101
|
Ljubimov VA, Ramesh A, Davani S, Danielpour M, Breunig JJ, Black KL. Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID-19 pandemic. Adv Drug Deliv Rev 2022; 181:114033. [PMID: 34808227 PMCID: PMC8604570 DOI: 10.1016/j.addr.2021.114033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
Neurosurgery as one of the most technologically demanding medical fields rapidly adapts the newest developments from multiple scientific disciplines for treating brain tumors. Despite half a century of clinical trials, survival for brain primary tumors such as glioblastoma (GBM), the most common primary brain cancer, or rare ones including primary central nervous system lymphoma (PCNSL), is dismal. Cancer therapy and research have currently shifted toward targeted approaches, and personalized therapies. The orchestration of novel and effective blood-brain barrier (BBB) drug delivery approaches, targeting of cancer cells and regulating tumor microenvironment including the immune system are the key themes of this review. As the global pandemic due to SARS-CoV-2 virus continues, neurosurgery and neuro-oncology must wrestle with the issues related to treatment-related immune dysfunction. The selection of chemotherapeutic treatments, even rare cases of hypersensitivity reactions (HSRs) that occur among immunocompromised people, and number of vaccinations they have to get are emerging as a new chapter for modern Nano neurosurgery.
Collapse
Affiliation(s)
- Vladimir A Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | | | | | - Moise Danielpour
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joshua J Breunig
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
102
|
Steiger S, Rossaint J, Zarbock A, Anders HJ. Secondary Immunodeficiency Related to Kidney Disease (SIDKD)-Definition, Unmet Need, and Mechanisms. J Am Soc Nephrol 2022; 33:259-278. [PMID: 34907031 PMCID: PMC8819985 DOI: 10.1681/asn.2021091257] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Kidney disease is a known risk factor for poor outcomes of COVID-19 and many other serious infections. Conversely, infection is the second most common cause of death in patients with kidney disease. However, little is known about the underlying secondary immunodeficiency related to kidney disease (SIDKD). In contrast to cardiovascular disease related to kidney disease, which has triggered countless epidemiologic, clinical, and experimental research activities or interventional trials, investments in tracing, understanding, and therapeutically targeting SIDKD have been sparse. As a call for more awareness of SIDKD as an imminent unmet medical need that requires rigorous research activities at all levels, we review the epidemiology of SIDKD and the numerous aspects of the abnormal immunophenotype of patients with kidney disease. We propose a definition of SIDKD and discuss the pathogenic mechanisms of SIDKD known thus far, including more recent insights into the unexpected immunoregulatory roles of elevated levels of FGF23 and hyperuricemia and shifts in the secretome of the intestinal microbiota in kidney disease. As an ultimate goal, we should aim to develop therapeutics that can reduce mortality due to infections in patients with kidney disease by normalizing host defense to pathogens and immune responses to vaccines.
Collapse
Affiliation(s)
- Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, Ludwig Maximilians University Hospital of Munich, Munich, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Ludwig Maximilians University Hospital of Munich, Munich, Germany
| |
Collapse
|
103
|
Sullivan MK, Lees JS, Drake TM, Docherty AB, Oates G, Hardwick HE, Russell CD, Merson L, Dunning J, Nguyen-Van-Tam JS, Openshaw P, Harrison EM, Baillie JK, Semple MG, Ho A, Mark PB. Acute kidney injury in patients hospitalized with COVID-19 from the ISARIC WHO CCP-UK Study: a prospective, multicentre cohort study. Nephrol Dial Transplant 2022; 37:271-284. [PMID: 34661677 PMCID: PMC8788218 DOI: 10.1093/ndt/gfab303] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is common in coronavirus disease 2019 (COVID-19). This study investigated adults hospitalized with COVID-19 and hypothesized that risk factors for AKI would include comorbidities and non-White race. METHODS A prospective multicentre cohort study was performed using patients admitted to 254 UK hospitals with COVID-19 between 17 January 2020 and 5 December 2020. RESULTS Of 85 687 patients, 2198 (2.6%) received acute kidney replacement therapy (KRT). Of 41 294 patients with biochemistry data, 13 000 (31.5%) had biochemical AKI: 8562 stage 1 (65.9%), 2609 stage 2 (20.1%) and 1829 stage 3 (14.1%). The main risk factors for KRT were chronic kidney disease (CKD) [adjusted odds ratio (aOR) 3.41: 95% confidence interval 3.06-3.81], male sex (aOR 2.43: 2.18-2.71) and Black race (aOR 2.17: 1.79-2.63). The main risk factors for biochemical AKI were admission respiratory rate >30 breaths per minute (aOR 1.68: 1.56-1.81), CKD (aOR 1.66: 1.57-1.76) and Black race (aOR 1.44: 1.28-1.61). There was a gradated rise in the risk of 28-day mortality by increasing severity of AKI: stage 1 aOR 1.58 (1.49-1.67), stage 2 aOR 2.41 (2.20-2.64), stage 3 aOR 3.50 (3.14-3.91) and KRT aOR 3.06 (2.75-3.39). AKI rates peaked in April 2020 and the subsequent fall in rates could not be explained by the use of dexamethasone or remdesivir. CONCLUSIONS AKI is common in adults hospitalized with COVID-19 and it is associated with a heightened risk of mortality. Although the rates of AKI have fallen from the early months of the pandemic, high-risk patients should have their kidney function and fluid status monitored closely.
Collapse
Affiliation(s)
- Michael K Sullivan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jennifer S Lees
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Thomas M Drake
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Annemarie B Docherty
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Georgia Oates
- Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Hayley E Hardwick
- HPRU in Infection and Emerging Diseases, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, UK
| | - Clark D Russell
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Laura Merson
- ISARIC Global Support Centre, University of Oxford, Oxford, UK
| | - Jake Dunning
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | | | - Peter Openshaw
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ewen M Harrison
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, UK
| | | | - Malcolm G Semple
- HPRU in Infection and Emerging Diseases, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, UK
| | - Antonia Ho
- Medical Research Council-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
104
|
Pesce E, Manfrini N, Cordiglieri C, Santi S, Bandera A, Gobbini A, Gruarin P, Favalli A, Bombaci M, Cuomo A, Collino F, Cricrì G, Ungaro R, Lombardi A, Mangioni D, Muscatello A, Aliberti S, Blasi F, Gori A, Abrignani S, De Francesco R, Biffo S, Grifantini R. Exosomes Recovered From the Plasma of COVID-19 Patients Expose SARS-CoV-2 Spike-Derived Fragments and Contribute to the Adaptive Immune Response. Front Immunol 2022; 12:785941. [PMID: 35111156 PMCID: PMC8801440 DOI: 10.3389/fimmu.2021.785941] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by beta-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has rapidly spread across the globe starting from February 2020. It is well established that during viral infection, extracellular vesicles become delivery/presenting vectors of viral material. However, studies regarding extracellular vesicle function in COVID-19 pathology are still scanty. Here, we performed a comparative study on exosomes recovered from the plasma of either MILD or SEVERE COVID-19 patients. We show that although both types of vesicles efficiently display SARS-CoV-2 spike-derived peptides and carry immunomodulatory molecules, only those of MILD patients are capable of efficiently regulating antigen-specific CD4+ T-cell responses. Accordingly, by mass spectrometry, we show that the proteome of exosomes of MILD patients correlates with a proper functioning of the immune system, while that of SEVERE patients is associated with increased and chronic inflammation. Overall, we show that exosomes recovered from the plasma of COVID-19 patients possess SARS-CoV-2-derived protein material, have an active role in enhancing the immune response, and possess a cargo that reflects the pathological state of patients in the acute phase of the disease.
Collapse
Affiliation(s)
- Elisa Pesce
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| | - Nicola Manfrini
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Cordiglieri
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| | - Spartaco Santi
- Unit of Bologna, Consiglio Nazionale delle Ricerche (CNR) Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Bologna, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
| | - Andrea Gobbini
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| | - Paola Gruarin
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| | - Andrea Favalli
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| | - Mauro Bombaci
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| | - Alessandro Cuomo
- Department of Experimental Oncology, Istituto Europeo di Oncologia (IEO), European Institute of Oncology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Federica Collino
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca’ Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - Giulia Cricrì
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca’ Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - Riccardo Ungaro
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Lombardi
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Davide Mangioni
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonio Muscatello
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Aliberti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Respiratory Unit and Cystic Fibrosis Adult Center, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Respiratory Unit and Cystic Fibrosis Adult Center, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
| | - Sergio Abrignani
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Raffaele De Francesco
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefano Biffo
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Renata Grifantini
- Istituto Nazionale Genetica Molecolare (INGM), Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| |
Collapse
|
105
|
Aiello S, Gastoldi S, Galbusera M, Ruggenenti P, Portalupi V, Rota S, Rubis N, Liguori L, Conti S, Tironi M, Gamba S, Santarsiero D, Benigni A, Remuzzi G, Noris M. C5a and C5aR1 are key drivers of microvascular platelet aggregation in clinical entities spanning from aHUS to COVID-19. Blood Adv 2022; 6:866-881. [PMID: 34852172 PMCID: PMC8945302 DOI: 10.1182/bloodadvances.2021005246] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/19/2021] [Indexed: 11/20/2022] Open
Abstract
Unrestrained activation of the complement system till the terminal products, C5a and C5b-9, plays a pathogenetic role in acute and chronic inflammatory diseases. In endothelial cells, complement hyperactivation may translate into cell dysfunction, favoring thrombus formation. The aim of this study was to investigate the role of the C5a/C5aR1 axis as opposed to C5b-9 in inducing endothelial dysfunction and loss of antithrombogenic properties. In vitro and ex vivo assays with serum from patients with atypical hemolytic uremic syndrome (aHUS), a prototype rare disease of complement-mediated microvascular thrombosis due to genetically determined alternative pathway dysregulation, and cultured microvascular endothelial cells, demonstrated that the C5a/C5aR1 axis is a key player in endothelial thromboresistance loss. C5a added to normal human serum fully recapitulated the prothrombotic effects of aHUS serum. Mechanistic studies showed that C5a caused RalA-mediated exocytosis of von Willebrand factor (vWF) and P-selectin from Weibel-Palade bodies, which favored further vWF binding on the endothelium and platelet adhesion and aggregation. In patients with severe COVID-19 who suffered from acute activation of complement triggered by severe acute respiratory syndrome coronavirus 2 infection, we found the same C5a-dependent pathogenic mechanisms. These results highlight C5a/C5aR1 as a common prothrombogenic effector spanning from genetic rare diseases to viral infections, and it may have clinical implications. Selective C5a/C5aR1 blockade could have advantages over C5 inhibition because the former preserves the formation of C5b-9, which is critical for controlling bacterial infections that often develop as comorbidities in severely ill patients. The ACCESS trial registered at www.clinicaltrials.gov as #NCT02464891 accounts for the results related to aHUS patients treated with CCX168.
Collapse
Affiliation(s)
- Sistiana Aiello
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Sara Gastoldi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Miriam Galbusera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Piero Ruggenenti
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Valentina Portalupi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Stefano Rota
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Nadia Rubis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Lucia Liguori
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Sara Conti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Matteo Tironi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Sara Gamba
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Donata Santarsiero
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| | - Marina Noris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; and
| |
Collapse
|
106
|
Augustine R, S A, Nayeem A, Salam SA, Augustine P, Dan P, Maureira P, Mraiche F, Gentile C, Hansbro PM, McClements L, Hasan A. Increased complications of COVID-19 in people with cardiovascular disease: Role of the renin-angiotensin-aldosterone system (RAAS) dysregulation. Chem Biol Interact 2022; 351:109738. [PMID: 34740598 PMCID: PMC8563522 DOI: 10.1016/j.cbi.2021.109738] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 01/28/2023]
Abstract
The rapid spread of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 2019 (COVID-19), has had a dramatic negative impact on public health and economies worldwide. Recent studies on COVID-19 complications and mortality rates suggest that there is a higher prevalence in cardiovascular diseases (CVD) patients. Past investigations on the associations between pre-existing CVDs and susceptibility to coronavirus infections including SARS-CoV and the Middle East Respiratory Syndrome coronavirus (MERS-CoV), have demonstrated similar results. However, the underlying mechanisms are poorly understood. This has impeded adequate risk stratification and treatment strategies for CVD patients with SARS-CoV-2 infections. Generally, dysregulation of the expression of angiotensin-converting enzyme (ACE) and the counter regulator, angiotensin-converting enzyme 2 (ACE2) is a hallmark of cardiovascular risk and CVD. ACE2 is the main host receptor for SARS-CoV-2. Although further studies are required, dysfunction of ACE2 after virus binding and dysregulation of the renin-angiotensin-aldosterone system (RAAS) signaling may worsen the outcomes of people affected by COVID-19 and with preexisting CVD. Here, we review the current knowledge and outline the gaps related to the relationship between CVD and COVID-19 with a focus on the RAAS. Improved understanding of the mechanisms regulating viral entry and the role of RAAS may direct future research with the potential to improve the prevention and management of COVID-19.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| | - Abhilash S
- Department of Microbiology, Majlis Arts and Science College, Puramannur, Malappuram, Kerala, 676552, India
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur, 680020, Kerala, India
| | - Shaheen Abdul Salam
- Department of Biosciences, MES College Marampally, Aluva, Ernakulam, 683107, Kerala, India
| | - Priya Augustine
- Department of Zoology, Kongunadu Arts and Science College, Coimbatore, Tamil Nadu, 641029, India
| | - Pan Dan
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, France; Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Pablo Maureira
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, France
| | - Fatima Mraiche
- College of Pharmacy, QU-Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, Australia; School of Medicine, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
107
|
Hwang YC, Lu RM, Su SC, Chiang PY, Ko SH, Ke FY, Liang KH, Hsieh TY, Wu HC. Monoclonal antibodies for COVID-19 therapy and SARS-CoV-2 detection. J Biomed Sci 2022; 29:1. [PMID: 34983527 PMCID: PMC8724751 DOI: 10.1186/s12929-021-00784-w] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is an exceptional public health crisis that demands the timely creation of new therapeutics and viral detection. Owing to their high specificity and reliability, monoclonal antibodies (mAbs) have emerged as powerful tools to treat and detect numerous diseases. Hence, many researchers have begun to urgently develop Ab-based kits for the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Ab drugs for use as COVID-19 therapeutic agents. The detailed structure of the SARS-CoV-2 spike protein is known, and since this protein is key for viral infection, its receptor-binding domain (RBD) has become a major target for therapeutic Ab development. Because SARS-CoV-2 is an RNA virus with a high mutation rate, especially under the selective pressure of aggressively deployed prophylactic vaccines and neutralizing Abs, the use of Ab cocktails is expected to be an important strategy for effective COVID-19 treatment. Moreover, SARS-CoV-2 infection may stimulate an overactive immune response, resulting in a cytokine storm that drives severe disease progression. Abs to combat cytokine storms have also been under intense development as treatments for COVID-19. In addition to their use as drugs, Abs are currently being utilized in SARS-CoV-2 detection tests, including antigen and immunoglobulin tests. Such Ab-based detection tests are crucial surveillance tools that can be used to prevent the spread of COVID-19. Herein, we highlight some key points regarding mAb-based detection tests and treatments for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Yu-Chyi Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Ruei-Min Lu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Chieh Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Pao-Yin Chiang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Feng-Yi Ke
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Kang-Hao Liang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Tzung-Yang Hsieh
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
108
|
Kajikawa T, Mastellos DC, Hasturk H, Kotsakis GA, Yancopoulou D, Lambris JD, Hajishengallis G. C3-targeted host-modulation approaches to oral inflammatory conditions. Semin Immunol 2022; 59:101608. [PMID: 35691883 DOI: 10.1016/j.smim.2022.101608] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Periodontitis is an inflammatory disease caused by biofilm accumulation and dysbiosis in subgingival areas surrounding the teeth. If not properly treated, this oral disease may result in tooth loss and consequently poor esthetics, deteriorated masticatory function and compromised quality of life. Epidemiological and clinical intervention studies indicate that periodontitis can potentially aggravate systemic diseases, such as, cardiovascular disease, type 2 diabetes mellitus, rheumatoid arthritis, and Alzheimer disease. Therefore, improvements in the treatment of periodontal disease may benefit not only oral health but also systemic health. The complement system is an ancient host defense system that plays pivotal roles in immunosurveillance and tissue homeostasis. However, complement has unwanted consequences if not controlled appropriately or excessively activated. Complement overactivation has been observed in patients with periodontitis and in animal models of periodontitis and drives periodontal inflammation and tissue destruction. This review places emphasis on a promising periodontal host-modulation therapy targeting the complement system, namely the complement C3-targeting drug, AMY-101. AMY-101 has shown safety and efficacy in reducing gingival inflammation in a recent Phase 2a clinical study. We also discuss the potential of AMY-101 to treat peri-implant inflammatory conditions, where complement also seems to be involved and there is an urgent unmet need for effective treatment.
Collapse
Affiliation(s)
- Tetsuhiro Kajikawa
- University of Pennsylvania, Penn Dental Medicine, Department of Basic and Translational Sciences, Philadelphia, PA, USA; Tohoku University Graduate School of Dentistry, Department of Periodontology and Endodontology, Sendai, Miyagi, Japan
| | - Dimitrios C Mastellos
- National Center for Scientific Research 'Demokritos', Division of Biodiagnostic Sciences and Technologies, INRASTES, Athens, Greece
| | - Hatice Hasturk
- The Forsyth Institute, Center for Clinical and Translational Research, Cambridge, MA, USA
| | - Georgios A Kotsakis
- University of Texas Health Science Center at San Antonio, School of Dentistry, Department of Periodontics, San Antonio, TX, USA
| | | | - John D Lambris
- University of Pennsylvania, Perelman School of Medicine, Department of Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - George Hajishengallis
- University of Pennsylvania, Penn Dental Medicine, Department of Basic and Translational Sciences, Philadelphia, PA, USA.
| |
Collapse
|
109
|
Fernández S, Moreno-Castaño AB, Palomo M, Martinez-Sanchez J, Torramadé-Moix S, Téllez A, Ventosa H, Seguí F, Escolar G, Carreras E, Nicolás JM, Richardson E, García-Bernal D, Carlo-Stella C, Moraleda JM, Richardson PG, Díaz-Ricart M, Castro P. Distinctive Biomarker Features in the Endotheliopathy of COVID-19 and Septic Syndromes. Shock 2022; 57:95-105. [PMID: 34172614 PMCID: PMC8662948 DOI: 10.1097/shk.0000000000001823] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Endotheliopathy is a key element in COVID-19 pathophysiology, contributing to both morbidity and mortality. Biomarkers distinguishing different COVID-19 phenotypes from sepsis syndrome remain poorly understood. OBJECTIVE To characterize circulating biomarkers of endothelial damage in different COVID-19 clinical disease stages compared with sepsis syndrome and normal volunteers. METHODS Patients with COVID-19 pneumonia (n = 49) were classified into moderate, severe, or critical (life-threatening) disease. Plasma samples were collected within 48 to 72 h of hospitalization to analyze endothelial activation markers, including soluble Vascular Cell Adhesion Molecule-1 (sVCAM-1), von Willebrand Factor (VWF), A disintegrin-like and metalloprotease with thrombospondin type 1 motif no. 13 (ADAMTS-13) activity, thrombomodulin (TM), and soluble TNF receptor I (sTNFRI); heparan sulfate (HS) for endothelial glycocalyx degradation; C5b9 deposits on endothelial cells in culture and soluble C5b9 for complement activation; circulating dsDNA for neutrophil extracellular traps (NETs) presence, and α2-antiplasmin and PAI-1 as parameters of fibrinolysis. We compared the level of each biomarker in all three COVID-19 groups and healthy donors as controls (n = 45). Results in critically ill COVID-19 patients were compared with other intensive care unit (ICU) patients with septic shock (SS, n = 14), sepsis (S, n = 7), and noninfectious systemic inflammatory response syndrome (NI-SIRS, n = 7). RESULTS All analyzed biomarkers were increased in COVID-19 patients versus controls (P < 0.001), except for ADAMTS-13 activity that was normal in both groups. The increased expression of sVCAM-1, VWF, sTNFRI, and HS was related to COVID-19 disease severity (P < 0.05). Several differences in these parameters were found between ICU groups: SS patients showed significantly higher levels of VWF, TM, sTNFRI, and NETS compared with critical COVID-19 patients and ADAMTS-13 activity was significantly lover in SS, S, and NI-SIRS versus critical COVID-19 (P < 0.001). Furthermore, α2-antiplasmin activity was higher in critical COVID-19 versus NI-SIRS (P < 0.01) and SS (P < 0.001), whereas PAI-1 levels were significantly lower in COVID-19 patients compared with NI-SIRS, S, and SS patients (P < 0.01). CONCLUSIONS COVID-19 patients present with increased circulating endothelial stress products, complement activation, and fibrinolytic dysregulation, associated with disease severity. COVID-19 endotheliopathy differs from SS, in which endothelial damage is also a critical feature of pathobiology. These biomarkers could help to stratify the severity of COVID-19 disease and may also provide information to guide specific therapeutic strategies to mitigate endotheliopathy progression.
Collapse
Affiliation(s)
- Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Ana B. Moreno-Castaño
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Marta Palomo
- Barcelona Endothelium Team, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Barcelona Endothelium Team, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Sergi Torramadé-Moix
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Adrián Téllez
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Helena Ventosa
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Ferran Seguí
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Ginés Escolar
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Enric Carreras
- Barcelona Endothelium Team, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Josep M. Nicolás
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| | - Edward Richardson
- Frank H. Netter M.D. School of Medicine at Quinnipiac University, North Haven, Connecticut
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - David García-Bernal
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Carmelo Carlo-Stella
- Department of Oncology and Hematology, Humanitas Clinical and Research Center-IRCCS, Rozzano-Milano, Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano-Milano, Italy
| | - José M. Moraleda
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Paul G. Richardson
- Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Harvard Medical School, Boston, Massachusetts
| | - Maribel Díaz-Ricart
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
110
|
Tiwari V, Bhandari G, Gupta A, Gupta P, Bhargava V, Malik M, Gupta A, Bhalla A, Rana D. Atypical HUS triggered by COVID-19: A case report. Indian J Nephrol 2022; 32:367-370. [PMID: 35967527 PMCID: PMC9364994 DOI: 10.4103/ijn.ijn_196_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/19/2021] [Accepted: 08/20/2021] [Indexed: 11/04/2022] Open
Abstract
We hereby present a case of an atypical hemolytic uremic syndrome (aHUS) precipitated by coronavirus disease 2019 (COVID-19). A 26-year-old male was diagnosed with COVID-19 and acute kidney injury. His kidney biopsy was suggestive of thrombotic microangiopathy. Five sessions of plasmapheresis were done but were discontinued in view of nonrecovery of kidney function. He was then referred for a kidney transplant. On genetic analysis, he was found to have mutations in the complement system (CFHR1 and CFHR3), which suggested this was a case of aHUS precipitated by COVID-19. In view of the high risk of recurrence of the primary disease in live-related kidney donor transplantation, he was advised for simultaneous liver and kidney transplants.
Collapse
|
111
|
Shi K, Liu Y, Zhang Q, Ran CP, Hou J, Zhang Y, Wang XB. Severe Type of COVID-19: Pathogenesis, Warning Indicators and Treatment. Chin J Integr Med 2021; 28:3-11. [PMID: 34962616 PMCID: PMC8713541 DOI: 10.1007/s11655-021-3313-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, is a major public health issue. The epidemic is unlikely to be contained until the global launch of safe and effective vaccines that could prevent serious illnesses and provide herd immunity. Although most patients have mild flu-like symptoms, some develop severe illnesses accompanied by multiple organ dysfunction. The identification of pathophysiology and early warning biomarkers of a severe type of COVID-19 contribute to the treatment and prevention of serious complications. Here, we review the pathophysiology, early warning indicators, and effective treatment of Chinese and Western Medicine for patients with a severe type of COVID-19.
Collapse
Affiliation(s)
- Ke Shi
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
| | - Yao Liu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Qun Zhang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Chong-Ping Ran
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Jie Hou
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
| | - Yi Zhang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Xian-Bo Wang
- Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China.
| |
Collapse
|
112
|
Odilov A, Volkov A, Abdullaev A, Gasanova T, Lipina T, Babichenko I. COVID-19: Multiorgan Dissemination of SARS-CoV-2 Is Driven by Pulmonary Factors. Viruses 2021; 14:v14010039. [PMID: 35062243 PMCID: PMC8777766 DOI: 10.3390/v14010039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Multi-organ failure is one of the common causes of fatal outcome in COVID-19 patients. However, the pathogenetic association of the SARS-CoV-2 viral load (VL) level with fatal dysfunctions of the lungs, liver, kidneys, heart, spleen and brain, as well as with the risk of death in COVID-19 patients remains poorly understood. SARS-CoV-2 VL in the lungs, heart, liver, kidneys, brain, spleen and lymph nodes have been measured by RT qPCR using the following formula: NSARS-CoV-2/NABL1 × 100. Dissemination of SARS-CoV-2 in 30.5% of cases was mono-organ, and in 63.9% of cases, it was multi-organ. The average SARS-CoV-2 VL in the exudative phase of diffuse alveolar damage (DAD) was 60 times higher than in the proliferative phase. The SARS-CoV-2 VL in the lungs ranged from 0 to 250,281 copies. The "pulmonary factors" of SARS-CoV-2 multi-organ dissemination are the high level of SARS-CoV-2 VL (≥4909) and the exudative phase of DAD. The frequency of SARS-CoV-2 dissemination to lymph nodes was 86.9%, heart-56.5%, spleen-52.2%, liver-47.8%, kidney-26%, and brain-13%. We found no link between the SARS-CoV-2 VL level in the liver, kidneys, and heart and the serum level of CPK, LDH, ALP, ALT, AST and Cr of COVID-19 patients. Isolated detection of SARS-CoV-2 RNA in the myocardium of COVID-19 patients who died from heart failure is possible. The pathogenesis of COVID-19-associated multi-organ failure requires further research in a larger cohort of patients.
Collapse
Affiliation(s)
- Akmaljon Odilov
- Department of Pathological Anatomy, Peoples′ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russia; (A.V.); (I.B.)
- Correspondence:
| | - Alexey Volkov
- Department of Pathological Anatomy, Peoples′ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russia; (A.V.); (I.B.)
- Department of Pathological Anatomy, Municipal Clinical Hospital Named after E.O. Mukhin, Moscow 111399, Russia
| | - Adhamjon Abdullaev
- Laboratory of Molecular Hematology, National Research Center for Hematology, Novy Zykovski lane 4a, Moscow 125167, Russia;
| | - Tatiana Gasanova
- Department of Virology, Lomonosov Moscow State University, Leninskie gori, 1, 40, Moscow 119234, Russia;
| | - Tatiana Lipina
- Department of Cell Biology and Histology, Faculty of Biology, Lomonosov Moscow State University, Leninskie gori, 1, 12, Moscow 119234, Russia;
| | - Igor Babichenko
- Department of Pathological Anatomy, Peoples′ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russia; (A.V.); (I.B.)
| |
Collapse
|
113
|
Ruggenenti P, Di Marco F, Cortinovis M, Lorini L, Sala S, Novelli L, Raimondi F, Gastoldi S, Galbusera M, Donadelli R, Mele C, Piras R, Noris M, Portalupi V, Cappelletti L, Carrara C, Tomatis F, Bernardi S, Perna A, Peracchi T, Diadei O, Benigni A, Remuzzi G. Eculizumab in patients with severe coronavirus disease 2019 (COVID-19) requiring continuous positive airway pressure ventilator support: Retrospective cohort study. PLoS One 2021; 16:e0261113. [PMID: 34928990 PMCID: PMC8687582 DOI: 10.1371/journal.pone.0261113] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/24/2021] [Indexed: 12/30/2022] Open
Abstract
Background Complement activation contributes to lung dysfunction in coronavirus disease 2019 (COVID-19). We assessed whether C5 blockade with eculizumab could improve disease outcome. Methods In this single-centre, academic, unblinded study two 900 mg eculizumab doses were added-on standard therapy in ten COVID-19 patients admitted from February 2020 to April 2020 and receiving Continuous-Positive-Airway-Pressure (CPAP) ventilator support from ≤24 hours. We compared their outcomes with those of 65 contemporary similar controls. Primary outcome was respiratory rate at one week of ventilator support. Secondary outcomes included the combined endpoint of mortality and discharge with chronic complications. Results Baseline characteristics of eculizumab-treated patients and controls were similar. At baseline, sC5b-9 levels, ex vivo C5b-9 and thrombi deposition were increased. Ex vivo tests normalised in eculizumab-treated patients, but not in controls. In eculizumab-treated patients respiratory rate decreased from 26.8±7.3 breaths/min at baseline to 20.3±3.8 and 18.0±4.8 breaths/min at one and two weeks, respectively (p<0.05 for both), but did not change in controls. Between-group changes differed significantly at both time-points (p<0.01). Changes in respiratory rate correlated with concomitant changes in ex vivo C5b-9 deposits at one (rs = 0.706, p = 0.010) and two (rs = 0.751, p = 0.032) weeks. Over a median (IQR) period of 47.0 (14.0–121.0) days, four eculizumab-treated patients died or had chronic complications versus 52 controls [HRCrude (95% CI): 0.26 (0.09–0.72), p = 0.010]. Between-group difference was significant even after adjustment for age, sex and baseline serum creatinine [HRAdjusted (95% CI): 0.30 (0.10–0.84), p = 0.023]. Six patients and 13 controls were discharged without complications [HRCrude (95% CI): 2.88 (1.08–7.70), p = 0.035]. Eculizumab was tolerated well. The main study limitations were the relatively small sample size and the non-randomised design. Conclusions In patients with severe COVID-19, eculizumab safely improved respiratory dysfunction and decreased the combined endpoint of mortality and discharge with chronic complications. Findings need confirmation in randomised controlled trials.
Collapse
Affiliation(s)
- Piero Ruggenenti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Fabiano Di Marco
- Unit of Pulmonary Medicine, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Monica Cortinovis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Luca Lorini
- Intensive Care Unit, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Silvia Sala
- Intensive Care Unit, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Luca Novelli
- Unit of Pulmonary Medicine, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Federico Raimondi
- Unit of Pulmonary Medicine, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Sara Gastoldi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Miriam Galbusera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Roberta Donadelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Caterina Mele
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Rossella Piras
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marina Noris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Valentina Portalupi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Laura Cappelletti
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Camillo Carrara
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Federica Tomatis
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
- School of Nephrology, Università degli Studi di Milano, Milan, Italy
| | - Silvia Bernardi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
- School of Nephrology, Università degli Studi di Milano, Milan, Italy
| | - Annalisa Perna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Tobia Peracchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Olimpia Diadei
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- * E-mail:
| |
Collapse
|
114
|
Xie TA, He ZJ, Liang C, Dong HN, Zhou J, Fan SJ, Guo XG. An integrative bioinformatics analysis for identifying hub genes associated with infection of lung samples in patients infected with SARS-CoV-2. Eur J Med Res 2021; 26:146. [PMID: 34920753 PMCID: PMC8677925 DOI: 10.1186/s40001-021-00609-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/14/2021] [Indexed: 12/22/2022] Open
Abstract
Background At the end of 2019, the world witnessed the emergence and ravages of a viral infection induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Also known as the coronavirus disease 2019 (COVID-19), it has been identified as a public health emergency of international concern (PHEIC) by the World Health Organization (WHO) because of its severity. Methods The gene data of 51 samples were extracted from the GSE150316 and GSE147507 data set and then processed by means of the programming language R, through which the differentially expressed genes (DEGs) that meet the standards were screened. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed on the selected DEGs to understand the functions and approaches of DEGs. The online tool STRING was employed to construct a protein–protein interaction (PPI) network of DEGs and, in turn, to identify hub genes. Results A total of 52 intersection genes were obtained through DEG identification. Through the GO analysis, we realized that the biological processes (BPs) that have the deepest impact on the human body after SARS-CoV-2 infection are various immune responses. By using STRING to construct a PPI network, 10 hub genes were identified, including IFIH1, DDX58, ISG15, EGR1, OASL, SAMD9, SAMD9L, XAF1, IFITM1, and TNFSF10. Conclusion The results of this study will hopefully provide guidance for future studies on the pathophysiological mechanism of SARS-CoV-2 infection. Supplementary Information The online version contains supplementary material available at 10.1186/s40001-021-00609-4.
Collapse
|
115
|
Senent Y, Inogés S, López-Díaz de Cerio A, Blanco A, Campo A, Carmona-Torre F, Sunsundegui P, González-Martín A, Ajona D, Okrój M, Prósper F, Pio R, Yuste JR, Tavira B. Persistence of High Levels of Serum Complement C5a in Severe COVID-19 Cases After Hospital Discharge. Front Immunol 2021; 12:767376. [PMID: 34868021 PMCID: PMC8636747 DOI: 10.3389/fimmu.2021.767376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/29/2021] [Indexed: 01/08/2023] Open
Abstract
Evidence supports a role of complement anaphylatoxin C5a in the pathophysiology of COVID-19. However, information about the evolution and impact of C5a levels after hospital discharge is lacking. We analyzed the association between circulating C5a levels and the clinical evolution of hospitalized patients infected with SARS-CoV-2. Serum C5a levels were determined in 32 hospitalized and 17 non-hospitalized patients from Clinica Universidad de Navarra. One hundred and eighty eight serial samples were collected during the hospitalization stay and up to three months during the follow-up. Median C5a levels were 27.71 ng/ml (25th to 75th percentile: 19.35-34.96) for samples collected during hospitalization, versus 16.76 ng/ml (12.90-25.08) for samples collected during the follow-up (p<0.001). There was a negative correlation between serum C5a levels and the number of days from symptom onset (p<0.001). C5a levels also correlated with a previously validated clinical risk score (p<0.001), and was associated with the severity of the disease (p<0.001). An overall reduction of C5a levels was observed after hospital discharge. However, elevated C5a levels persisted in those patients with high COVID-19 severity (i.e. those with a longest stay in the hospital), even after months from hospital discharge (p=0.020). Moreover, high C5a levels appeared to be associated with the presence of long-term respiratory symptoms (p=0.004). In conclusion, serum C5a levels remain high in severe cases of COVID-19, and are associated with the presence of respiratory symptoms after hospital discharge. These results may suggest a role for C5a in the long-term effects of COVID-19 infection.
Collapse
Affiliation(s)
- Yaiza Senent
- Program in Solid Tumors, Translational Oncology Group, Cima-University of Navarra, Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.,Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Susana Inogés
- Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, Clinica Universidad de Navarra, Pamplona, Spain.,Area of Cell Therapy and Department of Hematology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Ascensión López-Díaz de Cerio
- Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, Clinica Universidad de Navarra, Pamplona, Spain.,Area of Cell Therapy and Department of Hematology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Andres Blanco
- Department of Internal Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - Arantxa Campo
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Francisco Carmona-Torre
- Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Department of Internal Medicine, Clinica Universidad de Navarra, Pamplona, Spain.,Division of Infectious Diseases, Clinica Universidad de Navarra, Pamplona, Spain
| | - Patricia Sunsundegui
- Department of Internal Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - Antonio González-Martín
- Program in Solid Tumors, Translational Oncology Group, Cima-University of Navarra, Pamplona, Spain.,Department of Oncology, Clinica Universidad de Navarra, Madrid, Spain
| | - Daniel Ajona
- Program in Solid Tumors, Translational Oncology Group, Cima-University of Navarra, Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.,Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Program in Respiratory Tract Tumors, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Marcin Okrój
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Felipe Prósper
- Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, Clinica Universidad de Navarra, Pamplona, Spain.,Program in Respiratory Tract Tumors, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Program of Regenerative Medicine, Cima-University of Navarra, Pamplona, Spain
| | - Ruben Pio
- Program in Solid Tumors, Translational Oncology Group, Cima-University of Navarra, Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.,Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Program in Respiratory Tract Tumors, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - José Ramón Yuste
- Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Department of Internal Medicine, Clinica Universidad de Navarra, Pamplona, Spain.,Division of Infectious Diseases, Clinica Universidad de Navarra, Pamplona, Spain
| | - Beatriz Tavira
- Program in Solid Tumors, Translational Oncology Group, Cima-University of Navarra, Pamplona, Spain.,Respiratory Tract Cancer Group, Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| |
Collapse
|
116
|
Agostinis C, Mangogna A, Balduit A, Aghamajidi A, Ricci G, Kishore U, Bulla R. COVID-19, Pre-Eclampsia, and Complement System. Front Immunol 2021; 12:775168. [PMID: 34868042 PMCID: PMC8635918 DOI: 10.3389/fimmu.2021.775168] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is characterized by virus-induced injury leading to multi-organ failure, together with inflammatory reaction, endothelial cell (EC) injury, and prothrombotic coagulopathy with thrombotic events. Complement system (C) via its cross-talk with the contact and coagulation systems contributes significantly to the severity and pathological consequences due to SARS-CoV-2 infection. These immunopathological mechanisms overlap in COVID-19 and pre-eclampsia (PE). Thus, mothers contracting SARS-CoV-2 infection during pregnancy are more vulnerable to developing PE. SARS-CoV-2 infection of ECs, via its receptor ACE2 and co-receptor TMPRSS2, can provoke endothelial dysfunction and disruption of vascular integrity, causing hyperinflammation and hypercoagulability. This is aggravated by bradykinin increase due to inhibition of ACE2 activity by the virus. C is important for the progression of normal pregnancy, and its dysregulation can impact in the form of PE-like syndrome as a consequence of SARS-CoV-2 infection. Thus, there is also an overlap between treatment regimens of COVID-19 and PE. C inhibitors, especially those targeting C3 or MASP-2, are exciting options for treating COVID-19 and consequent PE. In this review, we examine the role of C, contact and coagulation systems as well as endothelial hyperactivation with respect to SARS-CoV-2 infection during pregnancy and likely development of PE.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Andrea Balduit
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy.,Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
117
|
Patrucco F, Zeppegno P, Baricich A, Gramaglia CM, Balbo PE, Falaschi Z, Carriero A, Cuneo D, Pirisi M, Bellan M. Long-lasting consequences of Coronavirus disease 19 pneumonia: a systematic review. Minerva Med 2021; 113:158-171. [PMID: 34856780 DOI: 10.23736/s0026-4806.21.07594-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Coronavirus Disease 19 (Covid-19) is an infectious disease caused by the newly discovered severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We have plenty of data about the clinical features of the disease's acute phase, while little is known about the long-term consequences on survivors. EVIDENCE ACQUISITION We aimed to review systematically emerging evidence about clinical and functional consequences of Covid-19 pneumonia months after hospital discharge. EVIDENCE SYNTHESIS Current evidence supports the idea that a high proportion of Covid-19 survivors complain of symptoms months after the acute illness phase, being fatigue and reduced tolerance to physical effort the most frequently reported symptom. The strongest association for these symptoms is with the female gender, while disease severity seems less relevant. Respiratory symptoms are associated with a decline in respiratory function and, conversely, seem to be more frequent in those who experienced a more severe acute pneumonia. Current evidence highlighted a persistent motor impairment which is, again, more prevalent among those survivors who experienced a more severe acute phase of the disease. Additionally, the persistence of symptoms is a primary determinant of mental health outcome, with anxiety, depression, sleep disturbances, and post-traumatic stress symptoms being commonly reported in Covid-19 survivors. CONCLUSIONS Current literature highlights the importance of a multidisciplinary approach to Coronavirus Disease 19 since the sequelae appear to involve different organs and systems. Given the pandemic outbreak's size, this is a critical public health issue: a better insight on this topic should inform clinical decisions about the modalities of follow-up for Covid-19 survivors.
Collapse
Affiliation(s)
- Filippo Patrucco
- Pneumology Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Patrizia Zeppegno
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Psychiatry Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Alessio Baricich
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy - .,Physical and Rehabilitation Medicine, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Carla M Gramaglia
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Psychiatry Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Piero E Balbo
- Pneumology Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Zeno Falaschi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Radiology Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Alessandro Carriero
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Radiology Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Daria Cuneo
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.,Physical and Rehabilitation Medicine, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Internal Medicine Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Internal Medicine Department, Ospedale Maggiore della Carità University Hospital, Novara, Italy
| |
Collapse
|
118
|
Verma S, Chaturvedi V, Ganguly NK, Mittal SA. Vitamin D deficiency: concern for rheumatoid arthritis and COVID-19? Mol Cell Biochem 2021; 476:4351-4362. [PMID: 34453644 PMCID: PMC8401347 DOI: 10.1007/s11010-021-04245-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Vitamin D is an immunomodulatory hormone with an established role in calcium and phosphate metabolism and skeletal mineralization. Evidence showing its immunological benefits by regulating essential components of the innate and adaptive immune system is prevalent. Vitamin D deficiency is reported worldwide and is thereby found to be associated with various immune-related diseases. Rheumatoid Arthritis and COVID-19 are two such diseases, sharing a similar hyperinflammatory response. Various studies have found an association of lower Vitamin D levels to be associated with both these diseases. However, contrasting data is also reported. We review here the available scientific data on risk factor association and supplementation benefits of Vitamin D in Rheumatoid Arthritis and COVID-19, intending to critically evaluate the literature.
Collapse
Affiliation(s)
- Sneha Verma
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Ved Chaturvedi
- Department of Rheumatology & Clinical Immunology, Sir Ganga Ram Hospital, New Delhi, India
| | - N K Ganguly
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | | |
Collapse
|
119
|
To KKW, Sridhar S, Chiu KHY, Hung DLL, Li X, Hung IFN, Tam AR, Chung TWH, Chan JFW, Zhang AJX, Cheng VCC, Yuen KY. Lessons learned 1 year after SARS-CoV-2 emergence leading to COVID-19 pandemic. Emerg Microbes Infect 2021; 10:507-535. [PMID: 33666147 PMCID: PMC8006950 DOI: 10.1080/22221751.2021.1898291] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 02/06/2023]
Abstract
Without modern medical management and vaccines, the severity of the Coronavirus Disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) might approach the magnitude of 1894-plague (12 million deaths) and 1918-A(H1N1) influenza (50 million deaths) pandemics. The COVID-19 pandemic was heralded by the 2003 SARS epidemic which led to the discovery of human and civet SARS-CoV-1, bat SARS-related-CoVs, Middle East respiratory syndrome (MERS)-related bat CoV HKU4 and HKU5, and other novel animal coronaviruses. The suspected animal-to-human jumping of 4 betacoronaviruses including the human coronaviruses OC43(1890), SARS-CoV-1(2003), MERS-CoV(2012), and SARS-CoV-2(2019) indicates their significant pandemic potential. The presence of a large reservoir of coronaviruses in bats and other wild mammals, culture of mixing and selling them in urban markets with suboptimal hygiene, habit of eating exotic mammals in highly populated areas, and the rapid and frequent air travels from these areas are perfect ingredients for brewing rapidly exploding epidemics. The possibility of emergence of a hypothetical SARS-CoV-3 or other novel viruses from animals or laboratories, and therefore needs for global preparedness should not be ignored. We reviewed representative publications on the epidemiology, virology, clinical manifestations, pathology, laboratory diagnostics, treatment, vaccination, and infection control of COVID-19 as of 20 January 2021, which is 1 year after person-to-person transmission of SARS-CoV-2 was announced. The difficulties of mass testing, labour-intensive contact tracing, importance of compliance to universal masking, low efficacy of antiviral treatment for severe disease, possibilities of vaccine or antiviral-resistant virus variants and SARS-CoV-2 becoming another common cold coronavirus are discussed.
Collapse
Affiliation(s)
- Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kelvin Hei-Yeung Chiu
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Derek Ling-Lung Hung
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Xin Li
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Anthony Raymond Tam
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Tom Wai-Hin Chung
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Anna Jian-Xia Zhang
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Vincent Chi-Chung Cheng
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| |
Collapse
|
120
|
Keshavarz F, Ghalamfarsa F, Javdansirat S, Hasanzadeh S, Azizi A, Sabz G, Salehi M, Ghalamfarsa G. Patients with Covid 19 have significantly reduced CH50 activity. Virusdisease 2021; 32:681-689. [PMID: 34631971 PMCID: PMC8486960 DOI: 10.1007/s13337-021-00710-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), is a new virus that emerged in China and immediately spread around the world. Evidence has been documented that the immune system is impressively involved in the pathogenesis of this disease, especially in causing inflammation. One of the important components of the immune system is the complement system whose increased activity has been shown in inflammatory diseases and consequently damage caused by the activity of its components. In the present study, serum levels of C3 and C4 factors as well as the activity level of complement system in the classical pathway were measured by CH50 test in patients with SARS-CoV-2. Participants in the study consisted of 53 hospitalized patients whose real-time PCR test was positive for SARS-CoV-2. The mean age of these patients was 42.06 ± 18.7 years, including 40% women and 60% men. The most common symptoms in these patients were cough (70%), fever (59%), dyspnea (53%) and chills (53%), respectively. Analysis of biochemical and hematological test results revealed that 26 (49%) patients had lymphopenia, 34 (64%) patients were positive for C-reactive protein (CRP) and 26 (49%) patients had ESR and LDH levels significantly higher than normal. In addition, 27 patients (51%) had vitamin D deficiency. The mean CH50 activity level in COVID-19 patients was significantly reduced compared to healthy individuals (84.9 versus 169.9 U/ml, p = < 0.0001). Comparison of the mean CH50 activity levels between different subgroups of patients indicated that COVID-19 patients with decreased peripheral blood lymphocyte count and positive CRP had a significant increase in activity compared to the other groups (p = 0.0002). The serum levels of C3 and C4 factors had no significant change between patients and healthy individuals. Conclusion: The activity level of complement system in the classical pathway decreases in COVID-19 patients compared to healthy individuals, due to increased activity of complement system factors in these patients.
Collapse
Affiliation(s)
- Fatemeh Keshavarz
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farideh Ghalamfarsa
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Javdansirat
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Sajad Hasanzadeh
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Arsalan Azizi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Gholamabbas Sabz
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Marziyeh Salehi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ghasem Ghalamfarsa
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
121
|
Abstract
PURPOSE OF REVIEW This review examines the global literature regarding rashes encountered in children and adults infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and aims to provide practicing pediatricians with an understanding of the relationship between instances of rashes and coronavirus disease 2019 (COVID-19) in children in order to effectively evaluate and treat patients. RECENT FINDINGS The true incidence of cutaneous reactions in children infected with SARS-CoV-2 is not known. Children's immune systems differ from those of adults and rashes as a manifestation of immune responses, in turn, differ in morphology and distribution. Rarely, children develop a severe multisystem inflammatory syndrome that has overlapping clinical features with Kawasaki disease. In addition, vaccinations produce rashes similar to natural infections. The rashes associated with COVID-19 vaccination are mild and transient, and should not preclude vaccination. Lastly, children who chronically wear masks are more likely to experience flaring of acne around the nose and mouth ('maskne') and facial conditions such as seborrheic dermatitis. SUMMARY There are ongoing worldwide registries, clinical and basic science studies to better understand the burden of skin disease and pathophysiology of rashes seen in patients infected with COVID-19. Robust vaccination programs should be encouraged as a way to contain viral spread among children and the greater population.
Collapse
Affiliation(s)
| | - James Gary Dinulos
- Seacoast Dermatology, PLLC, Portsmouth
- Department of Dermatology, Geisel School of Medicine at Dartmouth, Hanover New Hampshire
- Department of Dermatology, University of Connecticut School of Medicine, Framingham, Connecticut, USA
| |
Collapse
|
122
|
Gallo CG, Fiorino S, Posabella G, Antonacci D, Tropeano A, Pausini E, Pausini C, Guarniero T, Hong W, Giampieri E, Corazza I, Federico L, de Biase D, Zippi M, Zancanaro M. COVID-19, what could sepsis, severe acute pancreatitis, gender differences, and aging teach us? Cytokine 2021; 148:155628. [PMID: 34411989 PMCID: PMC8343368 DOI: 10.1016/j.cyto.2021.155628] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/02/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a potentially life-threatening disease, defined as Coronavirus Disease 19 (COVID-19). The most common signs and symptoms of this pathological condition include cough, fever, shortness of breath, and sudden onset of anosmia, ageusia, or dysgeusia. The course of COVID-19 is mild or moderate in more than 80% of cases, but it is severe or critical in about 14% and 5% of infected subjects respectively, with a significant risk of mortality. SARS-CoV-2 related infection is characterized by some pathogenetic events, resembling those detectable in other pathological conditions, such as sepsis and severe acute pancreatitis. All these syndromes are characterized by some similar features, including the coexistence of an exuberant inflammatory- as well as an anti-inflammatory-response with immune depression. Based on current knowledge concerning the onset and the development of acute pancreatitis and sepsis, we have considered these syndromes as a very interesting paradigm for improving our understanding of pathogenetic events detectable in patients with COVID-19. The aim of our review is: 1)to examine the pathogenetic mechanisms acting during the emergence of inflammatory and anti-inflammatory processes in human pathology; 2)to examine inflammatory and anti-inflammatory events in sepsis, acute pancreatitis, and SARS-CoV-2 infection and clinical manifestations detectable in patients suffering from these syndromes also according to the age and gender of these individuals; as well as to analyze the possible common and different features among these pathological conditions; 3)to obtain insights into our knowledge concerning COVID-19 pathogenesis. This approach may improve the management of patients suffering from this disease and it may suggest more effective diagnostic approaches and schedules of therapy, depending on the different phases and/or on the severity of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Claudio G Gallo
- Emilian Physiolaser Therapy Center, Castel S. Pietro Terme, Bologna, Italy.
| | - Sirio Fiorino
- Internal Medicine Unit, Budrio Hospital Azienda USL, Bologna, Italy
| | | | - Donato Antonacci
- Medical Science Department, "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo (FG), Italy
| | | | | | | | | | - Wandong Hong
- Department of Gastroenterology and Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang, The People's Republic of China
| | - Enrico Giampieri
- Experimental, Diagnostic and Specialty Medicine Department, University of Bologna, Bologna, Italy
| | - Ivan Corazza
- Experimental, Diagnostic and Specialty Medicine Department, University of Bologna, Bologna, Italy
| | - Lari Federico
- Internal Medicine Unit, Budrio Hospital Azienda USL, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Maddalena Zippi
- Unit of Gastroenterology and Digestive Endoscopy, Sandro Pertini Hospital, Rome, Italy
| | | |
Collapse
|
123
|
Al-Gburi S, Beissert S, Günther C. Molecular mechanisms of vasculopathy and coagulopathy in COVID-19. Biol Chem 2021; 402:1505-1518. [PMID: 34657406 DOI: 10.1515/hsz-2021-0245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/06/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 primarily affects the respiratory system and may lead to severe systemic complications, such as acute respiratory distress syndrome (ARDS), multiple organ failure, cytokine storm, and thromboembolic events. Depending on the immune status of the affected individual early disease control can be reached by a robust type-I-interferon (type-I-IFN) response restricting viral replication. If type-I-IFN upregulation is impaired, patients develop severe COVID-19 that involves profound alveolitis, endothelitis, complement activation, recruitment of immune cells, as well as immunothrombosis. In patients with proper initial disease control there can be a second flare of type-I-IFN release leading to post-COVID manifestation such as chilblain-like lesions that are characterized by thrombosis of small vessels in addition to an inflammatory infiltrate resembling lupus erythematosus (LE). Mechanistically, SARS-CoV-2 invades pneumocytes and endothelial cells by acting on angiotensin-II-converting enzyme 2 (ACE2). It is hypothesized, that viral uptake might downregulate ACE2 bioavailability and enhance angiotensin-II-derived pro-inflammatory and pro-thrombotic state. Since ACE2 is encoded on the X chromosome these conditions might also be influenced by gender-specific regulation. Taken together, SARS-CoV-2 infection affects the vascular compartment leading to variable thrombogenic or inflammatory response depending on the individual immune response status.
Collapse
Affiliation(s)
- Suzan Al-Gburi
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Stefan Beissert
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Claudia Günther
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| |
Collapse
|
124
|
Jarlhelt I, Nielsen SK, Jahn CXH, Hansen CB, Pérez-Alós L, Rosbjerg A, Bayarri-Olmos R, Skjoedt MO, Garred P. SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation. Front Immunol 2021; 12:767981. [PMID: 34804055 PMCID: PMC8596567 DOI: 10.3389/fimmu.2021.767981] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to constitute a serious public health threat worldwide. Protective antibody-mediated viral neutralization in response to SARS-CoV-2 infection has been firmly characterized. Where the effects of the antibody response are generally considered to be beneficial, an important biological question regarding potential negative outcomes of a SARS-CoV-2 antibody response has yet to be answered. We determined the distribution of IgG subclasses and complement activation levels in plasma from convalescent individuals using in-house developed ELISAs. The IgG response towards SARS-CoV-2 receptor-binding domain (RBD) after natural infection appeared to be mainly driven by IgG1 and IgG3 subclasses, which are the main ligands for C1q mediated classical complement pathway activation. The deposition of the complement components C4b, C3bc, and TCC as a consequence of SARS-CoV-2 specific antibodies were depending primarily on the SARS-CoV-2 RBD and significantly correlated with both IgG levels and disease severity, indicating that individuals with high levels of IgG and/or severe disease, might have a more prominent complement activation during viral infection. Finally, freshly isolated monocytes and a monocyte cell line (THP-1) were used to address the cellular mediated inflammatory response as a consequence of Fc-gamma receptor engagement by SARS-CoV-2 specific antibodies. Monocytic Fc gamma receptor charging resulted in a significant rise in the secretion of the pro-inflammatory cytokine TNF-α. Our results indicate that SARS-CoV-2 antibodies might drive significant inflammatory responses through the classical complement pathway and via cellular immune-complex activation that could have negative consequences during COVID-19 disease. We found that increased classical complement activation was highly associated to COVID-19 disease severity. The combination of antibody-mediated complement activation and subsequent cellular priming could constitute a significant risk of exacerbating COVID-19 severity.
Collapse
Affiliation(s)
- Ida Jarlhelt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sif Kaas Nielsen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Camilla Xenia Holtermann Jahn
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anne Rosbjerg
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Recombinant Protein and Antibody Laboratory, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Recombinant Protein and Antibody Laboratory, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mikkel-Ole Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Institute of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
125
|
Savitt AG, Manimala S, White T, Fandaros M, Yin W, Duan H, Xu X, Geisbrecht BV, Rubenstein DA, Kaplan AP, Peerschke EI, Ghebrehiwet B. SARS-CoV-2 Exacerbates COVID-19 Pathology Through Activation of the Complement and Kinin Systems. Front Immunol 2021; 12:767347. [PMID: 34804054 PMCID: PMC8602850 DOI: 10.3389/fimmu.2021.767347] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Infection with SARS-CoV-2 triggers the simultaneous activation of innate inflammatory pathways including the complement system and the kallikrein-kinin system (KKS) generating in the process potent vasoactive peptides that contribute to severe acute respiratory syndrome (SARS) and multi-organ failure. The genome of SARS-CoV-2 encodes four major structural proteins - the spike (S) protein, nucleocapsid (N) protein, membrane (M) protein, and the envelope (E) protein. However, the role of these proteins in either binding to or activation of the complement system and/or the KKS is still incompletely understood. In these studies, we used: solid phase ELISA, hemolytic assay and surface plasmon resonance (SPR) techniques to examine if recombinant proteins corresponding to S1, N, M and E: (a) bind to C1q, gC1qR, FXII and high molecular weight kininogen (HK), and (b) activate complement and/or the KKS. Our data show that the viral proteins: (a) bind C1q and activate the classical pathway of complement, (b) bind FXII and HK, and activate the KKS in normal human plasma to generate bradykinin and (c) bind to gC1qR, the receptor for the globular heads of C1q (gC1q) which in turn could serve as a platform for the activation of both the complement system and KKS. Collectively, our data indicate that the SARS-CoV-2 viral particle can independently activate major innate inflammatory pathways for maximal damage and efficiency. Therefore, if efficient therapeutic modalities for the treatment of COVID-19 are to be designed, a strategy that includes blockade of the four major structural proteins may provide the best option.
Collapse
Affiliation(s)
- Anne G Savitt
- Department of Microbiology & Immunology, Renaissance School of Medicine of Stony Brook University, Stony Brook, NY, United States.,Department of Medicine, Renaissance School of Medicine of Stony Brook University, Stony Brook, NY, United States
| | - Samantha Manimala
- Department of Medicine, Renaissance School of Medicine of Stony Brook University, Stony Brook, NY, United States
| | - Tiara White
- Department of Microbiology & Immunology, Renaissance School of Medicine of Stony Brook University, Stony Brook, NY, United States.,Department of Medicine, Renaissance School of Medicine of Stony Brook University, Stony Brook, NY, United States
| | - Marina Fandaros
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Wei Yin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Huiquan Duan
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, United States
| | - Xin Xu
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, United States
| | - Brian V Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, United States
| | - David A Rubenstein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Allen P Kaplan
- Pulmonary and Critical Care Division, The Medical University of South Carolina, Charleston, SC, United States
| | - Ellinor I Peerschke
- The Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Berhane Ghebrehiwet
- Department of Microbiology & Immunology, Renaissance School of Medicine of Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
126
|
Rahman EZ, Shah P, Ong JE, Goldberg M, Ong SS. Purtscher-like retinopathy in a patient with COVID-19 and disseminated intravascular coagulation. Am J Ophthalmol Case Rep 2021; 24:101229. [PMID: 34796309 PMCID: PMC8582232 DOI: 10.1016/j.ajoc.2021.101229] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/06/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022] Open
Abstract
Purpose To describe a unique case of Purtscher-like retinopathy after a severe, complicated COVID-19 course which included development of disseminated intravascular coagulation (DIC). Observations A 58-year-old male developed blurry vision in the left eye one week after being discharged from the hospital for severe COVID-19 pneumonia and DIC. He had been intubated and ventilated for 5 days. Fundus examination revealed optic nerve hyperemia in the right eye, optic nerve pallor in the left eye, arteriolar attenuation, multiple cotton wool spots and ill-defined areas of retinal whitening in the posterior pole in both eyes. His exam findings were most consistent with Purtscher-like retinopathy in both eyes. Conclusions and Importance While several cases of central retinal artery and vein occlusion have been described in COVID-19 patients thus far, there has not been any reported cases of Purtscher-like retinopathy. To the best of our knowledge, this is the first case of Purtscher-like retinopathy in a patient who developed DIC during a severe COVID-19 infection.
Collapse
Affiliation(s)
- Effie Z Rahman
- Department of Ophthalmology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Pooja Shah
- Department of Ophthalmology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Morton Goldberg
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sally S Ong
- Department of Ophthalmology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
127
|
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: 70] [Impact Index Per Article: 23.3] [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
|
128
|
Higashikuni Y, Liu W, Obana T, Sata M. Pathogenic Basis of Thromboinflammation and Endothelial Injury in COVID-19: Current Findings and Therapeutic Implications. Int J Mol Sci 2021; 22:ijms222112081. [PMID: 34769508 PMCID: PMC8584434 DOI: 10.3390/ijms222112081] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic with a great impact on social and economic activities, as well as public health. In most patients, the symptoms of COVID-19 are a high-grade fever and a dry cough, and spontaneously resolve within ten days. However, in severe cases, COVID-19 leads to atypical bilateral interstitial pneumonia, acute respiratory distress syndrome, and systemic thromboembolism, resulting in multiple organ failure with high mortality and morbidity. SARS-CoV-2 has immune evasion mechanisms, including inhibition of interferon signaling and suppression of T cell and B cell responses. SARS-CoV-2 infection directly and indirectly causes dysregulated immune responses, platelet hyperactivation, and endothelial dysfunction, which interact with each other and are exacerbated by cardiovascular risk factors. In this review, we summarize current knowledge on the pathogenic basis of thromboinflammation and endothelial injury in COVID-19. We highlight the distinct contributions of dysregulated immune responses, platelet hyperactivation, and endothelial dysfunction to the pathogenesis of COVID-19. In addition, we discuss potential therapeutic strategies targeting these mechanisms.
Collapse
Affiliation(s)
- Yasutomi Higashikuni
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (W.L.); (T.O.)
- Correspondence: (Y.H.); (M.S.)
| | - Wenhao Liu
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (W.L.); (T.O.)
| | - Takumi Obana
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (W.L.); (T.O.)
| | - Masataka Sata
- Department of Cardiovascular Medicine, The University of Tokushima, Tokushima 770-8503, Japan
- Correspondence: (Y.H.); (M.S.)
| |
Collapse
|
129
|
Paula Junior WD, Freitas CSD, Lima Neto FFD, Santana NA, Soares NZD, Fonseca VLM, Nascimento RCRMD, Grabe-Guimarães A. Immune response in acute respiratory syndrome induced by the new coronavirus. ABCS HEALTH SCIENCES 2021. [DOI: 10.7322/abcshs.2020256.1704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Coronaviruses (CoVs) belong to the family Coronaviridae, which are enveloped and have a single-stranded RNA genome. The new coronavirus (SARS-CoV-2) is the seventh known coronavirus that can infect humans and cause serious illness, such as acute respiratory syndrome. The coronaviruses already identified have contributed to the understanding of the clinical manifestations caused by SARS-CoV-2, as well as their associations with the immune system. The aim of the present study was to carry out a narrative review of the literature on the host's immune response to infection by the new coronavirus. The review contains basic and summarized information on the main mechanisms involved in the immune response to SARS-CoV-2. The characteristics of the infection were considered according to the following: from the initial contact with the host through binding to angiotensin-converting enzyme 2 (ACE-2); the recognition of the pathogen by innate immunity cells; its containment mechanisms, including the production of effector cytokines and chemokines important in the development of the inflammatory process; and the participation of the complement system until the activation of the adaptive immune response. The probable occurrence of a host dysfunctional immune response and the escape mechanisms of the virus were also addressed. Despite numerous studies on the pathogenesis of SARS-CoV-2 infection, knowledge about the host's immune response in COVID-19 is not fully understood. The present work established the relationship between the new coronavirus and the immune system, but further studies are needed for all the mechanisms of the process to be elucidated.
Collapse
|
130
|
COVID-19 as a potential trigger of complement-mediated atypical HUS. Blood 2021; 138:1777-1782. [PMID: 34482396 PMCID: PMC8421074 DOI: 10.1182/blood.2021012752] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/06/2021] [Indexed: 12/20/2022] Open
|
131
|
Manik M, Singh RK. Role of toll-like receptors in modulation of cytokine storm signaling in SARS-CoV-2-induced COVID-19. J Med Virol 2021; 94:869-877. [PMID: 34672376 PMCID: PMC8662021 DOI: 10.1002/jmv.27405] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 01/08/2023]
Abstract
Balanced immune regulation is crucial for recognizing an invading pathogen, its killing, and elimination. Toll‐like receptors (TLRs) are the key regulators of the innate immune system. It helps in identifying between self and nonself‐molecule and eventually eliminates the nonself. Endosomal TLR, mainly TLR3, TLR7, TLR8, and membrane‐bound TLR4, has a role in the induction of cytokine storms. TLR7/8 recognizes the ssRNA SARS‐COV‐2 and when it replicates to dsRNA, it is recognized by TLR3 and drives the TRIF‐mediated inflammatory signaling like NF‐κB, MAPK. Such signaling leads to significant transcription and translation of pro‐inflammatory genes, releasing inflammatory molecules into the systemic circulation, causing an imbalance in the system. So, whenever an imbalance occurs, a surge in the pro‐inflammatory mediators is observed in the blood, including cytokines like interleukin (IL)‐2, IL‐4, IL‐6, IL‐1β, IL‐8, interferon (IFN)‐γ, tumor necrosis factor (TNF)‐α. IL‐6 and IL‐1β are one of the driving factors for bringing the cytokine storm into the systemic circulation, which migrates into the other organs, causing multiple organ failures leading to the death of the individual with severe illness. The imbalanced and hyper responsive immune system leads to a surge leading to death of the infected patients in COVID‐19. It has been observed that cytokine surge is TLR induced, mainly through activation of TLR3, TLR4, TLR7, TLR8 receptors. The cytokine storm migrates into the other organ through systemic circulation. The inflammation and the organ damage occur due to the TLR mediated NF‐κB, MAPK pathway. Hence blocking these specific TLRs may alleviate the chance of SARS‐COV‐2 infection.
Collapse
Affiliation(s)
- Moumita Manik
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, Uttar Pradesh, India
| | - Rakesh K Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, Uttar Pradesh, India
| |
Collapse
|
132
|
Labarrere CA, Kassab GS. Pattern Recognition Proteins: First Line of Defense Against Coronaviruses. Front Immunol 2021; 12:652252. [PMID: 34630377 PMCID: PMC8494786 DOI: 10.3389/fimmu.2021.652252] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023] Open
Abstract
The rapid outbreak of COVID-19 caused by the novel coronavirus SARS-CoV-2 in Wuhan, China, has become a worldwide pandemic affecting almost 204 million people and causing more than 4.3 million deaths as of August 11 2021. This pandemic has placed a substantial burden on the global healthcare system and the global economy. Availability of novel prophylactic and therapeutic approaches are crucially needed to prevent development of severe disease leading to major complications both acutely and chronically. The success in fighting this virus results from three main achievements: (a) Direct killing of the SARS-CoV-2 virus; (b) Development of a specific vaccine, and (c) Enhancement of the host's immune system. A fundamental necessity to win the battle against the virus involves a better understanding of the host's innate and adaptive immune response to the virus. Although the role of the adaptive immune response is directly involved in the generation of a vaccine, the role of innate immunity on RNA viruses in general, and coronaviruses in particular, is mostly unknown. In this review, we will consider the structure of RNA viruses, mainly coronaviruses, and their capacity to affect the lungs and the cardiovascular system. We will also consider the effects of the pattern recognition protein (PRP) trident composed by (a) Surfactant proteins A and D, mannose-binding lectin (MBL) and complement component 1q (C1q), (b) C-reactive protein, and (c) Innate and adaptive IgM antibodies, upon clearance of viral particles and apoptotic cells in lungs and atherosclerotic lesions. We emphasize on the role of pattern recognition protein immune therapies as a combination treatment to prevent development of severe respiratory syndrome and to reduce pulmonary and cardiovascular complications in patients with SARS-CoV-2 and summarize the need of a combined therapeutic approach that takes into account all aspects of immunity against SARS-CoV-2 virus and COVID-19 disease to allow mankind to beat this pandemic killer.
Collapse
Affiliation(s)
| | - Ghassan S Kassab
- California Medical Innovations Institute, San Diego, CA, United States
| |
Collapse
|
133
|
Zou M, Su X, Wang L, Yi X, Qiu Y, Yin X, Zhou X, Niu X, Wang L, Su M. The Molecular Mechanism of Multiple Organ Dysfunction and Targeted Intervention of COVID-19 Based on Time-Order Transcriptomic Analysis. Front Immunol 2021; 12:729776. [PMID: 34504502 PMCID: PMC8421734 DOI: 10.3389/fimmu.2021.729776] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/04/2021] [Indexed: 12/22/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) pandemic is caused by the novel coronavirus that has spread rapidly around the world, leading to high mortality because of multiple organ dysfunction; however, its underlying molecular mechanism is unknown. To determine the molecular mechanism of multiple organ dysfunction, a bioinformatics analysis method based on a time-order gene co-expression network (TO-GCN) was performed. First, gene expression profiles were downloaded from the gene expression omnibus database (GSE161200), and a TO-GCN was constructed using the breadth-first search (BFS) algorithm to infer the pattern of changes in the different organs over time. Second, Gene Ontology enrichment analysis was used to analyze the main biological processes related to COVID-19. The initial gene modules for the immune response of different organs were defined as the research object. The STRING database was used to construct a protein-protein interaction network of immune genes in different organs. The PageRank algorithm was used to identify five hub genes in each organ. Finally, the Comparative Toxicogenomics Database played an important role in exploring the potential compounds that target the hub genes. The results showed that there were two types of biological processes: the body's stress response and cell-mediated immune response involving the lung, trachea, and olfactory bulb (olf) after being infected by COVID-19. However, a unique biological process related to the stress response is the regulation of neuronal signals in the brain. The stress response was heterogeneous among different organs. In the lung, the regulation of DNA morphology, angiogenesis, and mitochondrial-related energy metabolism are specific biological processes related to the stress response. In particular, an effect on tracheal stress response was made by the regulation of protein metabolism and rRNA metabolism-related biological processes, as biological processes. In the olf, the distinctive stress responses consist of neural signal transmission and brain behavior. In addition, myeloid leukocyte activation and myeloid leukocyte-mediated immunity in response to COVID-19 can lead to a cytokine storm. Immune genes such as SRC, RHOA, CD40LG, CSF1, TNFRSF1A, FCER1G, ICAM1, LAT, LCN2, PLAU, CXCL10, ICAM1, CD40, IRF7, and B2M were predicted to be the hub genes in the cytokine storm. Furthermore, we inferred that resveratrol, acetaminophen, dexamethasone, estradiol, statins, curcumin, and other compounds are potential target drugs in the treatment of COVID-19.
Collapse
Affiliation(s)
- Miao Zou
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Xiaoyun Su
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Luoying Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Xingcheng Yi
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Yue Qiu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Xirui Yin
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Xuan Zhou
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Xinhui Niu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Liuli Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Manman Su
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| |
Collapse
|
134
|
De Novo Crohn's Disease Triggered After COVID-19: Is COVID-19 More Than an Infectious Disease? ACG Case Rep J 2021; 8:e00652. [PMID: 34476279 PMCID: PMC8386903 DOI: 10.14309/crj.0000000000000652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19)-associated immune dysregulation is believed to trigger the onset of various autoimmune diseases. These occur either during active COVID-19 or soon after recovery. We report ileocolonic Crohn's disease in a 35-year-old woman after her recovery from a milder form of COVID-19. She achieved remission of her symptoms with oral corticosteroids and sulfasalazine.
Collapse
|
135
|
Manickavel S, Sinha N. A pandemic of acute respiratory distress syndrome-role of lung transplant in coronavirus disease-2019-associated respiratory failure. Indian J Thorac Cardiovasc Surg 2021; 37:359-365. [PMID: 34483505 PMCID: PMC8408360 DOI: 10.1007/s12055-021-01233-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
Lung transplantation may be appropriate, and may offer benefit only to a carefully selected subset of morbidly ill patients afflicted by coronavirus disease-2019. Identifying the appropriate recipient for the allocation of scarce resources, by safely navigating through the challenges that are unique to lung transplantation for coronavirus disease-2019-associated acute respiratory distress syndrome, demands a conscientious and meticulous approach. Categorizing the respiratory failure in coronavirus disease-2019 may facilitate the process of evaluation for the purpose of transplant. The progress in rescue transplants over the past two decades has greatly improved our ability to successfully perform high-risk lung transplantation.
Collapse
Affiliation(s)
| | - Neeraj Sinha
- University of Miami Miller School of Medicine, Miami, FL 33101 USA
| |
Collapse
|
136
|
Abstract
Severe COVID-19 is characterized by lung and multiorgan inflammation and coagulation in the presence of overactivation of the complement system. Complement is a double edged-sward in SARS-Cov-2 infection. On one hand, it can control the viral infection in milder cases, on the other hand in cases with severe and prolonged infection massive complement activation occurs, which can intensify lung and systemic inflammation and promote a procoagulant and prothrombotic state. Several uncontrolled studies and controlled clinical trials with different complement inhibitors have been performed and others are ongoing. Results are promising in some but negative in others. Further studies are required to elucidate the benefit to risk profile of complement inhibitors in COVID-19 patients at different stages of the disease and to clarify the best targets in the complement cascade.
Collapse
|
137
|
COVID-19-associated atypical hemolytic uremic syndrome and use of Eculizumab therapy. J Nephrol 2021; 35:317-321. [PMID: 34427905 PMCID: PMC8383924 DOI: 10.1007/s40620-021-01125-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/12/2021] [Indexed: 12/30/2022]
Abstract
There is a high incidence of acute kidney injury with COVID-19 infections. The etiologies of acute kidney injury could be ischemic acute tubular necrosis or a complex process of complement activation leading to thrombotic microangiopathy. We present a case of 32-year-old Hispanic male with a history of heart transplant, admitted with COVID-19 and atypical hemolytic uremic syndrome, which was successfully treated with Eculizumab.
Collapse
|
138
|
Complement and the prothrombotic state. Blood 2021; 139:1954-1972. [PMID: 34415298 DOI: 10.1182/blood.2020007206] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/08/2021] [Indexed: 11/20/2022] Open
Abstract
In 2007 and 2009 the regulatory approval of the first-in-class complement inhibitor Eculizumab has revolutionized the clinical management of two rare, life-threatening clinical conditions: paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). While being completely distinct diseases affecting blood cells and the glomerulus, PNH and aHUS remarkably share several features in their etiology and clinical presentation. An imbalance between complement activation and regulation at host surfaces underlies both diseases precipitating in severe thrombotic events that are largely resistant to anti-coagulant and/or anti-platelet therapies. Inhibition of the common terminal complement pathway by Eculizumab prevents the frequently occurring thrombotic events responsible for the high mortality and morbidity observed in patients not treated with anti-complement therapy. While many in vitro and ex vivo studies elaborate numerous different molecular interactions between complement activation products and hemostasis, this review focuses on the clinical evidence that links these two fields in humans. Several non-infectious conditions with known complement involvement are scrutinized for common patterns concerning a prothrombotic statues and the occurrence of certain complement activation levels. Next to PNH and aHUS, germline encoded CD59 or CD55 deficiency (the latter causing the disease Complement Hyperactivation, Angiopathic thrombosis, and Protein-Losing Enteropathy; CHAPLE), autoimmune hemolytic anemia (AIHA), (catastrophic) anti-phospholipid syndrome (APS, CAPS) and C3 glomerulopathy are considered. Parallels and distinct features among these conditions are discussed against the background of thrombosis, complement activation, and potential complement diagnostic and therapeutic avenues.
Collapse
|
139
|
Lean-ing Method in an Emergency Department of the Italian Epicenter of the COVID-19 Outbreak: When the Algorithm Makes Difference. APPLIED SYSTEM INNOVATION 2021. [DOI: 10.3390/asi4030055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The Lean method entails a set of standardized processes intending to optimize resources, reduce waste, and improve results. Lean has been proposed as an operative model for the COVID-19 outbreak. Herein, we summarized data resulted from the Lean model adoption in an Emergency Department of the Lombardy region, the Italian epicenter of the pandemic, to critically appraise its effectiveness and feasibility. The Lean algorithm was applied in the Humanitas Clinical and Research Hospital, Milan, north of Italy. At admission, patients underwent outdoor pre-triage for fever, respiratory, and gastrointestinal symptoms, with a focus on SpO2. Based on these data, they were directed to the most appropriate area for the COVID-19 first-level screening. High-risk patients were assisted by trained staff for second-level screening and planning of treatment. Out of 7.778 patients, 21.9% were suspected of SARS-CoV-2 infection. Mortality was 21.9% and the infection rate in health workers was 4.8%. The lean model has proved to be effective in optimizing the overall management of COVID-19 patients in an emergency setting. It allowed for screening of a large volume of patients, while also limiting the health workers’ infection rate. Further studies are necessary to validate the suggested approach.
Collapse
|
140
|
Karnaukhova E. C1-Inhibitor: Structure, Functional Diversity and Therapeutic Development. Curr Med Chem 2021; 29:467-488. [PMID: 34348603 DOI: 10.2174/0929867328666210804085636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/24/2021] [Accepted: 05/13/2021] [Indexed: 11/22/2022]
Abstract
Human C1-Inhibitor (C1INH), also known as C1-esterase inhibitor, is an important multifunctional plasma glycoprotein that is uniquely involved in a regulatory network of complement, contact, coagulation, and fibrinolytic systems. C1INH belongs to a superfamily of serine proteinase inhibitor (serpins) and exhibits its inhibitory activities towards several target proteases of plasmatic cascades, operating as a major anti-inflammatory protein in the circulation. In addition to its inhibitory activities, C1INH is also involved in non-inhibitory interactions with some endogenous proteins, polyanions, cells and infectious agents. While C1INH is essential for multiple physiological processes, it is better known for its deficiency with regards to Hereditary Angioedema (HAE), a rare autosomal dominant disease clinically manifested by recurrent acute attacks of increased vascular permeability and edema. Since the link was first established between functional C1INH deficiency in plasma and HAE in the 1960s, tremendous progress has been made in the biochemical characterization of C1INH and its therapeutic development for replacement therapies in patients with C1INH-dependent HAE. Various C1INH biological activities, recent advances in the HAE-targeted therapies, and availability of C1INH commercial products have prompted intensive investigation of the C1INH potential for treatment of clinical conditions other than HAE. This article provides an updated overview of the structure and biological activities of C1INH, its role in HAE pathogenesis, and recent advances in the research and therapeutic development of C1INH; it also considers some trends for using C1INH therapeutic preparations for applications other than angioedema, from sepsis and endotoxin shock to severe thrombotic complications in COVID-19 patients.
Collapse
Affiliation(s)
- Elena Karnaukhova
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993. United States
| |
Collapse
|
141
|
Nicosia RF, Ligresti G, Caporarello N, Akilesh S, Ribatti D. COVID-19 Vasculopathy: Mounting Evidence for an Indirect Mechanism of Endothelial Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1374-1384. [PMID: 34033751 PMCID: PMC8141344 DOI: 10.1016/j.ajpath.2021.05.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
Patients with coronavirus disease 2019 (COVID-19) who are critically ill develop vascular complications characterized by thrombosis of small, medium, and large vessels. Dysfunction of the vascular endothelium due to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been implicated in the pathogenesis of the COVID-19 vasculopathy. Although initial reports suggested that endothelial injury was caused directly by the virus, recent studies indicate that endothelial cells do not express angiotensin-converting enzyme 2, the receptor that SARS-CoV-2 uses to gain entry into cells, or express it at low levels and are resistant to the infection. These new findings, together with the observation that COVID-19 triggers a cytokine storm capable of injuring the endothelium and disrupting its antithrombogenic properties, favor an indirect mechanism of endothelial injury mediated locally by an augmented inflammatory reaction to infected nonendothelial cells, such as the bronchial and alveolar epithelium, and systemically by the excessive immune response to infection. Herein we review the vascular pathology of COVID-19 and critically discuss the potential mechanisms of endothelial injury in this disease.
Collapse
Affiliation(s)
- Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington.
| | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neuroscienze e Organi di Senso (SMBNOS), Universita' degli Studi Aldo Moro, Policlinico, Bari, Italy
| |
Collapse
|
142
|
Khan A, Shang N, Petukhova L, Zhang J, Shen Y, Hebbring SJ, Moncrieffe H, Kottyan LC, Namjou-Khales B, Knevel R, Raychaudhuri S, Karlson EW, Harley JB, Stanaway IB, Crosslin D, Denny JC, Elkind MS, Gharavi AG, Hripcsak G, Weng C, Kiryluk K. Medical Records-Based Genetic Studies of the Complement System. J Am Soc Nephrol 2021; 32:2031-2047. [PMID: 33941608 PMCID: PMC8455263 DOI: 10.1681/asn.2020091371] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/09/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Genetic variants in complement genes have been associated with a wide range of human disease states, but well-powered genetic association studies of complement activation have not been performed in large multiethnic cohorts. METHODS We performed medical records-based genome-wide and phenome-wide association studies for plasma C3 and C4 levels among participants of the Electronic Medical Records and Genomics (eMERGE) network. RESULTS In a GWAS for C3 levels in 3949 individuals, we detected two genome-wide significant loci: chr.1q31.3 (CFH locus; rs3753396-A; β=0.20; 95% CI, 0.14 to 0.25; P=1.52x10-11) and chr.19p13.3 (C3 locus; rs11569470-G; β=0.19; 95% CI, 0.13 to 0.24; P=1.29x10-8). These two loci explained approximately 2% of variance in C3 levels. GWAS for C4 levels involved 3998 individuals and revealed a genome-wide significant locus at chr.6p21.32 (C4 locus; rs3135353-C; β=0.40; 95% CI, 0.34 to 0.45; P=4.58x10-35). This locus explained approximately 13% of variance in C4 levels. The multiallelic copy number variant analysis defined two structural genomic C4 variants with large effect on blood C4 levels: C4-BS (β=-0.36; 95% CI, -0.42 to -0.30; P=2.98x10-22) and C4-AL-BS (β=0.25; 95% CI, 0.21 to 0.29; P=8.11x10-23). Overall, C4 levels were strongly correlated with copy numbers of C4A and C4B genes. In comprehensive phenome-wide association studies involving 102,138 eMERGE participants, we cataloged a full spectrum of autoimmune, cardiometabolic, and kidney diseases genetically related to systemic complement activation. CONCLUSIONS We discovered genetic determinants of plasma C3 and C4 levels using eMERGE genomic data linked to electronic medical records. Genetic variants regulating C3 and C4 levels have large effects and multiple clinical correlations across the spectrum of complement-related diseases in humans.
Collapse
Affiliation(s)
- Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Ning Shang
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Lynn Petukhova
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Jun Zhang
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Yufeng Shen
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Medical Center, New York, New York
| | - Scott J. Hebbring
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin
| | - Halima Moncrieffe
- Department of Pediatrics, Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Leah C. Kottyan
- Department of Pediatrics, Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Bahram Namjou-Khales
- Department of Pediatrics, Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Rachel Knevel
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Centre for Genetics and Genomics Versus Arthritis, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Elizabeth W. Karlson
- Division of Rheumatology, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - John B. Harley
- Department of Pediatrics, Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Ian B. Stanaway
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, Washington
| | - David Crosslin
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, Washington
| | - Joshua C. Denny
- Department of Biomedical Informatics, Vanderbilt University, Nashville, Tennessee
| | - Mitchell S.V. Elkind
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Ali G. Gharavi
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - George Hripcsak
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Chunhua Weng
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
143
|
Gupta A, Gupta GS. Status of mannose-binding lectin (MBL) and complement system in COVID-19 patients and therapeutic applications of antiviral plant MBLs. Mol Cell Biochem 2021; 476:2917-2942. [PMID: 33745077 PMCID: PMC7981598 DOI: 10.1007/s11010-021-04107-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by a virus called "Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)." In the majority of patients, infection with COVID-19 may be asymptomatic or may cause only mild symptoms. However, in some patients, there can also be immunological problems, such as macrophage activation syndrome (CSS) that results in cytokine storm syndrome (CSS) and acute respiratory distress syndrome (ARDS). Comprehension of host-microbe communications is the critical aspect in the advancement of new therapeutics against infectious illnesses. Endogenous animal lectins, a class of proteins, may perceive non-self glycans found on microorganisms. Serum mannose-binding lectin (sMBL), as a part of the innate immune framework, recognizes a wide range of microbial microorganisms and activates complement cascade via an antibody-independent pathway. Although the molecular basis for the intensity of SARS-CoV-2 infection is not generally understood, scientific literature indicates that COVID-19 is correlated with unregulated activation of the complement in terms of disease severity. Disseminated intravascular coagulation (DIC), inflammation, and immune paralysis contribute to unregulated complement activation. Pre-existing genetic defects in MBL and their association with complement play a major role in immune response dysregulation caused by SARS-CoV-2. In order to generate anti-complement-based therapies in Covid-19, an understanding of sMBL in immune response to SARS-CoV-2 and complement is therefore essential. This review highlights the role of endogenous sMBL and complement activation during SARS-CoV-2 infection and their therapeutic management by various agents, mainly plant lectins, since antiviral mannose-binding plant lectins (pMBLs) offer potential applications in the prevention and control of viral infections.
Collapse
Affiliation(s)
- Anita Gupta
- Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, India
| | - G S Gupta
- Department of Biophysics, Sector 25, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
144
|
Abstract
ABSTRACT The ongoing coronavirus disease 2019 (COVID-19) pandemic has swept over the world and causes thousands of deaths. Although the clinical features of COVID-19 become much clearer than before, there are still further problems with the pathophysiological process and treatments of severe patients. One primary problem is with the paradoxical immune states in severe patients with COVID-19. Studies indicate that Severe Acute Respiratory Syndrome Coronavirus 2 can attack the immune system, manifested as a state of immunosuppression with a decrease in lymphocytes, whereas a state of hyperinflammation, presenting as elevated cytokine levels, is also detected in COVID-19. Therefore, discussing the specific status of immunity in COVID-19 will contribute to the understanding of its pathophysiology and the search for appropriate treatments. Here, we review all the available literature concerning the different immune states in COVID-19 and the underlying pathophysiological mechanisms. In addition, the association between immune states and the development and severity of disease as well as the impact on the selection of immunotherapy strategies are discussed in our review.
Collapse
Affiliation(s)
- Ye Liu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Dongxue Xu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Jing Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| |
Collapse
|
145
|
Xia B, Shen X, He Y, Pan X, Liu FL, Wang Y, Yang F, Fang S, Wu Y, Duan Z, Zuo X, Xie Z, Jiang X, Xu L, Chi H, Li S, Meng Q, Zhou H, Zhou Y, Cheng X, Xin X, Jin L, Zhang HL, Yu DD, Li MH, Feng XL, Chen J, Jiang H, Xiao G, Zheng YT, Zhang LK, Shen J, Li J, Gao Z. SARS-CoV-2 envelope protein causes acute respiratory distress syndrome (ARDS)-like pathological damages and constitutes an antiviral target. Cell Res 2021; 31:847-860. [PMID: 34112954 PMCID: PMC8190750 DOI: 10.1038/s41422-021-00519-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/13/2021] [Indexed: 01/08/2023] Open
Abstract
Cytokine storm and multi-organ failure are the main causes of SARS-CoV-2-related death. However, the origin of excessive damages caused by SARS-CoV-2 remains largely unknown. Here we show that the SARS-CoV-2 envelope (2-E) protein alone is able to cause acute respiratory distress syndrome (ARDS)-like damages in vitro and in vivo. 2-E proteins were found to form a type of pH-sensitive cation channels in bilayer lipid membranes. As observed in SARS-CoV-2-infected cells, heterologous expression of 2-E channels induced rapid cell death in various susceptible cell types and robust secretion of cytokines and chemokines in macrophages. Intravenous administration of purified 2-E protein into mice caused ARDS-like pathological damages in lung and spleen. A dominant negative mutation lowering 2-E channel activity attenuated cell death and SARS-CoV-2 production. Newly identified channel inhibitors exhibited potent anti-SARS-CoV-2 activity and excellent cell protective activity in vitro and these activities were positively correlated with inhibition of 2-E channel. Importantly, prophylactic and therapeutic administration of the channel inhibitor effectively reduced both the viral load and secretion of inflammation cytokines in lungs of SARS-CoV-2-infected transgenic mice expressing human angiotensin-converting enzyme 2 (hACE-2). Our study supports that 2-E is a promising drug target against SARS-CoV-2.
Collapse
Affiliation(s)
- Bingqing Xia
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xurui Shen
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yang He
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Feng-Liang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yi Wang
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feipu Yang
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sui Fang
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Zilei Duan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaoli Zuo
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhuqing Xie
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xiangrui Jiang
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hao Chi
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuangqu Li
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Meng
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hu Zhou
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yubo Zhou
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xi Cheng
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoming Xin
- Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Lin Jin
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hai-Lin Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dan-Dan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming-Hua Li
- Kunming National High-level Biosafety Research Center for Non-human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao-Li Feng
- Kunming National High-level Biosafety Research Center for Non-human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jiekai Chen
- Center for Cell Fate and Lineage (CCLA), Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, Guangdong, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Joint School of Life Science, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hualiang Jiang
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming National High-level Biosafety Research Center for Non-human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Lei-Ke Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China.
| | - Jingshan Shen
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Jia Li
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, Guangdong, China.
| | - Zhaobing Gao
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, Guangdong, China.
- School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
146
|
Fan Y, Wang Y, Yu S, Chang J, Yan Y, Wang Y, Bian Y. Natural products provide a new perspective for anti-complement treatment of severe COVID-19: a review. Chin Med 2021; 16:67. [PMID: 34321065 PMCID: PMC8318062 DOI: 10.1186/s13020-021-00478-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/21/2021] [Indexed: 01/08/2023] Open
Abstract
Exaggerated immune response and cytokine storm are accounted for the severity of COVID-19, including organ dysfunction, especially progressive respiratory failure and generalized coagulopathy. Uncontrolled activation of complement contributes to acute and chronic inflammation, the generation of cytokine storm, intravascular coagulation and cell/tissue damage, which may be a favorable target for the treatment of multiple organ failure and reduction of mortality in critically ill patients with COVID-19. Cytokine storm suppression therapy can alleviate the symptoms of critically ill patients to some extent, but as a remedial etiological measure, its long-term efficacy is still questionable. Anti-complement therapy has undoubtedly become an important hotspot in the upstream regulation of cytokine storm. However, chemosynthetic complement inhibitors are expensive, and their drug resistance and long-term side effects require further investigation. New complement inhibitors with high efficiency and low toxicity can be obtained from natural products at low development cost. This paper puts forward some insights of the development of natural anti-complement products in traditional Chinese medicine, that may provide a bright perspective for suppressing cytokine storm in critically ill patients with COVID-19.
Collapse
Affiliation(s)
- Yadong Fan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 PoYangHu Road, JingHai, District, Tianjin, 301617, People's Republic of China
| | - Ying Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 PoYangHu Road, JingHai, District, Tianjin, 301617, People's Republic of China
| | - Shuang Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 PoYangHu Road, JingHai, District, Tianjin, 301617, People's Republic of China
| | - Jun Chang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 PoYangHu Road, JingHai, District, Tianjin, 301617, People's Republic of China
| | - Yiqi Yan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiyang Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 PoYangHu Road, JingHai, District, Tianjin, 301617, People's Republic of China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 PoYangHu Road, JingHai, District, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
147
|
Ruiz-Rodríguez JC, Molnar Z, Deliargyris EN, Ferrer R. The Use of CytoSorb Therapy in Critically Ill COVID-19 Patients: Review of the Rationale and Current Clinical Experiences. Crit Care Res Pract 2021; 2021:7769516. [PMID: 34336280 PMCID: PMC8324379 DOI: 10.1155/2021/7769516] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/08/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has led to the biggest global health crisis of our lifetime. There is accumulating evidence that a substantial number of critically ill COVID-19 patients exhibit a dysregulated host response manifesting as cytokine storm or cytokine release syndrome, which in turn contributes to the high observed rates of mortality. Just as in other hyperinflammatory conditions, extracorporeal cytokine removal may have potential beneficial effects in this subgroup of COVID-19 patients. The CytoSorb blood purification device is the most extensively investigated cytokine removal platform with considerable evidence suggesting that early intervention can provide rapid hemodynamic stabilization and improvement in vital organ functions. The purpose of this review is to provide an overview of the pathophysiological background of hyperinflammation in COVID-19 and to summarize the currently available evidence on the effects of hemoadsorption in these patients.
Collapse
Affiliation(s)
- Juan Carlos Ruiz-Rodríguez
- Department of Intensive Care, Hospital Universitari Vall d'Hebron, Shock Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Zsolt Molnar
- CytoSorbents Europe GmbH, Berlin, Germany
- Institute for Translational Medicine, School of Medicine, University of Pécs, Pécs, Hungary
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, Poznan, Poland
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | | | - Ricard Ferrer
- Department of Intensive Care, Hospital Universitari Vall d'Hebron, Shock Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| |
Collapse
|
148
|
Autoimmune- and complement-mediated hematologic condition recrudescence following SARS-CoV-2 vaccination. Blood Adv 2021; 5:2794-2798. [PMID: 34255033 PMCID: PMC8276576 DOI: 10.1182/bloodadvances.2021004957] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/20/2021] [Indexed: 12/22/2022] Open
Abstract
Severe exacerbation of underlying hematologic conditions can occur within 1 to 4 days after dose 2 of a 2-dose SARS-CoV-2 vaccine series. A mild exacerbation after dose 1 and/or a history of vaccine-related adverse events may portend a more serious event after dose 2.
A variety of autoimmune disorders have been reported after viral illnesses and specific vaccinations. Cases of de novo immune thrombocytopenia (ITP) have been reported after SARS-CoV-2 vaccination, although its effect on preexisting ITP has not been well characterized. In addition, although COVID-19 has been associated with complement dysregulation, the effect of SARS-CoV-2 vaccination on preexisting complementopathies is poorly understood. We sought to better understand SARS-CoV-2 vaccine-induced recurrence of autoimmune- and complement-mediated hematologic conditions. Three illustrative cases were identified at the University of Washington Medical Center and the Seattle Cancer Care Alliance from January through March 2021. We describe the recrudescence of 2 autoimmune conditions (ITP and acquired von Willebrand Disease [AvWD]/acquired hemophilia A) and 1 complementopathy (paroxysmal nocturnal hemoglobinuria [PNH]). We report the first known case of AvWD/acquired hemophilia A, and describe the first PNH exacerbation in the absence of complement inhibition after SARS-CoV-2 vaccination. Although SARS-CoV-2 vaccine-induced ITP is a known concern, our case clearly depicts how thrombocytopenia in the setting of preexisting ITP can sequentially worsen with each vaccine dose. Based on our experiences and these examples, we provide considerations for how to monitor and assess risk in patients with underlying autoimmune- and complement-mediated hematologic conditions.
Collapse
|
149
|
Sun Y, Tan J, Miao Y, Zhang Q. The role of PD-L1 in the immune dysfunction that mediates hypoxia-induced multiple organ injury. Cell Commun Signal 2021; 19:76. [PMID: 34256773 PMCID: PMC8276205 DOI: 10.1186/s12964-021-00742-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Hypoxia is a pathological condition common to many diseases, although multiple organ injuries induced by hypoxia are often overlooked. There is increasing evidence to suggest that the hypoxic environment may activate innate immune cells and suppress adaptive immunity, further stimulating inflammation and inhibiting immunosurveillance. We found that dysfunctional immune regulation may aggravate hypoxia-induced tissue damage and contribute to secondary injury. Among the diverse mechanisms of hypoxia-induced immune dysfunction identified to date, the role of programmed death-ligand 1 (PD-L1) has recently attracted much attention. Besides leading to tumour immune evasion, PD-L1 has also been found to participate in the progression of the immune dysfunction which mediates hypoxia-induced multiple organ injury. In this review, we aimed to summarise the role of immune dysfunction in hypoxia-induced multiple organ injury, the effects of hypoxia on the cellular expression of PD-L1, and the effects of upregulated PD-L1 expression on immune regulation. Furthermore, we summarise the latest information pertaining to the involvement, diagnostic value, and therapeutic potential of immunosuppression induced by PD-L1 in various types of hypoxia-related diseases, including cancers, ischemic stroke, acute kidney injury, and obstructive sleep apnoea. Video Abstract.
Collapse
Affiliation(s)
- Yang Sun
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052 China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052 China
| | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052 China
| |
Collapse
|
150
|
Lundstrom K, Barh D, Uhal BD, Takayama K, Aljabali AAA, Abd El-Aziz TM, Lal A, Redwan EM, Adadi P, Chauhan G, Sherchan SP, Azad GK, Rezaei N, Serrano-Aroca Á, Bazan NG, Hassan SS, Panda PK, Pal Choudhury P, Pizzol D, Kandimalla R, Baetas-da-Cruz W, Mishra YK, Palu G, Brufsky AM, Tambuwala MM, Uversky VN. COVID-19 Vaccines and Thrombosis-Roadblock or Dead-End Street? Biomolecules 2021; 11:1020. [PMID: 34356644 PMCID: PMC8301964 DOI: 10.3390/biom11071020] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 12/16/2022] Open
Abstract
Two adenovirus-based vaccines, ChAdOx1 nCoV-19 and Ad26.COV2.S, and two mRNA-based vaccines, BNT162b2 and mRNA.1273, have been approved by the European Medicines Agency (EMA), and are invaluable in preventing and reducing the incidence of coronavirus disease-2019 (COVID-19). Recent reports have pointed to thrombosis with associated thrombocytopenia as an adverse effect occurring at a low frequency in some individuals after vaccination. The causes of such events may be related to SARS-CoV-2 spike protein interactions with different C-type lectin receptors, heparan sulfate proteoglycans (HSPGs) and the CD147 receptor, or to different soluble splice variants of the spike protein, adenovirus vector interactions with the CD46 receptor or platelet factor 4 antibodies. Similar findings have been reported for several viral diseases after vaccine administration. In addition, immunological mechanisms elicited by viral vectors related to cellular delivery could play a relevant role in individuals with certain genetic backgrounds. Although rare, the potential COVID-19 vaccine-induced immune thrombotic thrombocytopenia (VITT) requires immediate validation, especially in risk groups, such as the elderly, chronic smokers, and individuals with pre-existing incidences of thrombocytopenia; and if necessary, a reformulation of existing vaccines.
Collapse
Affiliation(s)
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Purba Medinipur 721172, India
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Bruce D. Uhal
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Kazuo Takayama
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8397, Japan;
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan;
| | - Tarek Mohamed Abd El-Aziz
- Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt;
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Amos Lal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55902, USA;
| | - Elrashdy M. Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Parise Adadi
- Department of Food Science, University of Otago, Dunedin 9054, New Zealand;
| | - Gaurav Chauhan
- School of Engineering and Sciences, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico;
| | - Samendra P. Sherchan
- Department of Environmental Health Sciences, Tulane University, New Orleans, LA 70112, USA;
| | | | - Nima Rezaei
- Research Center for Immunodeficiency, Children’s Medical Center, Tehran University of Medical Sciences, Tehran 1416753955, Iran;
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 17177 Stockholm, Sweden
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Nicolas G. Bazan
- Neuroscience Center of Excellence, School of Medicine, LSU Health New Orleans, New Orleans, LA 70112, USA;
| | - Sk Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram 721140, India;
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, 75120 Uppsala, Sweden;
| | | | - Damiano Pizzol
- Italian Agency for Development Cooperation—Khartoum, Sudan Street 33, Al Amarat 11111, Sudan;
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, India;
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
| | - Wagner Baetas-da-Cruz
- Translational Laboratory in Molecular Physiology, Centre for Experimental Surgery, College of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil;
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, University of Southern Denmark, NanoSYD, Alsion 2, 6400 Sønderborg, Denmark;
| | - Giorgio Palu
- Department of Molecular Medicine, University of Padova, 35122 Padova, PD, Italy;
| | - Adam M. Brufsky
- UPMC Hillman Cancer Center, Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|