable in vitro experimental models that faithfully recapitulate key aspects of lung physiology. For such models to be informative, it is important these models incorporate human-derived, physiologically relevant versions of all cell types that normally form part of the lungs anti-viral response. This review proposes a guideline using human induced pluripotent stem cells (iPSCs) to create all the disease-relevant cell types. iPSCs can be differentiated into lung epithelium, innate immune cells, endothelial cells, and fibroblasts at a large scale, recapitulating in vivo functions and providing genetic tractability. We advocate for building comprehensive iPSC-derived in vitro models of both proximal and distal lung regions to better understand and model respiratory infections, including interactions with chronic lung diseases.
Collapse
Affiliation(s)
- Declan L Turner
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Sahel Amoozadeh
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Hannah Baric
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Ed Stanley
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, 3056, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia.
| |
Collapse
|
7
|
Pace E, Di Vincenzo S, Ferraro M, Lanata L, Scaglione F. Role of airway epithelium in viral respiratory infections: Can carbocysteine prevent or mitigate them? Immunology 2024; 172:329-342. [PMID: 38354831 DOI: 10.1111/imm.13762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Alterations in airway epithelial homeostasis increase viral respiratory infections risk. Viral infections frequently are associated with chronic obstructive pulmonary disease (COPD) exacerbations, events that dramatically promote disease progression. Mechanism promoting the main respiratory viruses entry and virus-evocated innate and adaptive immune responses have now been elucidated, and an oxidative stress central role in these pathogenic processes has been recognized. Presence of reactive oxygen species in macrophages and other cells allows them to eliminate virus, but its excess alters the balance between innate and adaptive immune responses and proteases/anti-proteases and leads to uncontrolled inflammation, tissue damage, and hypercoagulability. Different upper and lower airway cell types also play a role in viral entry and infection. Carbocysteine is a muco-active drug with anti-oxidant and anti-inflammatory properties used for the management of several chronic respiratory diseases. Although the use of anti-oxidants has been proposed as an effective strategy in COPD exacerbations management, the molecular mechanisms that explain carbocysteine efficacy have not yet been fully clarified. The present review describes the most relevant features of the common respiratory virus pathophysiology with a focus on epithelial cells and oxidative stress role and reports data supporting a putative role of carbocysteine in viral respiratory infections.
Collapse
Affiliation(s)
- Elisabetta Pace
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Serena Di Vincenzo
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Maria Ferraro
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | | | - Francesco Scaglione
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Chittasupho C, Umsumarng S, Srisawad K, Arjsri P, Phongpradist R, Samee W, Tingya W, Ampasavate C, Dejkriengkraikul P. Inhibition of SARS-CoV-2-Induced NLRP3 Inflammasome-Mediated Lung Cell Inflammation by Triphala-Loaded Nanoparticle Targeting Spike Glycoprotein S1. Pharmaceutics 2024; 16:751. [PMID: 38931873 PMCID: PMC11206841 DOI: 10.3390/pharmaceutics16060751] [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: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, poses a significant global health threat. The spike glycoprotein S1 of the SARS-CoV-2 virus is known to induce the production of pro-inflammatory mediators, contributing to hyperinflammation in COVID-19 patients. Triphala, an ancient Ayurvedic remedy composed of dried fruits from three plant species-Emblica officinalis (Family Euphorbiaceae), Terminalia bellerica (Family Combretaceae), and Terminalia chebula (Family Combretaceae)-shows promise in addressing inflammation. However, the limited water solubility of its ethanolic extract impedes its bioavailability. In this study, we aimed to develop nanoparticles loaded with Triphala extract, termed "nanotriphala", as a drug delivery system. Additionally, we investigated the in vitro anti-inflammatory properties of nanotriphala and its major compounds, namely gallic acid, chebulagic acid, and chebulinic acid, in lung epithelial cells (A549) induced by CoV2-SP. The nanotriphala formulation was prepared using the solvent displacement method. The encapsulation efficiency of Triphala in nanotriphala was determined to be 87.96 ± 2.60% based on total phenolic content. In terms of in vitro release, nanotriphala exhibited a biphasic release profile with zero-order kinetics over 0-8 h. A549 cells were treated with nanotriphala or its active compounds and then induced with 100 ng/mL of spike S1 subunit (CoV2-SP). The results demonstrate that chebulagic acid and chebulinic acid are the active compounds in nanotriphala, which significantly reduced cytokine release (IL-6, IL-1β, and IL-18) and suppressed the expression of inflammatory genes (IL-6, IL-1β, IL-18, and NLRP3) (p < 0.05). Mechanistically, nanotriphala and its active compounds notably attenuated the expression of inflammasome machinery proteins (NLRP3, ASC, and Caspase-1) (p < 0.05). In conclusion, the nanoparticle formulation of Triphala enhances its stability and exhibits anti-inflammatory properties against CoV2-SP-induction. This was achieved by suppressing inflammatory mediators and the NLRP3 inflammasome machinery. Thus, nanotriphala holds promise as a supportive preventive anti-inflammatory therapy for COVID-19-related chronic inflammation.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Sonthaya Umsumarng
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand;
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kamonwan Srisawad
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Punnida Arjsri
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
| | - Rungsinee Phongpradist
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Weerasak Samee
- Department of Pharmaceutical Chemistry, Srinakharinwirot University, Ongkharak, Nakhon Nayok 26120, Thailand;
| | - Wipawan Tingya
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Pornngarm Dejkriengkraikul
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
9
|
Starshinova A, Borozinets A, Kulpina A, Sereda V, Rubinstein A, Kudryavtsev I, Kudlay D. Bronchial Asthma and COVID-19: Etiology, Pathological Triggers, and Therapeutic Considerations. PATHOPHYSIOLOGY 2024; 31:269-287. [PMID: 38921725 PMCID: PMC11206645 DOI: 10.3390/pathophysiology31020020] [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: 02/24/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Bronchial asthma (BA) continues to be a difficult disease to diagnose. Various factors have been described in the development of BA, but to date, there is no clear evidence for the etiology of this chronic disease. The emergence of COVID-19 has contributed to the pandemic course of asthma and immunologic features. However, there are no unambiguous data on asthma on the background and after COVID-19. There is correlation between various trigger factors that provoke the development of bronchial asthma. It is now obvious that the SARS-CoV-2 virus is one of the provoking factors. COVID-19 has affected the course of asthma. Currently, there is no clear understanding of whether asthma progresses during or after COVID-19 infection. According to the results of some studies, a significant difference was identified between the development of asthma in people after COVID-19. Mild asthma and moderate asthma do not increase the severity of COVID-19 infection. Nevertheless, oral steroid treatment and hospitalization for severe BA were associated with higher COVID-19 severity. The influence of SARS-CoV-2 infection is one of the protective factors. It causes the development of severe bronchial asthma. The accumulated experience with omalizumab in patients with severe asthma during COVID-19, who received omalizumab during the pandemic, has strongly suggested that continued treatment with omalizumab is safe and may help prevent the severe course of COVID-19. Targeted therapy for asthma with the use of omalizumab may also help to reduce severe asthma associated with COVID-19. However, further studies are needed to prove the effect of omalizumab. Data analysis should persist, based on the results of the course of asthma after COVID-19 with varying degrees of severity.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Anastasia Borozinets
- Medical Department, I.M. Sechenov First Moscow State Medical University, 197022 Moscow, Russia
| | - Anastasia Kulpina
- Medical Department, Saint Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia;
| | - Vitaliy Sereda
- Medical Department, Saint Petersburg State University, 199034 St. Petersburg, Russia;
| | - Artem Rubinstein
- Department of immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia;
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
- Department of immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia;
| | - Dmitry Kudlay
- Institute of Immunology FMBA of Russia, 115478 Moscow, Russia;
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
10
|
Du L, Deiter F, Bouzidi MS, Billaud JN, Simmons G, Dabral P, Selvarajah S, Lingappa AF, Michon M, Yu SF, Paulvannan K, Manicassamy B, Lingappa VR, Boushey H, Greenland JR, Pillai SK. A viral assembly inhibitor blocks SARS-CoV-2 replication in airway epithelial cells. Commun Biol 2024; 7:486. [PMID: 38649430 PMCID: PMC11035691 DOI: 10.1038/s42003-024-06130-8] [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: 05/05/2023] [Accepted: 04/01/2024] [Indexed: 04/25/2024] Open
Abstract
The ongoing evolution of SARS-CoV-2 to evade vaccines and therapeutics underlines the need for innovative therapies with high genetic barriers to resistance. Therefore, there is pronounced interest in identifying new pharmacological targets in the SARS-CoV-2 viral life cycle. The small molecule PAV-104, identified through a cell-free protein synthesis and assembly screen, was recently shown to target host protein assembly machinery in a manner specific to viral assembly. In this study, we investigate the capacity of PAV-104 to inhibit SARS-CoV-2 replication in human airway epithelial cells (AECs). We show that PAV-104 inhibits >99% of infection with diverse SARS-CoV-2 variants in immortalized AECs, and in primary human AECs cultured at the air-liquid interface (ALI) to represent the lung microenvironment in vivo. Our data demonstrate that PAV-104 inhibits SARS-CoV-2 production without affecting viral entry, mRNA transcription, or protein synthesis. PAV-104 interacts with SARS-CoV-2 nucleocapsid (N) and interferes with its oligomerization, blocking particle assembly. Transcriptomic analysis reveals that PAV-104 reverses SARS-CoV-2 induction of the type-I interferon response and the maturation of nucleoprotein signaling pathway known to support coronavirus replication. Our findings suggest that PAV-104 is a promising therapeutic candidate for COVID-19 with a mechanism of action that is distinct from existing clinical management approaches.
Collapse
Affiliation(s)
- Li Du
- Vitalant Research Institute, 360 Spear St., San Francisco, CA, 94105, USA
- University of California, San Francisco, CA, 94143, USA
| | - Fred Deiter
- University of California, San Francisco, CA, 94143, USA
- Veterans Administration Health Care System, 4150 Clement St., San Francisco, CA, 94121, USA
| | - Mohamed S Bouzidi
- Vitalant Research Institute, 360 Spear St., San Francisco, CA, 94105, USA
- University of California, San Francisco, CA, 94143, USA
| | | | - Graham Simmons
- Vitalant Research Institute, 360 Spear St., San Francisco, CA, 94105, USA
- University of California, San Francisco, CA, 94143, USA
| | - Prerna Dabral
- Vitalant Research Institute, 360 Spear St., San Francisco, CA, 94105, USA
- University of California, San Francisco, CA, 94143, USA
| | | | | | - Maya Michon
- Prosetta Biosciences Inc, 670 5th St., San Francisco, CA, 94107, USA
| | - Shao Feng Yu
- Prosetta Biosciences Inc, 670 5th St., San Francisco, CA, 94107, USA
| | - Kumar Paulvannan
- Prosetta Biosciences Inc, 670 5th St., San Francisco, CA, 94107, USA
| | | | | | - Homer Boushey
- University of California, San Francisco, CA, 94143, USA
| | - John R Greenland
- University of California, San Francisco, CA, 94143, USA
- Veterans Administration Health Care System, 4150 Clement St., San Francisco, CA, 94121, USA
| | - Satish K Pillai
- Vitalant Research Institute, 360 Spear St., San Francisco, CA, 94105, USA.
- University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
11
|
Soltane R, Almulla N, Alasiri A, Elashmawy NF, Qumsani AT, Alshehrei FM, Keshek DEG, Alqadi T, AL-Ghamdi SB, Allayeh AK. A Comparative Analysis of MicroRNA Expression in Mild, Moderate, and Severe COVID-19: Insights from Urine, Serum, and Nasopharyngeal Samples. Biomolecules 2023; 13:1681. [PMID: 38136554 PMCID: PMC10742216 DOI: 10.3390/biom13121681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
COVID-19, caused by the SARS-CoV-2 virus, manifests with a wide range of clinical symptoms that vary from mild respiratory issues to severe respiratory distress. To effectively manage and predict the outcomes of the disease, it is important to understand the molecular mechanisms underlying its severity. This study focuses on analyzing and comparing the expression patterns of microRNAs (miRNAs) in serum, urine, and nasopharyngeal samples from patients with mild, moderate, and severe COVID-19. The aim is to identify potential associations with disease progression and discover suitable markers for diagnosis and prognosis. Our findings indicate the consistent upregulation of miR-21, miR-146a, and miR-155 in urine, serum, and nasopharyngeal samples from patients with mild COVID-19. In moderate cases, there were more significant changes in miRNA expression compared to mild cases. Specifically, miR-let-7 demonstrated upregulation, while miR-146b exhibited downregulation. The most notable alterations in miRNA expression profiles were observed in severe COVID-19 cases, with a significant upregulation of miR-223. Moreover, our analysis using Receiver-operating characteristic (ROC) curves demonstrated that miR-155, miR-let-7, and miR-223 exhibited high sensitivity and specificity, suggesting their potential as biomarkers for distinguishing COVID-19 patients from healthy individuals. Overall, this comparative analysis revealed distinct patterns in miRNA expression. The overlapping expression patterns of miRNAs in urine, serum, and nasopharyngeal samples suggest their potential utility in discriminating disease status.
Collapse
Affiliation(s)
- Raya Soltane
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (N.A.); (A.A.); (T.A.)
| | - Nuha Almulla
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (N.A.); (A.A.); (T.A.)
| | - Ahlam Alasiri
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (N.A.); (A.A.); (T.A.)
| | - Nabila F. Elashmawy
- Biology Department, College of Science, Jazan University, Jazan 82817, Saudi Arabia;
| | - Alaa T. Qumsani
- Department of Biology, Jumum College University, Umm Al-Qura University, P.O Box 7388, Makkah 21955, Saudi Arabia; (A.T.Q.); (F.M.A.); (D.E.-G.K.)
| | - Fatimah M. Alshehrei
- Department of Biology, Jumum College University, Umm Al-Qura University, P.O Box 7388, Makkah 21955, Saudi Arabia; (A.T.Q.); (F.M.A.); (D.E.-G.K.)
| | - Doaa El-Ghareeb Keshek
- Department of Biology, Jumum College University, Umm Al-Qura University, P.O Box 7388, Makkah 21955, Saudi Arabia; (A.T.Q.); (F.M.A.); (D.E.-G.K.)
- Agriculture Genetic Engineering Research Institute (AGERI), Agriculture Research Centre, Giza 12512, Egypt
| | - Taha Alqadi
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (N.A.); (A.A.); (T.A.)
| | | | - Abdou Kamal Allayeh
- Virology Lab 176, Environment and Climate Change Institute, National Research Centre, Giza 12622, Egypt
| |
Collapse
|
12
|
Tedbury PR, Manfredi C, Degenhardt F, Conway J, Horwath MC, McCracken C, Sorscher AJ, Moreau S, Wright C, Edwards C, Brewer J, Guarner J, de Wit E, Williamson BN, Suthar MS, Ong YT, Roback JD, Alter DN, Holter JC, Karlsen TH, Sacchi N, Romero-Gómez M, Invernizzi P, Fernández J, Buti M, Albillos A, Julià A, Valenti L, Asselta R, Banales JM, Bujanda L, de Cid R, the Severe COVID-19 GWAS group, Sarafianos SG, Hong JS, Sorscher EJ, Ehrhardt A. Mechanisms by which the cystic fibrosis transmembrane conductance regulator may influence SARS-CoV-2 infection and COVID-19 disease severity. FASEB J 2023; 37:e23220. [PMID: 37801035 PMCID: PMC10760435 DOI: 10.1096/fj.202300077r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
Patients with cystic fibrosis (CF) exhibit pronounced respiratory damage and were initially considered among those at highest risk for serious harm from SARS-CoV-2 infection. Numerous clinical studies have subsequently reported that individuals with CF in North America and Europe-while susceptible to severe COVID-19-are often spared from the highest levels of virus-associated mortality. To understand features that might influence COVID-19 among patients with cystic fibrosis, we studied relationships between SARS-CoV-2 and the gene responsible for CF (i.e., the cystic fibrosis transmembrane conductance regulator, CFTR). In contrast to previous reports, we found no association between CFTR carrier status (mutation heterozygosity) and more severe COVID-19 clinical outcomes. We did observe an unexpected trend toward higher mortality among control individuals compared with silent carriers of the common F508del CFTR variant-a finding that will require further study. We next performed experiments to test the influence of homozygous CFTR deficiency on viral propagation and showed that SARS-CoV-2 production in primary airway cells was not altered by the absence of functional CFTR using two independent protocols. On the contrary, experiments performed in vitro strongly indicated that virus proliferation depended on features of the mucosal fluid layer known to be disrupted by absent CFTR in patients with CF, including both low pH and increased viscosity. These results point to the acidic, viscous, and mucus-obstructed airways in patients with cystic fibrosis as unfavorable for the establishment of coronaviral infection. Our findings provide new and important information concerning relationships between the CF clinical phenotype and severity of COVID-19.
Collapse
Affiliation(s)
- Philip R. Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Candela Manfredi
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Joseph Conway
- Northeast Georgia Medical Center, Gainesville, Georgia, United States
| | - Michael C. Horwath
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Courtney McCracken
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Adam J. Sorscher
- Dartmouth University School of Medicine, Hanover, New Hampshire, United States
| | - Sandy Moreau
- Elliot Hospital, Manchester, New Hampshire, United States
| | | | - Carolina Edwards
- Northeast Georgia Medical Center, Gainesville, Georgia, United States
| | - Jo Brewer
- Northeast Georgia Medical Center, Gainesville, Georgia, United States
| | | | - Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, NIAID, National Institutes of Health, Hamilton, Montana, United States
| | - Brandi N. Williamson
- Laboratory of Virology, Division of Intramural Research, NIAID, National Institutes of Health, Hamilton, Montana, United States
| | - Mehul S. Suthar
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Yee T. Ong
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
| | - John D. Roback
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - David N. Alter
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jan C. Holter
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tom H. Karlsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute for Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Gastroenterology, Department of Transplantation Medicine, Division for Cancer Medicine, Surgery and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Manuel Romero-Gómez
- Hospital Universitario Virgen del Rocío de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
- University of Sevilla, Sevilla, Spain
- Digestive Diseases Unit, Virgen del Rocio University Hospital, Institute of Biomedicine of Seville, University of Seville, Seville, Spain
| | - Pietro Invernizzi
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Javier Fernández
- Hospital Clinic, University of Barcelona, and IDIBAPS, Barcelona, Spain
- European Foundation for the Study of Chronic Liver Failure (EF-CLIF), Barcelona, Spain
| | - Maria Buti
- Liver Unit. Hospital Universitario Valle Hebron and CIBEREHD del Instituto Carlos III. Barcelona, Spain
| | - Agustin Albillos
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Gastroenterology, Hospital Universitario Ramón y Cajal, University of Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Antonio Julià
- Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Barcelona, Spain
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Biological Resorce Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano, Milan Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Jesus M. Banales
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute – Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Luis Bujanda
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute – Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
| | - Rafael de Cid
- Genomes for Life-GCAT lab. German Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | | | - Stefan G. Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Jeong S. Hong
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Eric J. Sorscher
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Annette Ehrhardt
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
13
|
Luan Y, Luan Y, He H, Jue B, Yang Y, Qin B, Ren K. Glucose metabolism disorder: a potential accomplice of SARS-CoV-2. Int J Obes (Lond) 2023; 47:893-902. [PMID: 37542197 DOI: 10.1038/s41366-023-01352-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023]
Abstract
Globally, 265,713,467 confirmed cases of SARS-CoV-2 (CoV-2), including 5,260,888 deaths, have been reported by the WHO. It is important to study the mechanism of this infectious disease. A variety of evidences show the potential association between CoV-2 and glucose metabolism. Notably, people with type 2 diabetes mellitus (T2DM) and other metabolic complications were prone to have a higher risk of developing a more severe infection course than people who were metabolically normal. The correlations between glucose metabolism and CoV-2 progression have been widely revealed. This review will discuss the association between glucose metabolism disorders and CoV-2 progression, showing the promoting effect of diabetes and other diseases related to glucose metabolism disorders on the progression of CoV-2. We will further conclude the effects of key proteins and pathways in glucose metabolism regulation on CoV-2 progression and potential interventions by targeting glucose metabolism disorders for CoV-2 treatment. Therefore, this review will provide systematic insight into the treatment of CoV-2 from the perspective of glucose metabolism.
Collapse
Affiliation(s)
- Yi Luan
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100000, China
| | - Hongbo He
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Bolin Jue
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453000, China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Bo Qin
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
14
|
Guo TJF, Singhera GK, Leung JM, Dorscheid DR. Airway Epithelial-Derived Immune Mediators in COVID-19. Viruses 2023; 15:1655. [PMID: 37631998 PMCID: PMC10458661 DOI: 10.3390/v15081655] [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: 06/13/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The airway epithelium, which lines the conducting airways, is central to the defense of the lungs against inhaled particulate matter and pathogens such as SARS-CoV-2, the virus that causes COVID-19. Recognition of pathogens results in the activation of an innate and intermediate immune response which involves the release of cytokines and chemokines by the airway epithelium. This response can inhibit further viral invasion and influence adaptive immunity. However, severe COVID-19 is characterized by a hyper-inflammatory response which can give rise to clinical presentations including lung injury and lead to acute respiratory distress syndrome, viral pneumonia, coagulopathy, and multi-system organ failure. In response to SARS-CoV-2 infection, the airway epithelium can mount a maladaptive immune response which can delay viral clearance, perpetuate excessive inflammation, and contribute to the pathogenesis of severe COVID-19. In this article, we will review the barrier and immune functions of the airway epithelium, how SARS-CoV-2 can interact with the epithelium, and epithelial-derived cytokines and chemokines and their roles in COVID-19 and as biomarkers. Finally, we will discuss these immune mediators and their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Tony J. F. Guo
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
| | - Gurpreet K. Singhera
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Janice M. Leung
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Delbert R. Dorscheid
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
15
|
Chittasupho C, Srisawad K, Arjsri P, Phongpradist R, Tingya W, Ampasavate C, Dejkriengkraikul P. Targeting Spike Glycoprotein S1 Mediated by NLRP3 Inflammasome Machinery and the Cytokine Releases in A549 Lung Epithelial Cells by Nanocurcumin. Pharmaceuticals (Basel) 2023; 16:862. [PMID: 37375809 DOI: 10.3390/ph16060862] [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: 04/24/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic inflammation and tissue damage can result from uncontrolled inflammation during SARS-CoV-2 or COVID-19 infections, leading to post-acute COVID conditions or long COVID. Curcumin, found in turmeric, has potent anti-inflammatory properties but limited effectiveness. This study developed nanocurcumin, a curcumin nanoparticle, to enhance its physical and chemical stability and investigate its in vitro anti-inflammatory properties upon CoV2-SP induction in lung epithelial cells. Nanocurcumin was prepared by encapsulating curcumin extract in phospholipids. The particle size, polydispersity index, and zeta potential of nanocurcumin were measured using dynamic light scattering. The encapsulated curcumin content was determined using HPLC analysis. The encapsulation efficiency of curcumin was 90.74 ± 5.35% as determined by HPLC. Regarding the in vitro release of curcumin, nanocurcumin displayed a higher release content than non-nanoparticle curcumin. Nanocurcumin was further investigated for its anti-inflammatory properties using A549 lung epithelial cell line. As determined by ELISA, nanocurcumin showed inhibitory effects on inflammatory cytokine releases in CoV2-SP-stimulated conditions, as evidenced by a significant decrease in IL-6, IL-1β and IL-18 cytokine secretions compared with the spike-stimulated control group (p < 0.05). Additionally, as determined by RT-PCR, nanocurcumin significantly inhibited the CoV2-SP-stimulated expression of inflammatory genes (IL-6, IL-1β, IL-18, and NLRP3) compared with the spike-stimulated control group (p < 0.05). Regarding the inhibition of NLRP3 inflammasome machinery proteins by Western blot, nanocurcumin decreased the expressions of inflammasome machinery proteins including NLRP3, ASC, pro-caspase-1, and the active form of caspase-1 in CoV2-SP-stimulated A549 cells compared with the spike-stimulated control group (p < 0.05). Overall, the nanoparticle formulation of curcumin improved its solubility and bioavailability, demonstrating anti-inflammatory effects in a CoV2-SP-induced scenario by inhibiting inflammatory mediators and the NLRP3 inflammasome machinery. Nanocurcumin shows promise as an anti-inflammatory product for preventing COVID-19-related airway inflammation.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kamonwan Srisawad
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Punnida Arjsri
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Rungsinee Phongpradist
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wipawan Tingya
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
16
|
Pillai S, Du L, Deiter F, Bouzidi M, Billaud JN, Graham S, Prerna D, Selvarajah S, Lingappa A, Michon M, Yu S, Paulvannan K, Lingappa V, Boushey H, Greenland J. A Novel Viral Assembly Inhibitor Blocks SARS-CoV-2 Replication in Airway Epithelial Cells. RESEARCH SQUARE 2023:rs.3.rs-2887435. [PMID: 37292622 PMCID: PMC10246244 DOI: 10.21203/rs.3.rs-2887435/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The ongoing evolution of SARS-CoV-2 to evade vaccines and therapeutics underlines the need for novel therapies with high genetic barriers to resistance. The small molecule PAV-104, identified through a cell-free protein synthesis and assembly screen, was recently shown to target host protein assembly machinery in a manner specific to viral assembly. Here, we investigated the capacity of PAV-104 to inhibit SARS-CoV-2 replication in human airway epithelial cells (AECs). Our data demonstrate that PAV-104 inhibited > 99% of infection with diverse SARS-CoV-2 variants in primary and immortalized human AECs. PAV-104 suppressed SARS-CoV-2 production without affecting viral entry or protein synthesis. PAV-104 interacted with SARS-CoV-2 nucleocapsid (N) and interfered with its oligomerization, blocking particle assembly. Transcriptomic analysis revealed that PAV-104 reversed SARS-CoV-2 induction of the Type-I interferon response and the 'maturation of nucleoprotein' signaling pathway known to support coronavirus replication. Our findings suggest that PAV-104 is a promising therapeutic candidate for COVID-19.
Collapse
Affiliation(s)
| | - Li Du
- Vitalant Research Institute/UCSF
| | - Fred Deiter
- Veterans Administration Health Care System/UCSF
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhang J, Yang W, Roy S, Liu H, Roberts R, Wang L, Shi L, Ma W. Tight junction protein occludin is an internalization factor for SARS-CoV-2 infection and mediates virus cell-to-cell transmission. Proc Natl Acad Sci U S A 2023; 120:e2218623120. [PMID: 37068248 PMCID: PMC10151465 DOI: 10.1073/pnas.2218623120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/13/2023] [Indexed: 04/19/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spreads efficiently by spike-mediated, direct cell-to-cell transmission. However, the underlying mechanism is poorly understood. Herein, we demonstrate that the tight junction protein occludin (OCLN) is critical to this process. SARS-CoV-2 infection alters OCLN distribution and expression and causes syncytium formation that leads to viral spread. OCLN knockdown fails to alter SARS-CoV-2 binding but significantly lowers internalization, syncytium formation, and transmission. OCLN overexpression also has no effect on virus binding but enhances virus internalization, cell-to-cell transmission, and replication. OCLN directly interacts with the SARS-CoV-2 spike, and the endosomal entry pathway is involved in OCLN-mediated cell-to-cell fusion rather than in the cell surface entry pathway. All SARS-CoV-2 strains tested (prototypic, alpha, beta, gamma, delta, kappa, and omicron) are dependent on OCLN for cell-to-cell transmission, although the extent of syncytium formation differs between strains. We conclude that SARS-CoV-2 utilizes OCLN as an internalization factor for cell-to-cell transmission.
Collapse
Affiliation(s)
- Jialin Zhang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Wenyu Yang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Sawrab Roy
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Heidi Liu
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - R. Michael Roberts
- Division of Animal Sciences, College of Agriculture, Food, & Natural Resources, University of Missouri, Columbia, MO65211
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO65211
| | - Liping Wang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Lei Shi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Wenjun Ma
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| |
Collapse
|
18
|
Campos-Gómez J, Fernandez Petty C, Mazur M, Tang L, Solomon GM, Joseph R, Li Q, Peabody Lever JE, Hussain SS, Harrod KS, Onuoha EE, Kim H, Rowe SM. Mucociliary clearance augmenting drugs block SARS-CoV-2 replication in human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2023; 324:L493-L506. [PMID: 36809189 PMCID: PMC10042606 DOI: 10.1152/ajplung.00285.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 01/17/2023] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
The coronavirus disease (COVID-19) pandemic, caused by SARS-CoV-2 coronavirus, is devastatingly impacting human health. A prominent component of COVID-19 is the infection and destruction of the ciliated respiratory cells, which perpetuates dissemination and disrupts protective mucociliary transport (MCT) function, an innate defense of the respiratory tract. Thus, drugs that augment MCT could improve the barrier function of the airway epithelium and reduce viral replication and, ultimately, COVID-19 outcomes. We tested five agents known to increase MCT through distinct mechanisms for activity against SARS-CoV-2 infection using a model of human respiratory epithelial cells terminally differentiated in an air/liquid interphase. Three of the five mucoactive compounds tested showed significant inhibitory activity against SARS-CoV-2 replication. An archetype mucoactive agent, ARINA-1, blocked viral replication and therefore epithelial cell injury; thus, it was further studied using biochemical, genetic, and biophysical methods to ascertain the mechanism of action via the improvement of MCT. ARINA-1 antiviral activity was dependent on enhancing the MCT cellular response, since terminal differentiation, intact ciliary expression, and motion were required for ARINA-1-mediated anti-SARS-CoV2 protection. Ultimately, we showed that the improvement of cilia movement was caused by ARINA-1-mediated regulation of the redox state of the intracellular environment, which benefited MCT. Our study indicates that intact MCT reduces SARS-CoV-2 infection, and its pharmacologic activation may be effective as an anti-COVID-19 treatment.
Collapse
Affiliation(s)
- Javier Campos-Gómez
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
| | | | - Marina Mazur
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
| | - Liping Tang
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
| | - George M Solomon
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
| | - Reny Joseph
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
| | - Qian Li
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
| | - Jacelyn E Peabody Lever
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
- Medical Scientist Training Program, Heersink School of Medicine, University of Alabama at Birmingham, Alabama, United States
| | - Shah Saddad Hussain
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
| | - Kevin S Harrod
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Alabama, United States
| | - Ezinwanne E Onuoha
- Department of Biomedical Engineering, University of Alabama at Birmingham, Alabama, United States
| | - Harrison Kim
- Department of Radiology, University of Alabama at Birmingham, Alabama, United States
| | - Steven M Rowe
- Department of Medicine, University of Alabama at Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Alabama, United States
| |
Collapse
|
19
|
Lott N, Gebhard CE, Bengs S, Haider A, Kuster GM, Regitz-Zagrosek V, Gebhard C. Sex hormones in SARS-CoV-2 susceptibility: key players or confounders? Nat Rev Endocrinol 2023; 19:217-231. [PMID: 36494595 PMCID: PMC9734735 DOI: 10.1038/s41574-022-00780-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has a clear sex disparity in clinical outcomes. Hence, the interaction between sex hormones, virus entry receptors and immune responses has attracted major interest as a target for the prevention and treatment of SARS-CoV-2 infections. This Review summarizes the current understanding of the roles of androgens, oestrogens and progesterone in the regulation of virus entry receptors and disease progression of coronavirus disease 2019 (COVID-19) as well as their therapeutic value. Although many experimental and clinical studies have analysed potential mechanisms by which female sex hormones might provide protection against SARS-CoV-2 infectivity, there is currently no clear evidence for a sex-specific expression of virus entry receptors. In addition, reports describing an influence of oestrogen, progesterone and androgens on the course of COVID-19 vary widely. Current data also do not support the administration of oestradiol in COVID-19. The conflicting evidence and lack of consensus results from a paucity of mechanistic studies and clinical trials reporting sex-disaggregated data. Further, the influence of variables beyond biological factors (sex), such as sociocultural factors (gender), on COVID-19 manifestations has not been investigated. Future research will have to fill this knowledge gap as the influence of sex and gender on COVID-19 will be essential to understanding and managing the long-term consequences of this pandemic.
Collapse
Affiliation(s)
- Nicola Lott
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Achi Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Gabriela M Kuster
- Department of Cardiology and Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Vera Regitz-Zagrosek
- Charité, Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Department of Cardiology, Inselspital Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
20
|
Resnick JD, Beer MA, Pekosz A. Early Transcriptional Responses of Human Nasal Epithelial Cells to Infection with Influenza A and SARS-CoV-2 Virus Differ and Are Influenced by Physiological Temperature. Pathogens 2023; 12:480. [PMID: 36986402 PMCID: PMC10051809 DOI: 10.3390/pathogens12030480] [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: 12/30/2022] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza A (IAV) and SARS-CoV-2 (SCV2) viruses represent an ongoing threat to public health. Both viruses target the respiratory tract, which consists of a gradient of cell types, receptor expression, and temperature. Environmental temperature has been an understudied contributor to infection susceptibility and understanding its impact on host responses to infection could help uncover new insight into severe disease risk factors. As the nasal passageways are the initial site of respiratory virus infection, in this study we investigated the effect of temperature on host responses in human nasal epithelial cells (hNECs) utilizing IAV and SCV2 in vitro infection models. We demonstrate that temperature affected SCV2, but not IAV, viral replicative fitness and that SCV2-infected cultures were slower to mount an infection-induced response, likely due to suppression by the virus. Additionally, we show that that temperature not only changed the basal transcriptomic landscape of epithelial cells, but that it also impacted the response to infection. The induction of interferon and other innate immune responses was not drastically affected by temperature, suggesting that while the baseline antiviral response at different temperatures remained consistent, there may be metabolic or signaling changes that affect how well the cultures were able to adapt to new pressures, such as infection. Finally, we show that hNECs responded differently to IAV and SCV2 infection in ways that give insight into how the virus is able to manipulate the cell to allow for replication and release. Taken together, these data give new insight into the innate immune response to respiratory infections and can assist in identifying new treatment strategies for respiratory infections.
Collapse
Affiliation(s)
- Jessica D. Resnick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael A. Beer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
21
|
Resnick JD, Beer MA, Pekosz A. Early transcriptional responses of human nasal epithelial cells to infection with Influenza A and SARS-CoV-2 virus differ and are influenced by physiological temperature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531609. [PMID: 36945583 PMCID: PMC10028862 DOI: 10.1101/2023.03.07.531609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Influenza A (IAV) and SARS-CoV-2 (SCV2) viruses represent an ongoing threat to public health. Both viruses target the respiratory tract, which consists of a gradient of cell types, receptor expression, and temperature. Environmental temperature has been an un-derstudied contributor to infection susceptibility and understanding its impact on host responses to infection could help uncover new insights into severe disease risk factors. As the nasal passageways are the initial site of respiratory virus infection, in this study we investigated the effect of temperature on host responses in human nasal epithelial cells (hNECs) utilizing IAV and SCV2 in vitro infection models. We demonstrate that temperature affects SCV2, but not IAV, viral replicative fitness and that SCV2 infected cultures are slower to mount an infection-induced response, likely due to suppression by the virus. Additionally, we show that that temperature not only changes the basal transcriptomic landscape of epithelial cells, but that it also impacts the response to infection. The induction of interferon and other innate immune responses were not drastically affected by temperature, suggesting that while the baseline antiviral response at different temperatures remains consistent, there may be metabolic or signaling changes that affect how well the cultures are able to adapt to new pressures such as infection. Finally, we show that hNECs respond differently to IAV and SCV2 infection in ways that give insight into how the virus is able to manipulate the cell to allow for replication and release. Taken together, these data give new insight into the innate immune response to respiratory infections and can assist in identifying new treatment strategies for respiratory infections.
Collapse
Affiliation(s)
- Jessica D Resnick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- McKusick- Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael A Beer
- McKusick- Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Lee JH, Koh J, Jeon YK, Goo JM, Yoon SH. An Integrated Radiologic-Pathologic Understanding of COVID-19 Pneumonia. Radiology 2023; 306:e222600. [PMID: 36648343 PMCID: PMC9868683 DOI: 10.1148/radiol.222600] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 01/18/2023]
Abstract
This article reviews the radiologic and pathologic findings of the epithelial and endothelial injuries in COVID-19 pneumonia to help radiologists understand the fundamental nature of the disease. The radiologic and pathologic manifestations of COVID-19 pneumonia result from epithelial and endothelial injuries based on viral toxicity and immunopathologic effects. The pathologic features of mild and reversible COVID-19 pneumonia involve nonspecific pneumonia or an organizing pneumonia pattern, while the pathologic features of potentially fatal and irreversible COVID-19 pneumonia are characterized by diffuse alveolar damage followed by fibrosis or acute fibrinous organizing pneumonia. These pathologic responses of epithelial injuries observed in COVID-19 pneumonia are not specific to SARS-CoV-2 but rather constitute universal responses to viral pneumonia. Endothelial injury in COVID-19 pneumonia is a prominent feature compared with other types of viral pneumonia and encompasses various vascular abnormalities at different levels, including pulmonary thromboembolism, vascular engorgement, peripheral vascular reduction, a vascular tree-in-bud pattern, and lung perfusion abnormality. Chest CT with different imaging techniques (eg, CT quantification, dual-energy CT perfusion) can fully capture the various manifestations of epithelial and endothelial injuries. CT can thus aid in establishing prognosis and identifying patients at risk for deterioration.
Collapse
Affiliation(s)
- Jong Hyuk Lee
- From the Departments of Radiology (J.H.L., J.M.G., S.H.Y.) and
Pathology (J.K., Y.K.J.), Seoul National University Hospital, Seoul National
University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea;
Department of Radiology, Seoul National University College of Medicine, Seoul,
Korea (J.M.G.); Institute of Radiation Medicine, Seoul National University
Medical Research Center, Seoul, Korea (J.M.G.); and Cancer Research Institute,
Seoul National University, Seoul, Korea (J.M.G.)
| | - Jaemoon Koh
- From the Departments of Radiology (J.H.L., J.M.G., S.H.Y.) and
Pathology (J.K., Y.K.J.), Seoul National University Hospital, Seoul National
University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea;
Department of Radiology, Seoul National University College of Medicine, Seoul,
Korea (J.M.G.); Institute of Radiation Medicine, Seoul National University
Medical Research Center, Seoul, Korea (J.M.G.); and Cancer Research Institute,
Seoul National University, Seoul, Korea (J.M.G.)
| | - Yoon Kyung Jeon
- From the Departments of Radiology (J.H.L., J.M.G., S.H.Y.) and
Pathology (J.K., Y.K.J.), Seoul National University Hospital, Seoul National
University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea;
Department of Radiology, Seoul National University College of Medicine, Seoul,
Korea (J.M.G.); Institute of Radiation Medicine, Seoul National University
Medical Research Center, Seoul, Korea (J.M.G.); and Cancer Research Institute,
Seoul National University, Seoul, Korea (J.M.G.)
| | - Jin Mo Goo
- From the Departments of Radiology (J.H.L., J.M.G., S.H.Y.) and
Pathology (J.K., Y.K.J.), Seoul National University Hospital, Seoul National
University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea;
Department of Radiology, Seoul National University College of Medicine, Seoul,
Korea (J.M.G.); Institute of Radiation Medicine, Seoul National University
Medical Research Center, Seoul, Korea (J.M.G.); and Cancer Research Institute,
Seoul National University, Seoul, Korea (J.M.G.)
| | - Soon Ho Yoon
- From the Departments of Radiology (J.H.L., J.M.G., S.H.Y.) and
Pathology (J.K., Y.K.J.), Seoul National University Hospital, Seoul National
University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea;
Department of Radiology, Seoul National University College of Medicine, Seoul,
Korea (J.M.G.); Institute of Radiation Medicine, Seoul National University
Medical Research Center, Seoul, Korea (J.M.G.); and Cancer Research Institute,
Seoul National University, Seoul, Korea (J.M.G.)
| |
Collapse
|
23
|
Campos-Gomez J, Petty CF, Mazur M, Tang L, Solomon GM, Joseph R, Li Q, Lever JEP, Hussain S, Harrod K, Onuoha E, Kim H, Rowe SM. Mucociliary Clearance Augmenting Drugs Block SARS-Cov-2 Replication in Human Airway Epithelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526308. [PMID: 36778446 PMCID: PMC9915467 DOI: 10.1101/2023.01.30.526308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The coronavirus disease (COVID-19) pandemic, caused by SARS-CoV-2 coronavirus, is devastatingly impacting human health. A prominent component of COVID-19 is the infection and destruction of the ciliated respiratory cells, which perpetuates dissemination and disrupts protective mucociliary transport (MCT) function, an innate defense of the respiratory tract. Thus, drugs that augment MCT could improve barrier function of the airway epithelium, reduce viral replication and, ultimately, COVID-19 outcomes. We tested five agents known to increase MCT through distinct mechanisms for activity against SARS-CoV-2 infection using a model of human respiratory epithelial cells terminally differentiated in an air/liquid interphase. Three of the five mucoactive compounds tested showed significant inhibitory activity against SARS-CoV-2 replication. An archetype mucoactive agent, ARINA-1, blocked viral replication and therefore epithelial cell injury, thus, it was further studied using biochemical, genetic and biophysical methods to ascertain mechanism of action via improvement of MCT. ARINA-1 antiviral activity was dependent on enhancing the MCT cellular response, since terminal differentiation, intact ciliary expression and motion was required for ARINA-1-mediated anti-SARS-CoV2 protection. Ultimately, we showed that improvement of cilia movement was caused by ARINA-1-mediated regulation of the redox state of the intracellular environment, which benefited MCT. Our study indicates that Intact MCT reduces SARS-CoV-2 infection, and its pharmacologic activation may be effective as an anti-COVID-19 treatment.
Collapse
Affiliation(s)
- Javier Campos-Gomez
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Marina Mazur
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Liping Tang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - George M. Solomon
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Reny Joseph
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Qian Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jacelyn E. Peabody Lever
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- Medical Scientist Training Program, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shah Hussain
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kevin Harrod
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ezinwanne Onuoha
- Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Harrison Kim
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Steven M. Rowe
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
24
|
Dissook S, Umsumarng S, Mapoung S, Semmarath W, Arjsri P, Srisawad K, Dejkriengkraikul P. Luteolin-rich fraction from Perilla frutescens seed meal inhibits spike glycoprotein S1 of SARS-CoV-2-induced NLRP3 inflammasome lung cell inflammation via regulation of JAK1/STAT3 pathway: A potential anti-inflammatory compound against inflammation-induced long-COVID. Front Med (Lausanne) 2023; 9:1072056. [PMID: 36698809 PMCID: PMC9870545 DOI: 10.3389/fmed.2022.1072056] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Objective The multi-systemic inflammation as a result of COVID-19 can persevere long after the initial symptoms of the illness have subsided. These effects are referred to as Long-COVID. Our research focused on the contribution of the Spike protein S1 subunit of SARS-CoV-2 (Spike S1) on the lung inflammation mediated by NLRP3 inflammasome machinery and the cytokine releases, interleukin 6 (IL-6), IL-1beta, and IL-18, in lung epithelial cells. This study has attempted to identify the naturally- occurring agents that act against inflammation-related long-COVID. The seed meal of Perilla frutescens (P. frutescens), which contains two major dietary polyphenols (rosmarinic acid and luteolin), has been reported to exhibit anti-inflammation activities. Therefore, we have established the ethyl acetate fraction of P. frutescens seed meal (PFEA) and determined its anti-inflammatory effects on Spike S1 exposure in A549 lung cells. Methods PFEA was established using solvent-partitioned extraction. Rosmarinic acid (Ra) and luteolin (Lu) in PFEA were identified using the HPLC technique. The inhibitory effects of PFEA and its active compounds against Spike S1-induced inflammatory response in A549 cells were determined by RT-PCR and ELISA. The mechanistic study of anti-inflammatory properties of PFEA and Lu were determined using western blot technique. Results PFEA was found to contain Ra (388.70 ± 11.12 mg/g extract) and Lu (248.82 ± 12.34 mg/g extract) as its major polyphenols. Accordingly, A549 lung cells were pre-treated with PFEA (12.5-100 μg/mL) and its two major compounds (2.5-20 μg/mL) prior to the Spike S1 exposure at 100 ng/mL. PFEA dose-dependently exhibited anti-inflammatory properties upon Spike S1-exposed A549 cells through IL-6, IL-1β, IL-18, and NLRP3 gene suppressions, as well as IL-6, IL-1β, and IL-18 cytokine releases with statistical significance (p < 0.05). Importantly, Lu possesses superior anti-inflammatory properties when compared with Ra (p < 0.01). Mechanistically, PFEA and Lu effectively attenuated a Spike S1-induced inflammatory response through downregulation of the JAK1/STAT3-inflammasome-dependent inflammatory pathway as evidenced by the downregulation of NLRP3, ASC, and cleaved-caspase-1 of the NLRP3 inflammasome components and by modulating the phosphorylation of JAK1 and STAT3 proteins (p < 0.05). Conclusion The findings suggested that luteolin and PFEA can modulate the signaling cascades that regulate Spike S1-induced lung inflammation during the incidence of Long-COVID. Consequently, luteolin and P. frutescens may be introduced as potential candidates in the preventive therapeutic strategy for inflammation-related post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Sivamoke Dissook
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand
| | - Sonthaya Umsumarng
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand,Division of Veterinary Preclinical Sciences, Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sariya Mapoung
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand
| | - Warathit Semmarath
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand,Akkraratchkumari Veterinary College, Walailak University, Nakhon Si Thammarat, Thailand
| | - Punnida Arjsri
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand
| | - Kamonwan Srisawad
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand,Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand,Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,*Correspondence: Pornngarm Dejkriengkraikul,
| |
Collapse
|
25
|
Fluorogenic reporter enables identification of compounds that inhibit SARS-CoV-2. Nat Microbiol 2023; 8:121-134. [PMID: 36604514 PMCID: PMC9831272 DOI: 10.1038/s41564-022-01288-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 11/16/2022] [Indexed: 01/07/2023]
Abstract
The coronavirus SARS-CoV-2 causes the severe disease COVID-19. SARS-CoV-2 infection is initiated by interaction of the viral spike protein and host receptor angiotensin-converting enzyme 2 (ACE2). We report an improved bright and reversible fluorogenic reporter, named SURF (split UnaG-based reversible and fluorogenic protein-protein interaction reporter), that we apply to monitor real-time interactions between spike and ACE2 in living cells. SURF has a large dynamic range with a dark-to-bright fluorescence signal that requires no exogenous cofactors. Utilizing this reporter, we carried out a high-throughput screening of small-molecule libraries. We identified three natural compounds that block replication of SARS-CoV-2 in both Vero cells and human primary nasal and bronchial epithelial cells. Cell biological and biochemical experiments validated all three compounds and showed that they block the early stages of viral infection. Two of the inhibitors, bruceine A and gamabufotalin, were also found to block replication of the Delta and Omicron variants of SARS-CoV-2. Both bruceine A and gamabufotalin exhibited potent antiviral activity in K18-hACE2 and wild-type C57BL6/J mice, as evidenced by reduced viral titres in the lung and brain, and protection from alveolar and peribronchial inflammation in the lung, thereby limiting disease progression. We propose that our fluorescent assay can be applied to identify antiviral compounds with potential as therapeutic treatment for COVID-19 and other respiratory diseases.
Collapse
|
26
|
Lee HS, Volpe SJ, Chang EH. The Role of Viruses in the Inception of Chronic Rhinosinusitis. Clin Exp Otorhinolaryngol 2022; 15:310-318. [PMID: 36455880 PMCID: PMC9723285 DOI: 10.21053/ceo.2022.01004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/12/2022] [Accepted: 10/25/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic rhinosinusitis (CRS) is a complex inflammatory disorder that affects between 2% and 16% of adults in the United States, with estimated healthcare costs between 4 and 12 million USD. Viruses are a common etiologic factor for URIs, are frequently identified in the sinuses of patients with CRS, and trigger CRS exacerbations. Therefore, investigating the role of viruses provides an opportunity to identify their role in the pathogenesis of CRS. In this review, we identified the viruses frequently isolated in patients with CRS, as well as their associated immunologic responses and contributions to inflammation. Rhinovirus, parainfluenza virus, influenza virus, and respiratory syncytial virus are the viruses commonly found in patients with CRS. This information allows us to target pathways early in the pathogenesis of CRS, thereby playing a significant role in slowing the progression of this chronic disease.
Collapse
Affiliation(s)
- Hyeon Seung Lee
- Department of Otolaryngology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Sophia J Volpe
- Department of Otolaryngology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Eugene H Chang
- Department of Otolaryngology, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
27
|
Hardin LT, Xiao N. miRNAs: The Key Regulator of COVID-19 Disease. Int J Cell Biol 2022; 2022:1645366. [PMID: 36345541 PMCID: PMC9637033 DOI: 10.1155/2022/1645366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/30/2022] [Indexed: 01/12/2024] Open
Abstract
As many parts of the world continue to fight the innumerable waves of COVID-19 infection, SARS-CoV-2 continues to sculpt its antigenic determinants to enhance its virulence and evolvability. Several vaccines were developed and used around the world, and oral antiviral medications are being developed against SARS-CoV-2. However, studies showed that the virus is mutating in line with the antibody's neutralization escape; thus, new therapeutic alternatives are solicited. We hereby review the key role that miRNAs can play as epigenetic mediators of the cross-talk between SARS-CoV-2 and the host cells. The limitations resulting from the "virus intelligence" to escape and antagonize the host miRNAs as well as the possible mechanisms that could be used in the viral evasion strategies are discussed. Lastly, we suggest new therapeutic approaches based on viral miRNAs.
Collapse
Affiliation(s)
- Leyla Tahrani Hardin
- Department of Biomedical Sciences at the Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, 94103 CA, USA
| | - Nan Xiao
- Department of Biomedical Sciences at the Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, 94103 CA, USA
| |
Collapse
|
28
|
Pires De Souza GA, Le Bideau M, Boschi C, Wurtz N, Colson P, Aherfi S, Devaux C, La Scola B. Choosing a cellular model to study SARS-CoV-2. Front Cell Infect Microbiol 2022; 12:1003608. [PMID: 36339347 PMCID: PMC9634005 DOI: 10.3389/fcimb.2022.1003608] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/03/2022] [Indexed: 08/04/2023] Open
Abstract
As new pathogens emerge, new challenges must be faced. This is no different in infectious disease research, where identifying the best tools available in laboratories to conduct an investigation can, at least initially, be particularly complicated. However, in the context of an emerging virus, such as SARS-CoV-2, which was recently detected in China and has become a global threat to healthcare systems, developing models of infection and pathogenesis is urgently required. Cell-based approaches are crucial to understanding coronavirus infection biology, growth kinetics, and tropism. Usually, laboratory cell lines are the first line in experimental models to study viral pathogenicity and perform assays aimed at screening antiviral compounds which are efficient at blocking the replication of emerging viruses, saving time and resources, reducing the use of experimental animals. However, determining the ideal cell type can be challenging, especially when several researchers have to adapt their studies to specific requirements. This review strives to guide scientists who are venturing into studying SARS-CoV-2 and help them choose the right cellular models. It revisits basic concepts of virology and presents the currently available in vitro models, their advantages and disadvantages, and the known consequences of each choice.
Collapse
Affiliation(s)
- Gabriel Augusto Pires De Souza
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Marion Le Bideau
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Céline Boschi
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Nathalie Wurtz
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Philippe Colson
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Sarah Aherfi
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Christian Devaux
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Department of Biological Sciences (INSB), Centre National de la Recherche Scientifique, Marseille, France
| | - Bernard La Scola
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Aix-Marseille Université, Marseille, France
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| |
Collapse
|
29
|
Labarrere CA, Kassab GS. Glutathione deficiency in the pathogenesis of SARS-CoV-2 infection and its effects upon the host immune response in severe COVID-19 disease. Front Microbiol 2022; 13:979719. [PMID: 36274722 PMCID: PMC9582773 DOI: 10.3389/fmicb.2022.979719] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/14/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 19 (COVID-19) has numerous risk factors leading to severe disease with high mortality rate. Oxidative stress with excessive production of reactive oxygen species (ROS) that lower glutathione (GSH) levels seems to be a common pathway associated with the high COVID-19 mortality. GSH is a unique small but powerful molecule paramount for life. It sustains adequate redox cell signaling since a physiologic level of oxidative stress is fundamental for controlling life processes via redox signaling, but excessive oxidation causes cell and tissue damage. The water-soluble GSH tripeptide (γ-L-glutamyl-L-cysteinyl-glycine) is present in the cytoplasm of all cells. GSH is at 1-10 mM concentrations in all mammalian tissues (highest concentration in liver) as the most abundant non-protein thiol that protects against excessive oxidative stress. Oxidative stress also activates the Kelch-like ECH-associated protein 1 (Keap1)-Nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) redox regulator pathway, releasing Nrf2 to regulate the expression of genes that control antioxidant, inflammatory and immune system responses, facilitating GSH activity. GSH exists in the thiol-reduced and disulfide-oxidized (GSSG) forms. Reduced GSH is the prevailing form accounting for >98% of total GSH. The concentrations of GSH and GSSG and their molar ratio are indicators of the functionality of the cell and its alteration is related to various human pathological processes including COVID-19. Oxidative stress plays a prominent role in SARS-CoV-2 infection following recognition of the viral S-protein by angiotensin converting enzyme-2 receptor and pattern recognition receptors like toll-like receptors 2 and 4, and activation of transcription factors like nuclear factor kappa B, that subsequently activate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) expression succeeded by ROS production. GSH depletion may have a fundamental role in COVID-19 pathophysiology, host immune response and disease severity and mortality. Therapies enhancing GSH could become a cornerstone to reduce severity and fatal outcomes of COVID-19 disease and increasing GSH levels may prevent and subdue the disease. The life value of GSH makes for a paramount research field in biology and medicine and may be key against SARS-CoV-2 infection and COVID-19 disease.
Collapse
|
30
|
Giannakopoulos S, Strange DP, Jiyarom B, Abdelaal O, Bradshaw AW, Nerurkar VR, Ward MA, Bakse J, Yap J, Vanapruks S, Boisvert W, Tallquist MD, Shikuma C, Sadri-Ardekani H, Clapp P, Murphy S, Verma S. In vitro evidence against productive SARS-CoV-2 infection of human testicular cells: Bystander effects of infection mediate testicular injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.09.21.508904. [PMID: 36172118 PMCID: PMC9516847 DOI: 10.1101/2022.09.21.508904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The hallmark of severe COVID-19 involves systemic cytokine storm and multi-organ failure including testicular injury and germ cell depletion. The ACE2 receptor is also expressed in the resident testicular cells however, SARS-CoV-2 infection and mechanisms of testicular injury are not fully understood. The testicular injury can likely result either from direct virus infection of resident cells or by exposure to systemic inflammatory mediators or virus antigens. We here characterized SARS-CoV-2 infection in different human testicular 2D and 3D models including primary Sertoli cells, Leydig cells, mixed seminiferous tubule cells (STC), and 3D human testicular organoids (HTO). Data shows that SARS-CoV-2 does not establish a productive infection in any testicular cell types. However, exposure of STC and HTO to inflammatory supernatant from infected airway epithelial cells and COVID-19 plasma depicted a significant decrease in cell viability and death of undifferentiated spermatogonia. Further, exposure to only SARS-CoV-2 envelope protein, but not Spike or nucleocapsid proteins led to cytopathic effects on testicular cells that was dependent on the TLR2 receptor. A similar trend was observed in the K18h-ACE2 mouse model which revealed gross pathology in the absence of virus replication in the testis. Collectively, data strongly indicates that the testicular injury is not due to direct infection of SARS-CoV-2 but more likely an indirect effect of exposure to systemic inflammation or SARS-CoV-2 antigens. Data also provide novel insights into the mechanism of testicular injury and could explain the clinical manifestation of testicular symptoms associated with severe COVID-19.
Collapse
Affiliation(s)
- Stefanos Giannakopoulos
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Daniel P Strange
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Boonyanudh Jiyarom
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Omar Abdelaal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Aaron W Bradshaw
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Vivek R Nerurkar
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Monika A Ward
- Institute for Biogenesis Research, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Jackson Bakse
- Institute for Biogenesis Research, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Selena Vanapruks
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - William Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Michelle D Tallquist
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Cecilia Shikuma
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Hooman Sadri-Ardekani
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Urology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Philip Clapp
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sean Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Saguna Verma
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
31
|
Hesperetin from Root Extract of Clerodendrum petasites S. Moore Inhibits SARS-CoV-2 Spike Protein S1 Subunit-Induced NLRP3 Inflammasome in A549 Lung Cells via Modulation of the Akt/MAPK/AP-1 Pathway. Int J Mol Sci 2022; 23:ijms231810346. [PMID: 36142258 PMCID: PMC9498987 DOI: 10.3390/ijms231810346] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/24/2022] Open
Abstract
Inhibition of inflammatory responses from the spike glycoprotein of SARS-CoV-2 (Spike) by targeting NLRP3 inflammasome has recently been developed as an alternative form of supportive therapy besides the traditional anti-viral approaches. Clerodendrum petasites S. Moore (C. petasites) is a Thai traditional medicinal plant possessing antipyretic and anti-inflammatory activities. In this study, C. petasites ethanolic root extract (CpEE) underwent solvent-partitioned extraction to obtain the ethyl acetate fraction of C. petasites (CpEA). Subsequently, C. petasites extracts were determined for the flavonoid contents and anti-inflammatory properties against spike induction in the A549 lung cells. According to the HPLC results, CpEA significantly contained higher amounts of hesperidin and hesperetin flavonoids than CpEE (p < 0.05). A549 cells were then pre-treated with either C. petasites extracts or its active flavonoids and were primed with 100 ng/mL of spike S1 subunit (Spike S1) and determined for the anti-inflammatory properties. The results indicate that CpEA (compared with CpEE) and hesperetin (compared with hesperidin) exhibited greater anti-inflammatory properties upon Spike S1 induction through a significant reduction in IL-6, IL-1β, and IL-18 cytokine releases in A549 cells culture supernatant (p < 0.05). Additionally, CpEA and hesperetin significantly inhibited the Spike S1-induced inflammatory gene expressions (NLRP3, IL-1β, and IL-18, p < 0.05). Mechanistically, CpEA and hesperetin attenuated inflammasome machinery protein expressions (NLRP3, ASC, and Caspase-1), as well as inactivated the Akt/MAPK/AP-1 pathway. Overall, our findings could provide scientific-based evidence to support the use of C. petasites and hesperetin in the development of supportive therapies for the prevention of COVID-19-related chronic inflammation.
Collapse
|
32
|
Feng Y, Young CH, Lau SH, He M. Outbreak control management: Lessons from SARS-CoV-2 infections in 2020-2022 in Hong Kong, an international municipality with high-frequency travelers. MedComm (Beijing) 2022; 3:e158. [PMID: 35898696 PMCID: PMC9309738 DOI: 10.1002/mco2.158] [Citation(s) in RCA: 3] [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: 05/25/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022] Open
Abstract
The control management of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections is one of the most challenges in the 21st century. By May 8th, 2022, SARS-CoV-2 has infected over 510 million people with 6.2 million death worldwide and over 1.2 million people with 9133 deaths in the fifth wave of infection in Hong Kong. The government responded rapidly in the early days of the 2020 outbreak, and the results were encouraging to control COVID-19 outbreak unavailable of vaccine. The quick responses to the epidemic alerts, for example, public education and control policies, kept residents safe from infection in the city with such a high population density and large-scale travelers. Nevertheless, the extremely high infectivity, Omicron variant infections, and the shortcomings of transmission control measures led to uncontrollable outbreak in 2022. The weak immunity groups, elderly and children, experienced a high hospitalization rate and mortality rate because of low vaccination rate. Currently, the infection is under well controlled. This study timely summarizes the challenges, policy, and lessons of SARS-CoV-2 outbreak control from 2020 to 2022. More importantly, the lesson and policy revealed from this study may be beneficial and applied to other cities with the outbreak of highly infectious SARS-CoV-2.
Collapse
Affiliation(s)
- Yaxiu Feng
- Department of Biomedical SciencesCity University of Hong KongHong KongChina
| | - Ching Han Young
- Department of Biomedical SciencesCity University of Hong KongHong KongChina
- Cellomics International LimitedHong KongChina
| | - Siu Hin Lau
- Department of Biomedical SciencesCity University of Hong KongHong KongChina
| | - Ming‐Liang He
- Department of Biomedical SciencesCity University of Hong KongHong KongChina
- CityU Shenzhen Research InstituteNanshan, ShengzhenGuangdongChina
| |
Collapse
|
33
|
Baindara P, Sarker MB, Earhart AP, Mandal SM, Schrum AG. NOTCH signaling in COVID-19: a central hub controlling genes, proteins, and cells that mediate SARS-CoV-2 entry, the inflammatory response, and lung regeneration. Front Cell Infect Microbiol 2022; 12:928704. [PMID: 35992174 PMCID: PMC9386183 DOI: 10.3389/fcimb.2022.928704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/11/2022] [Indexed: 01/19/2023] Open
Abstract
In the lungs of infected individuals, the downstream molecular signaling pathways induced by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are incompletely understood. Here, we describe and examine predictions of a model in which NOTCH may represent a central signaling axis in lung infection in Coronavirus Disease 2019 (COVID-19). A pathway involving NOTCH signaling, furin, ADAM17, and ACE2 may be capable of increasing SARS-CoV-2 viral entry and infection. NOTCH signaling can also upregulate IL-6 and pro-inflammatory mediators induced to hyperactivation in COVID-19. Furthermore, if NOTCH signaling fails to turn down properly and stays elevated, airway regeneration during lung healing can be inhibited—a process that may be at play in COVID-19. With specific NOTCH inhibitor drugs in development and clinical trials for other diseases being conducted, the roles of NOTCH in all of these processes central to both infection and healing merit contemplation if such drugs might be applied to COVID-19 patients.
Collapse
Affiliation(s)
- Piyush Baindara
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- *Correspondence: Piyush Baindara, ; Santi M. Mandal, ; Adam G. Schrum,
| | - Md Bodruzzaman Sarker
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia MO, United States
| | - Alexander P. Earhart
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Santi M. Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, India
- *Correspondence: Piyush Baindara, ; Santi M. Mandal, ; Adam G. Schrum,
| | - Adam G. Schrum
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, Columbia MO, United States
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Biomedical, Biological, & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
- *Correspondence: Piyush Baindara, ; Santi M. Mandal, ; Adam G. Schrum,
| |
Collapse
|
34
|
Yip KM, Lee KM, Ng TB, Xu S, Yung KKL, Qu S, Cheung AKL, Sze SCW. An anti-inflammatory and anti-fibrotic proprietary Chinese medicine nasal spray designated as Allergic Rhinitis Nose Drops (ARND) with potential to prevent SARS-CoV-2 coronavirus infection by targeting RBD (Delta)- angiotensin converting enzyme 2 (ACE2) binding. Chin Med 2022; 17:88. [PMID: 35897044 PMCID: PMC9328017 DOI: 10.1186/s13020-022-00635-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/18/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Since the outbreak of COVID-19 has resulted in over 313,000,000 confirmed cases of infection and over 5,500,000 deaths, substantial research work has been conducted to discover agents/ vaccines against COVID-19. Undesired adverse effects were observed in clinical practice and common vaccines do not protect the nasal tissue. An increasing volume of direct evidence based on clinical studies of traditional Chinese medicines (TCM) in the treatment of COVID-19 has been reported. However, the safe anti-inflammatory and anti-fibrotic proprietary Chinese medicines nasal spray, designated as Allergic Rhinitis Nose Drops (ARND), and its potential of re-purposing for suppressing viral infection via SARS-CoV-2 RBD (Delta)- angiotensin converting enzyme 2 (ACE2) binding have not been elucidated. PURPOSE To characterize ARND as a potential SARS-CoV-2 entry inhibitor for its possible preventive application in anti-virus hygienic agent. METHODS Network pharmacology analysis of ARND was adopted to asacertain gene targets which were commonly affected by COVID-19. The inhibitory effect of ARND on viral infection was determined by an in vitro pseudovirus assay. Furthermore, ARND was confirmed to have a strong binding affinity with ACE2 and SARS-CoV-2 spike-RBD (Delta) by ELISA. Finally, inflammatory and fibrotic cell models were used in conjunction in this study. RESULTS The results suggested ARND not only inhibited pseudovirus infection and undermined the binding affinity between ACE2 and the Spike protein (Delta), but also attenuated the inflammatory response upon infection and may lead to a better prognosis with a lower risk of pulmonary fibrosis. The data in this study also provide a basis for further development of ARND as an antiviral hygienic product and further investigations on ARND in the live virus, in vivo and COVID-19 patients. ARND holds promise for use in the current COVID-19 outbreak as well as in future pandemics. CONCLUSION ARND could be considered as a safe anti-SARS-CoV-2 agent with potential to prevent SARS-CoV-2 coronavirus infection.
Collapse
Affiliation(s)
- Ka Man Yip
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China.,Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China
| | - Kwan Ming Lee
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China.,Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, Special Administrative Region, China
| | - Shujun Xu
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China.,Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China
| | - Ken Kin Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China. .,Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China.
| | - Shaogang Qu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China. .,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, Guangdong, China. .,Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Allen Ka Loon Cheung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China.
| | - Stephen Cho Wing Sze
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China. .,Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, Special Administrative Region, China.
| |
Collapse
|
35
|
Abstract
The dramatic global consequences of the coronavirus disease 2019 (COVID-19) pandemic soon fueled quests for a suitable model that would facilitate the development and testing of therapies and vaccines. In contrast to other rodents, hamsters are naturally susceptible to infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the Syrian hamster (Mesocricetus auratus) rapidly developed into a popular model. It recapitulates many characteristic features as seen in patients with a moderate, self-limiting course of the disease such as specific patterns of respiratory tract inflammation, vascular endothelialitis, and age dependence. Among 4 other hamster species examined, the Roborovski dwarf hamster (Phodopus roborovskii) more closely mimics the disease in highly susceptible patients with frequent lethal outcome, including devastating diffuse alveolar damage and coagulopathy. Thus, different hamster species are available to mimic different courses of the wide spectrum of COVID-19 manifestations in humans. On the other hand, fewer diagnostic tools and information on immune functions and molecular pathways are available than in mice, which limits mechanistic studies and inference to humans in several aspects. Still, under pandemic conditions with high pressure on progress in both basic and clinically oriented research, the Syrian hamster has turned into the leading non-transgenic model at an unprecedented pace, currently used in innumerable studies that all aim to combat the impact of the virus with its new variants of concern. As in other models, its strength rests upon a solid understanding of its similarities to and differences from the human disease, which we review here.
Collapse
|
36
|
Nie Z, Li Y, Li X, Xu Y, Yang G, Ke M, Qu X, Qin Y, Tan J, Fan Y, Zhu C. Layer-by-Layer Assembly of a Polysaccharide "Armor" on the Cell Surface Enabling the Prophylaxis of Virus Infection. ACS APPLIED MATERIALS & INTERFACES 2022; 14:acsami.2c03442. [PMID: 35639584 PMCID: PMC9173675 DOI: 10.1021/acsami.2c03442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Airborne pathogens, such as the world-spreading severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause global epidemics via transmission through the respiratory pathway. It is of great urgency to develop adequate interventions that can protect individuals against future pandemics. This study presents a nasal spray that forms a polysaccharide "armor" on the cell surface through the layer-by-layer self-assembly (LBL) method to minimize the risk of virus infection. The nasal spray has two separate components: chitosan and alginate. Harnessing the electrostatic interaction, inhaling the two polysaccharides alternatively enables the assembly of a barrier that reduces virus uptake into the cells. The results showed that this approach has no obvious cellular injury and endows cells with the ability to resist the infection of adenovirus and SARS-CoV-2 pseudovirus. Such a method can be a potential preventive strategy for protecting the respiratory tract against multiple viruses, especially the upcoming SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Zhiqiang Nie
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Yinghao Li
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
- Chongqing Institute of Zhong Zhi Yi
Gu, Shapingba District, Chongqing 400030, China
| | - Xinxin Li
- State Key Laboratory of Primate Biomedical Research,
Institute of Primate Translational Medicine, Kunming University of Science
and Technology, Kunming 650500, China
| | - Youqian Xu
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Guanyuan Yang
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Ming Ke
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Xiaohang Qu
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Yinhua Qin
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Ju Tan
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Yonghong Fan
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
| | - Chuhong Zhu
- Department of Anatomy, National and Regional
Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory
for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing,
Third Military Medical University, Chongqing 400038,
China
- State Key Laboratory of Primate Biomedical Research,
Institute of Primate Translational Medicine, Kunming University of Science
and Technology, Kunming 650500, China
- State Key Laboratory of Trauma, Burn and
Combined Injury, Chongqing 400038, China
| |
Collapse
|
37
|
Truzzi F, Whittaker A, D’Amen E, Tibaldi C, Abate A, Valerii MC, Spisni E, Dinelli G. Wheat Germ Spermidine and Clove Eugenol in Combination Stimulate Autophagy In Vitro Showing Potential in Supporting the Immune System against Viral Infections. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113425. [PMID: 35684363 PMCID: PMC9182079 DOI: 10.3390/molecules27113425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 12/02/2022]
Abstract
Impaired autophagy, responsible for increased inflammation, constitutes a risk factor for the more severe COVID-19 outcomes. Spermidine (SPD) is a known autophagy modulator and supplementation for COVID-19 risk groups (including the elderly) is recommended. However, information on the modulatory effects of eugenol (EUG) is scarce. Therefore, the effects of SPD and EUG, both singularly and in combination, on autophagy were investigated using different cell lines (HBEpiC, SHSY5Y, HUVEC, Caco-2, L929 and U937). SPD (0.3 mM), EUG (0.2 mM) and 0.3 mM SPD + 0.2 mM EUG, significantly increased autophagy using the hallmark measure of LC3-II protein accumulation in the cell lines without cytotoxic effects. Using Caco-2 cells as a model, several crucial autophagy proteins were upregulated at all stages of autophagic flux in response to the treatments. This effect was verified by the activation/differentiation and migration of U937 monocytes in a three-dimensional reconstituted intestinal model (Caco-2, L929 and U937 cells). Comparable benefits of SPD, EUG and SPD + EUG in inducing autophagy were shown by the protection of Caco-2 and L929 cells against lipopolysaccharide-induced inflammation. SPD + EUG is an innovative dual therapy capable of stimulating autophagy and reducing inflammation in vitro and could show promise for COVID-19 risk groups.
Collapse
Affiliation(s)
- Francesca Truzzi
- Department of Agricultural and Food Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (A.W.); (E.D.); (C.T.); (A.A.); (G.D.)
- Correspondence: ; Tel.: +39-051-2096674
| | - Anne Whittaker
- Department of Agricultural and Food Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (A.W.); (E.D.); (C.T.); (A.A.); (G.D.)
| | - Eros D’Amen
- Department of Agricultural and Food Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (A.W.); (E.D.); (C.T.); (A.A.); (G.D.)
| | - Camilla Tibaldi
- Department of Agricultural and Food Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (A.W.); (E.D.); (C.T.); (A.A.); (G.D.)
| | - Antonella Abate
- Department of Agricultural and Food Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (A.W.); (E.D.); (C.T.); (A.A.); (G.D.)
| | - Maria Chiara Valerii
- Department of Biological, Geological and Environmental Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (M.C.V.); (E.S.)
| | - Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (M.C.V.); (E.S.)
| | - Giovanni Dinelli
- Department of Agricultural and Food Sciences, Alma Mater Studiorum-University of Bologna, 40127 Bologna, Italy; (A.W.); (E.D.); (C.T.); (A.A.); (G.D.)
| |
Collapse
|
38
|
Baldassi D, Ambike S, Feuerherd M, Cheng CC, Peeler DJ, Feldmann DP, Porras-Gonzalez DL, Wei X, Keller LA, Kneidinger N, Stoleriu MG, Popp A, Burgstaller G, Pun SH, Michler T, Merkel OM. Inhibition of SARS-CoV-2 replication in the lung with siRNA/VIPER polyplexes. J Control Release 2022; 345:661-674. [PMID: 35364120 PMCID: PMC8963978 DOI: 10.1016/j.jconrel.2022.03.051] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 01/11/2023]
Abstract
SARS-CoV-2 has been the cause of a global pandemic since 2019 and remains a medical urgency. siRNA-based therapies are a promising strategy to fight viral infections. By targeting a specific region of the viral genome, siRNAs can efficiently downregulate viral replication and suppress viral infection. However, to achieve the desired therapeutic activity, siRNA requires a suitable delivery system. The VIPER (virus-inspired polymer for endosomal release) block copolymer has been reported as promising delivery system for both plasmid DNA and siRNA in the past years. It is composed of a hydrophilic block for condensation of nucleic acids as well as a hydrophobic, pH-sensitive block that, at acidic pH, exposes the membrane lytic peptide melittin, which enhances endosomal escape. In this study, we aimed at developing a formulation for pulmonary administration of siRNA to suppress SARS-CoV-2 replication in lung epithelial cells. After characterizing siRNA/VIPER polyplexes, the activity and safety profile were confirmed in a lung epithelial cell line. To further investigate the activity of the polyplexes in a more sophisticated cell culture system, an air-liquid interface (ALI) culture was established. siRNA/VIPER polyplexes reached the cell monolayer and penetrated through the mucus layer secreted by the cells. Additionally, the activity against wild-type SARS-CoV-2 in the ALI model was confirmed by qRT-PCR. To investigate translatability of our findings, the activity against SARS-CoV-2 was tested ex vivo in human lung explants. Here, siRNA/VIPER polyplexes efficiently inhibited SARS-CoV-2 replication. Finally, we verified the delivery of siRNA/VIPER polyplexes to lung epithelial cells in vivo, which represent the main cellular target of viral infection in the lung. In conclusion, siRNA/VIPER polyplexes efficiently delivered siRNA to lung epithelial cells and mediated robust downregulation of viral replication both in vitro and ex vivo without toxic or immunogenic side effects in vivo, demonstrating the potential of local siRNA delivery as a promising antiviral therapy in the lung.
Collapse
Affiliation(s)
- Domizia Baldassi
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-University of Munich, Butenandtstraße 5, 81377 Munich, Germany
| | - Shubhankar Ambike
- Institute of Virology, School of Medicine, Technical University of Munich / Helmholtz Zentrum Munich, Trogerstr.30, 81675 Munich, Germany
| | - Martin Feuerherd
- Institute of Virology, School of Medicine, Technical University of Munich / Helmholtz Zentrum Munich, Trogerstr.30, 81675 Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, School of Medicine, Technical University of Munich / Helmholtz Zentrum Munich, Trogerstr.30, 81675 Munich, Germany
| | - David J Peeler
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, United States
| | - Daniel P Feldmann
- Department of Oncology, Wayne State University School of Medicine, 4100 John R St, Detroit, MI 48201, United States
| | - Diana Leidy Porras-Gonzalez
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Xin Wei
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Lea-Adriana Keller
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-University of Munich, Butenandtstraße 5, 81377 Munich, Germany; Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Nikolaus Kneidinger
- Department of Medicine V, University Hospital, LMU, Munich, Member of the German Center for Lung Research (DZL), Germany
| | - Mircea Gabriel Stoleriu
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Pulmonary Hospital; Marchioninistraße 15, 81377 Munich and Robert-Koch-Allee 2, 82131 Gauting, Germany
| | - Andreas Popp
- Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Gerald Burgstaller
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, United States
| | - Thomas Michler
- Institute of Virology, School of Medicine, Technical University of Munich / Helmholtz Zentrum Munich, Trogerstr.30, 81675 Munich, Germany; Institute of Laboratory Medicine, University Hospital, LMU, Munich, Germany
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-University of Munich, Butenandtstraße 5, 81377 Munich, Germany; Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
39
|
Savchenko AA, Tikhonova E, Kudryavtsev I, Kudlay D, Korsunsky I, Beleniuk V, Borisov A. TREC/KREC Levels and T and B Lymphocyte Subpopulations in COVID-19 Patients at Different Stages of the Disease. Viruses 2022; 14:646. [PMID: 35337053 PMCID: PMC8954181 DOI: 10.3390/v14030646] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND T and B cell-mediated immunity can be assessed using T cell receptor excision circle (TREC) and Kappa-deleting recombination excision circle (KREC) analysis, respectively, and successful implementation of this method requires evaluation of the correlation between the TREC frequencies and T cell subsets as well as KREC levels and B lymphocyte subsets. The aim of the present study was to evaluate the correlation between the TREC/KREC concentrations and T/B lymphocyte subsets at different stages of COVID-19. METHODS We examined 33 patients in the acute stage of COVID-19 (including 8 patients with poor outcomes) and 33 COVID-19 survivors. TREC/KREC concentrations were measured using quantitative real-time PCR. T/B lymphocyte subsets were determined using flow cytometry. RESULTS Blood TREC and KREC levels were found to be significantly lower in the acute stage of COVID-19 compared to control values. Moreover, a zero blood TREC level was a predictor of a poor disease outcome. Reductions in CD3+CD4+CD45RO-CD62L- and CD3+CD8+CD45RO-CD62L- T cell counts (as well as in the main fractions of B1 and B2 B cells) indicated a favorable outcome in COVID-19 patients in the acute stage of the disease. Decreased CD3+CD4+CD45RO-CD62L+ and CD3+CD8+CD45RO-CD62L+ T cell frequencies and increased CD3+CD8+CD45RO-CD62L- cell counts were found to indicate a poor outcome in patients with acute COVID-19. These patients were also found to have increased B1 cell counts while demonstrating no changes in B2 cell counts. The levels of effector T cell subsets an naïve B cells were normal in COVID-19 survivors. The most pronounced correlations between TREC/KREC levels and T/B cell subsets counts were observed in COVID-19 survivors: there were positive correlations with naïve T and B lymphocytes and negative correlations with central and effector memory T cell subsets. CONCLUSIONS The assessment of correlations between TREC and T cell subsets as well as KREC levels and B cell subset counts in patients with acute COVID-19 and COVID-19 survivors has shown that blood concentrations of TREC and KREC are sensitive indicators of the stage of antigen-independent differentiation of adaptive immunity cells. The results of the TREC and KREC analysis correlated with the stages of COVID-19 and differed depending on the outcome of COVID-19.
Collapse
Affiliation(s)
- Andrei A. Savchenko
- Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.A.S.); (V.B.); (A.B.)
| | - Elena Tikhonova
- Ministry of Health of the Russian Federation, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia;
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Dmitry Kudlay
- National Research Center—Institute of Immunology, Federal Medical-Biological Agency, 115522 Moscow, Russia;
- Ministry of Health of the Russian Federation, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Ilya Korsunsky
- Moscow City Center for Pediatric Immunology and Allergy, G. Speransky Children’s Hospital No 9, 129329 Moscow, Russia;
| | - Vasily Beleniuk
- Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.A.S.); (V.B.); (A.B.)
| | - Alexandr Borisov
- Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.A.S.); (V.B.); (A.B.)
| |
Collapse
|
40
|
Paul S, Bravo Vázquez LA, Reyes-Pérez PR, Estrada-Meza C, Aponte Alburquerque RA, Pathak S, Banerjee A, Bandyopadhyay A, Chakraborty S, Srivastava A. The role of microRNAs in solving COVID-19 puzzle from infection to therapeutics: A mini-review. Virus Res 2022; 308:198631. [PMID: 34788642 PMCID: PMC8590742 DOI: 10.1016/j.virusres.2021.198631] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023]
Abstract
Nowadays, one of the major global health concerns is coronavirus disease 2019 (COVID-19), which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Even though numerous treatments and vaccines to combat this virus are currently under development, the detailed molecular mechanisms underlying the pathogenesis of this disease are yet to be elucidated to design future therapeutic tools against SARS-CoV-2 variants. MicroRNAs (miRNAs) are small (20-24 nucleotides), non-coding RNA molecules that regulate post-transcriptional gene expression. Recently, it has been demonstrated that both host and viral-encoded miRNAs are crucial for the successful infection of SARS-CoV-2. For instance, dysregulation of miRNAs that modulate multiple genes expressed in COVID-19 patients with comorbidities (e.g., type 2 diabetes, lung adenocarcinoma, and cerebrovascular disorders) could affect the severity of the disease. Thus, altered expression levels of circulating miRNAs might be helpful to diagnose this illness and forecast whether a COVID-19 patient could develop a severe state of the disease. Besides, researchers have found a number of miRNAs could inhibit the expression of proteins, such as ACE2, TMPRSS2, spike, and Nsp12, involved in the life cycle of SARS-CoV-2. Accordingly, miRNAs represent potential biomarkers and therapeutic targets for this devastating viral disease. Therefore, in this current review, we present the recent discoveries regarding the clinical relevance and biological roles of miRNAs in COVID-19.
Collapse
Affiliation(s)
- Sujay Paul
- Tecnológico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, CP 76130 Querétaro, México.
| | - Luis Alberto Bravo Vázquez
- Tecnológico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, CP 76130 Querétaro, México
| | - Paula Roxana Reyes-Pérez
- Tecnológico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, CP 76130 Querétaro, México
| | - Carolina Estrada-Meza
- Tecnológico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, CP 76130 Querétaro, México
| | - Rafael Arturo Aponte Alburquerque
- Tecnológico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, CP 76130 Querétaro, México
| | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Anindya Bandyopadhyay
- International Rice Research Institute, Manila, Philippines; Reliance Industries Ltd, Navi Mumbai, India
| | - Samik Chakraborty
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Aashish Srivastava
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
41
|
Shirbhate E, Pandey J, Patel VK, Kamal M, Jawaid T, Gorain B, Kesharwani P, Rajak H. Understanding the role of ACE-2 receptor in pathogenesis of COVID-19 disease: a potential approach for therapeutic intervention. Pharmacol Rep 2021; 73:1539-1550. [PMID: 34176080 PMCID: PMC8236094 DOI: 10.1007/s43440-021-00303-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) and its homologue, ACE2, are commonly allied with hypertension, renin-angiotensin-aldosterone system pathway, and other cardiovascular system disorders. The recent pandemic of COVID-19 has attracted the attention of numerous researchers on ACE2 receptors, where the causative viral particle, SARS-CoV-2, is established to exploit these receptors for permitting their entry into the human cells. Therefore, studies on the molecular origin and pathophysiology of the cell response in correlation to the role of ACE2 receptors to these viruses are bringing novel theories. The varying level of manifestation and importance of ACE proteins, underlying irregularities and disorders, intake of specific medications, and persistence of assured genomic variants at the ACE genes are potential questions raising nowadays while observing the marked alteration in response to the SARS-CoV-2-infected patients. Therefore, the present review has focused on several raised opinions associated with the role of the ACE2 receptor and its impact on COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India
| | - Jaiprakash Pandey
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India
| | - Vijay K Patel
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box No. 173, Al-Kharj, 11942, Kingdom of Saudi Arabia
| | - Talha Jawaid
- Department of Pharmacology, College of Medicine, Al Imam Bin Saud Islamic University, Riyadh, 13314, Kingdom of Saudi Arabia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Harish Rajak
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India.
| |
Collapse
|
42
|
Kamle S, Ma B, He CH, Akosman B, Zhou Y, Lee CM, El-Deiry WS, Huntington K, Liang O, Machan JT, Kang MJ, Shin HJ, Mizoguchi E, Lee CG, Elias JA. Chitinase 3-like-1 is a therapeutic target that mediates the effects of aging in COVID-19. JCI Insight 2021; 6:e148749. [PMID: 34747367 PMCID: PMC8663553 DOI: 10.1172/jci.insight.148749] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/29/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is caused by SARS-CoV-2 (SC2) and is more prevalent and severe in elderly and patients with comorbid diseases (CM). Because chitinase 3-like-1 (CHI3L1) is induced during aging and CM, the relationships between CHI3L1 and SC2 were investigated. Here, we demonstrate that CHI3L1 is a potent stimulator of the SC2 receptor angiotensin converting enzyme 2 (ACE2) and viral spike protein priming proteases (SPP), that ACE2 and SPP are induced during aging, and that anti-CHI3L1, kasugamycin, and inhibitors of phosphorylation abrogate these ACE2- and SPP-inductive events. Human studies also demonstrate that the levels of circulating CHI3L1 are increased in the elderly and patients with CM, where they correlate with COVID-19 severity. These studies demonstrate that CHI3L1 is a potent stimulator of ACE2 and SPP, that this induction is a major mechanism contributing to the effects of aging during SC2 infection, and that CHI3L1 co-opts the CHI3L1 axis to augment SC2 infection. CHI3L1 plays a critical role in the pathogenesis of and is an attractive therapeutic target in COVID-19.
Collapse
Affiliation(s)
| | - Bing Ma
- Molecular Microbiology and Immunology
| | | | | | - Yang Zhou
- Molecular Microbiology and Immunology
| | | | - Wafik S. El-Deiry
- Pathology and Laboratory Medicine
- Hematology-Oncology Division, Department of Medicine
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
| | - Kelsey Huntington
- Pathology and Laboratory Medicine
- Hematology-Oncology Division, Department of Medicine
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
| | - Olin Liang
- Hematology-Oncology Division, Department of Medicine
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
| | - Jason T. Machan
- Department of Biostatistics, Lifespan Health System, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hyeon Jun Shin
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emiko Mizoguchi
- Molecular Microbiology and Immunology
- Department of Immunology, Kurume University, School of Medicine, Kurume, Fukuoka, Japan
| | | | - Jack A. Elias
- Molecular Microbiology and Immunology
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
- Department of Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
43
|
Vázquez-Jiménez A, Avila-Ponce De León UE, Matadamas-Guzman M, Muciño-Olmos EA, Martínez-López YE, Escobedo-Tapia T, Resendis-Antonio O. On Deep Landscape Exploration of COVID-19 Patients Cells and Severity Markers. Front Immunol 2021; 12:705646. [PMID: 34603282 PMCID: PMC8481922 DOI: 10.3389/fimmu.2021.705646] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 is a disease with a spectrum of clinical responses ranging from moderate to critical. To study and control its effects, a large number of researchers are focused on two substantial aims. On the one hand, the discovery of diverse biomarkers to classify and potentially anticipate the disease severity of patients. These biomarkers could serve as a medical criterion to prioritize attention to those patients with higher prone to severe responses. On the other hand, understanding how the immune system orchestrates its responses in this spectrum of disease severities is a fundamental issue required to design new and optimized therapeutic strategies. In this work, using single-cell RNAseq of bronchoalveolar lavage fluid of nine patients with COVID-19 and three healthy controls, we contribute to both aspects. First, we presented computational supervised machine-learning models with high accuracy in classifying the disease severity (moderate and severe) in patients with COVID-19 starting from single-cell data from bronchoalveolar lavage fluid. Second, we identified regulatory mechanisms from the heterogeneous cell populations in the lungs microenvironment that correlated with different clinical responses. Given the results, patients with moderate COVID-19 symptoms showed an activation/inactivation profile for their analyzed cells leading to a sequential and innocuous immune response. In comparison, severe patients might be promoting cytotoxic and pro-inflammatory responses in a systemic fashion involving epithelial and immune cells without the possibility to develop viral clearance and immune memory. Consequently, we present an in-depth landscape analysis of how transcriptional factors and pathways from these heterogeneous populations can regulate their expression to promote or restrain an effective immune response directly linked to the patients prognosis.
Collapse
Affiliation(s)
- Aarón Vázquez-Jiménez
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Ugo Enrique Avila-Ponce De León
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biológicas, UNAM, Mexico City, Mexico
| | - Meztli Matadamas-Guzman
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico
| | - Erick Andrés Muciño-Olmos
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico
| | - Yoscelina E. Martínez-López
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Médicas y de la Salud, UNAM, Mexico City, Mexico
| | - Thelma Escobedo-Tapia
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Maestría y Doctorado en Ciencias Bioquímicas, UNAM, Mexico City, Mexico
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Coordinación de la Investigación Científica - Red de Apoyo a la Investigación, UNAM, Mexico City, Mexico
| |
Collapse
|
44
|
Post-mortem dissection of COVID-19: a pathogenic role for macrophages? Intensive Care Med 2021; 47:1322-1325. [PMID: 34471939 PMCID: PMC8409470 DOI: 10.1007/s00134-021-06509-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/15/2021] [Indexed: 11/23/2022]
|
45
|
Ultraviolet-A light reduces cellular cytokine release from human endotracheal cells infected with Coronavirus. Photodiagnosis Photodyn Ther 2021; 35:102457. [PMID: 34314863 PMCID: PMC8310417 DOI: 10.1016/j.pdpdt.2021.102457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 01/08/2023]
Abstract
Background An important clinical feature of coronavirus disease 2019 (COVID-19) is hypercytokinemia (cytokine storm). We previously showed that narrow band ultraviolet-A (NB-UVA) treatment salvages coronavirus (CoV)-229E-infected human tracheal cells, and that daily endotracheal NB-UVA therapy reduced severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) levels in human subjects, with improved clinical outcomes. Here, we examined NB-UVA effects on cytokine release during CoV-229E infection. Methods Primary human tracheal epithelial cells were transfected with CoV-229E, then exposed to 2 mW/cm2 NB-UVA for 20 minutes every 24h, either 3 or 4 times. Secreted cytokine/chemokine levels were analyzed in supernatants collected from CoV-229E-infected/UVA-exposed cells 24h after the last UVA treatment, and from matched non-infected/UVA-exposed controls, CoV-229E-infected/non-exposed controls, and non-infected/non-exposed (naïve) controls. Metabolic pathway/downstream prediction analyses were also performed. Results Pro-inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF), and chemokines IL-8, monocyte chemoattractant protein-1 (MCP1), and interferon gamma-induced protein 10 (IP-10), were significantly increased in CoV-229E-infected cells, and significantly decreased following NB-UVA treatment. Interferon (IFN)-α2, IFN-γ, and IL-10 were not upregulated in response to CoV-229E. Metabolic pathway predictions indicated hypercytokinemia as the top inflammatory response in CoV-229E-infected cells, whereas the top predicted pathway in CoV-229E-infected/UVA-exposed cells was the recovery stage of severe acute respiratory syndrome. Conclusions Human tracheal epithelial cells infected with CoV-229E showed reduced cytokine secretions including IL-6, TNF, IL-8, and MCP-1, following NB-UVA exposure. This reduction of cytokine levels in vitro, coupled with previously identified reduced cell death in CoV-229E-infected/UVA-exposed cells, suggests that determining UVA effects on cytokine storm in human SARS-Co-V2 patients is warranted.
Collapse
|
46
|
Sarr D, Gingerich AD, Asthiwi NM, Almutairi F, Sautto GA, Ecker J, Nagy T, Kilgore MB, Chandler JD, Ross TM, Tripp RA, Rada B. Dual oxidase 1 promotes antiviral innate immunity. Proc Natl Acad Sci U S A 2021; 118:e2017130118. [PMID: 34168077 PMCID: PMC8256044 DOI: 10.1073/pnas.2017130118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Indexed: 12/30/2022] Open
Abstract
Dual oxidase 1 (DUOX1) is an NADPH oxidase that is highly expre-ssed in respiratory epithelial cells and produces H2O2 in the airway lumen. While a line of prior in vitro observations suggested that DUOX1 works in partnership with an airway peroxidase, lactoperoxidase (LPO), to produce antimicrobial hypothiocyanite (OSCN-) in the airways, the in vivo role of DUOX1 in mammalian organisms has remained unproven to date. Here, we show that Duox1 promotes antiviral innate immunity in vivo. Upon influenza airway challenge, Duox1-/- mice have enhanced mortality, morbidity, and impaired lung viral clearance. Duox1 increases the airway levels of several cytokines (IL-1β, IL-2, CCL1, CCL3, CCL11, CCL19, CCL20, CCL27, CXCL5, and CXCL11), contributes to innate immune cell recruitment, and affects epithelial apoptosis in the airways. In primary human tracheobronchial epithelial cells, OSCN- is generated by LPO using DUOX1-derived H2O2 and inactivates several influenza strains in vitro. We also show that OSCN- diminishes influenza replication and viral RNA synthesis in infected host cells that is inhibited by the H2O2 scavenger catalase. Binding of the influenza virus to host cells and viral entry are both reduced by OSCN- in an H2O2-dependent manner in vitro. OSCN- does not affect the neuraminidase activity or morphology of the influenza virus. Overall, this antiviral function of Duox1 identifies an in vivo role of this gene, defines the steps in the infection cycle targeted by OSCN-, and proposes that boosting this mechanism in vivo can have therapeutic potential in treating viral infections.
Collapse
Affiliation(s)
- Demba Sarr
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Aaron D Gingerich
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Nuha Milad Asthiwi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Faris Almutairi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602
| | - Giuseppe A Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Jeffrey Ecker
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Tamás Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Matthew B Kilgore
- Department of Pediatrics, Division of Pulmonary, Allergy and Immunology, Cystic Fibrosis, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322
- Center for Cystic Fibrosis and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA 30322
| | - Joshua D Chandler
- Department of Pediatrics, Division of Pulmonary, Allergy and Immunology, Cystic Fibrosis, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322
- Center for Cystic Fibrosis and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA 30322
| | - Ted M Ross
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602;
| |
Collapse
|
47
|
Maruntelu I, Caragea AM, Tizu M, Constantinescu I. SARS-CoV-2 diagnosis: a single-centre experience. J Med Life 2021; 14:257-261. [PMID: 34104250 PMCID: PMC8169149 DOI: 10.25122/jml-2021-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) was declared a pandemic by the World Health Organization (WHO) on the 11th of March 2020. In Romania, there have been 983,217 confirmed cases and 24,386 deaths. We aim to show our experience at the Fundeni Clinical Institute in the diagnosis of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection in both patients and health care personnel. Swab samples were collected for extraction of the SARS-CoV-2 RNA from 29380 patients and health care personnel. We have combined three real-time reverse transcription-polymerase chain reaction (RT-PCR) assays for the qualitative detection of SARS-CoV-2. Also, the presence of IgG against SARS-CoV-2 nucleoprotein was analyzed in 1068 patients and clinical staff using the chemiluminescence method. Other 50 people were screened post-vaccination for the presence of SARS-CoV-2 antibodies against the spike (S) protein, using the chemiluminescence method as well. The majority of confirmed cases were in adults, 71.3% of cases being registered in people aged 30-69 years. Most patients diagnosed with SARS-CoV-2 infection (83%) were admitted to the gastroenterology, hematology, and surgery wards. Our study showed that one-third of people developed antibodies against the nucleocapsid of SARS-CoV-2. SARS-CoV-2 IgG seroprevalence does not vary by gender or age. Also, we noticed the presence of antibodies against the SARS-CoV-2 spike protein in all 50 people post-vaccination that were tested two weeks after the second dose. Due to the increasing number of infected patients with SARS-CoV-2, the new coronavirus pandemic involves a sustained testing effort for an accurate virological diagnosis in both direct and indirect diagnosis.
Collapse
Affiliation(s)
- Ion Maruntelu
- Department of Immunology and Immunology of Transplant, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Centre of Immunogenetics and Virology, Fundeni Clinical Institute, Bucharest, Romania
| | - Andreea Mirela Caragea
- Department of Immunology and Immunology of Transplant, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Centre of Immunogenetics and Virology, Fundeni Clinical Institute, Bucharest, Romania
| | - Maria Tizu
- Department of Immunology and Immunology of Transplant, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Centre of Immunogenetics and Virology, Fundeni Clinical Institute, Bucharest, Romania
| | - Ileana Constantinescu
- Department of Immunology and Immunology of Transplant, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Centre of Immunogenetics and Virology, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
48
|
Lange A, Lange J, Jaskuła E. Cytokine Overproduction and Immune System Dysregulation in alloHSCT and COVID-19 Patients. Front Immunol 2021; 12:658896. [PMID: 34149697 PMCID: PMC8206782 DOI: 10.3389/fimmu.2021.658896] [Citation(s) in RCA: 12] [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: 01/26/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022] Open
Abstract
The COVID-19 pathomechanism depends on (i) the pathogenicity of the virus, (ii) ability of the immune system to respond to the cytopathic effect of the virus infection, (iii) co-morbidities. Inflammatory cytokine production constitutes a hallmark of COVID-19 that is facilitated by inability of adaptive immunity to control virus invasion. The effect of cytokine release syndrome is deleterious, but the severity of it depends on other confounding factors: age and comorbidities. In this study, we analyze the literature data on the post-transplant course of allogeneic hematopoietic stem cell transplanted (alloHSCT) patients, which is affected by generated inflammatory cytokines. The sequence of events boosting cytokine production was analyzed in relation to clinical and laboratory data highlighting the impact of cytokine generation on the post-transplant course. The collected data were compared to those from studies on COVID-19 patients. The similarities are: (i) the damage/pathogen-associated molecular pattern (DAMP/PAMP) stage is similar except for the initiation hit being sterile in alloHSCT (toxic damage of conditioning regimen) and viral in COVID-19; (ii) genetic host-derived factors play a role; (iii) adaptive immunity fails, DAMP signal(s) increases, over-production of cytokines occurs; (iv) monocytes lacking HLADR expression emerge, being suppressor cells hampering adaptive immunity; (v) immune system homeostasis is broken, the patient's status deteriorates to bed dependency, leading to hypo-oxygenation and malnutrition, which in turn stimulates the intracellular alert pathways with vigorous transcription of cytokine genes. All starts with the interaction between DAMPs with appropriate receptors, which leads to the production of pro-inflammatory cytokines, the inflammatory process spreads, tissue is damaged, DAMPs are released and a vicious cycle occurs. Attempts to modify intracellular signaling pathways in patients with post-alloHSCT graft vs host disease have already been undertaken. The similarities documented in this study show that this approach may also be used in COVID-19 patients for tuning signal transduction processes to interrupt the cycle that powers the cytokine overproduction.
Collapse
Affiliation(s)
- Andrzej Lange
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- Lower Silesian Center for Cellular Transplantation with National Bone Marrow Donor Registry, Wroclaw, Poland
| | - Janusz Lange
- Lower Silesian Center for Cellular Transplantation with National Bone Marrow Donor Registry, Wroclaw, Poland
| | - Emilia Jaskuła
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- Lower Silesian Center for Cellular Transplantation with National Bone Marrow Donor Registry, Wroclaw, Poland
| |
Collapse
|