1
|
Li ZX, Wang YF, Chiang LT, Hsu LJ, Wang SM, Wang JR, Wu HL, Chen SH, Chang CF. Plasminogen deficiency reduces disease severity and immune responses in enterovirus A71-infected mice. Microbiol Spectr 2025:e0331124. [PMID: 40377310 DOI: 10.1128/spectrum.03311-24] [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: 01/17/2025] [Accepted: 04/04/2025] [Indexed: 05/18/2025] Open
Abstract
Enterovirus A71 (EV-A71) is a causative agent of hand, foot, and mouth diseases. EV-A71 infections may result in severe neurological complications in children. Although several receptors or attachment molecules for EV-A71 have been identified, EV-A71 can still infect host cells even after blocking these receptors with antibodies. We have previously identified plasminogen (PLG), a circulating zymogen of plasmin, as a cell membrane-associated EV-A71-interacting glycoprotein. We confirmed that anti-PLG antibodies could reduce the binding of EV-A71 to RD cells as anti-SCARB2 and anti-nucleolin. Knockdown of PLG reduced EV-A71 binding to RD cells, and preincubation of PLG with EV-A71 increased virus binding. Enzyme-linked immunosorbent assay and surface plasmon resonance assays demonstrated the direct binding of PLG to EV-A71. We further evaluated the biological characteristics of EV-A71-infected PLG knockout (heterozygous) and wild-type mice. We found that the clinical scores and mortality of WT mice were higher than those of PLG-knockout mice after EV-A71 infection. The viral loads in the spinal cord of PLG knockout mice were lower than those in WT mice 6 days post-infection. EV-A71-associated cytokines such as IL-1β, IL-6, MCP-1, IL-10, and IFN-γ were investigated. Serum IL-10 and MCP-1 expression were significantly higher in EV-71-infected WT mice than in PLG knockout mice, and MCP-1 may be one of the critical chemokines that induce intense inflammation and chemoattracts leukocytes. Our findings reveal a possible role for PLG in EV-A71 infection/pathogenesis and shed light on developing novel therapeutic approaches and drugs to prevent EV-A71 infection.IMPORTANCEUnderstanding the pathogenesis of enterovirus A71 (EV-A71) for developing novel drugs or therapeutic approaches has always been a significant issue. In this study, we demonstrated the interactions between plasminogen (PLG) and EV-A71, characterized the biological effects of EV-A71-infected PLG knockout mice, and evaluated their immune response. We found that EV-A71 caused more severe tissue damage than PLG knockout mice in skeletal muscle, spinal cord, and brain stem. Higher virus protein was observed in these tissues of WT mice. The reduced clinical scores, mortality, and cytokine expression suggested PLG may be involved in EV-A71 infection-induced cytokine storm. The findings and animal model in the current study provide the new drug target for anti-EV-A71 drug discovery.
Collapse
Affiliation(s)
- Zheng-Xun Li
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Ya-Fang Wang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan City, Taiwan
| | - Li-Ting Chiang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Li-Jin Hsu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Shih-Min Wang
- Department of Pediatric, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Jen-Ren Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Hua-Lin Wu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Shun-Hua Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chuan-Fa Chang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- University Center for Bioscience and Biotechnology, National Cheng Kung University, Tainan City, Taiwan
| |
Collapse
|
2
|
Liang Y, Wu J, Chen G, Du Y, Yan Y, Xie S, Qian W, Chen A, Yi C, Tian M. Risk factor analysis and prediction model construction for severe adenovirus pneumonia in children. Ital J Pediatr 2024; 50:210. [PMID: 39385312 PMCID: PMC11465519 DOI: 10.1186/s13052-024-01771-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/22/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Severe adenovirus pneumonia in children has a high mortality rate, but research on risk prediction models is lacking. Such models are essential as they allow individualized predictions and assess whether children will likely progress to severe disease. METHODS A retrospective analysis was performed on children with adenovirus pneumonia who were hospitalized at the Children's Hospital of Nanjing Medical University from January 2017 to March 2024. The patients were grouped according to clinical factors, and the groups were compared using Ridge regression and multiple logistic regression to identify risk factors associated with severe adenovirus pneumonia. A prediction model was constructed, and its value in clinical application was evaluated. RESULTS 699 patients were included in the study, with 284 in the severe group and 415 in the general group. Through the screening of 44 variables, the final risk factors for severe adenovirus pneumonia in children as the levels of neutrophils (OR = 1.086, 95% CI: 1.054‒1.119, P < 0.001), D-dimer (OR = 1.005, 95% CI: 1.003‒1.007, P < 0.001), fibrinogen degradation products (OR = 1.341, 95% CI: 1.034‒1.738, P = 0.027), B cells (OR = 1.076, 95%CI: 1.046‒1.107, P < 0.001), and lactate dehydrogenase (OR = 1.008, 95% CI: 1.005‒1.011, P < 0.001). The value of the area under the receiver operating characteristic curve was 0.974, the 95% CI was 0.963-0.985, and the P-value of the Hosmer-Lemeshow test was 0.547 (P > 0.05), indicating that the model had strong predictive power. CONCLUSION In this study, the clinical variables of children with adenovirus pneumonia were retrospectively analyzed to identify risk factors for severe disease. A prediction model for severe disease was constructed and evaluated, showing good application value.
Collapse
Affiliation(s)
- Yaowen Liang
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinhuan Wu
- Department of Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Gang Chen
- Children's Hospital of Nanjing Medical University; The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuchen Du
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Yan
- Department of Respiratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Shuqin Xie
- Department of Respiratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wenxian Qian
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Apeng Chen
- Department of Respiratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Changhua Yi
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China.
| | - Man Tian
- Department of Respiratory, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Clarke M, Falcione S, Boghozian R, Todoran R, Zhang Y, C. Real MG, StPierre A, Joy T, Jickling GC. Viral Infection and Ischemic Stroke: Emerging Trends and Mechanistic Insights. J Am Heart Assoc 2024; 13:e035892. [PMID: 39258541 PMCID: PMC11935600 DOI: 10.1161/jaha.124.035892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
Population studies have suggested that viral infections may be contributing to risk of ischemic stroke, although the mechanisms for this are unclear. In this review, we examine the epidemiological evidence supporting the involvement of viral diseases, including influenza, COVID-19, chronic herpesvirus infections, and hepatitis C in current trends of stroke incidence. To support these associations, we highlight the virus-host interactions that are critical in the context of stroke, including direct effects of acute and persistent viral infections on vascular function, inflammation, and thrombosis. Additionally, we evaluate the systemic changes that occur during viral infection that can predispose individuals to ischemic stroke, including alterations in blood pressure regulation, coagulation, and lipid metabolism. Our review emphasizes the need to further elucidate precise mechanisms involved in viral infections and stroke risk. Future research will inform the development of targeted interventions for stroke prevention in the context of viral diseases.
Collapse
Affiliation(s)
- Michael Clarke
- Faculty of Medicine and DentistryDepartment of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
| | - Sarina Falcione
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Roobina Boghozian
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Raluca Todoran
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Yiran Zhang
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Maria Guadalupe C. Real
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Alexis StPierre
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Twinkle Joy
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| | - Glen C. Jickling
- Faculty of Medicine and DentistryDepartment of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonABCanada
- Faculty of Medicine and DentistryDepartment of MedicineDivision of NeurologyUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
4
|
Li K, Feng KC, Simon M, Fu Y, Galanakis D, Mueller S, Rafailovich MH. Molecular Basis for Surface-Initiated Non-Thrombin-Generated Clot Formation Following Viral Infection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30703-30714. [PMID: 38848451 DOI: 10.1021/acsami.4c02918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
In this paper, we propose a model that connects two standard inflammatory responses to viral infection, namely, elevation of fibrinogen and the lipid drop shower, to the initiation of non-thrombin-generated clot formation. In order to understand the molecular basis for the formation of non-thrombin-generated clots following viral infection, human epithelial and Madin-Darby Canine Kidney (MDCK, epithelial) cells were infected with H1N1, OC43, and adenovirus, and conditioned media was collected, which was later used to treat human umbilical vein endothelial cells and human lung microvascular endothelial cells. After direct infection or after exposure to conditioned media from infected cells, tissue surfaces of both epithelial and endothelial cells, exposed to 8 mg/mL fibrinogen, were observed to initiate fibrillogenesis in the absence of thrombin. No fibers were observed after direct viral exposure of the endothelium or when the epithelium cells were exposed to SARS-CoV-2 isolated spike proteins. Heating the conditioned media to 60 °C had no effect on fibrillogenesis, indicating that the effect was not enzymatic but rather associated with relatively thermally stable inflammatory factors released soon after viral infection. Spontaneous fibrillogenesis had previously been reported and interpreted as being due to the release of the alpha C domains due to strong interactions of the interior of the fibrinogen molecule in contact with hydrophobic material surfaces rather than cleavage of the fibrinopeptides. Contact angle goniometry and immunohistochemistry were used to demonstrate that the lipids produced within the epithelium and released in the conditioned media, probably after the death of infected epithelial cells, formed a hydrophobic residue responsible for fibrillogenesis. Hence, the standard inflammatory response constitutes the ideal conditions for surface-initiated clot formation.
Collapse
Affiliation(s)
- Kao Li
- School of Biomedicine and Nursing, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, Shandong, China
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Kuan-Che Feng
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Marcia Simon
- Department of Oral Biology and Pathology, Stony Brook University Medical Center, Stony Brook, New York 11794, United States
| | - Yuyang Fu
- Dongying Stem Cell Bank Medical Technology Co., Ltd., Dongying 257000, Shandong, China
| | - Dennis Galanakis
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, New York 11720, United States
| | | | - Miriam H Rafailovich
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
5
|
Cau MF, Ferraresso F, Seadler M, Badior K, Zhang Y, Ketelboeter LM, Rodriguez GG, Chen T, Ferraresso M, Wietrzny A, Robertson M, Haugen A, Cullis PR, de Moya M, Dyer M, Kastrup CJ. siRNA-mediated reduction of a circulating protein in swine using lipid nanoparticles. Mol Ther Methods Clin Dev 2024; 32:101258. [PMID: 38779336 PMCID: PMC11109470 DOI: 10.1016/j.omtm.2024.101258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Genetic manipulation of animal models is a fundamental research tool in biology and medicine but is challenging in large animals. In rodents, models can be readily developed by knocking out genes in embryonic stem cells or by knocking down genes through in vivo delivery of nucleic acids. Swine are a preferred animal model for studying the cardiovascular and immune systems, but there are limited strategies for genetic manipulation. Lipid nanoparticles (LNPs) efficiently deliver small interfering RNA (siRNA) to knock down circulating proteins, but swine are sensitive to LNP-induced complement activation-related pseudoallergy (CARPA). We hypothesized that appropriately administering optimized siRNA-LNPs could knock down circulating levels of plasminogen, a blood protein synthesized in the liver. siRNA-LNPs against plasminogen (siPLG) reduced plasma plasminogen protein and hepatic plasminogen mRNA levels to below 5% of baseline values. Functional assays showed that reducing plasminogen levels modulated systemic blood coagulation. Clinical signs of CARPA were not observed, and occasional mild and transient hepatotoxicity was present in siPLG-treated animals at 5 h post-infusion, which returned to baseline by 7 days. These findings advance siRNA-LNPs in swine models, enabling genetic engineering of blood and hepatic proteins, which can likely expand to proteins in other tissues in the future.
Collapse
Affiliation(s)
- Massimo F. Cau
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Francesca Ferraresso
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Monica Seadler
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Youjie Zhang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | | | | | - Taylor Chen
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | | | | | - Madelaine Robertson
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Amber Haugen
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Pieter R. Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marc de Moya
- Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mitchell Dyer
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Surgery, Division of Vascular and Endovascular Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Christian J. Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Departments of Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
6
|
Maina TW, McDonald PO, Rani Samuel BE, Sardi MI, Yoon I, Rogers A, McGill JL. Feeding Saccharomyces cerevisiae fermentation postbiotic products alters immune function and the lung transcriptome of preweaning calves with an experimental viral-bacterial coinfection. J Dairy Sci 2024; 107:2253-2267. [PMID: 37806633 DOI: 10.3168/jds.2023-23866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023]
Abstract
Bovine respiratory disease causes morbidity and mortality in cattle of all ages. Supplementing with postbiotic products from Saccharomyces cerevisiae fermentation (SCFP) has been reported to improve growth and provide metabolic support required for immune activation in calves. The objective of this study was to determine effects of SCFP supplementation on the transcriptional response to coinfection with bovine respiratory syncytial virus (BRSV) and Pasteurella multocida in the lung using RNA sequencing. Twenty-three calves were enrolled and assigned to 2 treatment groups: control (n = 12) or SCFP-treated (n = 11, fed 1 g/d SmartCare in milk and 5 g/d NutriTek on starter grain; both from Diamond V Mills Inc.). Calves were infected with ∼104 median tissue culture infectious dose per milliliter of BRSV, followed 6 d later by intratracheal inoculation with ∼1010 cfu of Pasteurella multocida (strain P1062). Calves were euthanized on d 10 after viral infection. Blood cells were collected and assayed on d 0 and 10 after viral infection. Bronchoalveolar lavage (BAL) cells were collected and assayed on d 14 of the feeding period (preinfection) and d 10 after viral infection. Blood and BAL cells were assayed for proinflammatory cytokine production in response to stimulation with lipopolysaccharide (LPS) or a combination of polyinosinic:polycytidylic acid and imiquimod, and BAL cells were evaluated for phagocytic and reactive oxygen species production capacity. Antemortem and postmortem BAL and lesioned and nonlesioned lung tissue samples collected at necropsy were subjected to RNA extraction and sequencing. Sequencing reads were aligned to the bovine reference genome (UMD3.1) and edgeR version 3.32.1 used for differential gene expression analysis. Supplementation with SCFP did not affect the respiratory burst activity or phagocytic activity of either lung or blood immune cells. Immune cells from the peripheral blood of SCFP-supplemented calves produced increased quantities of IL-6 in response to toll-like receptor stimulation, whereas cells from the BAL of SCFP-treated calves secreted fewer proinflammatory cytokines and less tumor necrosis factor-α (TNF-α) and IL-6 in response to the same stimuli. Transcriptional responses in lung tissues and BAL samples from SCFP-fed calves differed from the control group. The top enriched pathways in SCFP-treated lungs were associated with decreased expression of inflammatory responses and increased expression of plasminogen and genes involved in glutathione metabolism, supporting effective lung repair. Our results indicate that supplementing with SCFP postbiotics modulates both systemic and mucosal immune responses, leading to increased resistance to bovine respiratory disease.
Collapse
Affiliation(s)
- Teresia W Maina
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010
| | - Paiton O McDonald
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI 48824
| | - Beulah E Rani Samuel
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010
| | | | - Ilkyu Yoon
- Diamond V Mills Inc., Cedar Rapids, IA 52404
| | - Adam Rogers
- Diamond V Mills Inc., Cedar Rapids, IA 52404
| | - Jodi L McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50010.
| |
Collapse
|
7
|
Alvarez I, Ducatez M, Guo Y, Lion A, Widgren A, Dubourdeau M, Baillif V, Saias L, Zohari S, Bergquist J, Meyer G, Valarcher JF, Hägglund S. Proteomic and Lipidomic Profiling of Calves Experimentally Co-Infected with Influenza D Virus and Mycoplasma bovis: Insights into the Host-Pathogen Interactions. Viruses 2024; 16:361. [PMID: 38543727 PMCID: PMC10975297 DOI: 10.3390/v16030361] [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: 01/22/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 05/23/2024] Open
Abstract
The role of Influenza D virus (IDV) in bovine respiratory disease remains unclear. An in vivo experiment resulted in increased clinical signs, lesions, and pathogen replication in calves co-infected with IDV and Mycoplasma bovis (M. bovis), compared to single-infected calves. The present study aimed to elucidate the host-pathogen interactions and profile the kinetics of lipid mediators in the airways of these calves. Bronchoalveolar lavage (BAL) samples collected at 2 days post-infection (dpi) were used for proteomic analyses by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Additionally, lipidomic analyses were performed by LC-MS/MS on BAL samples collected at 2, 7 and 14 dpi. Whereas M. bovis induced the expression of proteins involved in fibrin formation, IDV co-infection counteracted this coagulation mechanism and downregulated other acute-phase response proteins, such as complement component 4 (C4) and plasminogen (PLG). The reduced inflammatory response against M. bovis likely resulted in increased M. bovis replication and delayed M. bovis clearance, which led to a significantly increased abundance of oxylipids in co-infected calves. The identified induced oxylipids mainly derived from arachidonic acid; were likely oxidized by COX-1, COX-2, and LOX-5; and peaked at 7 dpi. This paper presents the first characterization of BAL proteome and lipid mediator kinetics in response to IDV and M. bovis infection in cattle and raises hypotheses regarding how IDV acts as a co-pathogen in bovine respiratory disease.
Collapse
Affiliation(s)
- Ignacio Alvarez
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| | - Mariette Ducatez
- IHAP, Université de Tolouse, INRAE, ENVT, 31076 Toulouse, France
| | - Yongzhi Guo
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| | - Adrien Lion
- IHAP, Université de Tolouse, INRAE, ENVT, 31076 Toulouse, France
| | - Anna Widgren
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden; (A.W.); (J.B.)
| | | | | | - Laure Saias
- Ambiotis SAS, 3 Rue des Satellites, 31400 Toulouse, France
| | - Siamak Zohari
- Department of Microbiology, Swedish Veterinary Agency, Ullsvägen 2B, 75189 Uppsala, Sweden;
| | - Jonas Bergquist
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden; (A.W.); (J.B.)
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Ulls väg 26, 75007 Uppsala, Sweden
| | - Gilles Meyer
- IHAP, Université de Tolouse, INRAE, ENVT, 31076 Toulouse, France
| | - Jean-Francois Valarcher
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| | - Sara Hägglund
- Division of Ruminant Medicine, Department of Clinical Sciences, Swedish University of Agriculture Sciences, 8 Almas Allé, 75007 Uppsala, Sweden (J.-F.V.); (S.H.)
| |
Collapse
|
8
|
Nagaja SA, John RS, Krishnan M. Efficacy of Tranexamic Acid in Preventing Alveolar Osteitis in Post-extraction Sockets of First Premolars. Cureus 2024; 16:e51816. [PMID: 38327915 PMCID: PMC10847889 DOI: 10.7759/cureus.51816] [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: 12/02/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024] Open
Abstract
OBJECTIVES The present study is a randomized trial for comparing the effectiveness of tranexamic acid as an antifibrinolytic agent in preventing alveolar osteitis in the post-extraction period in patients receiving orthodontic therapy that requires extraction. METHODOLOGY This research was carried out in the Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Chennai, India. A total of 40 patients were considered subjects for the research. Patients undergoing orthodontic treatment referred to the Department of Oral and Maxillofacial Surgery for the therapeutic extractions of the first premolars were considered for this study. Randomization was done to split the population into study and control. After the atraumatic extraction of the first premolars under local anesthesia using 2% lignocaine with 1:80000 adrenaline, a tranexamic acid solution of 500 mg soaked gauze over the extraction sockets was used as the intervention in the study group, and plain gauze was used on the control group. Patients were asked to hold the gauze in place for one hour. Participants were reviewed after three days for the incidence of alveolar osteitis and pain severity and healing of the extraction sockets. Results: The prevalence of Alveolitis sicca dolorosa was found to be 5% in the research group and 15% in the control group. Patients in the control group showed more pain than the patients in the research group. The period taken for healing ranged from 7 days to 10 days in the control group and 10 days to 12 days in the study group. Conclusion: This study gives an edge that tranexamic acid can be used as a local hemostatic agent in preventing fibrinolysis of clots and preventing alveolar osteitis.
Collapse
Affiliation(s)
- Sharanika A Nagaja
- Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Rubin S John
- Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Murugesan Krishnan
- Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
9
|
Jain R, Mathew D. Mechanisms influencing the high prevalence of COVID-19 in diabetics: A systematic review. MEDICAL RESEARCH ARCHIVES 2023; 11:4540. [PMID: 38933091 PMCID: PMC11198970 DOI: 10.18103/mra.v11i10.4540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Diabetics have an increased risk of contracting COVID-19 infection and tend to have more severe symptoms. This systematic review explores the potential mechanisms influencing the high prevalence of COVID-19 infections in individuals with diabetes. It reviews the emerging evidence about the interactions between viral and diabetic pathways, particularly how diabetes physiology could contribute to higher viral reception, viral entry and pathogenicity, and the severity of disease symptoms. Finally, it examines the challenges we face in studying these mechanisms and offers new strategies that might assist our fight against current and future pandemics.
Collapse
Affiliation(s)
- Roshni Jain
- Cell and Molecular Biology Program, University of Nevada, Reno, NV 89557
- Department of Biology, University of Nevada, Reno, NV 89557
| | - Dennis Mathew
- Cell and Molecular Biology Program, University of Nevada, Reno, NV 89557
- Department of Biology, University of Nevada, Reno, NV 89557
| |
Collapse
|
10
|
Yatsenko T, Skrypnyk M, Troyanovska O, Tobita M, Osada T, Takahashi S, Hattori K, Heissig B. The Role of the Plasminogen/Plasmin System in Inflammation of the Oral Cavity. Cells 2023; 12:cells12030445. [PMID: 36766787 PMCID: PMC9913802 DOI: 10.3390/cells12030445] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/03/2023] Open
Abstract
The oral cavity is a unique environment that consists of teeth surrounded by periodontal tissues, oral mucosae with minor salivary glands, and terminal parts of major salivary glands that open into the oral cavity. The cavity is constantly exposed to viral and microbial pathogens. Recent studies indicate that components of the plasminogen (Plg)/plasmin (Pm) system are expressed in tissues of the oral cavity, such as the salivary gland, and contribute to microbial infection and inflammation, such as periodontitis. The Plg/Pm system fulfills two major functions: (a) the destruction of fibrin deposits in the bloodstream or damaged tissues, a process called fibrinolysis, and (b) non-fibrinolytic actions that include the proteolytic modulation of proteins. One can observe both functions during inflammation. The virus that causes the coronavirus disease 2019 (COVID-19) exploits the fibrinolytic and non-fibrinolytic functions of the Plg/Pm system in the oral cavity. During COVID-19, well-established coagulopathy with the development of microthrombi requires constant activation of the fibrinolytic function. Furthermore, viral entry is modulated by receptors such as TMPRSS2, which is necessary in the oral cavity, leading to a derailed immune response that peaks in cytokine storm syndrome. This paper outlines the significance of the Plg/Pm system for infectious and inflammatory diseases that start in the oral cavity.
Collapse
Affiliation(s)
- Tetiana Yatsenko
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Maksym Skrypnyk
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Olga Troyanovska
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Morikuni Tobita
- Department of Oral and Maxillofacial Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Taro Osada
- Department of Gastroenterology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-Shi 279-0021, Japan
| | - Satoshi Takahashi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo 108-8639, Japan
| | - Koichi Hattori
- Center for Genome and Regenerative Medicine, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
- Correspondence: (K.H.); (B.H.); Tel.: +81-3-3813-3111 (switchboard 2115) (B.H.)
| | - Beate Heissig
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
- Correspondence: (K.H.); (B.H.); Tel.: +81-3-3813-3111 (switchboard 2115) (B.H.)
| |
Collapse
|
11
|
Sugimoto MA, Perucci LO, Tavares LP, Teixeira MM, Sousa LP. Fibrinolysis in COVID-19: Impact on Clot Lysis and Modulation of Inflammation. Curr Drug Targets 2022; 23:1578-1592. [PMID: 36221881 DOI: 10.2174/1389450123666221011102250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/26/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023]
Abstract
COVID-19 is a multisystem disease caused by SARS-CoV-2 and is associated with an imbalance between the coagulation and fibrinolytic systems. Overall, hypercoagulation, hypofibrinolysis and fibrin-clot resistance to fibrinolysis predispose patients to thrombotic and thromboembolic events. In the lungs, the virus triggers alveolar and interstitial fibrin deposition, endothelial dysfunction, and pulmonary intravascular coagulation, all events intrinsically associated with the activation of inflammation and organ injury. Adding to the pathogenesis of COVID-19, there is a positive feedback loop by which local fibrin deposition in the lungs can fuel inflammation and consequently dysregulates coagulation, a process known as immunothrombosis. Therefore, fibrinolysis plays a central role in maintaining hemostasis and tissue homeostasis during COVID-19 by cleaning fibrin clots and controlling feed-forward products of coagulation. In addition, components of the fibrinolytic system have important immunomodulatory roles, as evidenced by studies showing the contribution of Plasminogen/Plasmin (Plg/Pla) to the resolution of inflammation. Herein, we review clinical evidence for the dysregulation of the fibrinolytic system and discuss its contribution to thrombosis risk and exacerbated inflammation in severe COVID-19. We also discuss the current concept of an interplay between fibrinolysis and inflammation resolution, mirroring the well-known crosstalk between inflammation and coagulation. Finally, we consider the central role of the Plg/Pla system in resolving thromboinflammation, drawing attention to the overlooked consequences of COVID-19-associated fibrinolytic abnormalities to local and systemic inflammation.
Collapse
Affiliation(s)
- Michelle A Sugimoto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Division of Medicine, University College London, London, UK.,Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luiza O Perucci
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Nucleus of Research on Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luciana P Tavares
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Lirlândia P Sousa
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
12
|
Jadhav AN, Shushma G, Deshmukh VD. Efficacy of tranexamic acid in prevention of alveolar osteitis following surgical removal of impacted mandibular third molar. Natl J Maxillofac Surg 2022; 13:S85-S90. [PMID: 36393943 PMCID: PMC9651232 DOI: 10.4103/njms.njms_298_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 07/28/2021] [Accepted: 09/13/2021] [Indexed: 06/16/2023] Open
Abstract
CONTEXT Many preventive measures are described to avoid alveolar osteitis (AO) during third molar surgery (TMS), but very few are found to be effective. Tranexamic acid (TA), an antifibrinolytic agent, impedes the proteolytic degradation of fibrin and prevents blood clot disintegration. AIMS The study was conducted to determine the efficacy of intra-alveolar application of TA soaked in Gelfoam in prevention of AO. SETTINGS AND DESIGN This was a randomized control trial. MATERIALS AND METHODS A total of 200 patients (100 in control group and 100 in study group) reporting for TMS were allocated randomly. Following surgery, TA soaked in gel foam was placed in socket and sutured in the study group, while in the control group, closure was done by suturing. Patients followed subsequently to observe the incidence of AO, pain severity, and duration of healing after AO. STATISTICAL ANALYSIS Z-test, Mann-Whitney test, and t-test were applied, respectively, to compare the incidence of AO, severity of pain, and duration of healing between the two groups. RESULTS The incidence of AO in the control group was 18% and 6% in the study group. Patients in the control group experienced severe pain as compared to patients in the study group. The duration of healing varied from 12 to 16 days in the control group, but in the study group, it was <10 days. CONCLUSION TA significantly reduces the incidence of AO in addition to the reduced severity of pain and enhanced healing. We recommend the routine use of TA, owing to its astonishing rewards.
Collapse
Affiliation(s)
- Ajinath Nanasaheb Jadhav
- Department of Dentistry, JIIU'S Indian Institute of Medical Sciences and Research, Jalna, Maharashtra, India
| | - G. Shushma
- Department of Dentistry, Koppal Institute of Medical Sciences, Koppal, Bhagyanagar, Karnataka, India
| | - Vijay Dnyandev Deshmukh
- Department of Dentistry, JIIU'S Indian Institute of Medical Sciences and Research, Jalna, Maharashtra, India
| |
Collapse
|
13
|
Prudovsky I, Kacer D, Zucco VV, Palmeri M, Falank C, Kramer R, Carter D, Rappold J. Tranexamic acid: Beyond antifibrinolysis. Transfusion 2022; 62 Suppl 1:S301-S312. [PMID: 35834488 DOI: 10.1111/trf.16976] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
Tranexamic acid (TXA) is a popular antifibrinolytic drug widely used in hemorrhagic trauma patients and cardiovascular, orthopedic, and gynecological surgical patients. TXA binds plasminogen and prevents its maturation to the fibrinolytic enzyme plasmin. A number of studies have demonstrated the broad life-saving effects of TXA in trauma, superior to those of other antifibrinolytic agents. Besides preventing fibrinolysis and blood loss, TXA has been reported to suppress posttraumatic inflammation and edema. Although the efficiency of TXA transcends simple inhibition of fibrinolysis, little is known about its mechanisms of action besides the suppression of plasmin maturation. Understanding the broader effects of TXA at the cell, organ, and organism levels are required to elucidate its potential mechanisms of action transcending antifibrinolytic activity. In this article, we provide a brief review of the current clinical use of TXA and then focus on the effects of TXA beyond antifibrinolytics such as its anti-inflammatory activity, protection of the endothelial and epithelial monolayers, stimulation of mitochondrial respiration, and suppression of melanogenesis.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA
| | - Doreen Kacer
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA
| | - Victoria Vieira Zucco
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA
| | - Monica Palmeri
- Maine Medical Center Cardiovascular Institute, Maine Medical Center, Portland, Maine, USA
| | - Carolyne Falank
- Department of Trauma, Maine Medical Center, Maine Medical Center, Portland, Maine, USA
| | - Robert Kramer
- Maine Medical Center Cardiovascular Institute, Maine Medical Center, Portland, Maine, USA
| | - Damien Carter
- Department of Trauma, Maine Medical Center, Maine Medical Center, Portland, Maine, USA
| | - Joseph Rappold
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA.,Department of Trauma, Maine Medical Center, Maine Medical Center, Portland, Maine, USA
| |
Collapse
|
14
|
Gómez-Carballa A, Rivero-Calle I, Pardo-Seco J, Gómez-Rial J, Rivero-Velasco C, Rodríguez-Núñez N, Barbeito-Castiñeiras G, Pérez-Freixo H, Cebey-López M, Barral-Arca R, Rodriguez-Tenreiro C, Dacosta-Urbieta A, Bello X, Pischedda S, Currás-Tuala MJ, Viz-Lasheras S, Martinón-Torres F, Salas A. A multi-tissue study of immune gene expression profiling highlights the key role of the nasal epithelium in COVID-19 severity. ENVIRONMENTAL RESEARCH 2022; 210:112890. [PMID: 35202626 PMCID: PMC8861187 DOI: 10.1016/j.envres.2022.112890] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/11/2022] [Accepted: 02/02/2022] [Indexed: 05/08/2023]
Abstract
Coronavirus Disease-19 (COVID-19) symptoms range from mild to severe illness; the cause for this differential response to infection remains unknown. Unravelling the immune mechanisms acting at different levels of the colonization process might be key to understand these differences. We carried out a multi-tissue (nasal, buccal and blood; n = 156) gene expression analysis of immune-related genes from patients affected by different COVID-19 severities, and healthy controls through the nCounter technology. Mild and asymptomatic cases showed a powerful innate antiviral response in nasal epithelium, characterized by activation of interferon (IFN) pathway and downstream cascades, successfully controlling the infection at local level. In contrast, weak macrophage/monocyte driven innate antiviral response and lack of IFN signalling activity were present in severe cases. Consequently, oral mucosa from severe patients showed signals of viral activity, cell arresting and viral dissemination to the lower respiratory tract, which ultimately could explain the exacerbated innate immune response and impaired adaptative immune responses observed at systemic level. Results from saliva transcriptome suggest that the buccal cavity might play a key role in SARS-CoV-2 infection and dissemination in patients with worse prognosis. Co-expression network analysis adds further support to these findings, by detecting modules specifically correlated with severity involved in the abovementioned biological routes; this analysis also provides new candidate genes that might be tested as biomarkers in future studies. We also found tissue specific severity-related signatures mainly represented by genes involved in the innate immune system and cytokine/chemokine signalling. Local immune response could be key to determine the course of the systemic response and thus COVID-19 severity. Our findings provide a framework to investigate severity host gene biomarkers and pathways that might be relevant to diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Alberto Gómez-Carballa
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Irene Rivero-Calle
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jacobo Pardo-Seco
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - José Gómez-Rial
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Laboratorio de Inmunología. Servicio de Análisis Clínicos. Hospital Clínico Universitario (SERGAS), Galicia, Spain
| | - Carmen Rivero-Velasco
- Intensive Medicine Department, Hospital Clìnico Universitario de Santiago de Compostela, Galicia, Spain
| | - Nuria Rodríguez-Núñez
- Pneumology Department, Hospital Clìnico Universitario de Santiago de Compostela, Galicia, Spain
| | - Gema Barbeito-Castiñeiras
- Clinical Microbiology Unit, Complexo Hospitalario Universitario de Santiago Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
| | - Hugo Pérez-Freixo
- Preventive Medicine Department, Hospital Clínico Universitario de Santiago de Compostela, Spain
| | - Miriam Cebey-López
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Ruth Barral-Arca
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Carmen Rodriguez-Tenreiro
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Dacosta-Urbieta
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Xabier Bello
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Sara Pischedda
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - María José Currás-Tuala
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Sandra Viz-Lasheras
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Antonio Salas
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria (IDIS) de Santiago, Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela (USC), and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.
| |
Collapse
|
15
|
Insight towards the effect of the multi basic cleavage site of SARS-CoV-2 spike protein on cellular proteases. Virus Res 2022; 318:198845. [PMID: 35680004 PMCID: PMC9170277 DOI: 10.1016/j.virusres.2022.198845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/27/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection presents an immense global health problem. Spike (S) protein of coronavirus is the primary determinant of its entry into the host as it consists of both receptor binding and fusion domain. Besides tissue tropism, and host range, coronavirus pathogenesis are primarily controlled by the interaction of S protein with the cell receptor. Moreover, the proteolytic activation of S protein by host cell proteases plays a decisive role. The host-cell proteases have shown to be involved in the proteolysis of S protein and cleaving it into two functional subunits, S1 and S2, during the maturation process. In the present study, the interaction of the S protein of SARS-CoV-2 with different host proteases like furin, cathepsin B, and plasmin has been analyzed using molecular docking and molecular dynamics (MD) simulation. Incorporation of the furin cleavage site (R-R-A-R) in the S protein of SARS-CoV-2 has been studied by mutating the individual amino acid. MD simulation results suggest the polytropic nature of the S protein. Our analysis indicated that a single amino acid substitution in the polybasic cleavage site of S protein perturb the binding of cellular proteases. This mutation study might help to generate an attenuated SARS-CoV-2. Besides, targeting host proteases by inhibitors may result in a practical approach to stop the cellular spread of SARS-CoV-2 and develop its antiviral.
Collapse
|
16
|
Arslan FD, Başok BI, Terzioğlu M, Altan TK, Karaca Y, Senger SS, Çolak A. Evaluation of D-dimer levels measured by different analytical methods in COVID-19 patients. Blood Coagul Fibrinolysis 2022; 33:209-215. [PMID: 35239616 DOI: 10.1097/mbc.0000000000001129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Clinicians experience some challenges due to the lack of standardization of test, although D-dimer is a prognostic marker for COVID-19. We compared the clinical and analytical performances of D-dimer results obtained from different devices, kits and methods in patients with a diagnosis of COVID-19. Thirty-nine patients with a diagnosis of COVID-19 and 24 healthy individuals were included in the study. D-dimer levels were measured with Innovance D-DIMER kit (immunoturbidimetric method) on Sysmex CS-2500 and BCS XP and VIDAS D-Dimer Exclusion II kit (enzyme-linked fluorescence method) on mini VIDAS. The studies of precision, method comparison and clinic performance were performed. The variation coefficients in all systems were within the acceptable imprecision (7.8%). Bias%(12.5%) between BCS XP and Sysmex CS-2500 was lower than the acceptable Bias%(15.5%). Bias% values (19.2% and 33.3%, respectively) between Mini VIDAS with BCS XP and Sysmex CS-2500 were higher than the acceptable Bias%. The correlation coefficients among all systems were 0.89-0.98. For 500 ng/ml FEU, there was almost perfect agreement between BCS XP and Sysmex CS-2500, a moderate agreement between Mini VIDAS and BCS XP and Sysmex CS-2500. The cut-off values for distinguishing between individuals with and withoutCOVID-19 were Mini VIDAS, Sysmex CS-2500 and BCS XP 529, 380 and 390 ng/ml FEU, respectively. The immunoturbidimetric method can be used as an alternative to the enzyme-linked fluorescent method because of satisfactory agreement at the different thresholds proposed for venous thromboembolism. However, it is recommended to follow up COVID-19 with the D-dimer results obtained by the same assay system.
Collapse
Affiliation(s)
- Fatma Demet Arslan
- Department of Medical Biochemistry, University of Bakircay, School of Medicine
| | | | | | | | | | - Suheyla Serin Senger
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| | | |
Collapse
|
17
|
The Contribution of Viral Proteins to the Synergy of Influenza and Bacterial Co-Infection. Viruses 2022; 14:v14051064. [PMID: 35632805 PMCID: PMC9143653 DOI: 10.3390/v14051064] [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: 04/06/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
A severe course of acute respiratory disease caused by influenza A virus (IAV) infection is often linked with subsequent bacterial superinfection, which is difficult to cure. Thus, synergistic influenza-bacterial co-infection represents a serious medical problem. The pathogenic changes in the infected host are accelerated as a consequence of IAV infection, reflecting its impact on the host immune response. IAV infection triggers a complex process linked with the blocking of innate and adaptive immune mechanisms required for effective antiviral defense. Such disbalance of the immune system allows for easier initiation of bacterial superinfection. Therefore, many new studies have emerged that aim to explain why viral-bacterial co-infection can lead to severe respiratory disease with possible fatal outcomes. In this review, we discuss the key role of several IAV proteins-namely, PB1-F2, hemagglutinin (HA), neuraminidase (NA), and NS1-known to play a role in modulating the immune defense of the host, which consequently escalates the development of secondary bacterial infection, most often caused by Streptococcus pneumoniae. Understanding the mechanisms leading to pathological disorders caused by bacterial superinfection after the previous viral infection is important for the development of more effective means of prevention; for example, by vaccination or through therapy using antiviral drugs targeted at critical viral proteins.
Collapse
|
18
|
Khanal P, Patil VS, Bhandare VV, Dwivedi PS, Shastry C, Patil B, Gurav SS, Harish DR, Roy S. Computational investigation of benzalacetophenone derivatives against SARS-CoV-2 as potential multi-target bioactive compounds. Comput Biol Med 2022; 146:105668. [PMID: 35667894 PMCID: PMC9135652 DOI: 10.1016/j.compbiomed.2022.105668] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 02/08/2023]
Abstract
Benzalacetophenones, precursors of flavonoids are aromatic ketones and enones and possess the immunostimulant as well as antiviral activities. Thus, benzalacetophenones were screened against the COVID-19 that could be lethal in patients with compromised immunity. We considered ChEBI recorded benzalacetophenone derivative(s) and evaluated their activity against 3C-like protease (3CLpro), papain-like protease (PLpro), and spike protein of SARS-Cov-2 to elucidate their possible role as antiviral agents. The probable targets for each compound were retrieved from DIGEP-Pred at 0.5 pharmacological activity and all the modulated proteins were enriched to identify the probably regulated pathways, biological processes, cellular components, and molecular functions. In addition, molecular docking was performed using AutoDock 4 and the best-identified hits were subjected to all-atom molecular dynamics simulation and binding energy calculations using molecular mechanics Poisson-Boltzmann surface area (MMPBSA). The compound 4-hydroxycordoin showed the highest druglikeness score and regulated nine proteins of which five were down-regulated and four were upregulated. Similarly, enrichment analysis identified the modulation of multiple pathways concerned with the immune system as well as pathways related to infectious and non-infectious diseases. Likewise, 3'-(3-methyl-2-butenyl)-4′-O-β-d-glucopyranosyl-4,2′-dihydroxychalcone with 3CLpro, 4-hydroxycordoin with PLpro and mallotophilippen D with spike protein receptor-binding domain showed highest binding affinity, revealed stable interactions during the simulation, and scored binding free energy of −26.09 kcal/mol, −16.28 kcal/mol, and −39.2 kcal/mol, respectively. Predicted anti-SARS-CoV-2 activities of the benzalacetophenones reflected the requirement of wet lab studies to develop novel antiviral candidates.
Collapse
|
19
|
Xin Y, Chen S, Tang K, Wu Y, Guo Y. Identification of Nifurtimox and Chrysin as Anti-Influenza Virus Agents by Clinical Transcriptome Signature Reversion. Int J Mol Sci 2022; 23:ijms23042372. [PMID: 35216485 PMCID: PMC8876279 DOI: 10.3390/ijms23042372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/12/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022] Open
Abstract
The rapid development in the field of transcriptomics provides remarkable biomedical insights for drug discovery. In this study, a transcriptome signature reversal approach was conducted to identify the agents against influenza A virus (IAV) infection through dissecting gene expression changes in response to disease or compounds’ perturbations. Two compounds, nifurtimox and chrysin, were identified by a modified Kolmogorov–Smirnov test statistic based on the transcriptional signatures from 81 IAV-infected patients and the gene expression profiles of 1309 compounds. Their activities were verified in vitro with half maximal effective concentrations (EC50s) from 9.1 to 19.1 μM against H1N1 or H3N2. It also suggested that the two compounds interfered with multiple sessions in IAV infection by reversing the expression of 28 IAV informative genes. Through network-based analysis of the 28 reversed IAV informative genes, a strong synergistic effect of the two compounds was revealed, which was confirmed in vitro. By using the transcriptome signature reversion (TSR) on clinical datasets, this study provides an efficient scheme for the discovery of drugs targeting multiple host factors regarding clinical signs and symptoms, which may also confer an opportunity for decelerating drug-resistant variant emergence.
Collapse
Affiliation(s)
- Yijing Xin
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shubing Chen
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ke Tang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - You Wu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Guo
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: ; Tel.: +86-010-63161716
| |
Collapse
|
20
|
Perlas A, Argilaguet J, Bertran K, Sánchez-González R, Nofrarías M, Valle R, Ramis A, Cortey M, Majó N. Dual Host and Pathogen RNA-Seq Analysis Unravels Chicken Genes Potentially Involved in Resistance to Highly Pathogenic Avian Influenza Virus Infection. Front Immunol 2022; 12:800188. [PMID: 35003125 PMCID: PMC8727699 DOI: 10.3389/fimmu.2021.800188] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Highly pathogenic avian influenza viruses (HPAIVs) cause severe systemic disease and high mortality rates in chickens, leading to a huge economic impact in the poultry sector. However, some chickens are resistant to the disease. This study aimed at evaluating the mechanisms behind HPAIV disease resistance. Chickens of different breeds were challenged with H7N1 HPAIV or clade 2.3.4.4b H5N8 HPAIV, euthanized at 3 days post-inoculation (dpi), and classified as resistant or susceptible depending on the following criteria: chickens that presented i) clinical signs, ii) histopathological lesions, and iii) presence of HPAIV antigen in tissues were classified as susceptible, while chickens lacking all these criteria were classified as resistant. Once classified, we performed RNA-Seq from lung and spleen samples in order to compare the transcriptomic signatures between resistant and susceptible chickens. We identified minor transcriptomic changes in resistant chickens in contrast with huge alterations observed in susceptible chickens. Interestingly, six differentially expressed genes were downregulated in resistant birds and upregulated in susceptible birds. Some of these genes belong to the NF-kappa B and/or mitogen-activated protein kinase signaling pathways. Among these six genes, the serine protease-encoding gene PLAU was of particular interest, being the most significantly downregulated gene in resistant chickens. Expression levels of this protease were further validated by RT-qPCR in a larger number of experimentally infected chickens. Furthermore, HPAIV quasi-species populations were constructed using 3 dpi oral swabs. No substantial changes were found in the viral segments that interact with the innate immune response and with the host cell receptors, reinforcing the role of the immune system of the host in the clinical outcome. Altogether, our results suggest that an early inactivation of important host genes could prevent an exaggerated immune response and/or viral replication, conferring resistance to HPAIV in chickens.
Collapse
Affiliation(s)
- Albert Perlas
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Jordi Argilaguet
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Kateri Bertran
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Raúl Sánchez-González
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Miquel Nofrarías
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Rosa Valle
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Antonio Ramis
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Martí Cortey
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Natàlia Majó
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| |
Collapse
|
21
|
Gando S, Wada T. Pathomechanisms Underlying Hypoxemia in Two COVID-19-Associated Acute Respiratory Distress Syndrome Phenotypes: Insights From Thrombosis and Hemostasis. Shock 2022; 57:1-6. [PMID: 34172612 PMCID: PMC8662946 DOI: 10.1097/shk.0000000000001825] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND The pathomechanisms of hypoxemia and treatment strategies for type H and type L acute respiratory distress syndrome (ARDS) in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced coronavirus disease 2019 (COVID-19) have not been elucidated. MAIN TEXT SARS-CoV-2 mainly targets the lungs and blood, leading to ARDS, and systemic thrombosis or bleeding. Angiotensin II-induced coagulopathy, SARS-CoV-2-induced hyperfibrin(ogen)olysis, and pulmonary and/or disseminated intravascular coagulation due to immunothrombosis contribute to COVID-19-associated coagulopathy. Type H ARDS is associated with hypoxemia due to diffuse alveolar damage-induced high right-to-left shunts. Immunothrombosis occurs at the site of infection due to innate immune inflammatory and coagulofibrinolytic responses to SARS-CoV-2, resulting in microvascular occlusion with hypoperfusion of the lungs. Lung immunothrombosis in type L ARDS results from neutrophil extracellular traps containing platelets and fibrin in the lung microvasculature, leading to hypoxemia due to impaired blood flow and a high ventilation/perfusion (VA/Q) ratio. COVID-19-associated ARDS is more vascular centric than the other types of ARDS. D-dimer levels have been monitored for the progression of microvascular thrombosis in COVID-19 patients. Early anticoagulation therapy in critical patients with high D-dimer levels may improve prognosis, including the prevention and/or alleviation of ARDS. CONCLUSIONS Right-to-left shunts and high VA/Q ratios caused by lung microvascular thrombosis contribute to hypoxemia in type H and L ARDS, respectively. D-dimer monitoring-based anticoagulation therapy may prevent the progression to and/or worsening of ARDS in COVID-19 patients.
Collapse
Affiliation(s)
- Satoshi Gando
- Acute and Critical Center, Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
22
|
Castro P, Palomo M, Moreno-Castaño AB, Fernández S, Torramadé-Moix S, Pascual G, Martinez-Sanchez J, Richardson E, Téllez A, Nicolas JM, Carreras E, Richardson PG, Badimon JJ, Escolar G, Diaz-Ricart M. Is the Endothelium the Missing Link in the Pathophysiology and Treatment of COVID-19 Complications? Cardiovasc Drugs Ther 2022; 36:547-560. [PMID: 34097193 PMCID: PMC8181544 DOI: 10.1007/s10557-021-07207-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 02/08/2023]
Abstract
Patients with COVID-19 present a wide spectrum of disease severity, from asymptomatic cases in the majority to serious disease leading to critical care and even death. Clinically, four different scenarios occur within the typical disease timeline: first, an incubation and asymptomatic period; second, a stage with mild symptoms due mainly to the virus itself; third, in up to 20% of the patients, a stage with severe symptoms where a hyperinflammatory response with a cytokine storm driven by host immunity induces acute respiratory distress syndrome; and finally, a post-acute sequelae (PASC) phase, which present symptoms that can range from mild or annoying to actually quite incapacitating. Although the most common manifestation is acute respiratory failure of the lungs, other organs are also frequently involved. The clinical manifestations of the COVID-19 infection support a key role for endothelial dysfunction in the pathobiology of this condition. The virus enters into the organism via its interaction with angiotensin-converting enzyme 2-receptor that is present prominently in the alveoli, but also in endothelial cells, which can be directly infected by the virus. Cytokine release syndrome can also drive endothelial damage independently. Consequently, a distinctive feature of SARS-CoV-2 infection is vascular harm, with severe endothelial injury, widespread thrombosis, microangiopathy, and neo-angiogenesis in response to endothelial damage. Therefore, endothelial dysfunction seems to be the pathophysiological substrate for severe COVID-19 complications. Biomarkers of endothelial injury could constitute strong indicators of disease progression and severity. In addition, the endothelium could represent a very attractive target to both prevent and treat these complications. To establish an adequate therapy, the underlying pathophysiology and corresponding clinical stage should be clearly identified. In this review, the clinical features of COVID-19, the central role of the endothelium in COVID-19 and in other pathologies, and the potential of specific therapies aimed at protecting the endothelium in COVID-19 patients are addressed.
Collapse
Affiliation(s)
- Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Marta Palomo
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Ana Belen Moreno-Castaño
- Barcelona Endothelium Team, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | - Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Sergi Torramadé-Moix
- IDIBAPS, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | | | - Julia Martinez-Sanchez
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Edward Richardson
- Frank H. Netter M.D. School of Medicine At, Quinnipiac University, North Haven, CT, USA
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Adrián Téllez
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Josep M Nicolas
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Enric Carreras
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Paul G Richardson
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Juan José Badimon
- Cardiology Department, Cardiovascular Institute, Mount Sinai Hospital, New York, NY, USA
- AtheroThrombosis Research Unit, Cardiovascular Institute, Icahn School of Medicine At Mount Sinai, New York, NY, USA
| | - Gines Escolar
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | - Maribel Diaz-Ricart
- School of Medicine, University of Barcelona, Barcelona, Spain.
- IDIBAPS, Barcelona, Spain.
- Barcelona Endothelium Team, Barcelona, Spain.
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain.
| |
Collapse
|
23
|
Tabassum T, Rahman A, Araf Y, Ullah MA, Hosen MJ. Prospective selected biomarkers in COVID-19 diagnosis and treatment. Biomark Med 2021; 15:1435-1449. [PMID: 34538093 PMCID: PMC8454595 DOI: 10.2217/bmm-2021-0038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/28/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has become a global health concern, due to the high transmissible nature of its causal agent and lack of proper treatment. Early diagnosis and nonspecific medical supports of the patients appeared to be effective strategy so far to combat the pandemic caused by COVID-19 outbreak. Biomarkers can play pivotal roles in timely and proper diagnosis of COVID-19 patients, as well as for distinguishing them from other pulmonary infections. Besides, biomarkers can help in reducing the rate of mortality and evaluating viral pathogenesis with disease prognosis. This article intends to provide a broader overview of the roles and uses of different biomarkers in the early diagnosis of COVID-19, as well as in the classification of COVID-19 patients into multiple risk groups.
Collapse
Affiliation(s)
- Tahani Tabassum
- Department of Mathematics & Natural Sciences, Biotechnology Program, School of Data & Sciences, Brac University, Dhaka, Bangladesh
| | - Ahsab Rahman
- Department of Mathematics & Natural Sciences, Biotechnology Program, School of Data & Sciences, Brac University, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering & Biotechnology, School of Life Sciences, Shahjalal University of Science & Technology, Sylhet, Bangladesh
| | - Md A Ullah
- Department of Biotechnology & Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Dhaka, Bangladesh
| | - Mohammad J Hosen
- Department of Genetic Engineering & Biotechnology, School of Life Sciences, Shahjalal University of Science & Technology, Sylhet, Bangladesh
| |
Collapse
|
24
|
Gando S, Wada T. Thromboplasminflammation in COVID-19 Coagulopathy: Three Viewpoints for Diagnostic and Therapeutic Strategies. Front Immunol 2021; 12:649122. [PMID: 34177896 PMCID: PMC8226122 DOI: 10.3389/fimmu.2021.649122] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 05/28/2021] [Indexed: 01/08/2023] Open
Abstract
Thromboplasminflammation in coronavirus disease 2019 (COVID-19) coagulopathy consists of angiotensin II (Ang II)-induced coagulopathy, activated factor XII (FXIIa)- and kallikrein, kinin system-enhanced fibrinolysis, and disseminated intravascular coagulation (DIC). All three conditions induce systemic inflammation via each pathomechanism-developed production of inflammatory cytokines. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) downregulates angiotensin-converting enzyme 2, leading to an increase in Ang II levels. Ang II-induced coagulopathy comprising platelet activation, thrombin generation, plasminogen activator inhibitor-1 expression and endothelial injury causes thrombosis via the angiotensin II type 1 receptor. SARS-CoV-2 RNA and neutrophil extracellular trap (NET) DNA activate FXII, resulting in plasmin generation through FXIIa- and kallikrein-mediated plasminogen conversion to plasmin and bradykinin-induced tissue-type plasminogen activator release from the endothelium via the kinin B2 receptor. NETs induce immunothrombosis at the site of infection (lungs), through histone- and DNA-mediated thrombin generation, insufficient anticoagulation control, and inhibition of fibrinolysis. However, if the infection is sufficiently severe, immunothrombosis disseminates into the systemic circulation, and DIC, which is associated with the endothelial injury, occurs. Inflammation, and serine protease networks of coagulation and fibrinolysis, militate each other through complement pathways, which exacerbates three pathologies of COVID-19 coagulopathy. COVID-19 coagulopathy causes microvascular thrombosis and bleeding, resulting in multiple organ dysfunction and death in critically ill patients. Treatment targets for improving the prognosis of COVID-19 coagulopathy include thrombin, plasmin, and inflammation, and SARS-CoV-2 infection. Several drugs are candidates for controlling these conditions; however, further advances are required to establish robust treatments based on a clear understanding of molecular mechanisms of COVID-19 coagulopathy.
Collapse
Affiliation(s)
- Satoshi Gando
- Acute and Critical Center, Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan.,Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
25
|
Nizami DJ, Raman V, Paulose L, Hazari KS, Mallick AK. Role of laboratory biomarkers in assessing the severity of COVID-19 disease. A cross-sectional study. J Family Med Prim Care 2021; 10:2209-2215. [PMID: 34322414 PMCID: PMC8284239 DOI: 10.4103/jfmpc.jfmpc_145_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/17/2021] [Accepted: 03/22/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Corona virus disease 2019 (Covid-19) has high infectivity and mortality rate. Covid-19 patients can suddenly deteriorate and develop life threatening complications. Hence, there is a need to identify laboratory biomarkers in order to categorize high risk patients. The main purpose of the study is to investigate the role and correlation of laboratory parameters such as total leucocyte count (TLC), absolute lymphocyte count, platelet count, C-Reactive Proteins (CRP), serum ferritin, serum lactate dehydrogenase (LDH), serum procalcitonin and D-dimer in severe and non-severe Covid-19 patients. METHODOLOGY This retrospective cross-sectional study was conducted at Latifa Women and Child Hospital in the UAE after obtaining ethical committee clearance. Based on the symptoms and the criteria by National Institute of Health, USA, 109 patients were divided into three groups: Non-severe with 75, severe with 18 and critical with 16 patients. Laboratory data of these patients were assessed through the electronic medical records (SALAMA). Statistical analysis was done using Statistical Packages for Social Sciences (SPSS) version 25.0 (SPSS/PC; SPSS-25.0, Chicago, USA). Laboratory test profiles were expressed as mean (SD). Independent 't' test and ANOVA were used to study the significance of means. P value less than 0.05 was considered significant. RESULT Males were more severely affected than females. Severe and critically ill Covid-19 patients had a significantly higher TLC, serum LDH, ferritin and CRP and lower absolute lymphocyte count. PCT and D-dimer were significantly elevated in critical group. CONCLUSION Along with clinical presentation and radiological findings, biochemical parameter may also be considered as important predictors for assessing severity in covid-19 patients.
Collapse
Affiliation(s)
- Deba J. Nizami
- Department of Obstetrics and Gynecology, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | - Vidya Raman
- Department of Obstetrics and Gynecology, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | - L. Paulose
- Department of Obstetrics and Gynecology, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | - Komal S. Hazari
- Department of Obstetrics and Gynecology, Latifa Hospital, Dubai Health Authority, Dubai, UAE
| | - Ayaz K. Mallick
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| |
Collapse
|
26
|
Abstract
Plasminogen is an abundant plasma protein that exists in various zymogenic forms. Plasmin, the proteolytically active form of plasminogen, is known for its essential role in fibrinolysis. To date, therapeutic targeting of the fibrinolytic system has been for 2 purposes: to promote plasmin generation for thromboembolic conditions or to stop plasmin to reduce bleeding. However, plasmin and plasminogen serve other important functions, some of which are unrelated to fibrin removal. Indeed, for >40 years, the antifibrinolytic agent tranexamic acid has been administered for its serendipitously discovered skin-whitening properties. Plasmin also plays an important role in the removal of misfolded/aggregated proteins and can trigger other enzymatic cascades, including complement. In addition, plasminogen, via binding to one of its dozen cell surface receptors, can modulate cell behavior and further influence immune and inflammatory processes. Plasminogen administration itself has been reported to improve thrombolysis and to accelerate wound repair. Although many of these more recent findings have been derived from in vitro or animal studies, the use of antifibrinolytic agents to reduce bleeding in humans has revealed additional clinically relevant consequences, particularly in relation to reducing infection risk that is independent of its hemostatic effects. The finding that many viruses harness the host plasminogen to aid infectivity has suggested that antifibrinolytic agents may have antiviral benefits. Here, we review the broadening role of the plasminogen-activating system in physiology and pathophysiology and how manipulation of this system may be harnessed for benefits unrelated to its conventional application in thrombosis and hemostasis.
Collapse
|
27
|
Kiraz A, Guzeldag S, Eren E, Goksu M, Bayram A. Investigation of the relationship between inherited thrombophilia and novel coronavirus pneumonia. Future Virol 2021. [PMCID: PMC8059147 DOI: 10.2217/fvl-2020-0395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aim: This study aimed to investigate the relationship between severe novel coronavirus pneumonia (NCP) and hypercoagulable conditions that predispose patients to thrombosis such as the prothrombin gene ( F2) rs1799963 (G20210A), factor V Leiden ( F5) rs6025 (G1691A) and PAI-1 (rs1799768). Patients: NCP-diagnosed 62 previously healthy patients were enrolled for the investigation of the thrombophilia-related polymorphisms. Materials & methods: The frequency of genotypes were compared with healthy control group frequencies from other studies. Results: There were no statistically significant differences between the severe patient group and the healthy population regarding the investigated single nucleotide polymorphisms (SNPs). Conclusion: This study is the first to rule out the relationship of rs1799963, rs6025 and rs1799768 with severe NCP.
Collapse
Affiliation(s)
- Aslihan Kiraz
- Department of Medical Genetics, Kayseri City Education & Research Hospital, Kayseri 38080, Turkey
| | - Seda Guzeldag
- Department of Intensive Care Unit, Kayseri City Education & Research Hospital, Kayseri 38080, Turkey
| | - Esma Eren
- Department of Infectious Diseases & Clinical Microbiology, Kayseri City Education & Research Hospital, Kayseri 38080, Turkey
| | - Musa Goksu
- Department of Infectious Diseases & Clinical Microbiology, Kayseri City Education & Research Hospital, Kayseri 38080, Turkey
| | - Arslan Bayram
- Department of Medical Genetics, Etlik Zübeyde Hanim Women’s Diseases Education & Research Hospital, Ankara 06010, Turkey
| |
Collapse
|
28
|
Chen Z, Chen W, Wang Q, Qin Y, Wang X, Ma T, Zhang P, Li X, Wang X, Ding L, Li Z. Comparative analysis of sialic acid α2–3/6 galactose glycan-binding proteins in human saliva and serum. J Mol Struct 2021; 1230:129859. [DOI: 10.1016/j.molstruc.2020.129859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
29
|
Askary E, Poordast T, Shiravani Z, Ali MA, Hashemi A, Naseri R, Moradialamdarloo S, Karimi Z, Izanloo E, Najib FS. Coronavirus disease 2019 (COVID-19) manifestations during pregnancy in all three trimesters: A case series. Int J Reprod Biomed 2021; 19:191-204. [PMID: 33718763 PMCID: PMC7922300 DOI: 10.18502/ijrm.v19i2.8477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/23/2020] [Accepted: 12/05/2020] [Indexed: 12/15/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) pandemic has raised concerns about the susceptibility amongst different groups of the population. Pregnant women are one such group. This study was conducted to investigate the effect of COVID-19 on pregnancy and maternal/neonatal outcomes. Case presentation This case series was conducted on 16 pregnant women with COVID-19 from March 21 to May 11, 2020. Clinical characteristics, pregnancy complications, medication used, maternal/neonatal outcomes, and fatality rate were investigated through this study. The mean age of the patients was 30.06 yrs. Patients from all three trimesters were included (1 in first, 5 in second, and 10 in the third trimesters). The most common clinical symptoms were shortness of breath (n = 10), dry cough (n = 10), myalgia (n = 8), and chills (n = 7). Also, three cases had papulosquamous skin lesions with fissuring. The most common laboratory results were leukocytosis (n = 8), increased liver enzymes (n = 6), elevated CRP (n = 5), and thrombocytopenia (n = 4). There was one case of maternal mortality, five of premature labor pain (PLP), two of preeclampsia, and two of placenta accreta. Twelve pregnancies were terminated (nine cesarean sections, three vaginal deliveries). Among neonates, we had 6 cases of preterm labor. All neonates had negative PCR results. Conclusion Clinical manifestations and paraclinical results were similar to non-pregnant patients. There was no evidence of vertical transmission. PLP and premature rupture of membranes (PROM) were the most common complications in the second and third trimesters of pregnant COVID-19 women, which can lead to rupture of the uterus. Termination and delivery should be planned individually.
Collapse
Affiliation(s)
- Elham Askary
- Department of Obstetrics and Gynecology, School of Medicine, Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Poordast
- Department of Obstetrics and Gynecology, School of Medicine, Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Shiravani
- Department of Obstetrics and Gynecology, School of Medicine, Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ashraf Ali
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefeh Hashemi
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Naseri
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shaghayegh Moradialamdarloo
- Department of Obstetrics and Gynecology, School of Medicine, Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zinat Karimi
- Department of Obstetrics and Gynecology, School of Medicine, Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Izanloo
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Sadat Najib
- Department of Obstetrics and Gynecology, School of Medicine, Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
30
|
Robba C, Battaglini D, Ball L, Valbusa A, Porto I, Della Bona R, La Malfa G, Patroniti N, Brunetti I, Loconte M, Bassetti M, Giacobbe DR, Vena A, Silva CLM, Rocco PRM, Pelosi P. Coagulative Disorders in Critically Ill COVID-19 Patients with Acute Distress Respiratory Syndrome: A Critical Review. J Clin Med 2021; 10:E140. [PMID: 33401632 PMCID: PMC7795033 DOI: 10.3390/jcm10010140] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
In critically ill patients with acute respiratory distress syndrome (ARDS) coronavirus disease 2019 (COVID-19), a high incidence of thromboembolic and hemorrhagic events is reported. COVID-19 may lead to impairment of the coagulation cascade, with an imbalance in platelet function and the regulatory mechanisms of coagulation and fibrinolysis. Clinical manifestations vary from a rise in laboratory markers and subclinical microthrombi to thromboembolic events, bleeding, and disseminated intravascular coagulation. After an inflammatory trigger, the mechanism for activation of the coagulation cascade in COVID-19 is the tissue factor pathway, which causes endotoxin and tumor necrosis factor-mediated production of interleukins and platelet activation. The consequent massive infiltration of activated platelets may be responsible for inflammatory infiltrates in the endothelial space, as well as thrombocytopenia. The variety of clinical presentations of the coagulopathy confronts the clinician with the difficult questions of whether and how to provide optimal supportive care. In addition to coagulation tests, advanced laboratory tests such as protein C, protein S, antithrombin, tissue factor pathway inhibitors, D-dimers, activated factor Xa, and quantification of specific coagulation factors can be useful, as can thromboelastography or thromboelastometry. Treatment should be tailored, focusing on the estimated risk of bleeding and thrombosis. The aim of this review is to explore the pathophysiology and clinical evidence of coagulation disorders in severe ARDS-related COVID-19 patients.
Collapse
Affiliation(s)
- Chiara Robba
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Denise Battaglini
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
| | - Lorenzo Ball
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Alberto Valbusa
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
| | - Italo Porto
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Roberta Della Bona
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
| | - Giovanni La Malfa
- Dipartimento CardioToracoVascolare, Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy; (A.V.); (I.P.); (R.D.B.); (G.L.M.)
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Nicolò Patroniti
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Iole Brunetti
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
| | - Maurizio Loconte
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
| | - Matteo Bassetti
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, 16132 Genoa, Italy; (M.B.); (D.R.G.); (A.V.)
- Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy
| | - Daniele R. Giacobbe
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, 16132 Genoa, Italy; (M.B.); (D.R.G.); (A.V.)
| | - Antonio Vena
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, 16132 Genoa, Italy; (M.B.); (D.R.G.); (A.V.)
| | - Claudia Lucia M. Silva
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil;
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil;
| | - Paolo Pelosi
- Department of Anesthesia and Intensive Care, Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genoa, Italy; (D.B.); (L.B.); (N.P.); (I.B.); (M.L.); (P.P.)
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
31
|
Baker SK, Strickland S. A critical role for plasminogen in inflammation. J Exp Med 2020; 217:133866. [PMID: 32159743 PMCID: PMC7144526 DOI: 10.1084/jem.20191865] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/10/2019] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Plasminogen and its active form, plasmin, have diverse functions related to the inflammatory response in mammals. Due to these roles in inflammation, plasminogen has been implicated in the progression of a wide range of diseases with an inflammatory component. In this review, we discuss the functions of plasminogen in inflammatory regulation and how this system plays a role in the pathogenesis of diseases spanning organ systems throughout the body.
Collapse
Affiliation(s)
- Sarah K Baker
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY
| |
Collapse
|
32
|
Chen SM, Zou Z, Guo SY, Hou WT, Qiu XR, Zhang Y, Song LJ, Hu XY, Jiang YY, Shen H, An MM. Preventing Candida albicans from subverting host plasminogen for invasive infection treatment. Emerg Microbes Infect 2020; 9:2417-2432. [PMID: 33115324 PMCID: PMC7646593 DOI: 10.1080/22221751.2020.1840927] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Candida albicans is a common fungal pathogen in humans that colonizes the skin and mucosal surfaces of the majority healthy individuals. How C. albicans disseminates into the bloodstream and causes life-threatening systemic infections in immunocompromised patients remains unclear. Plasminogen system activation can degrade a variety of structural proteins in vivo and is involved in several homeostatic processes. Here, for the first time, we characterized that C. albicans could capture and “subvert” host plasminogen to invade host epithelial cell surface barriers through cell-wall localized Eno1 protein. We found that the “subverted” plasminogen system plays an important role in development of invasive infection caused by C. albicans in mice. Base on this finding, we discovered a mouse monoclonal antibody (mAb) 12D9 targeting C. albicans Eno1, with high affinity to the 254FYKDGKYDL262 motif in α-helices 6, β-sheet 6 (H6S6) loop and direct blocking activity for C. albicans capture host plasminogen. mAb 12D9 could prevent C. albicans from invading human epithelial and endothelial cells, and displayed antifungal activity and synergistic effect with anidulafungin or fluconazole in proof-of-concept in vivo studies, suggesting that blocking the function of cell surface Eno1 was effective for controlling invasive infection caused by Candida spp. In summary, our study provides the evidence of C. albicans invading host by “subverting” plasminogen system, suggesting a potential novel treatment strategy for invasive fungal infections.
Collapse
Affiliation(s)
- Si-Min Chen
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Zui Zou
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Shi-Yu Guo
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Wei-Tong Hou
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xi-Ran Qiu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yu Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Li-Jun Song
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Xin-Yu Hu
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yuan-Ying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hui Shen
- Department of Laboratory Diagnosis, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Mao-Mao An
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
33
|
José RJ, Williams A, Manuel A, Brown JS, Chambers RC. Targeting coagulation activation in severe COVID-19 pneumonia: lessons from bacterial pneumonia and sepsis. Eur Respir Rev 2020; 29:29/157/200240. [PMID: 33004529 PMCID: PMC7537941 DOI: 10.1183/16000617.0240-2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 08/20/2020] [Indexed: 12/15/2022] Open
Abstract
Novel coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), has rapidly spread throughout the world, resulting in a pandemic with high mortality. There are no effective treatments for the management of severe COVID-19 and current therapeutic trials are focused on antiviral therapy and attenuation of hyper-inflammation with anti-cytokine therapy. Severe COVID-19 pneumonia shares some pathological similarities with severe bacterial pneumonia and sepsis. In particular, it disrupts the haemostatic balance, which results in a procoagulant state locally in the lungs and systemically. This culminates in the formation of microthrombi, disseminated intravascular coagulation and multi-organ failure. The deleterious effects of exaggerated inflammatory responses and activation of coagulation have been investigated in bacterial pneumonia and sepsis and there is recognition that although these pathways are important for the host immune response to pathogens, they can lead to bystander tissue injury and are negatively associated with survival. In the past two decades, evidence from preclinical studies has led to the emergence of potential anticoagulant therapeutic strategies for the treatment of patients with pneumonia, sepsis and acute respiratory distress syndrome, and some of these anticoagulant approaches have been trialled in humans. Here, we review the evidence from preclinical studies and clinical trials of anticoagulant treatment strategies in bacterial pneumonia and sepsis, and discuss the importance of these findings in the context of COVID-19.
Collapse
Affiliation(s)
- Ricardo J José
- Centre for Inflammation and Tissue Repair, University College London, London, UK .,Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Andrew Williams
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Ari Manuel
- University Hospital Aintree, Liverpool, UK
| | - Jeremy S Brown
- Centre for Inflammation and Tissue Repair, University College London, London, UK.,Dept of Thoracic Medicine, University College London Hospital, London, UK
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| |
Collapse
|
34
|
Abstract
An ongoing global pandemic of viral pneumonia (coronavirus disease [COVID-19]), due to the virus SARS-CoV-2, has infected millions of people and remains a threat to many more. Most critically ill patients have respiratory failure and there is an international effort to understand mechanisms and predictors of disease severity. Coagulopathy, characterized by elevations in D-dimer and fibrin(ogen) degradation products (FDPs), is associated with critical illness and mortality in patients with COVID-19. Furthermore, increasing reports of microvascular and macrovascular thrombi suggest that hemostatic imbalances may contribute to the pathophysiology of SARS-CoV-2 infection. We review the laboratory and clinical findings of patients with COVID-19-associated coagulopathy, and prior studies of hemostasis in other viral infections and acute respiratory distress syndrome. We hypothesize that an imbalance between coagulation and inflammation may result in a hypercoagulable state. Although thrombosis initiated by the innate immune system is hypothesized to limit SARS-CoV-2 dissemination, aberrant activation of this system can cause endothelial injury resulting in loss of thromboprotective mechanisms, excess thrombin generation, and dysregulation of fibrinolysis and thrombosis. The role various components including neutrophils, neutrophil extracellular traps, activated platelets, microparticles, clotting factors, inflammatory cytokines, and complement play in this process remains an area of active investigation and ongoing clinical trials target these different pathways in COVID-19.
Collapse
Affiliation(s)
- Meaghan E Colling
- Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, and Ann Arbor Veterans Administration Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
35
|
Elrashdy F, Redwan EM, Uversky VN. Why COVID-19 Transmission Is More Efficient and Aggressive Than Viral Transmission in Previous Coronavirus Epidemics? Biomolecules 2020; 10:E1312. [PMID: 32933047 PMCID: PMC7565143 DOI: 10.3390/biom10091312] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causing a pandemic of coronavirus disease 2019 (COVID-19). The worldwide transmission of COVID-19 from human to human is spreading like wildfire, affecting almost every country in the world. In the past 100 years, the globe did not face a microbial pandemic similar in scale to COVID-19. Taken together, both previous outbreaks of other members of the coronavirus family (severe acute respiratory syndrome (SARS-CoV) and middle east respiratory syndrome (MERS-CoV)) did not produce even 1% of the global harm already inflicted by COVID-19. There are also four other CoVs capable of infecting humans (HCoVs), which circulate continuously in the human population, but their phenotypes are generally mild, and these HCoVs received relatively little attention. These dramatic differences between infection with HCoVs, SARS-CoV, MERS-CoV, and SARS-CoV-2 raise many questions, such as: Why is COVID-19 transmitted so quickly? Is it due to some specific features of the viral structure? Are there some specific human (host) factors? Are there some environmental factors? The aim of this review is to collect and concisely summarize the possible and logical answers to these questions.
Collapse
Affiliation(s)
- Fatma Elrashdy
- Department of Endemic Medicine and Hepatogastroenterology, Kasr Alainy School of Medicine, Cairo University, Cairo 11562, Egypt;
| | - Elrashdy M. Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Vladimir N. Uversky
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, 142290 Moscow, Russia
| |
Collapse
|
36
|
Allegra A, Innao V, Allegra AG, Musolino C. Coagulopathy and thromboembolic events in patients with SARS-CoV-2 infection: pathogenesis and management strategies. Ann Hematol 2020; 99:1953-1965. [PMID: 32671455 PMCID: PMC7363407 DOI: 10.1007/s00277-020-04182-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
In October 2019, a viral infectious disease appeared in the city of Wuhan in China. A new betacoronavirus, SARS-CoV-2, has been recognized as the responsible pathogen in this infection. Although coronavirus disease is principally expressed as a pulmonary infection, critical SARS-CoV-2 infection is frequently complicated with coagulopathy, and thromboembolic events are recognizable in several patients. Dehydration, acute inflammatory condition, protracted immobilization during disease, existence of multiple cardiovascular risk factors such as diabetes, obesity or hypertension, previous coronary artery disease, ischemic stroke, peripheral artery disease are frequent comorbidities in SARS-CoV-2 hospitalized subjects, which possibly augment thrombo-embolic risk. However, other causal factors can still be identified such as unrestricted angiotensin II action, the use of immunoglobulins, an increased production of adhesion molecules able to induce vascular inflammation and endothelial activation, complement stimulation, excessive production of neutrophil extracellular traps (NETs), and increased platelet count. Low-molecular-weight heparin should be chosen as early treatment because of its anti-inflammatory action and its ability to antagonize histones and so defend the endothelium. However, several therapeutic possibilities have also been proposed such as fibrinolytic treatment, drugs that target NETs, and complement inhibition. Nevertheless, although the violence of the pandemic may suggest the use of heroic treatments to reduce the frightening mortality that accompanies SARS-CoV-2 infection, we believe that experimental treatments should only be used within approved and controlled protocols, the only ones that can provide useful and specify information on the validity of the treatments.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy.
- COVID Centre AOU Policlinic G. Martino, Messina, Italy.
| | - Vanessa Innao
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy
| | - Andrea Gaetano Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy
| | - Caterina Musolino
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy
| |
Collapse
|
37
|
Medcalf RL, Keragala CB, Myles PS. Fibrinolysis and COVID-19: A plasmin paradox. J Thromb Haemost 2020; 18:2118-2122. [PMID: 32543119 PMCID: PMC7323332 DOI: 10.1111/jth.14960] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/24/2022]
Abstract
The COVID-19 pandemic has provided many challenges in the field of thrombosis and hemostasis. Among these is a novel form of coagulopathy that includes exceptionally high levels of D-dimer. D-dimer is a marker of poor prognosis, but does this also imply a causal relationship? These spectacularly raised D-dimer levels may actually signify the failing attempt of the fibrinolytic system to remove fibrin and necrotic tissue from the lung parenchyma, being consumed or overwhelmed in the process. Indeed, recent studies suggest that increasing fibrinolytic activity might offer hope for patients with critical disease and severe respiratory failure. However, the fibrinolytic system can also be harnessed by coronavirus to promote infectivity and where antifibrinolytic measures would also seem appropriate. Hence, there is a clinical paradox where plasmin formation can be either deleterious or beneficial in COVID-19, but not at the same time. Hence, it all comes down to timing.
Collapse
Affiliation(s)
- Robert L Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Charithani B Keragala
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, Alfred Hospital, Melbourne, Victoria, Australia
- Department of Anaesthesiology and Perioperative Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Coagulation dysfunction in COVID-19: The interplay between inflammation, viral infection and the coagulation system. Blood Rev 2020; 46:100745. [PMID: 32868115 PMCID: PMC7444609 DOI: 10.1016/j.blre.2020.100745] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 is a new pandemic, caused by Severe Acute Respiratory Syndrome-CoronaVirus-2 (SARS-Cov2) infection and characterized by a broad spectrum of clinical manifestations. Inflammation and the innate immune system have been recently recognized as pivotal players in the most severe forms, characterized by significantly elevated levels of pro-inflammatory cytokines. In this setting, several studies have also reported the presence of abnormalities in coagulation parameters and platelets count, possibly identifying a subgroup of patients with poor prognosis. Some reports of full-blown thromboembolic events are emerging. Among the possible mechanisms underlying coagulation dysfunction, the so-called "cytokine storm" seems to play a pivotal role. Other candidate factors include virus-specific mechanisms, related to the virus interaction with renin angiotensin system (RAS) and the fibrinolytic pathway, but also comorbidities affecting these patients. Coagulation dysfunction is therefore a candidate risk factor for adverse outcomes in COVID-19 and should be carefully addressed in clinical practice.
Collapse
|
39
|
Dai J, Du Y, Gao J, Zhao J, Wang L, Huang Y, Xia J, Luo Y, Li S, McNeil EB. Difference in Biomarkers Between COVID-19 Patients and Other Pulmonary Infection Patients. Infect Drug Resist 2020; 13:2609-2615. [PMID: 32801798 PMCID: PMC7397211 DOI: 10.2147/idr.s257936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/09/2020] [Indexed: 01/08/2023] Open
Abstract
Background The pandemic due to the novel coronavirus disease 2019 (COVID-19) has resulted in an increasing number of patients need to be tested. We aimed to determine if the use of integrated laboratory data can discriminate COVID-19 patients from other pulmonary infection patients. Methods This retrospective cohort study was conducted at Kunming Third People’s Hospital in China from January 20 to February 28, 2020. Medical records and laboratory data were extracted and combined for COVID-19 and other pulmonary infection patients on admission. A partial least square discriminant analysis (PLS-DA) model was constructed and calibrated to discriminate COVID-19 from other pulmonary infection patients. Results COVID-19 patients diagnosed and treated in Kunming were balanced in terms of sex and covered all age groups. Most of them were mild cases; only five were severe cases. The first two dimensions of the PLS-DA model could classify COVID-19 and other pulmonary infection patients with an accuracy of 96.6% (95.1% in the cross-validation model). Basophil count, the proportion of basophils, prothrombin time, prothrombin time activity, and international normalized ratio were the five most discriminant biomarkers. Conclusion Integration of biomarkers can discriminate COVID-19 patients from other pulmonary infections on admission to hospital and thus may be a supplement to nucleic acid tests.
Collapse
Affiliation(s)
- Jingyi Dai
- Department of Infectious Diseases, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Yingrong Du
- Department of Infectious Diseases, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Jianpeng Gao
- Department of Infectious Diseases, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Jun Zhao
- School of Public Health and Management, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Lin Wang
- Department of Clinical Laboratory, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Ying Huang
- Department of Infectious Diseases, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Jiawei Xia
- Department of Critical Care Medicine, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Yu Luo
- Department of Infectious Diseases, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Shenghao Li
- Department of Critical Care Medicine, Kunming Third People's Hospital, Kunming, Yunnan, People's Republic of China
| | - Edward B McNeil
- Epidemiology Unit, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
40
|
Baeza C, González A, Torres P, Pizzamiglio M, Arribas A, Aparicio C. Acute aortic thrombosis in COVID-19. JOURNAL OF VASCULAR SURGERY CASES INNOVATIONS AND TECHNIQUES 2020; 6:483-486. [PMID: 32838094 PMCID: PMC7366084 DOI: 10.1016/j.jvscit.2020.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/24/2020] [Indexed: 01/16/2023]
Abstract
Acute aortic occlusion is an infrequent disease but with significant mortality. The new pandemic of the SARS-CoV-2 coronavirus disease (COVID-19) represents a great challenge for health systems. This contagious disease is generating high infection and mortality rates in several countries. It is speculated that the inflammatory process accompanying the infection is triggered by massive macrophage activation and is associated with the development of coagulopathy. We present three cases of COVID-19 patients, treated in our hospital during a period of 2 weeks, who presented with an acute thrombosis of the infrarenal abdominal aorta.
Collapse
Affiliation(s)
- Ciro Baeza
- Department of Vascular Surgery, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Alejandro González
- Department of Vascular Surgery, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Patricia Torres
- Department of Vascular Surgery, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Mateo Pizzamiglio
- Department of Vascular Surgery, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Ana Arribas
- Department of Vascular Surgery, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - César Aparicio
- Department of Vascular Surgery, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| |
Collapse
|
41
|
Ji HL, Zhao R, Matalon S, Matthay MA. Elevated Plasmin(ogen) as a Common Risk Factor for COVID-19 Susceptibility. Physiol Rev 2020; 100:1065-1075. [PMID: 32216698 PMCID: PMC7191627 DOI: 10.1152/physrev.00013.2020] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 01/08/2023] Open
Abstract
Patients with hypertension, diabetes, coronary heart disease, cerebrovascular illness, chronic obstructive pulmonary disease, and kidney dysfunction have worse clinical outcomes when infected with SARS-CoV-2, for unknown reasons. The purpose of this review is to summarize the evidence for the existence of elevated plasmin(ogen) in COVID-19 patients with these comorbid conditions. Plasmin, and other proteases, may cleave a newly inserted furin site in the S protein of SARS-CoV-2, extracellularly, which increases its infectivity and virulence. Hyperfibrinolysis associated with plasmin leads to elevated D-dimer in severe patients. The plasmin(ogen) system may prove a promising therapeutic target for combating COVID-19.
Collapse
Affiliation(s)
- Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas Health Science Centre at Tyler, Tyler, Texas; Texas Lung Injury Institute, University of Texas Health Science Centre at Tyler, Tyler, Texas; Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Medicine and Anesthesia, University of California San Francisco, San Francisco, California
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas Health Science Centre at Tyler, Tyler, Texas; Texas Lung Injury Institute, University of Texas Health Science Centre at Tyler, Tyler, Texas; Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Medicine and Anesthesia, University of California San Francisco, San Francisco, California
| | - Sadis Matalon
- Department of Cellular and Molecular Biology, University of Texas Health Science Centre at Tyler, Tyler, Texas; Texas Lung Injury Institute, University of Texas Health Science Centre at Tyler, Tyler, Texas; Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Medicine and Anesthesia, University of California San Francisco, San Francisco, California
| | - Michael A Matthay
- Department of Cellular and Molecular Biology, University of Texas Health Science Centre at Tyler, Tyler, Texas; Texas Lung Injury Institute, University of Texas Health Science Centre at Tyler, Tyler, Texas; Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Medicine and Anesthesia, University of California San Francisco, San Francisco, California
| |
Collapse
|
42
|
Yuksel A, Karadogan D, Gurkan CG, Akyil FT, Toreyin ZN, Marim F, Arikan H, Eyuboglu TS, Emiralioglu N, Serifoglu I, Develi E, Celik S, Sertcelik UO, Gursoy TR, Elversli MF, Oncel A, Er B, Firincioglulari A, Gunaydin FE, Ozakinci H, Ozcelik N, Esendagli D, Aydin A, Kose N, Ercelik M, Gulhan PY, Yildiz E, Irmak I, Kara BY, Gurz S, Karakas FG, Akgun M. Unclear Issues Regarding COVID-19. Eurasian J Med 2020; 52:191-196. [PMID: 32612430 PMCID: PMC7311136 DOI: 10.5152/eurasianjmed.2020.20092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 04/29/2020] [Indexed: 01/08/2023] Open
Abstract
Scientists from all over the world have been intensively working to discover different aspects of Coronavirus disease 2019 (COVID-19) since the first cluster of cases was reported in China. Herein, we aimed to investigate unclear issues related to transmission and pathogenesis of disease as well as accuracy of diagnostic tests and treatment modalities. A literature search on PubMed, Ovid, and EMBASE databases was conducted, and articles pertinent to identified search terms were extracted. A snow-ball search strategy was followed in order to retrieve additional relevant articles. It was reported that viral spread may occur during the asymptomatic phase of infection, and viral load was suggested to be a useful marker to assess disease severity. In contrast to immune response against viral infections, cytotoxic T lymphocytes decline in SARS-CoV-2 infection, which can be partially explained by direct invasion of T lymphocytes or apoptosis activated by SARS-CoV-2. Dysregulation of the urokinase pathway, cleavage of the SARS-CoV-2 Spike protein by FXa and FIIa, and consumption coagulopathy were the proposed mechanisms of the coagulation dysfunction in COVID-19. False-negative rates of reverse transcriptase polymerase chain reaction varied between 3% and 41% across studies. The probability of the positive test was proposed to decrease with the number of days past from symptom onset. Safety issues related to infection spread limit the use of high flow nasal oxygen (HFNO) and continuous positive airway pressure (CPAP) in hypoxic patients. Further studies are required to elucidate the challenging issues, thus enhancing the management of COVID-19 patients.
Collapse
Affiliation(s)
- Aycan Yuksel
- Department of Chest Diseases, Ufuk University School of Medicine, Ankara, Turkey
| | - Dilek Karadogan
- Department of Chest Diseases, Recep Tayyip Erdogan University School of Medicine, Rize, Turkey
| | - Canan Gunduz Gurkan
- Department of Chest Diseases, Sureyyapasa Chest Diseases and Thoracic Surgery Training and Research Hospital, Istanbul, Turkey
| | - Fatma Tokgoz Akyil
- Department of Chest Diseases, Yedikule Chest Diseases and Thoracic Surgery Training and Research Hospital, Istanbul, Turkey
| | - Zehra Nur Toreyin
- Department of Occupational Health and Diseases, Adana City Research and Training Hospital, Adana, Turkey
| | - Feride Marim
- Department of Chest Diseases, Kutahya University of Health Sciences School of Medicine, Kutahya, Turkey
| | - Huseyin Arikan
- Yuzuncu Yil University, Dursun Odabas Medical Center, Internal Medicine Intensive Care Unit, Van, Turkey
| | | | - Nagehan Emiralioglu
- Department of Pediatric Pulmonology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Irem Serifoglu
- Department of Chest Diseases Kirikhan State Hospital, Hatay, Turkey
| | - Elif Develi
- Department of Physiotherapy and Rehabilitation, Yeditepe University Faculty of Health Sciences, Istanbul, Turkey
| | - Selman Celik
- Department of Nursing and Health Services, Yeditepe University Faculty of Health Sciences, Istanbul, Turkey
| | - Umran Ozden Sertcelik
- Department of Chest Diseases, Hacettepe University School of Medicine, Ankara, Turkey
| | - Tugba Ramasli Gursoy
- Department of Pediatric Pulmonology, Gazi University School of Medicine, Ankara, Turkey
| | | | - Asli Oncel
- Department of Chest Diseases, Hacettepe University School of Medicine, Ankara, Turkey
| | - Berrin Er
- Internal Medicine Intensive Care Unit, Hacettepe University School of Medicine, Ankara, Turkey
| | - Ali Firincioglulari
- Department of Chest Diseases, University of Health Sciences, Ankara Ataturk Chest Diseases and Thoracic Surgery Training and Research Hospital, Ankara, Turkey
| | - Fatma Esra Gunaydin
- Department of Allergy and Immunology, Uludag University School of Medicine, Bursa, Turkey
| | - Hilal Ozakinci
- Department of Pathology, Ankara University School of Medicine, Ankara, Turkey
| | - Neslihan Ozcelik
- Department of Chest Diseases, Recep Tayyip Erdogan University School of Medicine, Rize, Turkey
| | - Dorina Esendagli
- Department of Chest Diseases, Baskent University School of Medicine, Ankara, Turkey
| | - Asena Aydin
- Department of Chest Diseases, Kestel State Hospital, Bursa, Turkey
| | - Neslihan Kose
- Department of Chest Diseases, Bilecik State Hospital, Bilecik, Turkey
| | - Merve Ercelik
- Department of Chest Diseases, Duzce University School of Medicine, Düzce, Turkey
| | - Pinar Yildiz Gulhan
- Department of Chest Diseases, Duzce University School of Medicine, Düzce, Turkey
| | - Ethem Yildiz
- Department of Chest Diseases, Bingol State Hopital, Bingöl, Turkey
| | - Ilim Irmak
- Department of Chest Diseases, Hacettepe University School of Medicine, Ankara, Turkey
| | - Bilge Yilmaz Kara
- Department of Chest Diseases, Recep Tayyip Erdogan University School of Medicine, Rize, Turkey
| | - Selcuk Gurz
- Department of Thoracic Surgery, Ondokuz Mayis University School of Medicine, Samsun, Turkey
| | - Fatma Gulsum Karakas
- Department of Chest Diseases, Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Metin Akgun
- Department of Chest Diseases, Ataturk University School of Medicine, Erzurum, Turkey
| |
Collapse
|
43
|
Xiong M, Liang X, Wei YD. Changes in blood coagulation in patients with severe coronavirus disease 2019 (COVID-19): a meta-analysis. Br J Haematol 2020; 189:1050-1052. [PMID: 32304581 PMCID: PMC7264726 DOI: 10.1111/bjh.16725] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mi Xiong
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xue Liang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - You-Dong Wei
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China
| |
Collapse
|
44
|
Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost 2020. [PMID: 32073213 DOI: 10.1111/jth.v18.410.1111/jth.14768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND In the recent outbreak of novel coronavirus infection in Wuhan, China, significantly abnormal coagulation parameters in severe novel coronavirus pneumonia (NCP) cases were a concern. OBJECTIVES To describe the coagulation feature of patients with NCP. METHODS Conventional coagulation results and outcomes of 183 consecutive patients with confirmed NCP in Tongji hospital were retrospectively analyzed. RESULTS The overall mortality was 11.5%, the non-survivors revealed significantly higher D-dimer and fibrin degradation product (FDP) levels, longer prothrombin time and activated partial thromboplastin time compared to survivors on admission (P < .05); 71.4% of non-survivors and 0.6% survivors met the criteria of disseminated intravascular coagulation during their hospital stay. CONCLUSIONS The present study shows that abnormal coagulation results, especially markedly elevated D-dimer and FDP are common in deaths with NCP.
Collapse
Affiliation(s)
- Ning Tang
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dengju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Wang
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyong Sun
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost 2020; 18:844-847. [PMID: 32073213 PMCID: PMC7166509 DOI: 10.1111/jth.14768] [Citation(s) in RCA: 4025] [Impact Index Per Article: 805.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/18/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND In the recent outbreak of novel coronavirus infection in Wuhan, China, significantly abnormal coagulation parameters in severe novel coronavirus pneumonia (NCP) cases were a concern. OBJECTIVES To describe the coagulation feature of patients with NCP. METHODS Conventional coagulation results and outcomes of 183 consecutive patients with confirmed NCP in Tongji hospital were retrospectively analyzed. RESULTS The overall mortality was 11.5%, the non-survivors revealed significantly higher D-dimer and fibrin degradation product (FDP) levels, longer prothrombin time and activated partial thromboplastin time compared to survivors on admission (P < .05); 71.4% of non-survivors and 0.6% survivors met the criteria of disseminated intravascular coagulation during their hospital stay. CONCLUSIONS The present study shows that abnormal coagulation results, especially markedly elevated D-dimer and FDP are common in deaths with NCP.
Collapse
Affiliation(s)
- Ning Tang
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dengju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Wang
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyong Sun
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Mukund K, Mathee K, Subramaniam S. Plasmin Cascade Mediates Thrombotic Events in SARS-CoV-2 Infection via Complement and Platelet-Activating Systems. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2020; 1:220-227. [PMID: 34786557 PMCID: PMC8527892 DOI: 10.1109/ojemb.2020.3014798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 11/11/2022] Open
Abstract
Objective: Recently emerged beta-coronavirus SARS-CoV-2, has resulted in the current pandemic designated COVID-19. COVID-19 manifests as severe illness exhibiting systemic inflammatory response syndrome, acute respiratory distress syndrome (ARDS), thrombotic events, and shock, exacerbated further by co-morbidities and age. Recent clinical evidence suggests that the development of ARDS and subsequent pulmonary failure result from a complex interplay between cell types (endothelial, epithelial and immune) within the lung promoting inflammatory infiltration and a pro-coagulative state. How the complex molecular events mediated by SARS-CoV-2 in infected lung epithelial cells lead to thrombosis and pulmonary failure, is yet to be fully understood. Methods: We address these questions here, using publicly available transcriptomic data in the context of lung epithelia affected by SARS-CoV-2 and other respiratory infections, in vitro. We then extend our results to the understanding of in vivo lung, using a publicly available COVID-19 lung transcriptomic study. Results and Conclusions: Our analysis indicates that there exists a complex interplay between the fibrinolytic system particularly plasmin, and the complement and platelet-activating systems upon SARS-CoV-2 infection, with a potential for therapeutic intervention.
Collapse
Affiliation(s)
- Kavitha Mukund
- 1 Department of BioengineeringUniversity of California San Diego La Jolla CA 92093 USA
| | - Kalai Mathee
- 2 Department of Human and Molecular GeneticsHerbert Wertheim College of Medicine Miami FL 33199 USA
- 3 Biomolecular Sciences InstituteFlorida International University Miami FL 33199 USA
| | - Shankar Subramaniam
- 1 Department of BioengineeringUniversity of California San Diego La Jolla CA 92093 USA
- 4 Department of Cellular and Molecular MedicineUniversity of California San Diego La Jolla CA 92093 USA
- 5 Department of Computer Science and EngineeringUniversity of California San Diego La Jolla CA 92093 USA
| |
Collapse
|
47
|
Mitchell HD, Eisfeld AJ, Stratton KG, Heller NC, Bramer LM, Wen J, McDermott JE, Gralinski LE, Sims AC, Le MQ, Baric RS, Kawaoka Y, Waters KM. The Role of EGFR in Influenza Pathogenicity: Multiple Network-Based Approaches to Identify a Key Regulator of Non-lethal Infections. Front Cell Dev Biol 2019; 7:200. [PMID: 31616667 PMCID: PMC6763731 DOI: 10.3389/fcell.2019.00200] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022] Open
Abstract
Despite high sequence similarity between pandemic and seasonal influenza viruses, there is extreme variation in host pathogenicity from one viral strain to the next. Identifying the underlying mechanisms of variability in pathogenicity is a critical task for understanding influenza virus infection and effective management of highly pathogenic influenza virus disease. We applied a network-based modeling approach to identify critical functions related to influenza virus pathogenicity using large transcriptomic and proteomic datasets from mice infected with six influenza virus strains or mutants. Our analysis revealed two pathogenicity-related gene expression clusters; these results were corroborated by matching proteomics data. We also identified parallel downstream processes that were altered during influenza pathogenesis. We found that network bottlenecks (nodes that bridge different network regions) were highly enriched in pathogenicity-related genes, while network hubs (highly connected network nodes) were significantly depleted in these genes. We confirmed that this trend persisted in a distinct virus: Severe Acute Respiratory Syndrome Coronavirus (SARS). The role of epidermal growth factor receptor (EGFR) in influenza pathogenesis, one of the bottleneck regulators with corroborating signals across transcript and protein expression data, was tested and validated in additional mouse infection experiments. We demonstrate that EGFR is important during influenza infection, but the role it plays changes for lethal versus non-lethal infections. Our results show that by using association networks, bottleneck genes that lack hub characteristics can be used to predict a gene's involvement in influenza virus pathogenicity. We also demonstrate the utility of employing multiple network approaches for analyzing host response data from viral infections.
Collapse
Affiliation(s)
- Hugh D Mitchell
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Amie J Eisfeld
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Kelly G Stratton
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Natalie C Heller
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Lisa M Bramer
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Ji Wen
- Pacific Northwest National Laboratory, Richland, WA, United States
| | | | - Lisa E Gralinski
- Department of Microbiology and Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Amy C Sims
- Department of Microbiology and Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Mai Q Le
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Ralph S Baric
- Department of Microbiology and Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States.,Division of Virology, Department of Microbiology and Immunology, Institute of Medical Sciences, The University of Tokyo, Tokyo, Japan.,International Research Center for Infectious Diseases, Institute of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Katrina M Waters
- Pacific Northwest National Laboratory, Richland, WA, United States
| |
Collapse
|
48
|
Antoniak S. The coagulation system in host defense. Res Pract Thromb Haemost 2018; 2:549-557. [PMID: 30046760 PMCID: PMC6046589 DOI: 10.1002/rth2.12109] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The blood coagulation system and immune system of higher organisms are thought to have a common ancestral origin. During infections, the blood coagulation system is activated and components of the hemostatic system are directly involved in the immune response and immune system modulations. The current view is that the activation of coagulation is beneficial for infections with bacteria and viruses. It limits pathogen dissemination and supports pathogen killing and tissue repair. On the other hand, over-activation can lead to thrombosis with subsequent depletion of hemostatic factors and secondary bleeding. This review will summarize the current knowledge on blood coagulation and pathogen infection with focus on most recent studies of the role of the different parts of the blood coagulation system in selected bacterial and viral infections.
Collapse
Affiliation(s)
- Silvio Antoniak
- Program in Thrombosis and HemostasisDepartment of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| |
Collapse
|
49
|
Mamoun J. Dry Socket Etiology, Diagnosis, and Clinical Treatment Techniques. J Korean Assoc Oral Maxillofac Surg 2018; 44:52-58. [PMID: 29732309 PMCID: PMC5932271 DOI: 10.5125/jkaoms.2018.44.2.52] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/28/2017] [Accepted: 06/10/2017] [Indexed: 01/30/2023] Open
Abstract
Dry socket, also termed fibrinolytic osteitis or alveolar osteitis, is a complication of tooth exodontia. A dry socket lesion is a post-extraction socket that exhibits exposed bone that is not covered by a blood clot or healing epithelium and exists inside or around the perimeter of the socket or alveolus for days after the extraction procedure. This article describes dry socket lesions; reviews the basic clinical techniques of treating different manifestations of dry socket lesions; and shows how microscope level loupe magnification of 6× to 8× or greater, combined with co-axial illumination or a dental operating microscope, facilitate more precise treatment of dry socket lesions. The author examines the scientific validity of the proposed causes of dry socket lesions (such as bacteria, inflammation, fibrinolysis, or traumatic extractions) and the scientific validity of different terminologies used to describe dry socket lesions. This article also presents an alternative model of what causes dry socket lesions, based on evidence from dental literature. Although the clinical techniques for treating dry socket lesions seem empirically correct, more evidence is required to determine the causes of dry socket lesions.
Collapse
|
50
|
Lê VB, Riteau B, Alessi MC, Couture C, Jandrot-Perrus M, Rhéaume C, Hamelin MÈ, Boivin G. Protease-activated receptor 1 inhibition protects mice against thrombin-dependent respiratory syncytial virus and human metapneumovirus infections. Br J Pharmacol 2017; 175:388-403. [PMID: 29105740 DOI: 10.1111/bph.14084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Protease-activated receptor 1 (PAR1) has been demonstrated to be involved in the pathogenesis of viral diseases. However, its role remains controversial. The goal of our study was to investigate the contribution of PAR1 to respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) infections. EXPERIMENTAL APPROACH Pharmacological approaches were used to investigate the role of PAR1 during RSV and hMPV infection, in vitro using epithelial A549 cells and in vivo using a mouse model of virus infection. KEY RESULTS In vitro, the PAR1 antagonist RWJ-56110 reduced the replication of RSV and hMPV in A549 cells. In agreement with these results, RWJ-56110-treated mice were protected against RSV and hMPV infections, as indicated by less weight loss and mortality. This protective effect in mice correlated with decreased lung viral replication and inflammation. In contrast, hMPV-infected mice treated with the PAR1 agonist TFLLR-NH2 showed increased mortality, as compared to infected mice, which were left untreated. Thrombin generation was shown to occur downstream of PAR1 activation in infected mice via tissue factor exposure as part of the inflammatory response, and thrombin inhibition by argatroban reduced the pathogenicity of the infection with no additive effect to that induced by PAR1 inhibition. CONCLUSION AND IMPLICATIONS These data show that PAR1 plays a detrimental role during RSV and hMPV infections in mice via, at least, a thrombin-dependent mechanism. Thus, the use of PAR1 antagonists and thrombin inhibitors may have potential as a novel approach for the treatment of RSV and hMPV infections.
Collapse
Affiliation(s)
- Vuong Ba Lê
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Béatrice Riteau
- UMR INSERM U1062/INRA 1260/AMU, Aix Marseille University, Marseille, France
| | | | - Christian Couture
- Department of Anatomy-Pathology, Laval University Institute of Cardiology and Pneumology, Quebec City, Quebec, Canada
| | | | - Chantal Rhéaume
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Marie-Ève Hamelin
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Guy Boivin
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|