1
|
Gray A, Ou R, Djabourian R, Augustine JD, Sathyavagiswaran L. Deaths from Angioedema-Anaphylaxis and Covid-19. Acad Forensic Pathol 2025:19253621251320212. [PMID: 39991143 PMCID: PMC11843567 DOI: 10.1177/19253621251320212] [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: 10/15/2024] [Accepted: 01/08/2025] [Indexed: 02/25/2025]
Abstract
Angioedema is non-dependent, non-pitting edema at a variety of sites. Its forms can be divided into histamine-mediated and bradykinin-mediated types. Histamine-mediated forms can present similarly to anaphylaxis with urticaria and itching, while bradykinin-mediated angioedema is slower in onset, presents with greater facial oropharyngeal involvement, and has a higher risk of progression. Bradykinin-mediated angioedema is of particular importance as it will not respond to the usual medications used in histamine-mediated anaphylaxis (steroids, antihistamines, epinephrine, etc). In this report, we present two cases of angioedema-induced death with coexisting COVID-19 infections in two male decedents. The associated gross autopsy, histology, and laboratory values will be discussed in detail. This article will highlight the importance of early detection and adequate treatment for the type of angioedema being evaluated as well as a discussion regarding the relationship between COVID-19 and angioedema.
Collapse
Affiliation(s)
- Austin Gray
- Austin Gray, Los Angeles County Department of Medical Examiner, 1104 North Mission Road, Los Angeles, CA 90033, USA,
| | | | | | | | | |
Collapse
|
2
|
Li R, Zhang J, Ren L. A Meta-Analysis of the Impact of Using Angiotensin-Converting Enzyme Inhibitors (ACEIs) or Angiotensin II Receptor Blockers (ARBs) on Mortality, Severity, and Healthcare Resource Utilization in Patients with COVID-19. Adv Respir Med 2025; 93:4. [PMID: 39996621 PMCID: PMC11852372 DOI: 10.3390/arm93010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE The primary objective of this study is to explore the potential link between the utilization of angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin II receptor blockers (ARBs) and its impact on mortality, disease severity, and healthcare resource utilization in individuals diagnosed with COVID-19. We aim to establish a solid theoretical foundation for safe and effective clinical medications. METHODS We conducted a comprehensive search of various databases, including CNKI, PubMed, Science, Cell, Springer, Nature, Web of Science, and Embase. We also traced the literature of the included studies to ensure a thorough analysis of the available evidence. After applying a set of inclusion and exclusion criteria, we ultimately included a total of 41 articles in our analysis. To determine the overall effect size for dichotomous variables, we used the Mantel-Haenszel odds ratio in random effect models. For continuous variables, we calculated the inverse variance SMD using random effect models. To assess the outcomes and heterogeneity, we considered p-values (p < 0.05) and I2 values for all outcomes. We performed multivariate and univariate meta-regression analyses using the maximum likelihood approach with the CMA 3.0 software. RESULTS The results of our analysis indicated that the use of ACEIs or ARBs did not significantly influence mortality (OR = 1.10, 95% CI 0.83-1.46, p = 0.43, I2 = 84%), severity (OR = 0.99, 95% CI 0.68-1.45, p = 0.98, I2 = 84%), or healthcare resource utilization (SMD = 0.03, 95% CI 0.06-0.12, p = 0.54, I2 = 37%) in patients with COVID-19 compared to those not taking ACEIs or ARBs. The multivariate meta-regression analysis model explained 63%, 31%, and 100% of the sources of heterogeneity for the three outcome indicators. CONCLUSIONS The use of ACEIs and ARBs is not significantly correlated with mortality, severity, or healthcare resource utilization in patients with COVID-19, indicating safe clinical use of the medications.
Collapse
Affiliation(s)
| | | | - Liang Ren
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430032, China; (R.L.)
| |
Collapse
|
3
|
Berra S, Parolin D, Suffritti C, Folcia A, Zanichelli A, Gusso L, Cogliati C, Riva A, Gidaro A, Caccia S. Patterns of C1-Inhibitor Plasma Levels and Kinin-Kallikrein System Activation in Relation to COVID-19 Severity. Life (Basel) 2024; 14:1525. [PMID: 39768234 PMCID: PMC11679851 DOI: 10.3390/life14121525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Although more than four years have passed since the pandemic began, SARS-CoV-2 continues to be of concern. Therefore, research into the underlying mechanisms that contribute to the development of the disease, especially in more severe forms, remains a priority. Sustained activation of the complement (CS), contact (CAS), and fibrinolytic and kinin-kallikrein systems (KKS) has been shown to play a central role in the pathogenesis of the disease. Since the C1 esterase inhibitor (C1-INH) is a potent inhibitor of all these systems, its role in the disease has been investigated, but some issues remained unresolved. METHODS We evaluated the impact of C1-INH and KKS on disease progression in a cohort of 45 COVID-19 patients divided into groups according to disease severity. We measured plasma levels of total and functional C1-INH and its complexes with kallikrein (PKa), reflecting KKS activation and kallikrein spontaneous activity. RESULTS We observed increased total and functional plasma concentrations of C1-INH in COVID-19 patients. A direct correlation (positive Spearman's r) was observed between C1-INH levels, especially functional C1-INH, and the severity of the disease. Moreover, a significant reduction in the ratio of functional over total C1-INH was evident in patients exhibiting mild to intermediate clinical severity but not in critically ill patients. Accordingly, activation of the KKS, assessed as an increase in PKa:C1-INH complexes, was explicitly observed in the mild categories. CONCLUSIONS Our study's findings on the consumption of C1-INH and the activation of the KKS in the less severe stages of COVID-19 but not in the critical stage suggest a potential role for C1-INH in containing disease severity. These results underscore the importance of C1-INH in the early phases of the disease and its potential implications in COVID-19 progression and/or long-term effects.
Collapse
Affiliation(s)
- Silvia Berra
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Internal Medicine, Ospedale Fatebenefratelli, 20121 Milan, Italy
| | - Debora Parolin
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
| | - Chiara Suffritti
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, 20122 Milan, Italy
| | - Andrea Folcia
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Division of Oncology, Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Andrea Zanichelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
- Operative Unit of Medicine, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Luca Gusso
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Internal Medicine Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Chiara Cogliati
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Internal Medicine, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Infectious Diseases, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Antonio Gidaro
- Department of Internal Medicine, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
| |
Collapse
|
4
|
Gupta P, Dev K, Kaur G. Phytoconstituents as modulator of inflammatory pathways for COVID-19: A comprehensive review and recommendations. Phytother Res 2024; 38:5389-5416. [PMID: 39246209 DOI: 10.1002/ptr.8302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 09/10/2024]
Abstract
SARS-CoV-2 infection causes disruptions in inflammatory pathways, which fundamentally contribute to COVID-19 pathophysiology. The present review critically evaluates the gaps in scientific literature and presents the current status regarding the inflammatory signaling pathways in COVID-19. We propose that phytoconstituents can be used to treat COVID-19 associated inflammation, several already formulated in traditional medications. For this purpose, extensive literature analysis was conducted in the PubMed database to collect relevant in vitro, in vivo, and human patient studies where inflammation pathways were shown to be upregulated in COVID-19. Parallelly, scientific literature was screened for phytoconstituents with known cellular mechanisms implicated for inflammation or COVID-19 associated inflammation. Studies with insufficient evidence on cellular pathways for autophagy and mitophagy were considered out of scope and excluded from the study. The final analysis was visualized in figures and evaluated for accuracy. Our findings demonstrate the frequent participation of NF-κB, a transcription factor, in inflammatory signaling pathways linked to COVID-19. Moreover, the MAPK signaling pathway is also implicated in producing inflammatory molecules. Furthermore, it was also analyzed that the phytoconstituents with flavonoid and phenolic backbones could inhibit either the TLR4 receptor or its consecutive signaling molecules, thereby, decreasing NF-κB activity and suppressing cytokine production. Although, allopathy has treated the early phase of COVID-19, anti-inflammatory phytoconstituents and existing ayurvedic formulations may act on the COVID-19 associated inflammatory pathways and provide an additional treatment strategy. Therefore, we recommend the usage of flavonoids and phenolic phytoconstituents for the treatment of inflammation associated with COVID-19 infection and similar viral ailments.
Collapse
Affiliation(s)
- Pragati Gupta
- School of Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
| | - Kamal Dev
- School of Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
- Department of Pharmacology & Toxicology, Wright State University, Dayton, Ohio, USA
| | - Gurjot Kaur
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
- National Center cum Department of Human Genome Research Center and Studies, Panjab University, Chandigarh, Punjab, India
| |
Collapse
|
5
|
Kelleni MT. Angiotensin receptor blockers could be superior to angiotensin-converting enzyme inhibitors in COVID-19 management: the potential role of bradykinin. Ir J Med Sci 2024; 193:2077-2078. [PMID: 38530555 DOI: 10.1007/s11845-024-03679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 03/28/2024]
Affiliation(s)
- Mina T Kelleni
- Pharmacology Department, College of Medicine, Minia University, Minya, Egypt.
- Research Fellow INTI International University, Putra Nilai, Negeri Sembilan, Malaysia.
| |
Collapse
|
6
|
Barbosa MS, de Lima F, Peachazepi Moraes CR, Borba-Junior IT, Huber SC, Santos I, Bombassaro B, Dertkigil SSJ, Ilich A, Key NS, Annichino-Bizzacchi JM, Orsi FA, Mansour E, Velloso LA, De Paula EV. Angiopoietin2 is associated with coagulation activation and tissue factor expression in extracellular vesicles in COVID-19. Front Med (Lausanne) 2024; 11:1367544. [PMID: 38803346 PMCID: PMC11128612 DOI: 10.3389/fmed.2024.1367544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/12/2024] [Indexed: 05/29/2024] Open
Abstract
Coagulation activation in immunothrombosis involves various pathways distinct from classical hemostasis, offering potential therapeutic targets to control inflammation-induced hypercoagulability while potentially sparing hemostasis. The Angiopoietin/Tie2 pathway, previously linked to embryonic angiogenesis and sepsis-related endothelial barrier regulation, was recently associated with coagulation activation in sepsis and COVID-19. This study explores the connection between key mediators of the Angiopoietin/Tie2 pathway and coagulation activation. The study included COVID-19 patients with hypoxia and healthy controls. Blood samples were processed to obtain platelet-free plasma, and frozen until analysis. Extracellular vesicles (EVs) in plasma were characterized and quantified using flow cytometry, and their tissue factor (TF) procoagulant activity was measured using a kinetic chromogenic method. Several markers of hemostasis were assessed. Levels of ANGPT1, ANGPT2, and soluble Tie2 correlated with markers of coagulation and platelet activation. EVs from platelets and endothelial cells were increased in COVID-19 patients, and a significant increase in TF+ EVs derived from endothelial cells was observed. In addition, ANGPT2 levels were associated with TF expression and activity in EVs. In conclusion, we provide further evidence for the involvement of the Angiopoietin/Tie2 pathway in the coagulopathy of COVID-19 mediated in part by release of EVs as a potential source of TF activity.
Collapse
Affiliation(s)
- Mayck Silva Barbosa
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Franciele de Lima
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | | | | | - Stephany Cares Huber
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Irene Santos
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| | | | - Anton Ilich
- Blood Research Center, University of North Carolina, Chapel Hill, NC, United States
- Division of Hematology, Department of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Nigel S. Key
- Blood Research Center, University of North Carolina, Chapel Hill, NC, United States
- Division of Hematology, Department of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Joyce M. Annichino-Bizzacchi
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Fernanda Andrade Orsi
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Eli Mansour
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Licio A. Velloso
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| |
Collapse
|
7
|
Xiao MT, Ellsworth CR, Qin X. Emerging role of complement in COVID-19 and other respiratory virus diseases. Cell Mol Life Sci 2024; 81:94. [PMID: 38368584 PMCID: PMC10874912 DOI: 10.1007/s00018-024-05157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/03/2024] [Accepted: 02/03/2024] [Indexed: 02/19/2024]
Abstract
The complement system, a key component of innate immunity, provides the first line of defense against bacterial infection; however, the COVID-19 pandemic has revealed that it may also engender severe complications in the context of viral respiratory disease. Here, we review the mechanisms of complement activation and regulation and explore their roles in both protecting against infection and exacerbating disease. We discuss emerging evidence related to complement-targeted therapeutics in COVID-19 and compare the role of the complement in other respiratory viral diseases like influenza and respiratory syncytial virus. We review recent mechanistic studies and animal models that can be used for further investigation. Novel knockout studies are proposed to better understand the nuances of the activation of the complement system in respiratory viral diseases.
Collapse
Affiliation(s)
- Mark T Xiao
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Calder R Ellsworth
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
8
|
Coelho SVA, Augusto FM, de Arruda LB. Potential Pathways and Pathophysiological Implications of Viral Infection-Driven Activation of Kallikrein-Kinin System (KKS). Viruses 2024; 16:245. [PMID: 38400022 PMCID: PMC10892958 DOI: 10.3390/v16020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Microcirculatory and coagulation disturbances commonly occur as pathological manifestations of systemic viral infections. Research exploring the role of the kallikrein-kinin system (KKS) in flavivirus infections has recently linked microvascular dysfunctions to bradykinin (BK)-induced signaling of B2R, a G protein-coupled receptor (GPCR) constitutively expressed by endothelial cells. The relevance of KKS activation as an innate response to viral infections has gained increasing attention, particularly after the reports regarding thrombogenic events during COVID-19. BK receptor (B2R and B1R) signal transduction results in vascular permeability, edema formation, angiogenesis, and pain. Recent findings unveiling the role of KKS in viral pathogenesis include evidence of increased activation of KKS with elevated levels of BK and its metabolites in both intravascular and tissue milieu, as well as reports demonstrating that virus replication stimulates BKR expression. In this review, we will discuss the mechanisms triggered by virus replication and by virus-induced inflammatory responses that may stimulate KKS. We also explore how KKS activation and BK signaling may impact virus pathogenesis and further discuss the potential therapeutic application of BKR antagonists in the treatment of hemorrhagic and respiratory diseases.
Collapse
Affiliation(s)
- Sharton Vinícius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | | | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
9
|
Annane D, Pittock SJ, Kulkarni HS, Pickering BW, Khoshnevis MR, Siegel JL, Powell CA, Castro P, Fujii T, Dunn D, Smith K, Mitter S, Kazani S, Kulasekararaj A. Intravenous ravulizumab in mechanically ventilated patients hospitalised with severe COVID-19: a phase 3, multicentre, open-label, randomised controlled trial. THE LANCET. RESPIRATORY MEDICINE 2023; 11:1051-1063. [PMID: 36958364 PMCID: PMC10027334 DOI: 10.1016/s2213-2600(23)00082-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND The complement pathway is a potential target for the treatment of severe COVID-19. We evaluated the safety and efficacy of ravulizumab, a terminal complement C5 inhibitor, in patients hospitalised with severe COVID-19 requiring invasive or non-invasive mechanical ventilation. METHODS This phase 3, multicentre, open-label, randomised controlled trial (ALXN1210-COV-305) enrolled adult patients (aged ≥18 years) from 31 hospitals in France, Japan, Spain, the UK, and the USA. Eligible patients had a confirmed diagnosis of SARS-CoV-2 that required hospitalisation and either invasive or non-invasive mechanical ventilation, with severe pneumonia, acute lung injury, or acute respiratory distress syndrome confirmed by CT scan or x-ray. We randomly assigned participants (2:1) to receive intravenous ravulizumab plus best supportive care (BSC) or BSC alone using a web-based interactive response system. Randomisation was in permuted blocks of six with stratification by intubation status. Bodyweight-based intravenous doses of ravulizumab were administered on days 1, 5, 10, and 15. The primary efficacy endpoint was survival based on all-cause mortality at day 29 in the intention-to-treat (ITT) population. Safety endpoints were analysed in all randomly assigned patients in the ravulizumab plus BSC group who received at least one dose of ravulizumab, and in all randomly assigned patients in the BSC group. The trial is registered with ClinicalTrials.gov, NCT04369469, and was terminated at interim analysis due to futility. FINDINGS Between May 10, 2020, and Jan 13, 2021, 202 patients were enrolled in the study and randomly assigned to ravulizumab plus BSC or BSC. 201 patients were included in the ITT population (135 in the ravulizumab plus BSC group and 66 in the BSC group). The ravulizumab plus BSC group comprised 96 (71%) men and 39 (29%) women with a mean age of 63·2 years (SD 13·23); the BSC group comprised 43 (65%) men and 23 (35%) women with a mean age of 63·5 years (12·40). Most patients (113 [84%] of 135 in the ravulizumab plus BSC group and 53 [80%] of 66 in the BSC group) were on invasive mechanical ventilation at baseline. Overall survival estimates based on multiple imputation were 58% for patients receiving ravulizumab plus BSC and 60% for patients receiving BSC (Mantel-Haenszel analysis: risk difference -0·0205; 95% CI -0·1703 to 0·1293; one-sided p=0·61). In the safety population, 113 (89%) of 127 patients in the ravulizumab plus BSC group and 56 (84%) of 67 in the BSC group had a treatment-emergent adverse event. Of these events, infections and infestations (73 [57%] vs 24 [36%] patients) and vascular disorders (39 [31%] vs 12 [18%]) were observed more frequently in the ravulizumab plus BSC group than in the BSC group. Five patients had serious adverse events considered to be related to ravulizumab. These events were bacteraemia, thrombocytopenia, oesophageal haemorrhage, cryptococcal pneumonia, and pyrexia (in one patient each). INTERPRETATION Addition of ravulizumab to BSC did not improve survival or other secondary outcomes. Safety findings were consistent with the known safety profile of ravulizumab in its approved indications. Despite the lack of efficacy, the study adds value for future research into complement therapeutics in critical illnesses by showing that C5 inhibition can be accomplished in severely ill patients. FUNDING Alexion, AstraZeneca Rare Disease.
Collapse
Affiliation(s)
- Djillali Annane
- Department of Intensive Care, Raymond Poincaré Hospital (Assistance Publique-Hôpitaux de Paris), Garches, France; School of Medicine Simone Veil, University of Versailles Saint Quentin, University Paris-Saclay, Versailles, France; FHU SEPSIS, University Paris-Saclay, INSERM, Garches, France
| | - Sean J Pittock
- Department of Neurology, Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Hrishikesh S Kulkarni
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian W Pickering
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Matt R Khoshnevis
- Department of Medicine, Center for Critical Care, Houston Methodist Hospital, Houston, TX, USA
| | - Jason L Siegel
- Department of Critical Care Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Charles A Powell
- Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic of Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Tomoko Fujii
- Intensive Care Unit, Jikei University Hospital, Tokyo, Japan
| | - Derek Dunn
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | - Keisha Smith
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | | | | | - Austin Kulasekararaj
- Department of Haematological Medicine, King's College Hospital, National Institute of Health Research/Wellcome King's Clinical Research Facility and King's College London, London, UK.
| |
Collapse
|
10
|
Bailey M, Linden D, Earley O, Guo Parke H, McAuley DF, Peto T, Taggart C, Kidney J. Inhibition of bradykinin in SARS-CoV-2 infection: a randomised, double-blind trial of icatibant compared with placebo (ICASARS). BMJ Open 2023; 13:e074726. [PMID: 38035747 PMCID: PMC10689398 DOI: 10.1136/bmjopen-2023-074726] [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: 04/17/2023] [Accepted: 07/19/2023] [Indexed: 12/02/2023] Open
Abstract
SARS-CoV-2 binds to ACE2 receptors and enters cells. The symptoms are cough, breathlessness, loss of taste/smell and X-ray evidence of infiltrates on chest imaging initially caused by oedema, and subsequently by a lymphocytic pneumonitis. Coagulopathy, thrombosis and hypotension occur. Worse disease occurs with age, obesity, ischaemic heart disease, hypertension and diabetes.These features may be due to abnormal activation of the contact system. This triggers coagulation and the kallikrein-kinin system, leading to accumulation of bradykinin and its derivatives, which act on receptors B1R and B2R. Receptor activation causes cough, hypotension, oedema and release of the cytokine interleukin-6 (IL-6) which recruits lymphocytes. These effects are core features seen in early SARS CoV-2 infection. METHODS AND ANALYSIS In this study, hypoxic patients with COVID-19 with symptom onset ≤7 days will be randomised to either a bradykinin inhibitor (icatibant) or placebo. Patients and investigators will be blinded. The primary outcome will be blood oxygenation, measured by arterial blood sampling. The secondary outcome will be cardiovascular status. Retinal imaging will be performed to assess vessel size. Blood samples will be taken for measurement of inflammatory analyses including IL-6. As a separate substudy, we will also take comparator blood inflammatory samples from a COVID-19-negative cohort. ETHICS AND DISSEMINATION The study has received the following approvals: West Midlands-Edgbaston Research Ethics Committee. Medicines and Healthcare products Regulatory Agency has issued a clinical trial authorisation. Belfast Health and Social Care Trust is the study sponsor. Results will be made available to participants upon request and findings will be presented and published. TRIAL REGISTRATION NUMBER NCT05407597.
Collapse
Affiliation(s)
- Melanie Bailey
- Mater Hospital, Belfast Health and Social Care Trust, Belfast, UK
| | - Dermot Linden
- Mater Hospital, Belfast Health and Social Care Trust, Belfast, UK
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Olivia Earley
- Mater Hospital, Belfast Health and Social Care Trust, Belfast, UK
| | - Hong Guo Parke
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | | | - Tunde Peto
- Mater Hospital, Belfast Health and Social Care Trust, Belfast, UK
- Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, Belfast, UK
| | - Cliff Taggart
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Joe Kidney
- Mater Hospital, Belfast Health and Social Care Trust, Belfast, UK
| |
Collapse
|
11
|
Urwyler P, Leimbacher M, Charitos P, Moser S, Heijnen IAFM, Trendelenburg M, Thoma R, Sumer J, Camacho-Ortiz A, Bacci MR, Huber LC, Stüssi-Helbling M, Albrich WC, Sendi P, Osthoff M. Recombinant C1 inhibitor in the prevention of severe COVID-19: a randomized, open-label, multi-center phase IIa trial. Front Immunol 2023; 14:1255292. [PMID: 37965347 PMCID: PMC10641758 DOI: 10.3389/fimmu.2023.1255292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Background Conestat alfa (ConA), a recombinant human C1 inhibitor, may prevent thromboinflammation. Methods We conducted a randomized, open-label, multi-national clinical trial in which hospitalized adults at risk for progression to severe COVID-19 were assigned in a 2:1 ratio to receive either 3 days of ConA plus standard of care (SOC) or SOC alone. Primary and secondary endpoints were day 7 disease severity on the WHO Ordinal Scale, time to clinical improvement within 14 days, and safety, respectively. Results The trial was prematurely terminated because of futility after randomization of 84 patients, 56 in the ConA and 28 in the control arm. At baseline, higher WHO Ordinal Scale scores were more frequently observed in the ConA than in the control arm. On day 7, no relevant differences in the primary outcome were noted between the two arms (p = 0.11). The median time to defervescence was 3 days, and the median time to clinical improvement was 7 days in both arms (p = 0.22 and 0.56, respectively). Activation of plasma cascades and endothelial cells over time was similar in both groups. The incidence of adverse events (AEs) was higher in the intervention arm (any AE, 30% with ConA vs. 19% with SOC alone; serious AE, 27% vs. 15%; death, 11% vs. 0%). None of these were judged as being related to the study drug. Conclusion The study results do not support the use of ConA to prevent COVID-19 progression. Clinical trial registration https://clinicaltrials.gov, identifier NCT04414631.
Collapse
Affiliation(s)
- Pascal Urwyler
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Marina Leimbacher
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | | | - Stephan Moser
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Ingmar A. F. M. Heijnen
- Division of Medical Immunology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Marten Trendelenburg
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Reto Thoma
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Johannes Sumer
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Adrián Camacho-Ortiz
- Servicio de Infectologia, Hospital Universitario Dr. José Eleuterio González, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Marcelo R. Bacci
- Department of General Practice, Centro Universitário em Saúde do ABC, Santo André, Brazil
| | - Lars C. Huber
- Clinic for Internal Medicine, City Hospital Triemli, Zurich, Switzerland
| | | | - Werner C. Albrich
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Michael Osthoff
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
12
|
Bailey M, Linden D, Guo-Parke H, Earley O, Peto T, McAuley DF, Taggart C, Kidney J. Vascular risk factors for COVID-19 ARDS: endothelium, contact-kinin system. Front Med (Lausanne) 2023; 10:1208866. [PMID: 37448794 PMCID: PMC10336249 DOI: 10.3389/fmed.2023.1208866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
SARS-CoV-2 binds to ACE2 receptors, expressed within the lungs. Risk factors for hospitalization include hypertension, diabetes, ischaemic heart disease and obesity-conditions linked by the presence of endothelial pathology. Viral infection in this setting causes increased conversion of circulating Factor XII to its active form (FXIIa). This is the first step in the contact-kinin pathway, leading to synchronous activation of the intrinsic coagulation cascade and the plasma Kallikrein-Kinin system, resulting in clotting and inflammatory lung disease. Temporal trends are evident from blood results of hospitalized patients. In the first week of symptoms the activated partial thromboplastin time (APTT) is prolonged. This can occur when clotting factors are consumed as part of the contact (intrinsic) pathway. Platelet counts initially fall, reflecting their consumption in coagulation. Lymphopenia occurs after approximately 1 week, reflecting the emergence of a lymphocytic pneumonitis [COVID-19 acute respiratory distress syndrome (ARDS)]. Intrinsic coagulation also induces the contact-kinin pathway of inflammation. A major product of this pathway, bradykinin causes oedema with ground glass opacities (GGO) on imaging in early COVID-19. Bradykinin also causes release of the pleiotrophic cytokine IL-6, which causes lymphocyte recruitment. Thromobosis and lymphocytic pneumonitis are hallmark features of COVID-19 ARDS. In this review we examine the literature with particular reference to the contact-kinin pathway. Measurements of platelets, lymphocytes and APTT should be undertaken in severe infections to stratify for risk of developing ARDS.
Collapse
Affiliation(s)
- Melanie Bailey
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Dermot Linden
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Hong Guo-Parke
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Olivia Earley
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Tunde Peto
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Danny F. McAuley
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Clifford Taggart
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Joseph Kidney
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
| |
Collapse
|
13
|
Borba-Junior IT, Lima F, Sidarta-Oliveira D, Moraes CRP, Annichino-Bizzacchi JM, Bombassaro B, Palma AC, Costa FTM, Moretti ML, Mansour E, Velloso LA, Orsi FA, De Paula EV. Podoplanin and CLEC-2 levels in patients with COVID-19. Res Pract Thromb Haemost 2023; 7:100282. [PMID: 37361399 PMCID: PMC10284445 DOI: 10.1016/j.rpth.2023.100282] [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: 12/01/2022] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction Podoplanin (PDPN gene) and CLEC-2 are involved in inflammatory hemostasis and have also been related with the pathogenesis of thrombosis. Emerging evidence also suggest that podoplanin can exert protective effects in sepsis and in acute lung injury. In lungs, podoplanin is co-expressed with ACE2, which is the main entry receptor for SARS-CoV-2. Aim To explore the role of podoplanin and CLEC-2 in COVID-19. Methods Circulating levels of podoplanin and CLEC-2 were measured in 30 consecutive COVID-19 patients admitted due to hypoxia, and in 30 age- and sex-matched healthy individuals. Podoplanin expression in lungs from patients who died of COVID-19 was obtained from two independent public databases of single-cell RNAseq from which data from control lungs were also available. Results Circulating podoplanin levels were lower in COVID-19, while no difference was observed in CLEC-2 levels. Podoplanin levels were significantly inversely correlated with markers of coagulation, fibrinolysis and innate immunity. scRNAseq data confirmed that PDPN is co-expressed with ACE2 in pneumocytes, and showed that PDPN expression is lower in this cell compartment in lungs from patients with COVID-19. Conclusion Circulating levels of podoplanin are lower in COVID-19, and the magnitude of this reduction is correlated with hemostasis activation. We also demonstrate the downregulation of PDPN at the transcription level in pneumocytes. Together, our exploratory study questions whether an acquired podoplanin deficiency could be involved in the pathogenesis of acute lung injury in COVID-19, and warrant additional studies to confirm and refine these findings.
Collapse
Affiliation(s)
| | - Franciele Lima
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | | | - Joyce M. Annichino-Bizzacchi
- School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Bruna Bombassaro
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - André C. Palma
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | | | - Eli Mansour
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Lício Augusto Velloso
- School of Medical Sciences, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Center, University of Campinas, Campinas, Brazil
| | - Fernanda Andrade Orsi
- School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
14
|
Shen JK, Zhang HT. Function and structure of bradykinin receptor 2 for drug discovery. Acta Pharmacol Sin 2023; 44:489-498. [PMID: 36075965 PMCID: PMC9453710 DOI: 10.1038/s41401-022-00982-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
Type 2 bradykinin receptor (B2R) is an essential G protein-coupled receptor (GPCR) that regulates the cardiovascular system as a vasodepressor. Dysfunction of B2R is also closely related to cancers and hereditary angioedema (HAE). Although several B2R agonists and antagonists have been developed, icatibant is the only B2R antagonist clinically used for treating HAE. The recently determined structures of B2R have provided molecular insights into the functions and regulation of B2R, which shed light on structure-based drug design for the treatment of B2R-related diseases. In this review, we summarize the structure and function of B2R in relation to drug discovery and discuss future research directions to elucidate the remaining unknown functions of B2R dimerization.
Collapse
Affiliation(s)
- Jin-Kang Shen
- Hangzhou Institute of Innovative Medicine, Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hai-Tao Zhang
- Hangzhou Institute of Innovative Medicine, Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
15
|
de Lima F, Moraes CRP, Barbosa MS, Bombassaro B, Palma AC, Dertkigil SSJ, Moretti ML, Orsi FA, Annichino-Bizzacchi JM, Mansour E, Velloso LA, De Paula EV. Association of heme-oxygenase 1, hemopexin, and heme levels with markers of disease severity in COVID-19. Exp Biol Med (Maywood) 2023; 248:309-316. [PMID: 36740756 PMCID: PMC9902789 DOI: 10.1177/15353702221139185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heme-oxygenase 1 (HO-1) is an enzyme with well-known anti-inflammatory and antioxidant properties, whose levels have been previously associated with disease severity in the context of sterile and infectious diseases. Moreover, the heme/HO-1 pathway has been associated with prothrombotic changes in other diseases. Accordingly, the potential of modulating HO-1 levels for the treatment of COVID-19 was extensively speculated during the COVID-19 pandemic, but very few actual data were generated. The aim of our study was to explore the association of HO-1, heme, and hemopexin (HPX) levels with COVID-19 severity and with markers of inflammation and coagulation activation. The study was conducted in 30 consecutive patients with COVID-19 admitted due to hypoxemia, and 30 healthy volunteers matched by sex, age, and geographic region. HO-1 and HPX levels were measured by enzyme immunoassay (ELISA) and heme levels were measured by a colorimetric method. A comprehensive panel of coagulation and fibrinolysis activation was also used. Patients with COVID-19 presented increased levels of HO-1 when compared to controls (5741 ± 2696 vs 1953 ± 612 pg/mL, respectively, P < 0.0001), as well as a trend toward increased levels of HPX (3.724 ± 0.880 vs 3.254 ± 1.022 mg/mL, respectively; P = 0.06). In addition, HO-1 and HPX levels reduced from admission to day + 4. HO-1 levels were associated with duration of intensive care unit stay and with several markers of coagulation activation. In conclusion, modulation of HO-1 could be associated with the prothrombotic state observed in COVID-19, and HO-1 could also represent a relevant biomarker for COVID-19. New independent studies are warranted to explore and expand these findings.
Collapse
Affiliation(s)
- Franciele de Lima
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Franciele de Lima.
| | | | - Mayck Silva Barbosa
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, Brazil
| | - André C Palma
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | - Maria Luiza Moretti
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | - Joyce M Annichino-Bizzacchi
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| | - Eli Mansour
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | - Licio A Velloso
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| |
Collapse
|
16
|
Moraes CRP, Borba-Junior IT, De Lima F, Silva JRA, Bombassaro B, Palma AC, Mansour E, Velloso LA, Orsi FA, Costa FTM, De Paula EV. Association of Ang/Tie2 pathway mediators with endothelial barrier integrity and disease severity in COVID-19. Front Physiol 2023; 14:1113968. [PMID: 36895630 PMCID: PMC9988918 DOI: 10.3389/fphys.2023.1113968] [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: 12/01/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Endothelial barrier (EB) disruption contributes to acute lung injury in COVID-19, and levels of both VEGF-A and Ang-2, which are mediators of EB integrity, have been associated with COVID-19 severity. Here we explored the participation of additional mediators of barrier integrity in this process, as well as the potential of serum from COVID-19 patients to induce EB disruption in cell monolayers. In a cohort from a clinical trial consisting of thirty patients with COVID-19 that required hospital admission due to hypoxia we demonstrate that i) levels of soluble Tie2 were increase, and of soluble VE-cadherin were decreased when compared to healthy individuals; ii) sera from these patients induce barrier disruption in monolayers of endothelial cells; and iii) that the magnitude of this effect is proportional to disease severity and to circulating levels of VEGF-A and Ang-2. Our study confirms and extends previous findings on the pathogenesis of acute lung injury in COVID-19, reinforcing the concept that EB is a relevant component of this disease. Our results pave the way for future studies that can refine our understanding of the pathogenesis of acute lung injury in viral respiratory disorders, and contribute to the identification of new biomarkers and therapeutic targets for these conditions.
Collapse
Affiliation(s)
| | | | - Franciele De Lima
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - André C Palma
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Eli Mansour
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Lício Augusto Velloso
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | | | | | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
17
|
Lim EHT, van Amstel RBE, de Boer VV, van Vught LA, de Bruin S, Brouwer MC, Vlaar APJ, van de Beek D. Complement activation in COVID-19 and targeted therapeutic options: A scoping review. Blood Rev 2023; 57:100995. [PMID: 35934552 PMCID: PMC9338830 DOI: 10.1016/j.blre.2022.100995] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/07/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
Increasing evidence suggests that activation of the complement system plays a key role in the pathogenesis and disease severity of Coronavirus disease 2019 (COVID-19). We used a systematic approach to create an overview of complement activation in COVID-19 based on histopathological, preclinical, multiomics, observational and clinical interventional studies. A total of 1801 articles from PubMed, EMBASE and Cochrane was screened of which 157 articles were included in this scoping review. Histopathological, preclinical, multiomics and observational studies showed apparent complement activation through all three complement pathways and a correlation with disease severity and mortality. The complement system was targeted at different levels in COVID-19, of which C5 and C5a inhibition seem most promising. Adequately powered, double blind RCTs are necessary in order to further investigate the effect of targeting the complement system in COVID-19.
Collapse
Affiliation(s)
- Endry Hartono Taslim Lim
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Rombout Benjamin Ezra van Amstel
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Vieve Victoria de Boer
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Lonneke Alette van Vught
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, the Netherlands
| | - Sanne de Bruin
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Matthijs Christian Brouwer
- Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Alexander Petrus Johannes Vlaar
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands.
| | - Diederik van de Beek
- Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Liu X, Shi J, Wang D, Su Y, Xing Z, Sun F, Chen F. Therapeutic Polypeptides and Peptidomimetics: Powerful Tools for COVID-19 Treatment. Clin Drug Investig 2023; 43:13-22. [PMID: 36462104 PMCID: PMC9734822 DOI: 10.1007/s40261-022-01231-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 12/04/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has swept the whole world and brought about a public health crisis of unprecedented proportions. To combat the rapid transmission and possible deaths due to the disease, researchers and companies around the world are developing all possible strategies. Due to the advantages of safety, specificity, and fewer adverse effects, polypeptide and peptidomimetic drugs are considered promising strategies. This review comprehensively summarizes and discusses the progress in development of peptide drugs for use in the treatment of COVID-19. Based on the latest results in this field, we divided them into clinically approved drugs, clinical trial drugs, and clinically ineffective drugs, and outlined the molecular targets and mechanisms of action one by one to reveal their feasibility as promising therapeutic agents for COVID-19. Notably, monoclonal antibodies have shown beneficial effects in the early stages of infection, while Paxlovid can significantly reduce hospitalization and mortality among non-vaccinated patients. Among clinical experimental drugs, both the interleukin-1 receptor antagonist anakinra and the bradykinin B2 receptor antagonist icatibant are well tolerated and effective in patients with COVID-19, but long-term trials are needed to confirm the durability of efficacy.
Collapse
Affiliation(s)
- Xinyu Liu
- grid.449428.70000 0004 1797 7280Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067 China
| | - Jian Shi
- grid.449428.70000 0004 1797 7280Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067 China
| | - Deyang Wang
- grid.449428.70000 0004 1797 7280Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067 China
| | - Ying Su
- grid.411634.50000 0004 0632 4559Dongping County People’s Hospital, Tai-an, China
| | - Zhen Xing
- grid.411634.50000 0004 0632 4559Dongping County People’s Hospital, Tai-an, China
| | - Fei Sun
- Qilu Medical University, Zibo, China
| | - Fei Chen
- grid.449428.70000 0004 1797 7280Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067 China
| |
Collapse
|
19
|
Silva BRS, Jara CP, Sidarta-Oliveira D, Velloso LA, Velander WH, Araújo EP. Downregulation of the Protein C Signaling System Is Associated with COVID-19 Hypercoagulability-A Single-Cell Transcriptomics Analysis. Viruses 2022; 14:2753. [PMID: 36560757 PMCID: PMC9785999 DOI: 10.3390/v14122753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Because of the interface between coagulation and the immune response, it is expected that COVID-19-associated coagulopathy occurs via activated protein C signaling. The objective was to explore putative changes in the expression of the protein C signaling network in the liver, peripheral blood mononuclear cells, and nasal epithelium of patients with COVID-19. Single-cell RNA-sequencing data from patients with COVID-19 and healthy subjects were obtained from the COVID-19 Cell Atlas database. A functional protein-protein interaction network was constructed for the protein C gene. Patients with COVID-19 showed downregulation of protein C and components of the downstream protein C signaling cascade. The percentage of hepatocytes expressing protein C was lower. Part of the liver cell clusters expressing protein C presented increased expression of ACE2. In PBMC, there was increased ACE2, inflammatory, and pro-coagulation transcripts. In the nasal epithelium, PROC, ACE2, and PROS1 were expressed by the ciliated cell cluster, revealing co-expression of ACE-2 with transcripts encoding proteins belonging to the coagulation and immune system interface. Finally, there was upregulation of coagulation factor 3 transcript in the liver and PBMC. Protein C could play a mechanistic role in the hypercoagulability syndrome affecting patients with severe COVID-19.
Collapse
Affiliation(s)
- Bruna Rafaela Santos Silva
- Nursing School, University of Campinas, Tessalia Vieira de Camargo, 126, Campinas 13084-970, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Center, OCRC, University of Campinas, Carl Von Linnaeus, s/n, Campinas 13084-864, Brazil
| | - Carlos Poblete Jara
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE 68588-0643, USA
| | - Davi Sidarta-Oliveira
- Laboratory of Cell Signalling, Obesity and Comorbidities Center, OCRC, University of Campinas, Carl Von Linnaeus, s/n, Campinas 13084-864, Brazil
- School of Medical Sciences, University of Campinas, Tessalia Vieira de Camargo, 126, Campinas 13083-887, Brazil
| | - Licio A. Velloso
- Laboratory of Cell Signalling, Obesity and Comorbidities Center, OCRC, University of Campinas, Carl Von Linnaeus, s/n, Campinas 13084-864, Brazil
- School of Medical Sciences, University of Campinas, Tessalia Vieira de Camargo, 126, Campinas 13083-887, Brazil
| | - William H. Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE 68588-0643, USA
| | - Eliana P. Araújo
- Nursing School, University of Campinas, Tessalia Vieira de Camargo, 126, Campinas 13084-970, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Center, OCRC, University of Campinas, Carl Von Linnaeus, s/n, Campinas 13084-864, Brazil
| |
Collapse
|
20
|
Hausburg MA, Williams JS, Banton KL, Mains CW, Roshon M, Bar-Or D. C1 esterase inhibitor-mediated immunosuppression in COVID-19: Friend or foe? CLINICAL IMMUNOLOGY COMMUNICATIONS 2022; 2:83-90. [PMID: 38013973 PMCID: PMC9068237 DOI: 10.1016/j.clicom.2022.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/03/2022] [Accepted: 05/03/2022] [Indexed: 10/10/2023]
Abstract
From asymptomatic to severe, SARS-CoV-2, causative agent of COVID-19, elicits varying disease severities. Moreover, understanding innate and adaptive immune responses to SARS-CoV-2 is imperative since variants such as Omicron negatively impact adaptive antibody neutralization. Severe COVID-19 is, in part, associated with aberrant activation of complement and Factor XII (FXIIa), initiator of contact system activation. Paradoxically, a protein that inhibits the three known pathways of complement activation and FXIIa, C1 esterase inhibitor (C1-INH), is increased in COVID-19 patient plasma and is associated with disease severity. Here we review the role of C1-INH in the regulation of innate and adaptive immune responses. Additionally, we contextualize regulation of C1-INH and SERPING1, the gene encoding C1-INH, by other pathogens and SARS viruses and propose that viral proteins bind to C1-INH to inhibit its function in severe COVID-19. Finally, we review the current clinical trials and published results of exogenous C1-INH treatment in COVID-19 patients.
Collapse
Key Words
- C1 esterase inhibitor
- C1 esterase inhibitor, C1-INH
- C1-INH
- COVID-19
- Complement
- FXII
- Inflammation
- Middle East respiratory syndrome coronavirus, MERS-CoV
- Mycobacterium tuberculosis, Mtb
- Severe acute respiratory syndrome coronavirus, SARS-CoV
- acquired C1-INH deficiency, AEE
- activated plasma kallikrein, PKa
- antibody-mediated rejection, AMR
- bradykinin, BK
- contact system, CS
- coronavirus disease 2019, COVID-19
- exogenous C1-INH, exC1-INH
- hereditary angioedema, HAE
- high-molecular-weight kininogen, HK
- human immunodeficiency virus, HIV
- interferon, IFN
- interleukin, IL
- ischemia/reperfusion injury, IRI
- mannose-binding lectin, MBL
- prekallikrein, PK
- recombinant C1-INH, rhC1-INH
- serine protease inhibitor, serpin
- tuberculosis, TB
Collapse
Affiliation(s)
- Melissa A Hausburg
- Department of Trauma Research, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113, USA
- Department of Trauma Research, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228, USA
- Department of Trauma Research, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907, USA
| | - Jason S Williams
- Department of Trauma Research, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113, USA
- Department of Trauma Research, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228, USA
- Department of Trauma Research, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907, USA
| | - Kaysie L Banton
- Department of Trauma Research, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113, USA
| | - Charles W Mains
- Centura Health Trauma Systems, Centura Health, 9100 E Mineral Circle, Centennial, CO 80112, USA
| | - Michael Roshon
- Centura Health Trauma Systems, Centura Health, 9100 E Mineral Circle, Centennial, CO 80112, USA
- Department of Emergency Services, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907, USA
| | - David Bar-Or
- Department of Trauma Research, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113, USA
- Department of Trauma Research, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228, USA
- Department of Trauma Research, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907, USA
- Department of Molecular Biology, Rocky Vista University, 8401 S Chambers Rd, Parker, CO 80134, USA
| |
Collapse
|
21
|
Urwyler P, Moser S, Trendelenburg M, Sendi P, Osthoff M. Targeting thromboinflammation in COVID-19 - A narrative review of the potential of C1 inhibitor to prevent disease progression. Mol Immunol 2022; 150:99-113. [PMID: 36030710 PMCID: PMC9393183 DOI: 10.1016/j.molimm.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/07/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 is associated with a clinical spectrum ranging from asymptomatic carriers to critically ill patients with complications including thromboembolic events, myocardial injury, multisystemic inflammatory syndromes and death. Since the beginning of the pandemic several therapeutic options emerged, with a multitude of randomized trials, changing the medical landscape of COVID-19. The effect of various monoclonal antibodies, antiviral, anti-inflammatory and anticoagulation drugs have been studied, and to some extent, implemented into clinical practice. In addition, a multitude of trials improved the understanding of the disease and emerging evidence points towards a significant role of the complement system, kallikrein-kinin, and contact activation system as drivers of disease in severe COVID-19. Despite their involvement in COVID-19, treatments targeting these plasmatic cascades have neither been systematically studied nor introduced into clinical practice, and randomized studies with regards to these treatments are scarce. Given the multiple-action, multiple-target nature of C1 inhibitor (C1-INH), the natural inhibitor of these cascades, this drug may be an interesting candidate to prevent disease progression and combat thromboinflammation in COVID-19. This narrative review will discuss the current evidence with regards to the involvement of these plasmatic cascades as well as endothelial cells in COVID-19. Furthermore, we summarize the evidence of C1-INH in COVID-19 and potential benefits and pitfalls of C1-INH treatment in COVID-19.
Collapse
Affiliation(s)
- Pascal Urwyler
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Stephan Moser
- Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marten Trendelenburg
- Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland; Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Michael Osthoff
- Department of Clinical Research and Department of Biomedicine, University of Basel, Basel, Switzerland; Division of Internal Medicine, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
22
|
Pham N, Hu F, Evelo CT, Kutmon M. Tissue-specific pathway activities: A retrospective analysis in COVID-19 patients. Front Immunol 2022; 13:963357. [PMID: 36189295 PMCID: PMC9519890 DOI: 10.3389/fimmu.2022.963357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/24/2022] [Indexed: 12/04/2022] Open
Abstract
The ACE2 receptors essential for SARS-CoV-2 infections are expressed not only in the lung but also in many other tissues in the human body. To better understand the disease mechanisms and progression, it is essential to understand how the virus affects and alters molecular pathways in the different affected tissues. In this study, we mapped the proteomics data obtained from Nie X. et al. (2021) to the pathway models of the COVID-19 Disease Map project and WikiPathways. The differences in pathway activities between COVID-19 and non-COVID-19 patients were calculated using the Wilcoxon test. As a result, 46% (5,235) of the detected proteins were found to be present in at least one pathway. Only a few pathways were altered in multiple tissues. As an example, the Kinin-Kallikrein pathway, an important inflammation regulatory pathway, was found to be less active in the lung, spleen, testis, and thyroid. We can confirm previously reported changes in COVID-19 patients such as the change in cholesterol, linolenic acid, and arachidonic acid metabolism, complement, and coagulation pathways in most tissues. Of all the tissues, we found the thyroid to be the organ with the most changed pathways. In this tissue, lipid pathways, energy pathways, and many COVID-19 specific pathways such as RAS and bradykinin pathways, thrombosis, and anticoagulation have altered activities in COVID-19 patients. Concluding, our results highlight the systemic nature of COVID-19 and the effect on other tissues besides the lung.
Collapse
Affiliation(s)
- Nhung Pham
- Department of Bioinformatics (BiGCaT), School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, Netherlands
| | - Finterly Hu
- Department of Bioinformatics (BiGCaT), School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, Netherlands
| | - Chris T. Evelo
- Department of Bioinformatics (BiGCaT), School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, Netherlands
| | - Martina Kutmon
- Department of Bioinformatics (BiGCaT), School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, Netherlands
- *Correspondence: Martina Kutmon,
| |
Collapse
|
23
|
Martens CP, Van Mol P, Wauters J, Wauters E, Gangnus T, Noppen B, Callewaert H, Feyen JH, Liesenborghs L, Heylen E, Jansen S, Pereira LCV, Kraisin S, Guler I, Engelen MM, Ockerman A, Van Herck A, Vos R, Vandenbriele C, Meersseman P, Hermans G, Wilmer A, Martinod K, Burckhardt BB, Vanhove M, Jacquemin M, Verhamme P, Neyts J, Vanassche T. Dysregulation of the kallikrein-kinin system in bronchoalveolar lavage fluid of patients with severe COVID-19. EBioMedicine 2022; 83:104195. [PMID: 35939907 PMCID: PMC9352453 DOI: 10.1016/j.ebiom.2022.104195] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/06/2022] [Accepted: 07/15/2022] [Indexed: 11/21/2022] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds to the angiotensin-converting enzyme 2 (ACE2) receptor, a critical component of the kallikrein-kinin system. Its dysregulation may lead to increased vascular permeability and release of inflammatory chemokines. Interactions between the kallikrein-kinin and the coagulation system might further contribute to thromboembolic complications in COVID-19. Methods In this observational study, we measured plasma and tissue kallikrein hydrolytic activity, levels of kinin peptides, and myeloperoxidase (MPO)-DNA complexes as a biomarker for neutrophil extracellular traps (NETs), in bronchoalveolar lavage (BAL) fluid from patients with and without COVID-19. Findings In BAL fluid from patients with severe COVID-19 (n = 21, of which 19 were mechanically ventilated), we observed higher tissue kallikrein activity (18·2 pM [1·2-1535·0], median [range], n = 9 vs 3·8 [0·0-22·0], n = 11; p = 0·030), higher levels of the kinin peptide bradykinin-(1-5) (89·6 [0·0-2425·0], n = 21 vs 0·0 [0·0-374·0], n = 19, p = 0·001), and higher levels of MPO-DNA complexes (699·0 ng/mL [66·0-142621·0], n = 21 vs 70·5 [9·9-960·0], n = 19, p < 0·001) compared to patients without COVID-19. Interpretation Our observations support the hypothesis that dysregulation of the kallikrein-kinin system might occur in mechanically ventilated patients with severe pulmonary disease, which might help to explain the clinical presentation of patients with severe COVID-19 developing pulmonary oedema and thromboembolic complications. Therefore, targeting the kallikrein-kinin system should be further explored as a potential treatment option for patients with severe COVID-19. Funding Research Foundation-Flanders (G0G4720N, 1843418N), KU Leuven COVID research fund.
Collapse
|
24
|
Camargo RL, Bombassaro B, Monfort-Pires M, Mansour E, Palma AC, Ribeiro LC, Ulaf RG, Bernardes AF, Nunes TA, Agrela MV, Dertkigil RP, Dertkigil SS, Araujo EP, Nadruz W, Moretti ML, Velloso LA, Sposito AC. Plasma Angiotensin II Is Increased in Critical Coronavirus Disease 2019. Front Cardiovasc Med 2022; 9:847809. [PMID: 35811697 PMCID: PMC9263116 DOI: 10.3389/fcvm.2022.847809] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) employs angiotensin-converting enzyme 2 (ACE2) as its receptor for cell entrance, and studies have suggested that upon viral binding, ACE2 catalytic activity could be inhibited; therefore, impacting the regulation of the renin-angiotensin-aldosterone system (RAAS). To date, only few studies have evaluated the impact of SARS-CoV-2 infection on the blood levels of the components of the RAAS. The objective of this study was to determine the blood levels of ACE, ACE2, angiotensin-II, angiotensin (1-7), and angiotensin (1-9) at hospital admission and discharge in a group of patients presenting with severe or critical evolution of coronavirus disease 2019 (COVID-19). We showed that ACE, ACE2, angiotensin (1-7), and angiotensin (1-9) were similar in patients with critical and severe COVID-19. However, at admission, angiotensin-II levels were significantly higher in patients presenting as critical, compared to patients presenting with severe COVID-19. We conclude that blood levels of angiotensin-II are increased in hospitalized patients with COVID-19 presenting the critical outcome of the disease. We propose that early measurement of Ang-II could be a useful biomarker for identifying patients at higher risk for extremely severe progression of the disease.
Collapse
Affiliation(s)
- Rafael L Camargo
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Milena Monfort-Pires
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Eli Mansour
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Andre C Palma
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Luciana C Ribeiro
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Raisa G Ulaf
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Ana Flavia Bernardes
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Thyago A Nunes
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Marcus V Agrela
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Rachel P Dertkigil
- Department of Radiology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Sergio S Dertkigil
- Department of Radiology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Eliana P Araujo
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.,School of Nursing, University of Campinas, Campinas, Brazil
| | - Wilson Nadruz
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.,Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Maria Luiza Moretti
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.,Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Andrei C Sposito
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.,Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| |
Collapse
|
25
|
Agirbasli M. The effects of antihypertensive medications on severity and outcomes of COVID19. J Hum Hypertens 2022; 36:875-879. [PMID: 35810205 PMCID: PMC9281571 DOI: 10.1038/s41371-022-00722-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/18/2022] [Accepted: 06/28/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Mehmet Agirbasli
- Department of Cardiology, School of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey.
| |
Collapse
|
26
|
Bueno LCM, Paim LR, Minin EOZ, da Silva LM, Mendes PR, Kiyota TA, Schreiber AZ, Bombassaro B, Mansour E, Moretti ML, Chow JTS, Salmena L, Coelho-Filho OR, Velloso LA, Nadruz W, Schreiber R. Increased Serum Mir-150-3p Expression Is Associated with Radiological Lung Injury Improvement in Patients with COVID-19. Viruses 2022; 14:v14071363. [PMID: 35891345 PMCID: PMC9323362 DOI: 10.3390/v14071363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/25/2022] [Accepted: 06/21/2022] [Indexed: 12/04/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the SARS-CoV-2 virus, responsible for an atypical pneumonia that can progress to acute lung injury. MicroRNAs are small non-coding RNAs that control specific genes and pathways. This study evaluated the association between circulating miRNAs and lung injury associated with COVID-19. Methods: We evaluated lung injury by computed tomography at hospital admission and discharge and the serum expression of 754 miRNAs using the TaqMan OpenArray after hospital discharge in 27 patients with COVID-19. In addition, miR-150-3p was validated by qRT-PCR on serum samples collected at admission and after hospital discharge. Results: OpenArray analysis revealed that seven miRNAs were differentially expressed between groups of patients without radiological lung improvement compared to those with lung improvement at hospital discharge, with three miRNAs being upregulated (miR-548c-3p, miR-212-3p, and miR-548a-3p) and four downregulated (miR-191-5p, miR-151a-3p, miR-92a-3p, and miR-150-3p). Bioinformatics analysis revealed that five of these miRNAs had binding sites in the SARS-CoV-2 genome. Validation of miR-150-3p by qRT-PCR confirmed the OpenArray results. Conclusions: The present study shows the potential association between the serum expression of seven miRNAs and lung injury in patients with COVID-19. Furthermore, increased expression of miR-150 was associated with pulmonary improvement at hospital discharge.
Collapse
Affiliation(s)
- Larissa C. M. Bueno
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Layde R. Paim
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Eduarda O. Z. Minin
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Luís Miguel da Silva
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Paulo R. Mendes
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Tatiana A. Kiyota
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Angelica Z. Schreiber
- Department of Clinical Pathology, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil;
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, SP, Brazil;
| | - Eli Mansour
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Maria Luiza Moretti
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Jonathan Tak-Sum Chow
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada; (J.T.-S.C.); (L.S.)
| | - Leonardo Salmena
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada; (J.T.-S.C.); (L.S.)
| | - Otavio R. Coelho-Filho
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Licio A. Velloso
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada; (J.T.-S.C.); (L.S.)
| | - Wilson Nadruz
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
| | - Roberto Schreiber
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas 13083-887, SP, Brazil; (L.C.M.B.); (L.R.P.); (E.O.Z.M.); (L.M.d.S.); (P.R.M.); (T.A.K.); (E.M.); (M.L.M.); (O.R.C.-F.); (L.A.V.); (W.N.)
- Correspondence:
| |
Collapse
|
27
|
Henderson MW, Lima F, Moraes CRP, Ilich A, Huber SC, Barbosa MS, Santos I, Palma AC, Nunes TA, Ulaf RG, Ribeiro LC, Bernardes AF, Bombassaro B, Dertkigil SSJ, Moretti ML, Strickland S, Annichino-Bizzacchi JM, Orsi FA, Mansour E, Velloso LA, Key NS, De Paula EV. Contact and intrinsic coagulation pathways are activated and associated with adverse clinical outcomes in COVID-19. Blood Adv 2022; 6:3367-3377. [PMID: 35235941 PMCID: PMC8893951 DOI: 10.1182/bloodadvances.2021006620] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/19/2022] [Indexed: 12/27/2022] Open
Abstract
Coagulation activation is a prominent feature of severe acute respiratory syndrome coronavirus 2 (COVID-19) infection. Activation of the contact system and intrinsic pathway has increasingly been implicated in the prothrombotic state observed in both sterile and infectious inflammatory conditions. We therefore sought to assess activation of the contact system and intrinsic pathway in individuals with COVID-19 infection. Baseline plasma levels of protease:serpin complexes indicative of activation of the contact and intrinsic pathways were measured in samples from inpatients with COVID-19 and healthy individuals. Cleaved kininogen, a surrogate for bradykinin release, was measured by enzyme-linked immunosorbent assay, and extrinsic pathway activation was assessed by microvesicle tissue factor-mediated factor Xa (FXa; MVTF) generation. Samples were collected within 24 hours of COVID-19 diagnosis. Thirty patients with COVID-19 and 30 age- and sex-matched controls were enrolled. Contact system and intrinsic pathway activation in COVID-19 was demonstrated by increased plasma levels of FXIIa:C1 esterase inhibitor (C1), kallikrein:C1, FXIa:C1, FXIa:α1-antitrypsin, and FIXa:antithrombin (AT). MVTF levels were also increased in patients with COVID-19. Because FIXa:AT levels were associated with both contact/intrinsic pathway complexes and MVTF, activation of FIX likely occurs through both contact/intrinsic and extrinsic pathways. Among the protease:serpin complexes measured, FIXa:AT complexes were uniquely associated with clinical indices of disease severity, specifically total length of hospitalization, length of intensive care unit stay, and extent of lung computed tomography changes. We conclude that the contact/intrinsic pathway may contribute to the pathogenesis of the prothrombotic state in COVID-19. Larger prospective studies are required to confirm whether FIXa:AT complexes are a clinically useful biomarker of adverse clinical outcomes.
Collapse
Affiliation(s)
- Michael W. Henderson
- University of North Carolina (UNC) Blood Research Center, UNC at Chapel Hill, Chapel Hill, NC
| | - Franciele Lima
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Anton Ilich
- University of North Carolina (UNC) Blood Research Center, UNC at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, UNC at Chapel Hill, Chapel Hill, NC
| | | | - Mayck Silva Barbosa
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Andre C. Palma
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Thyago Alves Nunes
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Raisa Gusso Ulaf
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Luciana Costa Ribeiro
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Ana Flavia Bernardes
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Sergio San Juan Dertkigil
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Maria Luiza Moretti
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY; and
| | - Joyce M. Annichino-Bizzacchi
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, and
| | - Fernanda Andrade Orsi
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Eli Mansour
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Licio A. Velloso
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Nigel S. Key
- University of North Carolina (UNC) Blood Research Center, UNC at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, UNC at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, UNC at Chapel Hill, Chapel Hill, NC
| | - Erich Vinicius De Paula
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, and
| |
Collapse
|
28
|
Nilsson B, Persson B, Eriksson O, Fromell K, Hultström M, Frithiof R, Lipcsey M, Huber-Lang M, Ekdahl KN. How the Innate Immune System of the Blood Contributes to Systemic Pathology in COVID-19-Induced ARDS and Provides Potential Targets for Treatment. Front Immunol 2022; 13:840137. [PMID: 35350780 PMCID: PMC8957861 DOI: 10.3389/fimmu.2022.840137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
Most SARS-CoV-2 infected patients experience influenza-like symptoms of low or moderate severity. But, already in 2020 early during the pandemic it became obvious that many patients had a high incidence of thrombotic complications, which prompted treatment with high doses of low-molecular-weight heparin (LMWH; typically 150-300IU/kg) to prevent thrombosis. In some patients, the disease aggravated after approximately 10 days and turned into a full-blown acute respiratory distress syndrome (ARDS)-like pulmonary inflammation with endothelialitis, thrombosis and vascular angiogenesis, which often lead to intensive care treatment with ventilator support. This stage of the disease is characterized by dysregulation of cytokines and chemokines, in particular with high IL-6 levels, and also by reduced oxygen saturation, high risk of thrombosis, and signs of severe pulmonary damage with ground glass opacities. The direct link between SARS-CoV-2 and the COVID-19-associated lung injury is not clear. Indirect evidence speaks in favor of a thromboinflammatory reaction, which may be initiated by the virus itself and by infected damaged and/or apoptotic cells. We and others have demonstrated that life-threatening COVID-19 ARDS is associated with a strong activation of the intravascular innate immune system (IIIS). In support of this notion is that activation of the complement and kallikrein/kinin (KK) systems predict survival, the necessity for usage of mechanical ventilation, acute kidney injury and, in the case of MBL, also coagulation system activation with thromboembolism. The general properties of the IIIS can easily be translated into mechanisms of COVID-19 pathophysiology. The prognostic value of complement and KKsystem biomarkers demonstrate that pharmaceuticals, which are licensed or have passed the phase I trial stage are promising candidate drugs for treatment of COVID-19. Examples of such compounds include complement inhibitors AMY-101 and eculizumab (targeting C3 and C5, respectively) as well as kallikrein inhibitors ecallantide and lanadelumab and the bradykinin receptor (BKR) 2 antagonist icatibant. In this conceptual review we discuss the activation, crosstalk and the therapeutic options that are available for regulation of the IIIS.
Collapse
Affiliation(s)
- Bo Nilsson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Barbro Persson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Eriksson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Karin Fromell
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.,Unit for Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Miklos Lipcsey
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.,Hedenstierna Laboratory, Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Kristina N Ekdahl
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
29
|
Jakwerth CA, Feuerherd M, Guerth FM, Oelsner M, Schellhammer L, Giglberger J, Pechtold L, Jerin C, Kugler L, Mogler C, Haller B, Erb A, Wollenberg B, Spinner CD, Buch T, Protzer U, Schmidt-Weber CB, Zissler UM, Chaker AM. Early reduction of SARS-CoV-2-replication in bronchial epithelium by kinin B 2 receptor antagonism. J Mol Med (Berl) 2022; 100:613-627. [PMID: 35247068 PMCID: PMC8897552 DOI: 10.1007/s00109-022-02182-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/14/2022]
Abstract
Abstract SARS-CoV-2 has evolved to enter the host via the ACE2 receptor which is part of the kinin-kallikrein pathway. This complex pathway is only poorly understood in context of immune regulation but critical to control infection. This study examines SARS-CoV-2-infection and epithelial mechanisms of the kinin-kallikrein-system at the kinin B2 receptor level in SARS-CoV-2-infection that is of direct translational relevance. From acute SARS-CoV-2-positive study participants and -negative controls, transcriptomes of nasal curettages were analyzed. Primary airway epithelial cells (NHBEs) were infected with SARS-CoV-2 and treated with the approved B2R-antagonist icatibant. SARS-CoV-2 RNA RT-qPCR, cytotoxicity assays, plaque assays, and transcriptome analyses were performed. The treatment effect was further studied in a murine airway inflammation model in vivo. Here, we report a broad and strong upregulation of kallikreins and the kinin B2 receptor (B2R) in the nasal mucosa of acutely symptomatic SARS-CoV-2-positive study participants. A B2R-antagonist impeded SARS-CoV-2 replication and spread in NHBEs, as determined in plaque assays on Vero-E6 cells. B2R-antagonism reduced the expression of SARS-CoV-2 entry receptor ACE2, G protein–coupled receptor signaling, and ion transport in vitro and in a murine airway inflammation in vivo model. In summary, this study provides evidence that treatment with B2R-antagonists protects airway epithelial cells from SARS-CoV-2 by inhibiting its replication and spread, through the reduction of ACE2 levels and the interference with several cellular signaling processes. Future clinical studies need to shed light on the airway protection potential of approved B2R-antagonists, like icatibant, in the treatment of early-stage COVID-19. Graphical Abstract ![]()
Key messages Induction of kinin B2 receptor in the nose of SARS-CoV-2-positive patients. Treatment with B2R-antagonist protects airway epithelial cells from SARS-CoV-2. B2R-antagonist reduces ACE2 levels in vivo and ex vivo. Protection by B2R-antagonist is mediated by inhibiting viral replication and spread.
Supplementary information The online version contains supplementary material available at 10.1007/s00109-022-02182-7.
Collapse
Affiliation(s)
- Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Martin Feuerherd
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, German Center of Infectiology Research (DZIF), Munich partner site, Munich, Germany
| | - Ferdinand M Guerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Madlen Oelsner
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Linda Schellhammer
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Johanna Giglberger
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Lisa Pechtold
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Claudia Jerin
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Luisa Kugler
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, Medical School, Technical University of Munich, Munich, Germany
| | - Anna Erb
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Christoph D Spinner
- Department of Internal Medicine II, University Hospital Rechts Der Isar, Medical School, Technical University of Munich, Munich, Germany
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, German Center of Infectiology Research (DZIF), Munich partner site, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany
| | - Adam M Chaker
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German, Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, and Member of the Helmholtz I&I Initiative, Biedersteiner Str. 29, 80202, Munich, Germany.,Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| |
Collapse
|
30
|
Labandeira-Garcia JL, Labandeira CM, Valenzuela R, Pedrosa MA, Quijano A, Rodriguez-Perez AI. Drugs Modulating Renin-Angiotensin System in COVID-19 Treatment. Biomedicines 2022; 10:502. [PMID: 35203711 PMCID: PMC8962306 DOI: 10.3390/biomedicines10020502] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023] Open
Abstract
A massive worldwide vaccination campaign constitutes the main tool against the COVID-19 pandemic. However, drug treatments are also necessary. Antivirals are the most frequently considered treatments. However, strategies targeting mechanisms involved in disease aggravation may also be effective. A major role of the tissue renin-angiotensin system (RAS) in the pathophysiology and severity of COVID-19 has been suggested. The main link between RAS and COVID-19 is angiotensin-converting enzyme 2 (ACE2), a central RAS component and the primary binding site for SARS-CoV-2 that facilitates the virus entry into host cells. An initial suggestion that the susceptibility to infection and disease severity may be enhanced by angiotensin type-1 receptor blockers (ARBs) and ACE inhibitors (ACEIs) because they increase ACE2 levels, led to the consideration of discontinuing treatments in thousands of patients. More recent experimental and clinical data indicate that ACEIs and, particularly, ARBs can be beneficial for COVID-19 outcome, both by reducing inflammatory responses and by triggering mechanisms (such as ADAM17 inhibition) counteracting viral entry. Strategies directly activating RAS anti-inflammatory components such as soluble ACE2, Angiotensin 1-7 analogues, and Mas or AT2 receptor agonists may also be beneficial. However, while ACEIs and ARBs are cheap and widely used, the second type of strategies are currently under study.
Collapse
Affiliation(s)
- Jose L. Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Carmen M. Labandeira
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Neurology Service, Hospital Alvaro Cunqueiro, University Hospital Complex, 36213 Vigo, Spain
| | - Rita Valenzuela
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Maria A. Pedrosa
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Aloia Quijano
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
| | - Ana I. Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
31
|
Thrombosis and fibrosis: mutually inclusive targets to combat in COVID-19. Future Sci OA 2022; 8:FSO777. [PMID: 35194516 PMCID: PMC8687043 DOI: 10.2144/fsoa-2021-0127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/26/2021] [Indexed: 11/23/2022] Open
|
32
|
Simko F, Baka T. Commentary: Effect of Angiotensin-Converting-Enzyme Inhibitor and Angiotensin II Receptor Antagonist Treatment on ACE2 Expression and SARS-CoV-2 Replication in Primary Airway Epithelial Cells. Front Pharmacol 2022; 13:842512. [PMID: 35153802 PMCID: PMC8832014 DOI: 10.3389/fphar.2022.842512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/12/2022] [Indexed: 12/23/2022] Open
Affiliation(s)
- Fedor Simko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- *Correspondence: Fedor Simko,
| | - Tomas Baka
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
33
|
Farahani M, Niknam Z, Mohammadi Amirabad L, Amiri-Dashatan N, Koushki M, Nemati M, Danesh Pouya F, Rezaei-Tavirani M, Rasmi Y, Tayebi L. Molecular pathways involved in COVID-19 and potential pathway-based therapeutic targets. Biomed Pharmacother 2022; 145:112420. [PMID: 34801852 PMCID: PMC8585639 DOI: 10.1016/j.biopha.2021.112420] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 01/08/2023] Open
Abstract
Deciphering the molecular downstream consequences of severe acute respiratory syndrome coronavirus (SARS-CoV)- 2 infection is important for a greater understanding of the disease and treatment planning. Furthermore, greater understanding of the underlying mechanisms of diagnostic and therapeutic strategies can help in the development of vaccines and drugs against COVID-19. At present, the molecular mechanisms of SARS-CoV-2 in the host cells are not sufficiently comprehended. Some of the mechanisms are proposed considering the existing similarities between SARS-CoV-2 and the other members of the β-CoVs, and others are explained based on studies advanced in the structure and function of SARS-CoV-2. In this review, we endeavored to map the possible mechanisms of the host response following SARS-CoV-2 infection and surveyed current research conducted by in vitro, in vivo and human observations, as well as existing suggestions. We addressed the specific signaling events that can cause cytokine storm and demonstrated three forms of cell death signaling following virus infection, including apoptosis, pyroptosis, and necroptosis. Given the elicited signaling pathways, we introduced possible pathway-based therapeutic targets; ADAM17 was especially highlighted as one of the most important elements of several signaling pathways involved in the immunopathogenesis of COVID-19. We also provided the possible drug candidates against these targets. Moreover, the cytokine-cytokine receptor interaction pathway was found as one of the important cross-talk pathways through a pathway-pathway interaction analysis for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Masoumeh Farahani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Niknam
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nasrin Amiri-Dashatan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Koushki
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Nemati
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Fahima Danesh Pouya
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| |
Collapse
|
34
|
Crosstalk between the renin-angiotensin, complement and kallikrein-kinin systems in inflammation. Nat Rev Immunol 2021; 22:411-428. [PMID: 34759348 PMCID: PMC8579187 DOI: 10.1038/s41577-021-00634-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 12/28/2022]
Abstract
During severe inflammatory and infectious diseases, various mediators modulate the equilibrium of vascular tone, inflammation, coagulation and thrombosis. This Review describes the interactive roles of the renin–angiotensin system, the complement system, and the closely linked kallikrein–kinin and contact systems in cell biological functions such as vascular tone and leakage, inflammation, chemotaxis, thrombosis and cell proliferation. Specific attention is given to the role of these systems in systemic inflammation in the vasculature and tissues during hereditary angioedema, cardiovascular and renal glomerular disease, vasculitides and COVID-19. Moreover, we discuss the therapeutic implications of these complex interactions, given that modulation of one system may affect the other systems, with beneficial or deleterious consequences. The renin–angiotensin, complement and kallikrein–kinin systems comprise a multitude of mediators that modulate physiological responses during inflammatory and infectious diseases. This Review investigates the complex interactions between these systems and how these are dysregulated in various conditions, including cardiovascular diseases and COVID-19, as well as their therapeutic implications.
Collapse
|
35
|
Cooper SL, Boyle E, Jefferson SR, Heslop CRA, Mohan P, Mohanraj GGJ, Sidow HA, Tan RCP, Hill SJ, Woolard J. Role of the Renin-Angiotensin-Aldosterone and Kinin-Kallikrein Systems in the Cardiovascular Complications of COVID-19 and Long COVID. Int J Mol Sci 2021; 22:8255. [PMID: 34361021 PMCID: PMC8347967 DOI: 10.3390/ijms22158255] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 01/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the virus responsible for the COVID-19 pandemic. Patients may present as asymptomatic or demonstrate mild to severe and life-threatening symptoms. Although COVID-19 has a respiratory focus, there are major cardiovascular complications (CVCs) associated with infection. The reported CVCs include myocarditis, heart failure, arrhythmias, thromboembolism and blood pressure abnormalities. These occur, in part, because of dysregulation of the Renin-Angiotensin-Aldosterone System (RAAS) and Kinin-Kallikrein System (KKS). A major route by which SARS-CoV-2 gains cellular entry is via the docking of the viral spike (S) protein to the membrane-bound angiotensin converting enzyme 2 (ACE2). The roles of ACE2 within the cardiovascular and immune systems are vital to ensure homeostasis. The key routes for the development of CVCs and the recently described long COVID have been hypothesised as the direct consequences of the viral S protein/ACE2 axis, downregulation of ACE2 and the resulting damage inflicted by the immune response. Here, we review the impact of COVID-19 on the cardiovascular system, the mechanisms by which dysregulation of the RAAS and KKS can occur following virus infection and the future implications for pharmacological therapies.
Collapse
Affiliation(s)
- Samantha L. Cooper
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Centre of Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Eleanor Boyle
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Sophie R. Jefferson
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Calum R. A. Heslop
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Pirathini Mohan
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Gearry G. J. Mohanraj
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Hamza A. Sidow
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Rory C. P. Tan
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Stephen J. Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Centre of Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Centre of Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
36
|
Scialo F, Vitale M, Daniele A, Nigro E, Perrotta F, Gelzo M, Iadevaia C, Cerqua FS, Costigliola A, Allocca V, Amato F, Pastore L, Castaldo G, Bianco A. SARS-CoV-2: One Year in the Pandemic. What Have We Learned, the New Vaccine Era and the Threat of SARS-CoV-2 Variants. Biomedicines 2021; 9:611. [PMID: 34072088 PMCID: PMC8226851 DOI: 10.3390/biomedicines9060611] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023] Open
Abstract
Since the beginning of 2020, the new pandemic caused by SARS-CoV-2 and named coronavirus disease 19 (COVID 19) has changed our socio-economic life. In just a few months, SARS-CoV-2 was able to spread worldwide at an unprecedented speed, causing hundreds of thousands of deaths, especially among the weakest part of the population. Indeed, especially at the beginning of this pandemic, many reports highlighted how people, suffering from other pathologies, such as hypertension, cardiovascular diseases, and diabetes, are more at risk of severe outcomes if infected. Although this pandemic has put the entire academic world to the test, it has also been a year of intense research and many important contributions have advanced our understanding of SARS-CoV-2 origin, its molecular structure and its mechanism of infection. Unfortunately, despite this great effort, we are still a long way from fully understanding how SARS-CoV-2 dysregulates organismal physiology and whether the current vaccines will be able to protect us from possible future pandemics. Here, we discuss the knowledge we have gained during this year and which questions future research should address.
Collapse
Affiliation(s)
- Filippo Scialo
- Dipartimento di Scienze Mediche Traslazionali, University of Campania “L. Vanvitelli”, 80131 Naples, Italy;
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
| | - Maria Vitale
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Aurora Daniele
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, University of Campania “L. Vanvitelli”, 80131 Naples, Italy
| | - Ersilia Nigro
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, University of Campania “L. Vanvitelli”, 80131 Naples, Italy
| | - Fabio Perrotta
- U.O.C Pneumologia Azienda Ospedaliera Sant’Anna e San Sebastiano, 81100 Caserta, Italy;
| | - Monica Gelzo
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Carlo Iadevaia
- Pneumology Vanvitelly-COVID Unit A.O. dei Colli Hospital Monaldi, 80131 Naples, Italy; (C.I.); (F.S.C.); (A.C.); (V.A.)
| | - Francesco Saverio Cerqua
- Pneumology Vanvitelly-COVID Unit A.O. dei Colli Hospital Monaldi, 80131 Naples, Italy; (C.I.); (F.S.C.); (A.C.); (V.A.)
| | - Adriano Costigliola
- Pneumology Vanvitelly-COVID Unit A.O. dei Colli Hospital Monaldi, 80131 Naples, Italy; (C.I.); (F.S.C.); (A.C.); (V.A.)
| | - Valentino Allocca
- Pneumology Vanvitelly-COVID Unit A.O. dei Colli Hospital Monaldi, 80131 Naples, Italy; (C.I.); (F.S.C.); (A.C.); (V.A.)
| | - Felice Amato
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Lucio Pastore
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Giuseppe Castaldo
- CEINGE, Biotecnologie Avanzate, 80131 Naples, Italy; (M.V.); (A.D.); (E.N.); (M.G.); (F.A.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Andrea Bianco
- Dipartimento di Scienze Mediche Traslazionali, University of Campania “L. Vanvitelli”, 80131 Naples, Italy;
- Pneumology Vanvitelly-COVID Unit A.O. dei Colli Hospital Monaldi, 80131 Naples, Italy; (C.I.); (F.S.C.); (A.C.); (V.A.)
| |
Collapse
|
37
|
Angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers: potential allies in the COVID-19 pandemic instead of a threat? Clin Sci (Lond) 2021; 135:1009-1014. [PMID: 33881142 PMCID: PMC8062870 DOI: 10.1042/cs20210182] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the leading player of the protective renin–angiotensin system (RAS) pathway but also the entry receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). RAS inhibitors seemed to interfere with the ACE2 receptor, and their safety was addressed in COVID-19 patients. Pedrosa et al. (Clin. Sci. (Lond.) (2021), 135, 465–481) showed in rats that captopril and candesartan up-regulated ACE2 expression and the protective RAS pathway in lung tissue. In culture of pneumocytes, the captopril/candesartan-induced ACE2 up-regulation was associated with inhibition of ADAM17 activity, counterbalancing increased ACE2 expression, which was associated with reduced SARS-CoV-2 spike protein entry. If confirmed in humans, these results could become the pathophysiological background for justifying RAS inhibitors as cornerstone cardiovascular protectives even during COVID-19 pandemic.
Collapse
|