1
|
Gunasekara S, Tamil Selvan M, Murphy CL, Shatnawi S, Cowan S, More S, Ritchey J, Miller CA, Rudd JM. Characterization of Neutrophil Functional Responses to SARS-CoV-2 Infection in a Translational Feline Model for COVID-19. Int J Mol Sci 2024; 25:10054. [PMID: 39337543 PMCID: PMC11432149 DOI: 10.3390/ijms251810054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
There is a complex interplay between viral infection and host innate immune response regarding disease severity and outcomes. Neutrophil hyperactivation, including excessive release of neutrophil extracellular traps (NETs), is linked to exacerbated disease in acute COVID-19, notably in hospitalized patients. Delineating protective versus detrimental neutrophil responses is essential to developing targeted COVID-19 therapies and relies on high-quality translational animal models. In this study, we utilize a previously established feline model for COVID-19 to investigate neutrophil dysfunction in which experimentally infected cats develop clinical disease that mimics acute COVID-19. Specific pathogen-free cats were inoculated with SARS-CoV-2 (B.1.617.2; Delta variant) (n = 24) or vehicle (n = 6). Plasma, bronchoalveolar lavage fluid, and lung tissues were collected at various time points over 12 days post-inoculation. Systematic and temporal evaluation of the kinetics of neutrophil activation was conducted by measuring markers of activation including myeloperoxidase (MPO), neutrophil elastase (NE), and citrullinated histone H3 (citH3) in SARS-CoV-2-infected cats at 4 and 12 days post-inoculation (dpi) and compared to vehicle-inoculated controls. Cytokine profiling supported elevated innate inflammatory responses with specific upregulation of neutrophil activation and NET formation-related markers, namely IL-8, IL-18, CXCL1, and SDF-1, in infected cats. An increase in MPO-DNA complexes and cell-free dsDNA in infected cats compared to vehicle-inoculated was noted and supported by histopathologic severity in respiratory tissues. Immunofluorescence analyses further supported correlation of NET markers with tissue damage, especially 4 dpi. Differential gene expression analyses indicated an upregulation of genes associated with innate immune and neutrophil activation pathways. Transcripts involved in activation and NETosis pathways were upregulated by 4 dpi and downregulated by 12 dpi, suggesting peak activation of neutrophils and NET-associated markers in the early acute stages of infection. Correlation analyses conducted between NET-specific markers and clinical scores as well as histopathologic scores support association between neutrophil activation and disease severity during SARS-CoV-2 infection in this model. Overall, this study emphasizes the effect of neutrophil activation and NET release in SARS-CoV-2 infection in a feline model, prompting further investigation into therapeutic strategies aimed at mitigating excessive innate inflammatory responses in COVID-19.
Collapse
Affiliation(s)
- Sachithra Gunasekara
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Miruthula Tamil Selvan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Chelsea L Murphy
- Department of Mathematical Sciences, College of Arts and Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Shoroq Shatnawi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Shannon Cowan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jerry Ritchey
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Craig A Miller
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jennifer M Rudd
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
2
|
Mazzuca C, Vitiello L, Travaglini S, Maurizi F, Finamore P, Santangelo S, Rigon A, Vadacca M, Angeletti S, Scarlata S. Immunological and homeostatic pathways of alpha -1 antitrypsin: a new therapeutic potential. Front Immunol 2024; 15:1443297. [PMID: 39224588 PMCID: PMC11366583 DOI: 10.3389/fimmu.2024.1443297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
α -1 antitrypsin (A1AT) is a 52 kDa acute-phase glycoprotein belonging to the serine protease inhibitor superfamily (SERPIN). It is primarily synthesized by hepatocytes and to a lesser extent by monocytes, macrophages, intestinal epithelial cells, and bronchial epithelial cells. A1AT is encoded by SERPINA1 locus, also known as PI locus, highly polymorphic with at least 100 allelic variants described and responsible for different A1AT serum levels and function. A1AT inhibits a variety of serine proteinases, but its main target is represented by Neutrophil Elastase (NE). However, recent attention has been directed towards its immune-regulatory and homeostatic activities. A1AT exerts immune-regulatory effects on different cell types involved in innate and adaptive immunity. Additionally, it plays a role in metal and lipid metabolism, contributing to homeostasis. An adequate comprehension of these mechanisms could support the use of A1AT augmentation therapy in many disorders characterized by a chronic immune response. The aim of this review is to provide an up-to-date understanding of the molecular mechanisms and regulatory pathways responsible for immune-regulatory and homeostatic activities of A1AT. This knowledge aims to support the use of A1AT in therapeutic applications. Furthermore, the review summarizes the current state of knowledge regarding the application of A1AT in clinical and laboratory settings human and animal models.
Collapse
Affiliation(s)
- Carmen Mazzuca
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
- Pediatric Allergology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Laura Vitiello
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele University, Rome, Italy
| | - Silvia Travaglini
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Fatima Maurizi
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Panaiotis Finamore
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Simona Santangelo
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| | - Amelia Rigon
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Marta Vadacca
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Simone Scarlata
- Unit of Internal Medicine and Geriatrics, Respiratory Pathophysiology and Thoracic Endoscopy, Fondazione Policlinico Campus Bio Medico University Hospital- Rome, Rome, Italy
| |
Collapse
|
3
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int J Mol Sci 2024; 25:7553. [PMID: 39062796 PMCID: PMC11277036 DOI: 10.3390/ijms25147553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Proteases are produced and released in the mucosal cells of the respiratory tract and have important physiological functions, for example, maintaining airway humidification to allow proper gas exchange. The infectious mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), takes advantage of host proteases in two ways: to change the spatial conformation of the spike (S) protein via endoproteolysis (e.g., transmembrane serine protease type 2 (TMPRSS2)) and as a target to anchor to epithelial cells (e.g., angiotensin-converting enzyme 2 (ACE2)). This infectious process leads to an imbalance in the mucosa between the release and action of proteases versus regulation by anti-proteases, which contributes to the exacerbation of the inflammatory and prothrombotic response in COVID-19. In this article, we describe the most important proteases that are affected in COVID-19, and how their overactivation affects the three main physiological systems in which they participate: the complement system and the kinin-kallikrein system (KKS), which both form part of the contact system of innate immunity, and the renin-angiotensin-aldosterone system (RAAS). We aim to elucidate the pathophysiological bases of COVID-19 in the context of the imbalance between the action of proteases and anti-proteases to understand the mechanism of aprotinin action (a panprotease inhibitor). In a second-part review, titled "Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions", we explain in depth the pharmacodynamics, pharmacokinetics, toxicity, and use of aprotinin as an antiviral drug.
Collapse
Affiliation(s)
- Juan Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
4
|
Bai X, Gao J, Guan X, Narum DE, Fornis LB, Griffith DE, Gao B, Sandhaus RA, Huang H, Chan ED. Analysis of alpha-1-antitrypsin (AAT)-regulated, glucocorticoid receptor-dependent genes in macrophages reveals a novel host defense function of AAT. Physiol Rep 2024; 12:e16124. [PMID: 39016119 PMCID: PMC11252833 DOI: 10.14814/phy2.16124] [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: 04/01/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/18/2024] Open
Abstract
Alpha-1-antitrypsin (AAT) plays a homeostatic role in attenuating excessive inflammation and augmenting host defense against microbes. We demonstrated previously that AAT binds to the glucocorticoid receptor (GR) resulting in significant anti-inflammatory and antimycobacterial consequences in macrophages. Our current investigation aims to uncover AAT-regulated genes that rely on GR in macrophages. We incubated control THP-1 cells (THP-1control) and THP-1 cells knocked down for GR (THP-1GR-KD) with AAT, performed bulk RNA sequencing, and analyzed the findings. In THP-1control cells, AAT significantly upregulated 408 genes and downregulated 376 genes. Comparing THP-1control and THP-1GR-KD cells, 125 (30.6%) of the AAT-upregulated genes and 154 (41.0%) of the AAT-downregulated genes were significantly dependent on GR. Among the AAT-upregulated, GR-dependent genes, CSF-2 that encodes for granulocyte-monocyte colony-stimulating factor (GM-CSF), known to be host-protective against nontuberculous mycobacteria, was strongly upregulated by AAT and dependent on GR. We further quantified the mRNA and protein of several AAT-upregulated, GR-dependent genes in macrophages and the mRNA of several AAT-downregulated, GR-dependent genes. We also discussed the function(s) of selected AAT-regulated, GR-dependent gene products largely in the context of mycobacterial infections. In conclusion, AAT regulated several genes that are dependent on GR and play roles in host immunity against mycobacteria.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of MedicineRocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Junfeng Gao
- Department of Immunology and Genomic MedicineNational Jewish HealthDenverColoradoUSA
| | - Xiaoyu Guan
- Department of Biostatistics and InformaticsUniversity of Colorado School of Public Health Anschutz Medical CampusAuroraColoradoUSA
| | - Drew E. Narum
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
| | | | - David E. Griffith
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of MedicineNational Jewish HealthDenverColoradoUSA
| | - Bifeng Gao
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Robert A. Sandhaus
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of MedicineNational Jewish HealthDenverColoradoUSA
| | - Hua Huang
- Department of Immunology and Genomic MedicineNational Jewish HealthDenverColoradoUSA
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Edward D. Chan
- Department of MedicineRocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
5
|
Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int J Mol Sci 2024; 25:5492. [PMID: 38791530 PMCID: PMC11122108 DOI: 10.3390/ijms25105492] [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: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.
Collapse
Affiliation(s)
- Coby J. Cheetham
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| |
Collapse
|
6
|
Pintanel-Raymundo M, Menao-Guillén S, Perales-Afán JJ, García-Gutiérrez A, Moreno-Gázquez I, Julián-Ansón M, Ramos-Álvarez M, Olivera-González S, Gutiérrez-Cía I, Torralba-Cabeza MA. Analysis of the expression of the Serpina1 gene in SARS-CoV-2 infection: study of a new biomarker. Rev Clin Esp 2024; 224:253-258. [PMID: 38608729 DOI: 10.1016/j.rceng.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/13/2024] [Indexed: 04/14/2024]
Abstract
INTRODUCTION The SERPINA1 gene encodes the protein Alpha-1 Antitrypsin (AAT1). Possible imbalances between the concentrations of proteases and antiproteases (AAT1) can lead to the development of serious pulmonary and extrapulmonary pathologies. In this work we study the importance of this possible imbalance in patients with COVID-19. OBJECTIVES To correlate the severity of the symptoms of SARS-COV-2 infection with the AAT1 concentrations at diagnosis of the disease. METHODS An observational, prospective, cross-sectional, non-interventional, analytical study was carried out where 181 cases with COVID-19 admitted to the "Lozano Blesa" University Clinical Hospital of Zaragoza were selected. The concentration of AAT1 was studied in all of them and this was correlated with the clinical aspects and biochemical parameters at hospital admission. RESULTS 141 cases corresponded to patients with severe COVID and 40 patients with mild COVID. AAT1 levels were positively correlated with the days of hospitalization, severity, C-Reactive Protein, ferritin, admission to Intensive Care, and death, and presented a negative correlation with the number of lymphocytes/mm3. AAT1 concentrations higher than 237.5 mg/dL allowed the patient to be classified as "severe" (S72%; E78%) and 311.5 mg/dL were associated with the risk of admission to Intensive Care or Exitus (S67%; E79%). CONCLUSIONS Levels of the SERPINA1 gene expression product, AAT1, correlate with the severity of COVID-19 patients at diagnosis of the disease, being useful as a prognostic biomarker.
Collapse
Affiliation(s)
| | - S Menao-Guillén
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain; Insitututo de Investigación Sanitaria de Aaragón, España
| | - J J Perales-Afán
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain; Insitututo de Investigación Sanitaria de Aaragón, España
| | - A García-Gutiérrez
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - I Moreno-Gázquez
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - M Julián-Ansón
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - M Ramos-Álvarez
- Servicio de Bioquímica, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - S Olivera-González
- Servicio de Medicina Interna, Hospital Marina Salud de Denia, Alicante, Spain
| | - I Gutiérrez-Cía
- Servicio de Cuidados Intensivos, Hospital Universitario "Lozano Blesa", Zaragoza, Spain
| | - M A Torralba-Cabeza
- Facultad de Medicina, Universidad de Zaragoza, Spain; Insitututo de Investigación Sanitaria de Aaragón, España; Unidad de Enfermedades Minoritarias, Servicio de Medicina Interna, Hospital Universitario "Lozano Blesa", Zaragoza, Spain; Grupo de Trabajo de Enfermedades Minoritarias de la Sociedad Española de Medicina Interna, España.
| |
Collapse
|
7
|
Lotke R, Petersen M, Sauter D. Restriction of Viral Glycoprotein Maturation by Cellular Protease Inhibitors. Viruses 2024; 16:332. [PMID: 38543698 PMCID: PMC10975521 DOI: 10.3390/v16030332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 05/23/2024] Open
Abstract
The human genome is estimated to encode more than 500 proteases performing a wide range of important physiological functions. They digest proteins in our food, determine the activity of hormones, induce cell death and regulate blood clotting, for example. During viral infection, however, some proteases can switch sides and activate viral glycoproteins, allowing the entry of virions into new target cells and the spread of infection. To reduce unwanted effects, multiple protease inhibitors regulate the proteolytic processing of self and non-self proteins. This review summarizes our current knowledge of endogenous protease inhibitors, which are known to limit viral replication by interfering with the proteolytic activation of viral glycoproteins. We describe the underlying molecular mechanisms and highlight the diverse strategies by which protease inhibitors reduce virion infectivity. We also provide examples of how viruses evade the restriction imposed by protease inhibitors. Finally, we briefly outline how cellular protease inhibitors can be modified and exploited for therapeutic purposes. In summary, this review aims to summarize our current understanding of cellular protease inhibitors as components of our immune response to a variety of viral pathogens.
Collapse
Affiliation(s)
| | | | - Daniel Sauter
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
8
|
Zhou X, Jin J, Lv T, Song Y. A Narrative Review: The Role of NETs in Acute Respiratory Distress Syndrome/Acute Lung Injury. Int J Mol Sci 2024; 25:1464. [PMID: 38338744 PMCID: PMC10855305 DOI: 10.3390/ijms25031464] [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: 11/09/2023] [Revised: 12/14/2023] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
Nowadays, acute respiratory distress syndrome (ARDS) still has a high mortality rate, and the alleviation and treatment of ARDS remains a major research focus. There are various causes of ARDS, among which pneumonia and non-pulmonary sepsis are the most common. Trauma and blood transfusion can also cause ARDS. In ARDS, the aggregation and infiltration of neutrophils in the lungs have a great influence on the development of the disease. Neutrophils regulate inflammatory responses through various pathways, and the release of neutrophils through neutrophil extracellular traps (NETs) is considered to be one of the most important mechanisms. NETs are mainly composed of DNA, histones, and granuloproteins, all of which can mediate downstream signaling pathways that can activate inflammatory responses, generate immune clots, and cause damage to surrounding tissues. At the same time, the components of NETs can also promote the formation and release of NETs, thus forming a vicious cycle that continuously aggravates the progression of the disease. NETs are also associated with cytokine storms and immune balance. Since DNA is the main component of NETs, DNase I is considered a viable drug for removing NETs. Other therapeutic methods to inhibit the formation of NETs are also worthy of further exploration. This review discusses the formation and mechanism of NETs in ARDS. Understanding the association between NETs and ARDS may help to develop new perspectives on the treatment of ARDS.
Collapse
Affiliation(s)
| | | | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210093, China; (X.Z.); (J.J.)
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210093, China; (X.Z.); (J.J.)
| |
Collapse
|
9
|
Wu A, Shi K, Wang J, Zhang R, Wang Y. Targeting SARS-CoV-2 entry processes: The promising potential and future of host-targeted small-molecule inhibitors. Eur J Med Chem 2024; 263:115923. [PMID: 37981443 DOI: 10.1016/j.ejmech.2023.115923] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/16/2023] [Accepted: 10/28/2023] [Indexed: 11/21/2023]
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has had a huge impact on global health. To respond to rapidly mutating viruses and to prepare for the next pandemic, there is an urgent need to develop small molecule therapies that target critical stages of the SARS-CoV-2 life cycle. Inhibiting the entry process of the virus can effectively control viral infection and play a role in prevention and treatment. Host factors involved in this process, such as ACE2, TMPRSS2, ADAM17, furin, PIKfyve, TPC2, CTSL, AAK1, V-ATPase, HSPG, and NRP1, have been found to be potentially good targets with stability. Through further exploration of the cell entry process of SARS-CoV-2, small-molecule drugs targeting these host factors have been developed. This review focuses on the structural functions of potential host cell targets during the entry of SARS-CoV-2 into host cells. The research progress, chemical structure, structure-activity relationship, and clinical value of small-molecule inhibitors against COVID-19 are reviewed to provide a reference for the development of small-molecule drugs against COVID-19.
Collapse
Affiliation(s)
- Aijia Wu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Kunyu Shi
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Ruofei Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, National Clinical Research Center for Geriatrics, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
10
|
J NH, Venkataraman A, Thiruvengadam K, B B, M K, S S, Balaji S, S E, Smuk M, Hanna LE, Prendergast AJ. Evaluation of platelet indices as markers of tuberculosis among children in India. ERJ Open Res 2024; 10:00734-2023. [PMID: 38410718 PMCID: PMC10895425 DOI: 10.1183/23120541.00734-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/15/2023] [Indexed: 02/28/2024] Open
Abstract
Children with tuberculosis have increased platelet count and platelet/lymphocyte ratio along with decreased mean platelet volume, suggesting that these indices may be useful as adjunct tools in diagnosis of paediatric tuberculosis https://bit.ly/3Ga4AWT.
Collapse
Affiliation(s)
- Nancy Hilda J
- ICMR - National Institute for Research in Tuberculosis, Chennai, India
- Joint first authors
| | - Aishwarya Venkataraman
- ICMR - National Institute for Research in Tuberculosis, Chennai, India
- Blizard Institute, Queen Mary University of London, London, UK
- Joint first authors
| | | | - Brindha B
- ICMR - National Institute for Research in Tuberculosis, Chennai, India
| | - Karthick M
- ICMR - National Institute for Research in Tuberculosis, Chennai, India
| | - Subha S
- ICMR - National Institute for Research in Tuberculosis, Chennai, India
| | - Sarath Balaji
- Institute of Child Health, Madras Medical College, Chennai, India
| | - Elilarasi S
- Institute of Child Health, Madras Medical College, Chennai, India
| | - Melanie Smuk
- Blizard Institute, Queen Mary University of London, London, UK
| | - Luke Elizabeth Hanna
- ICMR - National Institute for Research in Tuberculosis, Chennai, India
- Joint senior authors
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK
- Joint senior authors
| |
Collapse
|
11
|
Ishii S, Sakaguchi W, Yamamura M, Nagumo T, Koeda S, Akiyama H, Kinuta M, Nishikubo S, Tsukinoki K. Association between salivary proteases and protease inhibitors linked with viral infections and oral inflammatory diseases. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101572. [PMID: 37495185 DOI: 10.1016/j.jormas.2023.101572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 07/24/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Despite the role of transmembrane protease, serine 2 (TMPRSS2) in facilitating the entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the primary cause of the global COVID-19 pandemic, the interaction of extracellular and intracellular proteases in this process remains poorly elucidated. Thus, we monitored the salivary expression concentration (SEC) of TMPRSS2 and its inhibitor, alpha-1 antitrypsin (A1AT), and investigated whether oral inflammatory diseases affected the SEC of both proteins. MATERIALS AND METHODS We collected saliva samples before and after surgical treatment of inflammatory cystic diseases (radicular and inflammatory dentigerous cysts) in 25 patients. The SEC of TMPRSS2 and A1AT was measured using enzyme-linked immunosorbent assay. SEC in multiple patient status groups and subgroups of each status were investigated. Finally, the correlation between TMPRSS2 and A1AT SEC was analyzed. RESULTS The TMPRSS2 and A1AT SEC did not significantly change pre- or post-treatment. The TMPRSS2 SEC was significantly higher before and after treatment in patients aged >50 years, patients with radicular cysts, and patients with the basic disease. A1AT SEC was significantly decreased after treatment in the acute inflammation, large-sized, and patients without basic disease groups. No significant correlation was observed between the SEC of either protein before and after treatment. DISCUSSION Individual-specific SEC for TMPRSS2 may be influenced by age, lesion type, and basic disease; however, oral inflammatory diseases may not have a direct effect. Moreover, the extent of oral inflammatory diseases and the presence of basic diseases may be associated with A1AT SEC. Furthermore, the SEC between the two proteins may be independent.
Collapse
Affiliation(s)
- Shigeru Ishii
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Wakako Sakaguchi
- Department of Environmental Pathology, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka City, Kanagawa, 238-8580, Japan.
| | - Makiko Yamamura
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Tatsuhito Nagumo
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Satoko Koeda
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Hiroki Akiyama
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Mikihisa Kinuta
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Shuichi Nishikubo
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Keiichi Tsukinoki
- Department of Environmental Pathology, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka City, Kanagawa, 238-8580, Japan.
| |
Collapse
|
12
|
Zuniga-Hertz JP, Chitteti R, Dispenza J, Cuomo R, Bonds JA, Kopp EL, Simpson S, Okerblom J, Maurya S, Rana BK, Miyonahara A, Niesman IR, Maree J, Belza G, Hamilton HD, Stanton C, Gonzalez DJ, Poirier MA, Moeller-Bertram T, Patel HH. Meditation-induced bloodborne factors as an adjuvant treatment to COVID-19 disease. Brain Behav Immun Health 2023; 32:100675. [PMID: 37600600 PMCID: PMC10432704 DOI: 10.1016/j.bbih.2023.100675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 07/12/2023] [Accepted: 08/06/2023] [Indexed: 08/22/2023] Open
Abstract
The COVID-19 pandemic has resulted in significant morbidity and mortality worldwide. Management of the pandemic has relied mainly on SARS-CoV-2 vaccines, while alternative approaches such as meditation, shown to improve immunity, have been largely unexplored. Here, we probe the relationship between meditation and COVID-19 disease and directly test the impact of meditation on the induction of a blood environment that modulates viral infection. We found a significant inverse correlation between length of meditation practice and SARS-CoV-2 infection as well as accelerated resolution of symptomology of those infected. A meditation "dosing" effect was also observed. In cultured human lung cells, blood from experienced meditators induced factors that prevented entry of pseudotyped viruses for SARS-CoV-2 spike protein of both the wild-type Wuhan-1 virus and the Delta variant. We identified and validated SERPINA5, a serine protease inhibitor, as one possible protein factor in the blood of meditators that is necessary and sufficient for limiting pseudovirus entry into cells. In summary, we conclude that meditation can enhance resiliency to viral infection and may serve as a possible adjuvant therapy in the management of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Juan P. Zuniga-Hertz
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ramamurthy Chitteti
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - Raphael Cuomo
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jacqueline A. Bonds
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Elena L. Kopp
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sierra Simpson
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jonathan Okerblom
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Svetlana Maurya
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Brinda K. Rana
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Atsushi Miyonahara
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ingrid R. Niesman
- San Diego State University, Electron Microscope Facility, 5500 Campanile Dr, San Diego, CA, 92182, USA
| | - Jacqueline Maree
- VitaMed Research, 44630 Monterey Ave., Palm Desert, CA, 92260, USA
| | - Gianna Belza
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | | | - David J. Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | | | - Hemal H. Patel
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
13
|
Bai X, Schountz T, Buckle AM, Talbert JL, Sandhaus RA, Chan ED. Alpha-1-antitrypsin antagonizes COVID-19: a review of the epidemiology, molecular mechanisms, and clinical evidence. Biochem Soc Trans 2023; 51:1361-1375. [PMID: 37294003 PMCID: PMC10317171 DOI: 10.1042/bst20230078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023]
Abstract
Alpha-1-antitrypsin (AAT), a serine protease inhibitor (serpin), is increasingly recognized to inhibit SARS-CoV-2 infection and counter many of the pathogenic mechanisms of COVID-19. Herein, we reviewed the epidemiologic evidence, the molecular mechanisms, and the clinical evidence that support this paradigm. As background to our discussion, we first examined the basic mechanism of SARS-CoV-2 infection and contend that despite the availability of vaccines and anti-viral agents, COVID-19 remains problematic due to viral evolution. We next underscored that measures to prevent severe COVID-19 currently exists but teeters on a balance and that current treatment for severe COVID-19 remains grossly suboptimal. We then reviewed the epidemiologic and clinical evidence that AAT deficiency increases risk of COVID-19 infection and of more severe disease, and the experimental evidence that AAT inhibits cell surface transmembrane protease 2 (TMPRSS2) - a host serine protease required for SARS-CoV-2 entry into cells - and that this inhibition may be augmented by heparin. We also elaborated on the panoply of other activities of AAT (and heparin) that could mitigate severity of COVID-19. Finally, we evaluated the available clinical evidence for AAT treatment of COVID-19.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, U.S.A
- Department of Academic Affairs, National Jewish Health, Denver, CO, U.S.A
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, U.S.A
| | - Tony Schountz
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, U.S.A
| | - Ashley M. Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- PTNG Bio, Melbourne, Australia
| | - Janet L. Talbert
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, U.S.A
| | | | - Edward D. Chan
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, U.S.A
- Department of Academic Affairs, National Jewish Health, Denver, CO, U.S.A
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, U.S.A
| |
Collapse
|
14
|
Han L, Wu X, Wang O, Luan X, Velander WH, Aynardi M, Halstead ES, Bonavia AS, Jin R, Li G, Li Y, Wang Y, Dong C, Lei Y. Mesenchymal stromal cells and alpha-1 antitrypsin have a strong synergy in modulating inflammation and its resolution. Theranostics 2023; 13:2843-2862. [PMID: 37284443 PMCID: PMC10240832 DOI: 10.7150/thno.83942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/25/2023] [Indexed: 06/08/2023] Open
Abstract
Rationale: Trauma, surgery, and infection can cause severe inflammation. Both dysregulated inflammation intensity and duration can lead to significant tissue injuries, organ dysfunction, mortality, and morbidity. Anti-inflammatory drugs such as steroids and immunosuppressants can dampen inflammation intensity, but they derail inflammation resolution, compromise normal immunity, and have significant adverse effects. The natural inflammation regulator mesenchymal stromal cells (MSCs) have high therapeutic potential because of their unique capabilities to mitigate inflammation intensity, enhance normal immunity, and accelerate inflammation resolution and tissue healing. Furthermore, clinical studies have shown that MSCs are safe and effective. However, they are not potent enough, alone, to completely resolve severe inflammation and injuries. One approach to boost the potency of MSCs is to combine them with synergistic agents. We hypothesized that alpha-1 antitrypsin (A1AT), a plasma protein used clinically and has an excellent safety profile, was a promising candidate for synergism. Methods: This investigation examined the efficacy and synergy of MSCs and A1AT to mitigate inflammation and promote resolution, using in vitro inflammatory assay and in vivo mouse acute lung injury model. The in vitro assay measured cytokine releases, inflammatory pathways, reactive oxygen species (ROS), and neutrophil extracellular traps (NETs) production by neutrophils and phagocytosis in different immune cell lines. The in vivo model monitored inflammation resolution, tissue healing, and animal survival. Results: We found that the combination of MSCs and A1AT was much more effective than each component alone in i) modulating cytokine releases and inflammatory pathways, ii) inhibiting ROS and NETs production by neutrophils, iii) enhancing phagocytosis and, iv) promoting inflammation resolution, tissue healing, and animal survival. Conclusion: These results support the combined use of MSCs, and A1AT is a promising approach for managing severe, acute inflammation.
Collapse
Affiliation(s)
- Li Han
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University; University Park, PA, 16802, USA
| | - Xinran Wu
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln; Lincoln, NE, 68588, USA
| | - Xiao Luan
- Biomedical Center of Qingdao University; Qingdao, Shandong, 266000, China
| | - William H. Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln; Lincoln, NE, 68588, USA
| | - Michael Aynardi
- Department of Orthopedics Surgery, Pennsylvania State University College of Medicine; Hershey, PA, 17033, USA
| | - E. Scott Halstead
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Anthony S. Bonavia
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Rong Jin
- Department of Neurosurgery, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Guohong Li
- Department of Neurosurgery, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Yulong Li
- Department of Emergency Medicine, University of Nebraska Medical Center; Omaha, NE, 68105, USA
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
| | - Yuguo Lei
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University; University Park, PA, 16802, USA
| |
Collapse
|
15
|
Tian WJ, Wang XJ. Broad-Spectrum Antivirals Derived from Natural Products. Viruses 2023; 15:v15051100. [PMID: 37243186 DOI: 10.3390/v15051100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Scientific advances have led to the development and production of numerous vaccines and antiviral drugs, but viruses, including re-emerging and emerging viruses, such as SARS-CoV-2, remain a major threat to human health. Many antiviral agents are rarely used in clinical treatment, however, because of their inefficacy and resistance. The toxicity of natural products may be lower, and some natural products have multiple targets, which means less resistance. Therefore, natural products may be an effective means to solve virus infection in the future. New techniques and ideas are currently being developed for the design and screening of antiviral drugs thanks to recent revelations about virus replication mechanisms and the advancement of molecular docking technology. This review will summarize recently discovered antiviral drugs, mechanisms of action, and screening and design strategies for novel antiviral agents.
Collapse
Affiliation(s)
- Wen-Jun Tian
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, China Agricultural University, Beijing 100193, China
| | - Xiao-Jia Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, China Agricultural University, Beijing 100193, China
| |
Collapse
|
16
|
Ceja-Gálvez HR, Renteria-Flores FI, Nicoletti F, Hernández-Bello J, Macedo-Ojeda G, Muñoz-Valle JF. Severe COVID-19: Drugs and Clinical Trials. J Clin Med 2023; 12:2893. [PMID: 37109231 PMCID: PMC10142549 DOI: 10.3390/jcm12082893] [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: 02/27/2023] [Revised: 04/08/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
By January of 2023, the COVID-19 pandemic had led to a reported total of 6,700,883 deaths and 662,631,114 cases worldwide. To date, there have been no effective therapies or standardized treatment schemes for this disease; therefore, the search for effective prophylactic and therapeutic strategies is a primary goal that must be addressed. This review aims to provide an analysis of the most efficient and promising therapies and drugs for the prevention and treatment of severe COVID-19, comparing their degree of success, scope, and limitations, with the aim of providing support to health professionals in choosing the best pharmacological approach. An investigation of the most promising and effective treatments against COVID-19 that are currently available was carried out by employing search terms including "Convalescent plasma therapy in COVID-19" or "Viral polymerase inhibitors" and "COVID-19" in the Clinicaltrials.gov and PubMed databases. From the current perspective and with the information available from the various clinical trials assessing the efficacy of different therapeutic options, we conclude that it is necessary to standardize certain variables-such as the viral clearance time, biomarkers associated with severity, hospital stay, requirement of invasive mechanical ventilation, and mortality rate-in order to facilitate verification of the efficacy of such treatments and to better assess the repeatability of the most effective and promising results.
Collapse
Affiliation(s)
- Hazael Ramiro Ceja-Gálvez
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Francisco Israel Renteria-Flores
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Jorge Hernández-Bello
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Gabriela Macedo-Ojeda
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
17
|
Molecular Mechanisms of Neutrophil Extracellular Trap (NETs) Degradation. Int J Mol Sci 2023; 24:ijms24054896. [PMID: 36902325 PMCID: PMC10002918 DOI: 10.3390/ijms24054896] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Although many studies have been exploring the mechanisms driving NETs formation, much less attention has been paid to the degradation and elimination of these structures. The NETs clearance and the effective removal of extracellular DNA, enzymatic proteins (neutrophil elastase, proteinase 3, myeloperoxidase) or histones are necessary to maintain tissue homeostasis, to prevent inflammation and to avoid the presentation of self-antigens. The persistence and overabundance of DNA fibers in the circulation and tissues may have dramatic consequences for a host leading to the development of various systemic and local damage. NETs are cleaved by a concerted action of extracellular and secreted deoxyribonucleases (DNases) followed by intracellular degradation by macrophages. NETs accumulation depends on the ability of DNase I and DNAse II to hydrolyze DNA. Furthermore, the macrophages actively engulf NETs and this event is facilitated by the preprocessing of NETs by DNase I. The purpose of this review is to present and discuss the current knowledge about the mechanisms of NETs degradation and its role in the pathogenesis of thrombosis, autoimmune diseases, cancer and severe infections, as well as to discuss the possibilities for potential therapeutic interventions. Several anti-NETs approaches had therapeutic effects in animal models of cancer and autoimmune diseases; nevertheless, the development of new drugs for patients needs further study for an effective development of clinical compounds that are able to target NETs.
Collapse
|
18
|
Rodríguez-García C, Rodríguez-Ruiz E, Ruano-Raviña A, Cruz R, Piñeiro-Lamas M, Casal A, Lapunzina P, Carracedo Á, Valdés L, Valdés L. Is SARS-COV-2 associated with alpha-1 antitrypsin deficiency? J Thorac Dis 2023; 15:711-717. [PMID: 36910046 PMCID: PMC9992632 DOI: 10.21037/jtd-22-1062] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/23/2022] [Indexed: 01/31/2023]
Affiliation(s)
| | - Emilio Rodríguez-Ruiz
- Clinical University Hospital of Santiago, Santiago de Compostela, Spain.,Simulation, Life Support and Intensive Care Research Unit of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alberto Ruano-Raviña
- Preventive Medicine and Public Health Unit, University of Santiago de Compostela, Santiago de Compostela, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiology and Public Health - CIBERESP), Madrid, Spain.,Health Research Institute (IDIS), Santiago de Compostela, Spain
| | - Raquel Cruz
- Health Research Institute (IDIS), Santiago de Compostela, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain.,Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Piñeiro-Lamas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiology and Public Health - CIBERESP), Madrid, Spain.,Health Research Institute (IDIS), Santiago de Compostela, Spain
| | - Ana Casal
- Pulmonology Department, Clinical University Hospital of Santiago, Santiago de Compostela, Spain
| | - Pablo Lapunzina
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain.,Institute of Medical and Molecular Genetics (INGEMM), University Hospital La Paz - IDIPAZ, Madrid, Spain
| | - Ángel Carracedo
- Health Research Institute (IDIS), Santiago de Compostela, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain.,Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.,Galician Public Foundation for Genomic Medicine, Galician Health Service (SERGAS), Santiago de Compostela, Spain
| | - Luis Valdés
- Pulmonology Department, Clinical University Hospital of Santiago, Santiago de Compostela, Spain.,Health Research Institute (IDIS), Santiago de Compostela, Spain.,Medicine Department, University of Santiago, Santiago de Compostela, Spain
| | | |
Collapse
|
19
|
Calle Rubio M, López-Campos JL, Miravitlles M, Michel de la Rosa FJ, Hernández Pérez JM, Montero Martínez C, Montoro Ronsano JB, Casas Maldonado F, Rodríguez Hermosa JL, Tabernero Huguet EM, Martínez Sesmero JM, Martínez Rivera C, Callejas González FJ, Torres Durán M. COVID-19's impact on care practice for alpha-1-antitrypsin deficiency patients. BMC Health Serv Res 2023; 23:98. [PMID: 36717880 PMCID: PMC9885054 DOI: 10.1186/s12913-023-09094-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Patients with alpha-1 antitrypsin deficiency (AATD), commonly categorized as a rare disease, have been affected by the changes in healthcare management brought about by COVID-19. This study's aim was to identify the changes that have taken place in AATD patient care as a result of the COVID-19 pandemic in Spain and to propose experts' recommendations aimed at ensuring humanized and quality care for people with AATD in the post-pandemic situation. METHODS A qualitative descriptive case study with a holistic single-case design was conducted, using focus groups with experts in AATD clinical management, including 15 health professionals with ties to the Spanish health system (12 pneumologists and 2 hospital pharmacists from 11 different hospitals in Spain) and 1 patient representative. RESULTS COVID-19 has had a major impact on numerous aspects of AATD clinical patient management in Spain, including diagnostic, treatment, and follow-up phases. The experts concluded that there is a need to strengthen coordination between Primary Care and Hospital Care and improve the coordination processes across all the organizations and actors involved in the healthcare system. Regarding telemedicine and telecare, experts have concluded that it is necessary to promote this methodology and to develop protocols and training programs. Experts have recommended developing personalized and precision medicine, and patient participation in decision-making, promoting self-care and patient autonomy to optimize their healthcare and improve their quality of life. The possibility of monitoring and treating AATD patients from home has also been proposed by experts. Another result of the study was the recommendation of the need to ensure that plasma donations are made on a regular basis by a sufficient number of healthy individuals. CONCLUSION The study advances knowledge by highlighting the challenges faced by health professionals and changes in AATD patient management in the context of the COVID-19 pandemic. It also proposes experts' recommendations aimed at ensuring humanized and quality care for people with AATD in the post-pandemic situation. This work could serve as a reference study for physicians on their daily clinical practice with AATD patients and may also provide guidance on the changes to be put in place for the post-pandemic situation.
Collapse
Affiliation(s)
- Myriam Calle Rubio
- grid.414780.ePulmonology Department, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), San Carlos Clinical Hospital, Madrid, Spain ,grid.4795.f0000 0001 2157 7667Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - José Luis López-Campos
- grid.411109.c0000 0000 9542 1158Medical-Surgical Unit for Respiratory Diseases, Biomedicine Institute of Sevilla (IBiS), Virgen del Rocío University Hospital, Seville, Spain
| | - Marc Miravitlles
- grid.411083.f0000 0001 0675 8654Pulmonology Department, Research Institute of Vall d’Hebron (VHIR), Vall d’Hebron University Hospital, Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Network of Centers for Biomedical Research On Respiratory Diseases (CIBERES), Carlos III Health Institute, Madrid, Spain
| | | | - José María Hernández Pérez
- grid.413448.e0000 0000 9314 1427Network of Centers for Biomedical Research On Respiratory Diseases (CIBERES), Carlos III Health Institute, Madrid, Spain ,Pulmonology Department, Nuestra Señora de Candelaria University Hospital, Santa Cruz de Tenerife, Spain
| | - Carmen Montero Martínez
- grid.411066.40000 0004 1771 0279Pulmonology Department, A Coruña University Hospital, A Coruña, Spain
| | - José Bruno Montoro Ronsano
- grid.411083.f0000 0001 0675 8654Department of Pharmacy, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Francisco Casas Maldonado
- grid.411380.f0000 0000 8771 3783Pulmonology Department, San Cecilio Clinical University Hospital, Granada, Spain ,grid.4489.10000000121678994School of Health Science, University of Granada, Granada, Spain
| | - Juan Luis Rodríguez Hermosa
- grid.414780.ePulmonology Department, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), San Carlos Clinical Hospital, Madrid, Spain ,grid.4795.f0000 0001 2157 7667Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | - Carlos Martínez Rivera
- grid.413448.e0000 0000 9314 1427Network of Centers for Biomedical Research On Respiratory Diseases (CIBERES), Carlos III Health Institute, Madrid, Spain ,grid.411438.b0000 0004 1767 6330Pulmonology Department, Research Institute of Germans Trias I Pujol (IGTiP), Germans Trias I Pujol University Hospital, Barcelona, Spain
| | | | - María Torres Durán
- Pulmonology Department, Health Research Institute of Galicia Sur (IISGS), Álvaro Cunqueiro Hospital, Estrada Clara Campoamor, 342. 36312 Vigo, Spain
| |
Collapse
|
20
|
Thabet RH, Massadeh NA, Badarna OB, Al-Momani OM. Highlights on molecular targets in the management of COVID-19: Possible role of pharmacogenomics. J Int Med Res 2023; 51:3000605231153764. [PMID: 36717541 PMCID: PMC9893104 DOI: 10.1177/03000605231153764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
By the end of 2022, there had been a reduction in new cases and deaths caused by coronavirus disease 2019 (COVID-19). At the same time, new variants of the severe acute respiratory syndrome coronavirus 2 virus were being discovered. Critically ill patients with COVID-19 have been found to have high serum levels of proinflammatory cytokines, especially interleukin (IL)-6. COVID-19-related mortality has been attributed in most cases to the cytokine storm caused by increased levels of inflammatory cytokines. Dexamethasone in low doses and immunomodulators such as IL-6 inhibitors are recommended to overcome the cytokine storm. This current narrative review highlights the place of other therapeutic choices such as proteasome inhibitors, protease inhibitors and nuclear factor kappa B inhibitors in the treatment of patients with COVID-19.
Collapse
Affiliation(s)
- Romany H. Thabet
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt,Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan,Romany H. Thabet, Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Shafiq Irshidat Street, Irbid 21163, Jordan.
| | - Noor A. Massadeh
- Internship, Princess Basma Hospital, Ministry of Health, Irbid, Jordan
| | - Omar B. Badarna
- Internship, Princess Basma Hospital, Ministry of Health, Irbid, Jordan
| | - Omar M. Al-Momani
- Internship, Princess Basma Hospital, Ministry of Health, Irbid, Jordan
| |
Collapse
|
21
|
Farhan FS, Nori W, Al Kadir ITA, Hameed BH. Can Fetal Heart Lie? Intrapartum CTG Changes in COVID-19 Mothers. J Obstet Gynaecol India 2022; 72:479-484. [PMID: 35634476 PMCID: PMC9128777 DOI: 10.1007/s13224-022-01663-6] [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: 10/03/2021] [Accepted: 05/01/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND COVID-19 infection has raised multiple concerns in pregnant mothers; many questioned the risk of vertical transmission and the implication on the feto-maternal outcome. Cardiotocogrm (CTG) is the principal method to observe intrapartum fetal well-being. This paper aims to verify intrapartum CTG changes seen in seropositive COVID-19 mothers versus healthy controls and looks into their relation to subsequent delivery mode and neonatal outcome. METHODS A case-control study recruited 90 pregnant women at the labor word of AL Yarmouk Teaching Hospital. All were term pregnancy admitted for delivery. They were grouped into 2: seropositive COVID-19 confirmed by real-time RT-PCR test (30/90) and healthy controls (60/90). We recorded their demographic criteria, laboratory results, CTG changes, delivery mode, and indication. RESULTS COVID-19 cases showed significantly higher pulse rate, temperature, and leukocyte counts. Cesarian deliveries (CS) were higher in cases versus healthy controls (70 % vs. 53.3 %) and P = 0.45. Analysis of the CS indications showed that abnormal fetal heart tracing accounts for 33.3 % versus 15.6 % (P-value = 0.015) for cases versus healthy controls. 60 % of COVID-19 cases exhibited abnormal CTG changes versus 19.4 % in healthy controls. These changes were primarily fetal tachycardia and reduced variabilities. CONCLUSIONS The higher incidence of abnormal CTG in COVID-19 cases, alongside infection signs and symptoms, underlies the exaggerated inflammatory reactions inside the pregnant mother. These inflammatory reactions are the main causes of CTG changes and higher CS rates. Therefore, obstetricians are advised to optimize the maternal condition to rectify reactive CTG changes rather than proceeding into urgent CS. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13224-022-01663-6.
Collapse
Affiliation(s)
- Fatin Shallal Farhan
- Department of Gynaecology And Obstetrics, Mustansiriyah University \ College of Medicine, Baghdad, Iraq
| | - Wassan Nori
- Department of Gynaecology And Obstetrics, Mustansiriyah University \ College of Medicine, Baghdad, Iraq
| | | | - Ban Hadi Hameed
- Department of Gynaecology And Obstetrics, Mustansiriyah University \ College of Medicine, Baghdad, Iraq
| |
Collapse
|
22
|
Progress on COVID-19 Chemotherapeutics Discovery and Novel Technology. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238257. [PMID: 36500347 PMCID: PMC9736643 DOI: 10.3390/molecules27238257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/29/2022]
Abstract
COVID-19 is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel highly contagious and pathogenic coronavirus that emerged in late 2019. SARS-CoV-2 spreads primarily through virus-containing droplets and small particles of air pollution, which greatly increases the risk of inhaling these virus particles when people are in close proximity. COVID-19 is spreading across the world, and the COVID-19 pandemic poses a threat to human health and public safety. To date, there are no specific vaccines or effective drugs against SARS-CoV-2. In this review, we focus on the enzyme targets of the virus and host that may be critical for the discovery of chemical compounds and natural products as antiviral drugs, and describe the development of potential antiviral drugs in the preclinical and clinical stages. At the same time, we summarize novel emerging technologies applied to the research on new drug development and the pathological mechanisms of COVID-19.
Collapse
|
23
|
Jiménez-Rodríguez BM, Triviño-Ibáñez EM, Gutiérrez-Fernández J, Romero-Ortiz AD, Ramos-Urbina EM, Morales-García C. Abnormal Alpha-1 Antitrypsin Levels and Other Risk Factors Associated with Lung Function Impairment at 6 and 12 Months after Hospitalization Due to COVID-19: A Cohort Study. Healthcare (Basel) 2022; 10:healthcare10122341. [PMID: 36553867 PMCID: PMC9777719 DOI: 10.3390/healthcare10122341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Respiratory function deficits are common sequelae for COVID-19. In this study, we aimed to identify the medical conditions that may influence lung function impairment at 12 months after SARS-CoV2 infection and to analyze the role of alpha-1 antytripsin (AAT) deficiciency (AATD). A cohort study was conducted on hospitalized COVID-19 pneumonia patients in Granada (Spain) during the first infection wave who were referred to a post-COVID-19 hospital clinic. The patients were monitored with three follow-up visits from May 2020 to May 2021. Previous medical history, hospital admission data, baseline parameters and physical examination data were collected at the first visit. Pulmonary function tests were performed at 6 and 12 months together with the determination of AAT level and AATD genotype. After 12 months, 49 out of 157 patients (31.2%) continued to have lung function impairment. A multivariate analysis showed a statistically significant association of lung function impairment with: higher Charlson index; pneumonia with a central and/or mixed distribution; anemia on admission; time in intensive care; need for corticosteroid boluses; abnormal respiratory sounds at 6 months; elevated lactate dehydrogenase at 12 months; abnormal AAT; and MZ genotype. Our results suggest that these medical conditions predispose COVID-19 patients to develop long-term lung function sequelae.
Collapse
Affiliation(s)
- Beatriz María Jiménez-Rodríguez
- Department of Pneumology, University Hospital Virgen de las Nieves, 18014 Granada, Spain
- Department of Microbiology, School of Medicine and PhD Program in Clinical Medicine and Public Health, University of Granada-IBS, 18010 Granada, Spain
- Correspondence: ; Tel.: +34-958020246
| | - Eva Maria Triviño-Ibáñez
- Biomedical Research Institute of Granada-IBS, 18012 Granada, Spain
- Department of Nuclear Medicine, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| | - José Gutiérrez-Fernández
- Department of Microbiology, School of Medicine and PhD Program in Clinical Medicine and Public Health, University of Granada-IBS, 18010 Granada, Spain
- Biomedical Research Institute of Granada-IBS, 18012 Granada, Spain
- Department of Microbiology, University Hospital Virgen de las Nieves, 18014 Granada, Spain
| | - Ana Dolores Romero-Ortiz
- Department of Pneumology, University Hospital Virgen de las Nieves, 18014 Granada, Spain
- Biomedical Research Institute of Granada-IBS, 18012 Granada, Spain
| | | | - Concepción Morales-García
- Department of Pneumology, University Hospital Virgen de las Nieves, 18014 Granada, Spain
- Biomedical Research Institute of Granada-IBS, 18012 Granada, Spain
| |
Collapse
|
24
|
Akbasheva OE, Spirina LV, Dyakov DA, Masunova NV. Proteolysis and Deficiency of α1-Proteinase Inhibitor in SARS-CoV-2 Infection. BIOCHEMISTRY (MOSCOW) SUPPLEMENT. SERIES B, BIOMEDICAL CHEMISTRY 2022; 16:271-291. [PMID: 36407837 PMCID: PMC9668222 DOI: 10.1134/s1990750822040035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022]
Abstract
The SARS-CoV-2 pandemic had stimulated the emergence of numerous publications on the α1-proteinase inhibitor (α1-PI, α1-antitrypsin), especially when it was found that the regions of high mortality corresponded to the regions with deficient α1-PI alleles. By analogy with the data obtained in the last century, when the first cause of the genetic deficiency of α1-antitrypsin leading to elastase activation in pulmonary emphysema was proven, it can be supposed that proteolysis hyperactivation in COVID-19 may be associated with the impaired functions of α1-PI. The purpose of this review was to systematize the scientific data and critical directions for translational studies on the role of α1-PI in SARS-CoV-2-induced proteolysis hyperactivation as a diagnostic marker and a therapeutic target. This review describes the proteinase-dependent stages of viral infection: the reception and penetration of the virus into a cell and the imbalance of the plasma aldosterone-angiotensin-renin, kinin, and blood clotting systems. The role of ACE2, TMPRSS, ADAM17, furin, cathepsins, trypsin- and elastase-like serine proteinases in the virus tropism, the activation of proteolytic cascades in blood, and the COVID-19-dependent complications is considered. The scientific reports on α1-PI involvement in the SARS-CoV-2-induced inflammation, the relationship with the severity of infection and comorbidities were analyzed. Particular attention is paid to the acquired α1-PI deficiency in assessing the state of patients with proteolysis overactivation and chronic non-inflammatory diseases, which are accompanied by the risk factors for comorbidity progression and the long-term consequences of COVID-19. Essential data on the search and application of protease inhibitor drugs in the therapy for bronchopulmonary and cardiovascular pathologies were analyzed. The evidence of antiviral, anti-inflammatory, anticoagulant, and anti-apoptotic effects of α1-PI, as well as the prominent data and prospects for its application as a targeted drug in the SARS-CoV-2 acquired pneumonia and related disorders, are presented.
Collapse
Affiliation(s)
| | - L. V. Spirina
- Siberian State Medical University, 634050 Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, 634009 Tomsk, Russia
| | - D. A. Dyakov
- Siberian State Medical University, 634050 Tomsk, Russia
| | | |
Collapse
|
25
|
Bruchelt G, Treuner J, Schmidt K. Proposal for the use of an inhalation drug containing 2-5 oligoadenylates for treatment of COVID-19. Med Hypotheses 2022; 168:110969. [PMID: 36317071 PMCID: PMC9605919 DOI: 10.1016/j.mehy.2022.110969] [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: 02/27/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Interferons (IFN), first described 1957 by Isaacs and Lindemann, are antiviral proteins generated in cells after viral infections. One of several interferon-induced effector mechanisms is the so called 2-5A / RNaseL system: Interferon is produced in the virus-affected cells and released. After binding to cell membrane receptors of adjacent cells, 2-5 A synthetase (oligoadenylate synthetase, OAS) is generated, attaches to dsRNA section areas of the viral RNA and catalyses the production of 2-5 oligoadenylates from ATP. In 2-5 oligoadenylates, several adenosine residues (3-4 and more) are combined via phosphodiester binding in the unusual 2'-5' positions of the riboses. 2-5 oligoadenylates activate a RNaseL which degrades the viral RNA. Recently, characteristic gene mutations and other disturbances concerning the interferon system were detected in patients with severe COVID-19, leading to problems of 2-5 oligoadenylate synthesis and the activation of RNAseL. In order to circumvent these problems, we hypothesize that a direct application of 2-5 oligoadenylates, included in an inhalation spray, may be effective in treatment of severe COVID-19 infections of the respiratory system. In contrast to some other anti-COVID-19 drugs, oligoadenylates act inside the cells (like e.g. Paxlovid) and are therefore independent of cell surface mutations of the virus. For confirmation of our hypothesis, proof of concept investigations in vitro are suggested, before a possible clinical application can be considered.
Collapse
|
26
|
Parr DG, Chorostowska-Wynimko J, Corsico A, Esquinas C, McElvaney GN, Sark AD, Sucena M, Tanash H, Turner AM, Miravitlles M. IMpact of COVID-19 in Patients With Severe Alpha-1 Antitrypsin Deficiency: The IMCA1 Study of the EARCO Clinical Research Collaboration. Arch Bronconeumol 2022; 58:840-842. [PMID: 35879122 PMCID: PMC9272568 DOI: 10.1016/j.arbres.2022.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022]
Affiliation(s)
- David G Parr
- Department of Respiratory Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK.
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Disease, Warsaw, Poland
| | - Angelo Corsico
- Respiratory Diseases Division, IRCCS Policlinico San Matteo Foundation and University of Pavia, Italy
| | - Cristina Esquinas
- Pneumology Department, Hospital Universitari Vall d'Hebron/Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Gerard N McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Annelot D Sark
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Sucena
- Pulmonology Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Hanan Tanash
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Sweden
| | - Alice M Turner
- Institute for Applied Health Research, University of Birmingham, Birmingham, UK
| | - Marc Miravitlles
- Pneumology Department, Hospital Universitari Vall d'Hebron/Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| |
Collapse
|
27
|
Nygren D, Mölstad U, Thulesius H, Hillman M, Broman LM, Tanash H, Landin-Olsson M, Rasmussen M, Thunander M. Low Prevalence of Mild Alpha-1-Antitrypsin Deficiency in Hospitalized COVID-19-Patients. Int J Gen Med 2022; 15:5843-5848. [PMID: 35789772 PMCID: PMC9250346 DOI: 10.2147/ijgm.s370434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/31/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction Alpha-1-antitrypsin (AAT) has been shown to inhibit SARS-CoV-2 cell entry and suggested as a therapeutic agent for COVID-19. Furthermore, epidemiological association of high prevalence of Alpha-1-antitrypsin deficiency (AATD) and regional severity of COVID-19-impact has been hypothesized. In our study setting, the estimated prevalence rates of mild (PI*MZ, PI*SS or PI*MS) and moderate-to-severe AATD (PI*ZZ or PI*SZ) are high, 9% and 0.2%, respectively. Our primary aim was to examine the prevalence rate of AATD among hospitalized COVID-19-patients. Methods In this prospective observational study, enrollment occurred from December 2020 to January 2021 in two COVID-19-units at Skåne University Hospital, Lund, Sweden. Case definition was a patient hospitalized due to COVID-19. Patients were screened for AATD with PI-typing and if results were inconclusive, PCR for the S- and Z-genes were performed. Patients were categorized as severe or moderate COVID-19 and 30-day-mortality data were collected. The primary outcome was prevalence rate of AATD. The secondary outcome investigated association between presence of mild AATD and severe COVID-19. Results We enrolled 61 patients with COVID-19. Two patients out of 61 (3%) had mild AATD (PI*MZ) and none had moderate-to-severe AATD. 30/61 (49%) had severe COVID-19. Both patients with mild AATD developed severe COVID-19. Yet, presence of AATD was not significantly associated with severe COVID-19 (p=0.24). Conclusion Mild AATD (PI*MS or PI*MZ) was rare in a small cohort of hospitalized patients with COVID-19 in a study setting with a high background prevalence of AATD.
Collapse
Affiliation(s)
- David Nygren
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden,Correspondence: David Nygren, Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden, Tel +4646171192, Fax +4646176002, Email
| | - Ulrica Mölstad
- Department of Research and Development, Health Care Region Kronoberg, Växjö, Sweden
| | - Hans Thulesius
- Department of Research and Development, Health Care Region Kronoberg, Växjö, Sweden,Department of Medicine and Optometry, Linnaeus University, Växjö, Sweden,Department of Clinical Sciences, Family Medicine, Lund University, Malmö, Sweden
| | - Magnus Hillman
- Diabetes Research Laboratory, Biomedical Center, Lund University, Lund, Sweden,Department of Clinical Sciences, Endocrinology and Diabetes, Lund University, Lund, Sweden
| | - Lars Mikael Broman
- ECMO Centre Karolinska, Pediatric Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hanan Tanash
- Department of Clinical Sciences, Respiratory Medicine, Lund University, Malmö, Sweden,Department of Respiratory Medicine, Skåne University Hospital, Malmö, Sweden
| | - Mona Landin-Olsson
- Diabetes Research Laboratory, Biomedical Center, Lund University, Lund, Sweden,Department of Clinical Sciences, Endocrinology and Diabetes, Lund University, Lund, Sweden
| | - Magnus Rasmussen
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Thunander
- Department of Research and Development, Health Care Region Kronoberg, Växjö, Sweden,Department of Clinical Sciences, Endocrinology and Diabetes, Lund University, Lund, Sweden,Department of Internal Medicine, Endocrinology, Växjö Central Hospital, Växjö, Sweden
| |
Collapse
|
28
|
Akbasheva OE, Spirina LV, Dyakov DA, Masunova NV. [Proteolysis and deficiency of α1-proteinase inhibitor in SARS-CoV-2 infection]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:157-176. [PMID: 35717581 DOI: 10.18097/pbmc20226803157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The SARS-CoV-2 pandemia had stimulated the numerous publications emergence on the α1-proteinase inhibitor (α1-PI, α1-antitrypsin), primarily when it was found that high mortality in some regions corresponded to the regions with deficient α1-PI alleles. By analogy with the last century's data, when the root cause of the α1-antitrypsin, genetic deficiency leading to the elastase activation in pulmonary emphysema, was proven. It is evident that proteolysis hyperactivation in COVID-19 may be associated with α1-PI impaired functions. The purpose of this review is to systematize scientific data, critical directions for translational studies on the role of α1-PI in SARS-CoV-2-induced proteolysis hyperactivation as a diagnostic marker and a target in therapy. This review describes the proteinase-dependent stages of a viral infection: the reception and virus penetration into the cell, the plasma aldosterone-angiotensin-renin, kinins, blood clotting systems imbalance. The ACE2, TMPRSS, ADAM17, furin, cathepsins, trypsin- and elastase-like serine proteinases role in the virus tropism, proteolytic cascades activation in blood, and the COVID-19-dependent complications is presented. The analysis of scientific reports on the α1-PI implementation in the SARS-CoV-2-induced inflammation, the links with the infection severity, and comorbidities were carried out. Particular attention is paid to the acquired α1-PI deficiency in assessing the patients with the proteolysis overactivation and chronic non-inflammatory diseases that are accompanied by the risk factors for the comorbidities progression, and the long-term consequences of COVID-19 initiation. Analyzed data on the search and proteases inhibitory drugs usage in the bronchopulmonary cardiovascular pathologies therapy are essential. It becomes evident the antiviral, anti-inflammatory, anticoagulant, anti-apoptotic effect of α1-PI. The prominent data and prospects for its application as a targeted drug in the SARS-CoV-2 acquired pneumonia and related disorders are presented.
Collapse
Affiliation(s)
| | - L V Spirina
- Siberian State Medical University, Tomsk, Russia; Cancer Research Institute, Tomsk National Research Medical Center, Tomsk, Russia
| | - D A Dyakov
- Siberian State Medical University, Tomsk, Russia
| | - N V Masunova
- Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
29
|
Bai X, Buckle AM, Vladar EK, Janoff EN, Khare R, Ordway D, Beckham D, Fornis LB, Majluf-Cruz A, Fugit RV, Freed BM, Kim S, Sandhaus RA, Chan ED. Enoxaparin augments alpha-1-antitrypsin inhibition of TMPRSS2, a promising drug combination against COVID-19. Sci Rep 2022; 12:5207. [PMID: 35338216 PMCID: PMC8953970 DOI: 10.1038/s41598-022-09133-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
The cell surface serine protease Transmembrane Protease 2 (TMPRSS2) is required to cleave the spike protein of SARS-CoV-2 for viral entry into cells. We determined whether negatively-charged heparin enhanced TMPRSS2 inhibition by alpha-1-antitrypsin (AAT). TMPRSS2 activity was determined in HEK293T cells overexpressing TMPRSS2. We quantified infection of primary human airway epithelial cells (hAEc) with human coronavirus 229E (HCoV-229E) by immunostaining for the nucleocapsid protein and by the plaque assay. Detailed molecular modeling was undertaken with the heparin-TMPRSS2-AAT ternary complex. Enoxaparin enhanced AAT inhibition of both TMPRSS2 activity and infection of hAEc with HCoV-229E. Underlying these findings, detailed molecular modeling revealed that: (i) the reactive center loop of AAT adopts an inhibitory-competent conformation compared with the crystal structure of TMPRSS2 bound to an exogenous (nafamostat) or endogenous (HAI-2) TMPRSS2 inhibitor and (ii) negatively-charged heparin bridges adjacent electropositive patches at the TMPRSS2-AAT interface, neutralizing otherwise repulsive forces. In conclusion, enoxaparin enhances AAT inhibition of both TMPRSS2 and coronavirus infection. Such host-directed therapy is less likely to be affected by SARS-CoV-2 mutations. Furthermore, given the known anti-inflammatory activities of both AAT and heparin, this form of treatment may target both the virus and the excessive inflammatory consequences of severe COVID-19.
Collapse
Affiliation(s)
- Xiyuan Bai
- grid.422100.50000 0000 9751 469XDepartment of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO USA ,grid.240341.00000 0004 0396 0728Department of Academic Affairs and Medicine, National Jewish Health, Denver, CO USA ,grid.430503.10000 0001 0703 675XDivision of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.240341.00000 0004 0396 0728National Jewish Health, D509, Neustadt Building, 1400 Jackson Street, Denver, CO 80206 USA
| | - Ashley M. Buckle
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC Australia
| | - Eszter K. Vladar
- grid.430503.10000 0001 0703 675XDivision of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Edward N. Janoff
- grid.422100.50000 0000 9751 469XDepartment of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO USA ,grid.430503.10000 0001 0703 675XDivision of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Reeti Khare
- grid.240341.00000 0004 0396 0728Mycobacteriology Laboratory, Advance Diagnostics, National Jewish Health, Denver, CO USA
| | - Diane Ordway
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunlogy, and Pathology, Colorado State University, Fort Collins, CO USA
| | - David Beckham
- grid.430503.10000 0001 0703 675XDivision of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Lorelenn B. Fornis
- grid.240341.00000 0004 0396 0728Department of Academic Affairs and Medicine, National Jewish Health, Denver, CO USA
| | - Abraham Majluf-Cruz
- grid.419157.f0000 0001 1091 9430Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Randolph V. Fugit
- grid.422100.50000 0000 9751 469XDepartment of Pharmacy, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO USA
| | - Brian M. Freed
- grid.430503.10000 0001 0703 675XDepartment of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Soohyun Kim
- grid.258676.80000 0004 0532 8339Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea ,grid.258676.80000 0004 0532 8339College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Robert A. Sandhaus
- grid.240341.00000 0004 0396 0728Department of Academic Affairs and Medicine, National Jewish Health, Denver, CO USA
| | - Edward D. Chan
- grid.422100.50000 0000 9751 469XDepartment of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO USA ,grid.240341.00000 0004 0396 0728Department of Academic Affairs and Medicine, National Jewish Health, Denver, CO USA ,grid.430503.10000 0001 0703 675XDivision of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.240341.00000 0004 0396 0728National Jewish Health, D509, Neustadt Building, 1400 Jackson Street, Denver, CO 80206 USA
| |
Collapse
|
30
|
Boraldi F, Lofaro FD, Cossarizza A, Quaglino D. The "Elastic Perspective" of SARS-CoV-2 Infection and the Role of Intrinsic and Extrinsic Factors. Int J Mol Sci 2022; 23:ijms23031559. [PMID: 35163482 PMCID: PMC8835950 DOI: 10.3390/ijms23031559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
Elastin represents the structural component of the extracellular matrix providing elastic recoil to tissues such as skin, blood vessels and lungs. Elastogenic cells secrete soluble tropoelastin monomers into the extracellular space where these monomers associate with other matrix proteins (e.g., microfibrils and glycoproteins) and are crosslinked by lysyl oxidase to form insoluble fibres. Once elastic fibres are formed, they are very stable, highly resistant to degradation and have an almost negligible turnover. However, there are circumstances, mainly related to inflammatory conditions, where increased proteolytic degradation of elastic fibres may lead to consequences of major clinical relevance. In severely affected COVID-19 patients, for instance, the massive recruitment and activation of neutrophils is responsible for the profuse release of elastases and other proteolytic enzymes which cause the irreversible degradation of elastic fibres. Within the lungs, destruction of the elastic network may lead to the permanent impairment of pulmonary function, thus suggesting that elastases can be a promising target to preserve the elastic component in COVID-19 patients. Moreover, intrinsic and extrinsic factors additionally contributing to damaging the elastic component and to increasing the spread and severity of SARS-CoV-2 infection are reviewed.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
| | - Francesco Demetrio Lofaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
- Correspondence:
| |
Collapse
|
31
|
Wettstein L, Kirchhoff F, Münch J. The Transmembrane Protease TMPRSS2 as a Therapeutic Target for COVID-19 Treatment. Int J Mol Sci 2022; 23:1351. [PMID: 35163273 PMCID: PMC8836196 DOI: 10.3390/ijms23031351] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 01/25/2023] Open
Abstract
TMPRSS2 is a type II transmembrane protease with broad expression in epithelial cells of the respiratory and gastrointestinal tract, the prostate, and other organs. Although the physiological role of TMPRSS2 remains largely elusive, several endogenous substrates have been identified. TMPRSS2 serves as a major cofactor in SARS-CoV-2 entry, and primes glycoproteins of other respiratory viruses as well. Consequently, inhibiting TMPRSS2 activity is a promising strategy to block viral infection. In this review, we provide an overview of the role of TMPRSS2 in the entry processes of different respiratory viruses. We then review the different classes of TMPRSS2 inhibitors and their clinical development, with a focus on COVID-19 treatment.
Collapse
Affiliation(s)
| | | | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (L.W.); (F.K.)
| |
Collapse
|
32
|
Cambier S, Metzemaekers M, Carvalho AC, Nooyens A, Jacobs C, Vanderbeke L, Malengier-Devlies B, Gouwy M, Heylen E, Meersseman P, Hermans G, Wauters E, Wilmer A, Consortium C, Schols D, Matthys P, Opdenakker G, Marques RE, Wauters J, Vandooren J, Proost P. Atypical response to bacterial co-infection and persistent neutrophilic broncho-alveolar inflammation distinguish critical COVID-19 from influenza. JCI Insight 2021; 7:155055. [PMID: 34793331 PMCID: PMC8765057 DOI: 10.1172/jci.insight.155055] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Neutrophils are recognized as important circulating effector cells in the pathophysiology of severe coronavirus disease 2019 (COVID-19). However, their role within the inflamed lungs is incompletely understood. Here, we collected broncho-alveolar lavage (BAL) fluids and parallel blood samples of critically ill COVID-19 patients requiring invasive mechanical ventilation and compared BAL fluid parameters with those of mechanically ventilated influenza patients, as a non-COVID-19 viral pneumonia cohort. Compared to influenza, BAL fluids of COVID-19 patients contained increased numbers of hyperactivated degranulating neutrophils and elevated concentrations of the cytokines IL-1β, IL-1RA, IL-17A, TNF-α and G-CSF, the chemokines CCL7, CXCL1, CXCL8, CXCL11 and CXCL12α, and the protease inhibitors elafin, secretory leukocyte protease inhibitor (SLPI) and tissue inhibitor of metalloproteinases 1 (TIMP-1). In contrast, α-1 antitrypsin levels and net proteolytic activity were comparable in COVID-19 and influenza BAL fluids. During antibiotics treatment for bacterial co-infections, increased BAL fluid levels of several activating and chemotactic factors for monocytes, lymphocytes and NK cells were detected in COVID-19 patients whereas concentrations tended to decrease in influenza patients, highlighting the persistent immunological response to co-infections in COVID-19. Finally, the high proteolytic activity in COVID-19 lungs suggests considering protease inhibitors as a treatment option.
Collapse
Affiliation(s)
- Seppe Cambier
- Laboratory of Molecular Immunology, KU Leuven, Leuven, Belgium
| | | | | | - Amber Nooyens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Cato Jacobs
- Laboratory for Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium
| | - Lore Vanderbeke
- Laboratory of Clinical Bacteriology and Mycology, KU Leuven, Leuven, Belgium
| | | | - Mieke Gouwy
- Laboratory of Molecular Immunology, KU Leuven, Leuven, Belgium
| | - Elisabeth Heylen
- Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Philippe Meersseman
- Laboratory for Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium
| | - Greet Hermans
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Els Wauters
- Laboratory of Respiratory Diseases and Thoracic Surgery, KU Leuven, Leuven, Belgium
| | - Alexander Wilmer
- Laboratory for Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium
| | - Contagious Consortium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | | | | | | | - Joost Wauters
- Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | | | - Paul Proost
- Laboratory of Molecular Immunology, KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Herth FJF, Sandhaus RA, Turner AM, Sucena M, Welte T, Greulich T. Alpha 1 Antitrypsin Therapy in Patients with Alpha 1 Antitrypsin Deficiency: Perspectives from a Registry Study and Practical Considerations for Self-Administration During the COVID-19 Pandemic. Int J Chron Obstruct Pulmon Dis 2021; 16:2983-2996. [PMID: 34754184 PMCID: PMC8570922 DOI: 10.2147/copd.s325211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/27/2021] [Indexed: 01/06/2023] Open
Abstract
Alpha 1 Antitrypsin deficiency (AATD) is a hereditary condition characterized by low serum Alpha 1 Antitrypsin (AAT) levels and a predisposition towards early-onset emphysema. Infusion of AAT is the only disease-modifying therapy that can sufficiently raise plasma AAT levels above the putative protective threshold and reduce the decline in lung density loss. Several randomized controlled trials (RCTs) and registry studies support the clinical efficacy of AAT therapy in slowing the progression of AATD-related emphysema and improving survival outcomes. The COVID-19 pandemic has prompted physicians to develop additional strategies for delivering AAT therapy, which are not only more convenient for the patient, but are “COVID-19 friendly”, thereby reducing the risk of exposing these vulnerable patients. Intravenous (IV) self-administration of AAT therapy is likely to be beneficial in certain subgroups of patients with AATD and can remove the need for weekly hospital visits, thereby improving independence and well-being. Increasing the awareness of self-administration in AATD through the development of formal guidelines and training programs is required among both physicians and patients and will play an essential role, especially post-COVID-19, in encouraging physicians to consider self-administration for AATD in suitable patients. This review summarizes the benefits of AAT therapy on the clinical endpoints of mortality and quality of life (QoL) and discusses the benefits of self-administration therapy compared with conventional therapy administered by a healthcare professional. In addition, this review highlights the challenges of providing AAT therapy during the COVID-19 pandemic and the potential considerations for its implementation thereafter.
Collapse
Affiliation(s)
- Felix J F Herth
- Department of Pneumology and Critical Care Medicine, University of Heidelberg, Heidelberg, Germany
| | - Robert A Sandhaus
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Alice M Turner
- Institute of Applied Health Research, University of Birmingham, Birmingham, England
| | - Maria Sucena
- Pulmonology Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Tobias Welte
- Department of Pulmonary and Infectious Diseases, Hannover Medical School, Hannover, Germany
| | - Timm Greulich
- Department of Internal Medicine and Pneumology, University Hospital Marburg, Marburg, Germany
| |
Collapse
|
34
|
Patients with COVID-19: in the dark-NETs of neutrophils. Cell Death Differ 2021; 28:3125-3139. [PMID: 34031543 PMCID: PMC8142290 DOI: 10.1038/s41418-021-00805-z] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 infection poses a major threat to the lungs and multiple other organs, occasionally causing death. Until effective vaccines are developed to curb the pandemic, it is paramount to define the mechanisms and develop protective therapies to prevent organ dysfunction in patients with COVID-19. Individuals that develop severe manifestations have signs of dysregulated innate and adaptive immune responses. Emerging evidence implicates neutrophils and the disbalance between neutrophil extracellular trap (NET) formation and degradation plays a central role in the pathophysiology of inflammation, coagulopathy, organ damage, and immunothrombosis that characterize severe cases of COVID-19. Here, we discuss the evidence supporting a role for NETs in COVID-19 manifestations and present putative mechanisms, by which NETs promote tissue injury and immunothrombosis. We present therapeutic strategies, which have been successful in the treatment of immunο-inflammatory disorders and which target dysregulated NET formation or degradation, as potential approaches that may benefit patients with severe COVID-19.
Collapse
|
35
|
Liu LP, Gholam MF, Elshikha AS, Kawakibi T, Elmoujahid N, Moussa HH, Song S, Alli AA. Transgenic Mice Overexpressing Human Alpha-1 Antitrypsin Exhibit Low Blood Pressure and Altered Epithelial Transport Mechanisms in the Inactive and Active Cycles. Front Physiol 2021; 12:710313. [PMID: 34630137 PMCID: PMC8493122 DOI: 10.3389/fphys.2021.710313] [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: 05/15/2021] [Accepted: 08/20/2021] [Indexed: 11/13/2022] Open
Abstract
Human alpha-1 antitrypsin (hAAT) is a versatile protease inhibitor, but little is known about its targets in the aldosterone-sensitive distal nephron and its role in electrolyte balance and blood pressure control. We analyzed urinary electrolytes, osmolality, and blood pressure from hAAT transgenic (hAAT-Tg) mice and C57B/6 wild-type control mice maintained on either a normal salt or high salt diet. Urinary sodium, potassium, and chloride concentrations as well as urinary osmolality were lower in hAAT-Tg mice maintained on a high salt diet during both the active and inactive cycles. hAAT-Tg mice showed a lower systolic blood pressure compared to C57B6 mice when maintained on a normal salt diet but this was not observed when they were maintained on a high salt diet. Cathepsin B protein activity was less in hAAT-Tg mice compared to wild-type controls. Protein expression of the alpha subunit of the sodium epithelial channel (ENaC) alpha was also reduced in the hAAT-Tg mice. Natriuretic peptide receptor C (NPRC) protein expression in membrane fractions of the kidney cortex was reduced while circulating levels of atrial natriuretic peptide (ANP) were greater in hAAT-Tg mice compared to wild-type controls. This study characterizes the electrolyte and blood pressure phenotype of hAAT-Tg mice during the inactive and active cycles and investigates the mechanism by which ENaC activation is inhibited in part by a mechanism involving decreased cathepsin B activity and increased ANP levels in the systemic circulation.
Collapse
Affiliation(s)
- Lauren P Liu
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mohammed F Gholam
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ahmed Samir Elshikha
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Tamim Kawakibi
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Nasseem Elmoujahid
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Hassan H Moussa
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Sihong Song
- Department of Pharmaceutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States.,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
36
|
Vianello A, Guarnieri G, Braccioni F, Molena B, Lococo S, Achille A, Lionello F, Salviati L, Caminati M, Senna G. Correlation between α1-Antitrypsin Deficiency and SARS-CoV-2 Infection: Epidemiological Data and Pathogenetic Hypotheses. J Clin Med 2021; 10:4493. [PMID: 34640510 PMCID: PMC8509830 DOI: 10.3390/jcm10194493] [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: 09/02/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022] Open
Abstract
The most common hereditary disorder in adults, α1-antitrypsin deficiency (AATD), is characterized by reduced plasma levels or the abnormal functioning of α1-antitrypsin (AAT), a major human blood serine protease inhibitor, which is encoded by the SERine Protein INhibitor-A1 (SERPINA1) gene and produced in the liver. Recently, it has been hypothesized that the geographic differences in COVID-19 infection and fatality rates may be partially explained by ethnic differences in SERPINA1 allele frequencies. In our review, we examined epidemiological data on the correlation between the distribution of AATD, SARS-CoV-2 infection, and COVID-19 mortality rates. Moreover, we described shared pathogenetic pathways that may provide a theoretical basis for our epidemiological findings. We also considered the potential use of AAT augmentation therapy in patients with COVID-19.
Collapse
Affiliation(s)
- Andrea Vianello
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy; (G.G.); (F.B.); (B.M.); (S.L.); (A.A.); (F.L.)
| | - Gabriella Guarnieri
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy; (G.G.); (F.B.); (B.M.); (S.L.); (A.A.); (F.L.)
| | - Fausto Braccioni
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy; (G.G.); (F.B.); (B.M.); (S.L.); (A.A.); (F.L.)
| | - Beatrice Molena
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy; (G.G.); (F.B.); (B.M.); (S.L.); (A.A.); (F.L.)
| | - Sara Lococo
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy; (G.G.); (F.B.); (B.M.); (S.L.); (A.A.); (F.L.)
| | - Alessia Achille
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy; (G.G.); (F.B.); (B.M.); (S.L.); (A.A.); (F.L.)
| | - Federico Lionello
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35122 Padova, Italy; (G.G.); (F.B.); (B.M.); (S.L.); (A.A.); (F.L.)
| | - Leonardo Salviati
- Department of Pediatrics, University of Padova, 35122 Padova, Italy;
| | - Marco Caminati
- Asthma Center and Allergy Unit, University of Verona, 37129 Verona, Italy; (M.C.); (G.S.)
| | - Gianenrico Senna
- Asthma Center and Allergy Unit, University of Verona, 37129 Verona, Italy; (M.C.); (G.S.)
| |
Collapse
|
37
|
Marzouk S, Attia N, Mashal M. Insights into the potential role of alpha1-antitrypsin in COVID-19 patients: Mechanisms, current update, and future perspectives. THE CLINICAL RESPIRATORY JOURNAL 2021; 15:1019-1024. [PMID: 34087062 PMCID: PMC8239917 DOI: 10.1111/crj.13406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 02/02/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
In this work, we provide an up-to-date summary of the available molecular- and cell-related mechanisms by which alpha1-antitrypsin (AAT) protein could be of benefit in treating COVID-19 patients. As well, we demonstrate the current status in terms of the ongoing clinical trials using AAT in COVID-19 patients. Finally, we touch on the potential role gene therapy and stem cell-based gene therapy could have in such emerging and serious condition caused by the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Saber Marzouk
- Department of PharmacyMinistry of HealthMallawi, El MiniaEgypt
| | - Noha Attia
- The American University of Antigua‐College of MedicineUniversity ParkCoolidgeAntigua and Barbuda
- NanoBioCel GroupLaboratory of PharmaceuticsSchool of PharmacyUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
- Department of Medical Histology and Cell BiologyFaculty of MedicineUniversity of AlexandriaAlexandriaEgypt
| | - Mohamed Mashal
- The American University of Antigua‐College of MedicineUniversity ParkCoolidgeAntigua and Barbuda
- NanoBioCel GroupLaboratory of PharmaceuticsSchool of PharmacyUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
| |
Collapse
|
38
|
Rahbar Saadat Y, Hosseiniyan Khatibi SM, Zununi Vahed S, Ardalan M. Host Serine Proteases: A Potential Targeted Therapy for COVID-19 and Influenza. Front Mol Biosci 2021; 8:725528. [PMID: 34527703 PMCID: PMC8435734 DOI: 10.3389/fmolb.2021.725528] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/11/2021] [Indexed: 12/23/2022] Open
Abstract
The ongoing pandemic illustrates limited therapeutic options for controlling SARS-CoV-2 infections, calling a need for additional therapeutic targets. The viral spike S glycoprotein binds to the human receptor angiotensin-converting enzyme 2 (ACE2) and then is activated by the host proteases. Based on the accessibility of the cellular proteases needed for SARS-S activation, SARS-CoV-2 entrance and activation can be mediated by endosomal (such as cathepsin L) and non-endosomal pathways. Evidence indicates that in the non-endosomal pathway, the viral S protein is cleaved by the furin enzyme in infected host cells. To help the virus enter efficiently, the S protein is further activated by the serine protease 2 (TMPRSS2), provided that the S has been cleaved by furin previously. In this review, important roles for host proteases within host cells will be outlined in SARS-CoV-2 infection and antiviral therapeutic strategies will be highlighted. Although there are at least five highly effective vaccines at this time, the appearance of the new viral mutations demands the development of therapeutic agents. Targeted inhibition of host proteases can be used as a therapeutic approach for viral infection.
Collapse
|
39
|
Neutrophil Extracellular Traps (NETs) in Severe SARS-CoV-2 Lung Disease. Int J Mol Sci 2021; 22:ijms22168854. [PMID: 34445556 PMCID: PMC8396177 DOI: 10.3390/ijms22168854] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
Neutrophil extracellular traps (NETs), built from mitochondrial or nuclear DNA, proteinases, and histones, entrap and eliminate pathogens in the course of bacterial or viral infections. Neutrophils’ activation and the formation of NETs have been described as major risk factors for acute lung injury, multi-organ damage, and mortality in COVID-19 disease. NETs-related lung injury involves both epithelial and endothelial cells, as well as the alveolar-capillary barrier. The markers for NETs formation, such as circulating DNA, neutrophil elastase (NE) activity, or myeloperoxidase-DNA complexes, were found in lung specimens of COVID-19 victims, as well as in sera and tracheal aspirates obtained from COVID-19 patients. DNA threads form large conglomerates causing local obstruction of the small bronchi and together with NE are responsible for overproduction of mucin by epithelial cells. Various components of NETs are involved in the pathogenesis of cytokine storm in SARS-CoV-2 pulmonary disease. NETs are responsible for the interplay between inflammation and thrombosis in the affected lungs. The immunothrombosis, stimulated by NETs, has a poor prognostic significance. Better understanding of the role of NETs in the course of COVID-19 can help to develop novel approaches to the therapeutic interventions in this condition.
Collapse
|
40
|
Boosted Pro-Inflammatory Activity in Human PBMCs by Lipopolysaccharide and SARS-CoV-2 Spike Protein Is Regulated by α-1 Antitrypsin. Int J Mol Sci 2021; 22:ijms22157941. [PMID: 34360706 PMCID: PMC8347018 DOI: 10.3390/ijms22157941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
For the treatment of severe COVID-19, supplementation with human plasma-purified α-1 antitrypsin (AAT) to patients is currently considered. AAT inhibits host proteases that facilitate viral entry and possesses broad anti-inflammatory and immunomodulatory activities. Researchers have demonstrated that an interaction between SARS-CoV-2 spike protein (S) and lipopolysaccharides (LPS) enhances pro-inflammatory responses in vitro and in vivo. Hence, we wanted to understand the potential anti-inflammatory activities of plasma-derived and recombinant AAT (recAAT) in a model of human total peripheral blood mononuclear cells (PBMCs) exposed to a combination of CHO expressed trimeric spike protein and LPS, ex vivo. We confirmed that cytokine production was enhanced in PBMCs within six hours when low levels of LPS were combined with purified spike proteins (“spike”). In the presence of 0.5 mg/mL recAAT, however, LPS/spike-induced TNF-α and IL-1β mRNA expression and protein release were significantly inhibited (by about 46–50%) relative to LPS/spike alone. Although without statistical significance, recAAT also reduced production of IL-6 and IL-8. Notably, under the same experimental conditions, the plasma-derived AAT preparation Respreeza (used in native and oxidized forms) did not show significant effects. Our findings imply that an early pro-inflammatory activation of human PBMCs is better controlled by the recombinant version of AAT than the human plasma-derived AAT used here. Considering the increasing clinical interest in AAT therapy as useful to ameliorate the hyper-inflammation seen during COVID-19 infection, different AAT preparations require careful evaluation.
Collapse
|
41
|
Bianchera A, Alomari E, Bruno S. Augmentation therapy with alpha 1-antitrypsin: present and future of production, formulation, and delivery. Curr Med Chem 2021; 29:385-410. [PMID: 34036902 DOI: 10.2174/0929867328666210525161942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
Alpha 1-antitrypsin is one of the first protein therapeutics introduced on the market - more than 30 years ago - and, to date, it is indicated only for the treatment of the severe forms of a genetic condition known as alpha-1 antitrypsin deficiency. The only approved preparations are derived from plasma, posing potential problems associated with its limited supply and high processing costs. Moreover, augmentation therapy with alpha 1-antitrypsin is still limited to intravenous infusions, a cumbersome regimen for patients. Here, we review the recent literature on its possible future developments, focusing on i) the recombinant alternatives to the plasma-derived protein, ii) novel formulations, and iii) novel administration routes. Regulatory issues and the still unclear noncanonical functions of alpha 1-antitrypsin - possibly associated with the glycosylation pattern found only in the plasma-derived protein - have hindered the introduction of new products. However, potentially new therapeutic indications other than the treatment of alpha-1 antitrypsin deficiency might open the way to new sources and new formulations.
Collapse
Affiliation(s)
- Annalisa Bianchera
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Esraa Alomari
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Stefano Bruno
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| |
Collapse
|
42
|
Schneider CV, Strnad P. SARS-CoV-2 infection in alpha1-antitrypsin deficiency. Respir Med 2021; 184:106466. [PMID: 34010739 PMCID: PMC8116136 DOI: 10.1016/j.rmed.2021.106466] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/17/2022]
Abstract
Alpha1-antitrypsin deficiency arises due to mutations in alpha1-antitrypsin (AAT) gene and represents the most prominent genetic predisposition to chronic obstructive pulmonary disease and emphysema. Since AAT plays important immunomodulatory and tissue-protective roles and since it was suggested to protect from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we assessed this association in United Kingdom Biobank, a community-based cohort with >500,000 participants. The most common, mild AATD genotypes were associated neither with increased SARS-CoV-2 infection rates nor with increased SARS-CoV-2 fatalities, while the numbers of severe AATD cases were too low to allow definitive conclusions.
Collapse
Affiliation(s)
- Carolin V Schneider
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Pavel Strnad
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany; Coordinating Center for Alpha-1 Antitrypsin Deficiency-related Liver Disease of the European Reference Network (ERN) "Rare Liver" and the European Association for the Study of the Liver (EASL) Registry Group "Alpha-1 Liver", Germany.
| |
Collapse
|
43
|
Mahler M, Meroni PL, Infantino M, Buhler KA, Fritzler MJ. Circulating Calprotectin as a Biomarker of COVID-19 Severity. Expert Rev Clin Immunol 2021; 17:431-443. [PMID: 33750254 PMCID: PMC8054493 DOI: 10.1080/1744666x.2021.1905526] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although demographic and clinical parameters such as sex, age, comorbidities, genetic background and various biomarkers have been identified as risk factors, there is an unmet need to predict the risk and onset of severe inflammatory disease leading to poor clinical outcomes. In addition, very few mechanistic biomarkers are available to inform targeted treatment of severe (auto)-inflammatory conditions associated with COVID-19. Calprotectin, also known as S100A8/S100A9, MRP8/14 (Myeloid-Related Protein) or L1, is a heterodimer involved in neutrophil-related inflammatory processes. In COVID-19 patients, calprotectin levels were reported to be associated with poor clinical outcomes such as significantly reduced survival time, especially in patients with severe pulmonary disease. AREAS COVERED Pubmed was searched using the following keywords: Calprotectin + COVID19, S100A8/A9 + COVID19, S100A8 + COVID-19, S100A9 + COVID-19, MRP8/14 + COVID19; L1 + COVID-19 between May 2020 and 8 March 2021. The results summarized in this review provide supporting evidence and propose future directions that define calprotectin as an important biomarker in COVID-19. EXPERT OPINION Calprotectin represents a promising serological biomarker for the risk assessment of COVID-19 patients.
Collapse
Affiliation(s)
- Michael Mahler
- Research and Development, Inova Diagnostics, San Diego, CA, USA
| | - Pier-Luigi Meroni
- Allergy, Clinical Immunology and Rheumatology Unit, Immunorheumatology Research Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Maria Infantino
- Immunology and Allergology Laboratory Unit, Rheumatology Unit, S. Giovanni Di Dio Hospital, Florence, Italy
| | - Katherine A. Buhler
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marvin J. Fritzler
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
44
|
Alpha-1 antitrypsin inhibits TMPRSS2 protease activity and SARS-CoV-2 infection. Nat Commun 2021; 12:1726. [PMID: 33741941 PMCID: PMC7979852 DOI: 10.1038/s41467-021-21972-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 02/18/2021] [Indexed: 12/18/2022] Open
Abstract
SARS-CoV-2 is a respiratory pathogen and primarily infects the airway epithelium. As our knowledge about innate immune factors of the respiratory tract against SARS-CoV-2 is limited, we generated and screened a peptide/protein library derived from bronchoalveolar lavage for inhibitors of SARS-CoV-2 spike-driven entry. Analysis of antiviral fractions revealed the presence of α1-antitrypsin (α1AT), a highly abundant circulating serine protease inhibitor. Here, we report that α1AT inhibits SARS-CoV-2 entry at physiological concentrations and suppresses viral replication in cell lines and primary cells including human airway epithelial cultures. We further demonstrate that α1AT binds and inactivates the serine protease TMPRSS2, which enzymatically primes the SARS-CoV-2 spike protein for membrane fusion. Thus, the acute phase protein α1AT is an inhibitor of TMPRSS2 and SARS-CoV-2 entry, and may play an important role in the innate immune defense against the novel coronavirus. Our findings suggest that repurposing of α1AT-containing drugs has prospects for the therapy of COVID-19. Here, via screening of a polypeptide library from bronchoalveolar lavage, the authors identify and characterize α1-antitrypsin (α1AT) as SARS-CoV-2 inhibitor and show that α1AT binds and inactivates the serine protease TMPRSS2, which enzymatically primes the SARS-CoV-2 spike protein for membrane fusion.
Collapse
|