1
|
Wu W, Aisha A, Chang F, Jianghui Z, Shuai T, Zheng L, Chen F. Establishment and analysis of an animal model for nutcracker syndrome. Int Urol Nephrol 2025; 57:419-426. [PMID: 39422826 DOI: 10.1007/s11255-024-04245-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND This study investigated the effects of renal vein congestion of varying durations on kidney damage by establishing an animal model of Nutcracker Syndrome (NCS). In addition, we assessed whether renal damage improved after relieving the congestion. METHOD We established a rat model of NCS and relieved renal congestion through secondary surgery, with renal vein reperfusion confirmed by animal ultrasound. Histopathological changes in congested kidneys were evaluated at 3, 6, and 9 days using HE and Masson staining, and the expression of renal injury factors was determined using q-PCR. RESULTS Renal tissue pathology changes were evident with prolonged obstruction time. Compared to the sham-operated group, no apparent fibrosis was observed in the kidneys at 3 days post-congestion, whereas fibrosis was most severe at 9 days post-congestion, with no significant improvement observed even after blood flow restoration. Six days post-congestion relief, some alleviation of renal fibrosis occurred. q-PCR results indicated a significant reduction in the expression of fibrosis markers after 3 or 6 days of renal vein ligation release. Following blood flow restoration on the 9th day post-congestion, no significant changes in fibrosis markers were noted. CONCLUSION Prolonged renal vein congestion in a rat model of NCS resulted in severe kidney damage and significant fibrosis after 9 days. While relieving congestion within 6 days led to some improvement, no reduction in fibrosis occurred after 9 days, underscoring the importance of congestion duration in renal pathology.
Collapse
Affiliation(s)
- Wensong Wu
- Department of Urology, The Third Central Clinical College of Tianjin Medical University, Tianjin, 300170, China
| | - Awuti Aisha
- Department of Urology, The Third Central Clinical College of Tianjin Medical University, Tianjin, 300170, China
| | - Fan Chang
- Department of Urology, The Third Central Hospital of Tianjin, 83 Jintang Road, Tianjin, 300170, China
- Department of Urology, Nankai University Affinity the Third Central Hospital, Tianjin, 300170, China
| | - Zhang Jianghui
- Department of Urology, The Third Central Hospital of Tianjin, 83 Jintang Road, Tianjin, 300170, China
- Department of Urology, Nankai University Affinity the Third Central Hospital, Tianjin, 300170, China
| | - Tang Shuai
- Department of Urology, The Third Central Hospital of Tianjin, 83 Jintang Road, Tianjin, 300170, China
- Department of Urology, Nankai University Affinity the Third Central Hospital, Tianjin, 300170, China
| | - Lv Zheng
- Department of Urology, The Third Central Hospital of Tianjin, 83 Jintang Road, Tianjin, 300170, China
- Department of Urology, Nankai University Affinity the Third Central Hospital, Tianjin, 300170, China
| | - Fangmin Chen
- Department of Urology, The Third Central Hospital of Tianjin, 83 Jintang Road, Tianjin, 300170, China.
- Department of Urology, Nankai University Affinity the Third Central Hospital, Tianjin, 300170, China.
| |
Collapse
|
2
|
Wang K, Nie Y, Maguire C, Syphurs C, Sheen H, Karoly M, Lapp L, Gygi JP, Jayavelu ND, Patel RK, Hoch A, Corry D, Kheradmand F, McComsey GA, Fernandez-Sesma A, Simon V, Metcalf JP, Higuita NIA, Messer WB, Davis MM, Nadeau KC, Kraft M, Bime C, Schaenman J, Erle D, Calfee CS, Atkinson MA, Brackenridge SC, Hafler DA, Shaw A, Rahman A, Hough CL, Geng LN, Ozonoff A, Haddad EK, Reed EF, van Bakel H, Kim-Schultz S, Krammer F, Wilson M, Eckalbar W, Bosinger S, Langelier CR, Sekaly RP, Montgomery RR, Maecker HT, Krumholz H, Melamed E, Steen H, Pulendran B, Augustine AD, Cairns CB, Rouphael N, Becker PM, Fourati S, Shannon CP, Smolen KK, Peters B, Kleinstein SH, Levy O, Altman MC, Iwasaki A, Diray-Arce J, Ehrlich LIR, Guan L. Unraveling SARS-CoV-2 Host-Response Heterogeneity through Longitudinal Molecular Subtyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624784. [PMID: 39651165 PMCID: PMC11623532 DOI: 10.1101/2024.11.22.624784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Hospitalized COVID-19 patients exhibit diverse immune responses during acute infection, which are associated with a wide range of clinical outcomes. However, understanding these immune heterogeneities and their links to various clinical complications, especially long COVID, remains a challenge. In this study, we performed unsupervised subtyping of longitudinal multi-omics immunophenotyping in over 1,000 hospitalized patients, identifying two critical subtypes linked to mortality or mechanical ventilation with prolonged hospital stay and three severe subtypes associated with timely acute recovery. We confirmed that unresolved systemic inflammation and T-cell dysfunctions were hallmarks of increased severity and further distinguished patients with similar acute respiratory severity by their distinct immune profiles, which correlated with differences in demographic and clinical complications. Notably, one critical subtype (SubF) was uniquely characterized by early excessive inflammation, insufficient anticoagulation, and fatty acid dysregulation, alongside higher incidences of hematologic, cardiac, and renal complications, and an elevated risk of long COVID. Among the severe subtypes, significant differences in viral clearance and early antiviral responses were observed, with one subtype (SubC) showing strong early T-cell cytotoxicity but a poor humoral response, slower viral clearance, and greater risks of chronic organ dysfunction and long COVID. These findings provide crucial insights into the complex and context-dependent nature of COVID-19 immune responses, highlighting the importance of personalized therapeutic strategies to improve both acute and long-term outcomes.
Collapse
|
3
|
Berra S, Parolin D, Suffritti C, Folcia A, Zanichelli A, Gusso L, Cogliati C, Riva A, Gidaro A, Caccia S. Patterns of C1-Inhibitor Plasma Levels and Kinin-Kallikrein System Activation in Relation to COVID-19 Severity. Life (Basel) 2024; 14:1525. [PMID: 39768234 PMCID: PMC11679851 DOI: 10.3390/life14121525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Although more than four years have passed since the pandemic began, SARS-CoV-2 continues to be of concern. Therefore, research into the underlying mechanisms that contribute to the development of the disease, especially in more severe forms, remains a priority. Sustained activation of the complement (CS), contact (CAS), and fibrinolytic and kinin-kallikrein systems (KKS) has been shown to play a central role in the pathogenesis of the disease. Since the C1 esterase inhibitor (C1-INH) is a potent inhibitor of all these systems, its role in the disease has been investigated, but some issues remained unresolved. METHODS We evaluated the impact of C1-INH and KKS on disease progression in a cohort of 45 COVID-19 patients divided into groups according to disease severity. We measured plasma levels of total and functional C1-INH and its complexes with kallikrein (PKa), reflecting KKS activation and kallikrein spontaneous activity. RESULTS We observed increased total and functional plasma concentrations of C1-INH in COVID-19 patients. A direct correlation (positive Spearman's r) was observed between C1-INH levels, especially functional C1-INH, and the severity of the disease. Moreover, a significant reduction in the ratio of functional over total C1-INH was evident in patients exhibiting mild to intermediate clinical severity but not in critically ill patients. Accordingly, activation of the KKS, assessed as an increase in PKa:C1-INH complexes, was explicitly observed in the mild categories. CONCLUSIONS Our study's findings on the consumption of C1-INH and the activation of the KKS in the less severe stages of COVID-19 but not in the critical stage suggest a potential role for C1-INH in containing disease severity. These results underscore the importance of C1-INH in the early phases of the disease and its potential implications in COVID-19 progression and/or long-term effects.
Collapse
Affiliation(s)
- Silvia Berra
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Internal Medicine, Ospedale Fatebenefratelli, 20121 Milan, Italy
| | - Debora Parolin
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
| | - Chiara Suffritti
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, 20122 Milan, Italy
| | - Andrea Folcia
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Division of Oncology, Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Andrea Zanichelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
- Operative Unit of Medicine, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Luca Gusso
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Internal Medicine Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Chiara Cogliati
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Internal Medicine, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
- Department of Infectious Diseases, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Antonio Gidaro
- Department of Internal Medicine, Ospedale Luigi Sacco, 20157 Milan, Italy
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (S.B.); (D.P.); (C.S.); (A.F.); (L.G.); (C.C.); (A.R.)
| |
Collapse
|
4
|
Azoulay E, Zuber J, Bousfiha AA, Long Y, Tan Y, Luo S, Essafti M, Annane D. Complement system activation: bridging physiology, pathophysiology, and therapy. Intensive Care Med 2024; 50:1791-1803. [PMID: 39254734 DOI: 10.1007/s00134-024-07611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/10/2024] [Indexed: 09/11/2024]
Abstract
The complement system is a set of over 50 proteins that constitutes an essential part of the innate immune system. Complement system activation involves an organized proteolytic cascade. Overactivation of complement system activation is the main pathogenic mechanism of several diseases and contributes to the manifestations of many other conditions. This review describes the normal complement system and the role for complement dysregulation in critical illnesses, notably sepsis and acute respiratory distress syndrome. Complement activation is involved in the immune system response to pathogens but, when excessive, can contribute to tissue damage, runaway inflammation, and capillary leakage syndrome. Complement overactivation may play a key role in severe forms of coronavirus disease 2019 (COVID-19). Two diseases whose manifestations are mainly caused by complement overactivation, namely, atypical hemolytic and uremic syndrome (aHUS) and myasthenia gravis, are discussed. A diagnostic algorithm for aHUS is provided. Early complement-inhibiting therapy has been proven effective. When renal transplantation is required, complement-inhibiting drugs can be used prophylactically to prevent aHUS recurrence. Similarly, acetylcholine-receptor autoantibody-positive generalized myasthenia gravis involves complement system overactivation and responds to complement inhibition. The two main complement inhibitors used in to date routine are eculizumab and ravulizumab. The main adverse event is Neisseria infection, which is rare and preventable, but can be fatal. The complement system is crucial to health but, when overactivated, can cause or contribute to disease. Effective complement inhibitors are now available, although additional data are required to determine optimal regimens. Further research is also needed to better understand the complement system, develop advanced diagnostic tools, and identify markers that allow the personalization of treatment strategies.
Collapse
Affiliation(s)
- Elie Azoulay
- Intensive Care Unit, Saint-Louis University Hospital, AP-HP, Paris Cité University, Paris, France.
| | - Julien Zuber
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker University Hospital, AP-HP, Paris, France
| | - Ahmed Aziz Bousfiha
- Department of Pediatric Infectious and Immunological Diseases, IbnRochd University Hospital, Casablanca, Morocco
- Laboratory of Clinical Immunology, Inflammation and Allergy (LICIA), Casablanca, Morocco
- School of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Yun Long
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China
- Institute of Nephrology, Peking University, Beijing, PR China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China
| | - Sushan Luo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, PR China
- Huashan Rare Diseases Center, Huashan Hospital, Fudan University, Shanghai, PR China
- National Center for Neurological Diseases, Shanghai, PR China
| | - Meriem Essafti
- Intensive Care Department, Mother-Children Center, Mohamed VI University Hospital, Marrakech, Morocco
| | - Djillali Annane
- Department of Intensive Care, Raymond Poincaré Hospital, AP-HP, Garches, France
- Simone Veil School of Medicine, Versailles-Saint Quentin University, Paris-Saclay University, Versaillles, France
- Institut Hospitalo-Universitaire PROMETHEUS & Fédération Hospitalo-Universitaire SEPSIS, Paris-Saclay University, Saclay, France
- INSERM, Garches, France
| |
Collapse
|
5
|
Jiang H, Guo Y, Wang Q, Wang Y, Peng D, Fang Y, Yan L, Ruan Z, Zhang S, Zhao Y, Zhang W, Shang W, Feng Z. The dysfunction of complement and coagulation in diseases: the implications for the therapeutic interventions. MedComm (Beijing) 2024; 5:e785. [PMID: 39445002 PMCID: PMC11496570 DOI: 10.1002/mco2.785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
The complement system, comprising over 30 proteins, is integral to the immune system, and the coagulation system is critical for vascular homeostasis. The activation of the complement and coagulation systems involves an organized proteolytic cascade, and the overactivation of these systems is a central pathogenic mechanism in several diseases. This review describes the role of complement and coagulation system activation in critical illness, particularly sepsis. The complexities of sepsis reveal significant knowledge gaps that can be compared to a profound abyss, highlighting the urgent need for further investigation and exploration. It is well recognized that the inflammatory network, coagulation, and complement systems are integral mechanisms through which multiple factors contribute to increased susceptibility to infection and may result in a disordered immune response during septic events in patients. Given the overlapping pathogenic mechanisms in sepsis, immunomodulatory therapies currently under development may be particularly beneficial for patients with sepsis who have concurrent infections. Herein, we present recent findings regarding the molecular relationships between the coagulation and complement pathways in the advancement of sepsis, and propose potential intervention targets related to the crosstalk between coagulation and complement, aiming to provide more valuable treatment of sepsis.
Collapse
Affiliation(s)
- Honghong Jiang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yiming Guo
- Department of Biological Science, The Dietrich School of Arts and SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Qihang Wang
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Yiran Wang
- Department of Obstetrics and GynecologyThe sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Dingchuan Peng
- School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yigong Fang
- Institute of Acupuncture and MoxibustionChina Academy of Chinese Medical SciencesBeijingChina
| | - Lei Yan
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Zhuolin Ruan
- Department of Obstetrics and Gynecology,Chinese PLA General HospitalBeijingChina
| | - Sheng Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yong Zhao
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Wendan Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Wei Shang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zhichun Feng
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| |
Collapse
|
6
|
Seki K, Sueyoshi K, Miyoshi Y, Nakamura Y, Ishihara T, Kondo Y, Kuroda Y, Yonekura A, Iwabuchi K, Okamoto K, Tanaka H. Complement activation and lung injury in Japanese patients with COVID-19: a prospective observational study. Sci Rep 2024; 14:24895. [PMID: 39438600 PMCID: PMC11496683 DOI: 10.1038/s41598-024-76365-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Background The production of inflammatory cytokines is reportedly increased in patients with coronavirus disease 2019 (COVID-19), causing the migration of neutrophils and monocytes to lung tissues. This disrupts the air-blood barrier by damaging the bronchial epithelial and vascular endothelial cells. As multiorgan dysfunction in sepsis is considered to be partly caused by complement activation, which can cause lung injury in patients with COVID-19. There are limited studies examining the link between complement activation in patients with COVID-19. This study aimed to AQinvestigate the association of complement activation with the pathophysiology of COVID-19. Twenty-seven patients with COVID-19 were enrolled in this study and classified into two groups depending on the indication for mechanical ventilation. Plasma complement factors (C3a, C5a, Ba, and sC5b-9), complement regulators (sCD59 and factor H), interleukin-6 (IL-6), and syndecan-1 levels were measured using Enzyme-linked immunosorbent assay (ELISA). Results: All complement factors and regulators, IL-6, and syndecan-1 levels were significantly elevated in patients with COVID-19 compared with those in healthy controls. C5a and sC5b-9 levels were decreased significantly in the invasive mechanical ventilation (IMV) group compared with those in the non-IMV group. Syndecan-1 levels were significantly increased in the IMV group compared with those in the non-IMV group. Conclusions: Complement activation is an exacerbating factor for lung injury in patients with COVID-19. Complement factors are nonessential predictors of mechanical ventilation; however, syndecan-1 could be a biomarker of COVID-19 severity in patients.
Collapse
Affiliation(s)
- Kentaro Seki
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Koichiro Sueyoshi
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan.
| | - Yukari Miyoshi
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Yuki Nakamura
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Tadashi Ishihara
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Yutaka Kondo
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Yoko Kuroda
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Ayumi Yonekura
- Institute for Environmental and Gender Specific Medicine, Juntendo University, Urayasu, Chiba, Japan
| | - Kazuhisa Iwabuchi
- Institute for Environmental and Gender Specific Medicine, Juntendo University, Urayasu, Chiba, Japan
| | - Ken Okamoto
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Hiroshi Tanaka
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| |
Collapse
|
7
|
Sababathy M, Ramanathan G, Ganesan S, Sababathy S, Yasmin A, Ramasamy R, Foo J, Looi Q, Nur-Fazila S. Multipotent mesenchymal stromal/stem cell-based therapies for acute respiratory distress syndrome: current progress, challenges, and future frontiers. Braz J Med Biol Res 2024; 57:e13219. [PMID: 39417447 PMCID: PMC11484355 DOI: 10.1590/1414-431x2024e13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/30/2024] [Indexed: 10/19/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a critical, life-threatening condition marked by severe inflammation and impaired lung function. Mesenchymal stromal/stem cells (MSCs) present a promising therapeutic avenue due to their immunomodulatory, anti-inflammatory, and regenerative capabilities. This review comprehensively evaluates MSC-based strategies for ARDS treatment, including direct administration, tissue engineering, extracellular vesicles (EVs), nanoparticles, natural products, artificial intelligence (AI), gene modification, and MSC preconditioning. Direct MSC administration has demonstrated therapeutic potential but necessitates optimization to overcome challenges related to effective cell delivery, homing, and integration into damaged lung tissue. Tissue engineering methods, such as 3D-printed scaffolds and MSC sheets, enhance MSC survival and functionality within lung tissue. EVs and MSC-derived nanoparticles offer scalable and safer alternatives to cell-based therapies. Likewise, natural products and bioactive compounds derived from plants can augment MSC function and resilience, offering complementary strategies to enhance therapeutic outcomes. In addition, AI technologies could aid in optimizing MSC delivery and dosing, and gene editing tools like CRISPR/Cas9 allow precise modification of MSCs to enhance their therapeutic properties and target specific ARDS mechanisms. Preconditioning MSCs with hypoxia, growth factors, or pharmacological agents further enhances their therapeutic potential. While MSC therapies hold significant promise for ARDS, extensive research and clinical trials are essential to determine optimal protocols and ensure long-term safety and effectiveness.
Collapse
Affiliation(s)
- M. Sababathy
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - G. Ramanathan
- Faculty of Computer Science and Information Technology, University Malaya, Kuala Lumpur, Malaysia
| | - S. Ganesan
- School of Pharmacy, Management and Science University, Shah Alam, Selangor, Malaysia
| | - S. Sababathy
- Faculty of Medicine and Defence Health, National Defence University of Malaysia, Sungai Besi, Kuala Lumpur, Malaysia
| | - A.R. Yasmin
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Vaccines and Biomolecules, Institute of Bioscience, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - R. Ramasamy
- Department of Pathology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - J.B. Foo
- Center for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Q.H. Looi
- My Cytohealth Sdn. Bhd., Bandar Seri Petaling, Kuala Lumpur, Malaysia
| | - S.H. Nur-Fazila
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Vaccines and Biomolecules, Institute of Bioscience, University Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
8
|
Ellsworth CR, Chen Z, Xiao MT, Qian C, Wang C, Khatun MS, Liu S, Islamuddin M, Maness NJ, Halperin JA, Blair RV, Kolls JK, Tomlinson S, Qin X. Enhanced complement activation and MAC formation accelerates severe COVID-19. Cell Mol Life Sci 2024; 81:405. [PMID: 39284944 PMCID: PMC11405604 DOI: 10.1007/s00018-024-05430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/31/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
Emerging evidence indicates that activation of complement system leading to the formation of the membrane attack complex (MAC) plays a detrimental role in COVID-19. However, their pathogenic roles have never been experimentally investigated before. We used three knock out mice strains (1. C3-/-; 2. C7-/-; and 3. Cd59ab-/-) to evaluate the role of complement in severe COVID-19 pathogenesis. C3 deficient mice lack a key common component of all three complement activation pathways and are unable to generate C3 and C5 convertases. C7 deficient mice lack a complement protein needed for MAC formation. Cd59ab deficient mice lack an important inhibitor of MAC formation. We also used anti-C5 antibody to block and evaluate the therapeutic potential of inhibiting MAC formation. We demonstrate that inhibition of complement activation (in C3-/-) and MAC formation (in C3-/-. C7-/-, and anti-C5 antibody) attenuates severe COVID-19; whereas enhancement of MAC formation (Cd59ab-/-) accelerates severe COVID-19. The degree of MAC but not C3 deposits in the lungs of C3-/-, C7-/- mice, and Cd59ab-/- mice as compared to their control mice is associated with the attenuation or acceleration of SARS-CoV-2-induced disease. Further, the lack of terminal complement activation for the formation of MAC in C7 deficient mice protects endothelial dysfunction, which is associated with the attenuation of diseases and pathologic changes. Our results demonstrated the causative effect of MAC in severe COVID-19 and indicate a potential avenue for modulating the complement system and MAC formation in the treatment of severe COVID-19.
Collapse
Affiliation(s)
- Calder R Ellsworth
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Zheng Chen
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mark T Xiao
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Chaosi Qian
- Department of Microbiology and Immunology, Medical University of South Carolina, and Ralph Johnson VA Medical Center, Charleston, SC, USA
| | - Chenxiao Wang
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mst Shamima Khatun
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shumei Liu
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mohammad Islamuddin
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Jose A Halperin
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Robert V Blair
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Jay K Kolls
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, and Ralph Johnson VA Medical Center, Charleston, SC, USA
| | - Xuebin Qin
- Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703, Three Rivers Road, Covington, LA, 70433, USA.
| |
Collapse
|
9
|
Sayed AA. Back to Basics: The Diagnostic Value of a Complete Blood Count in the Clinical Management of COVID-19. Diagnostics (Basel) 2024; 14:1933. [PMID: 39272717 PMCID: PMC11393994 DOI: 10.3390/diagnostics14171933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Since the beginning of the COVID-19 pandemic, scientists have struggled significantly to understand the complexity of COVID-19 pathophysiology. COVID-19 has demonstrated a notoriously unpredictable clinical course. This unpredictability constituted a significant obstacle to clinicians in predicting the disease course among COVID-19 patients, more specifically, in predicting who would develop severe cases and possibly die from the infection. This brief report aims to assess the diagnostic value of using a complete blood count (CBC) and applying high-dimensional analysis, i.e., principal component analysis (PCA), on it to differentiate between patients with mild and severe COVID-19 infection. The data of 855 patients were retrieved from multiple centres in Saudi Arabia. Descriptive statistics, such as counts, percentages, and medians (interquartile ranges) were used to describe patients' characteristics and CBC parameters. Analytical statistics, such as the Mann-Whitney U test, were used to compare between survivors and non-survivors. PCA was applied using the CBC parameters, and the results were compared between survivors and non-survivors. Patients in this study had a median age of 41, with an almost equal ratio of men to women. Most participants were Saudis, and non-survivors were 13.22% of the total cohort. The median values of all CBC indices were within reference ranges; however, some statistically significant differences were observed between survivors and non-survivors. Non-survivors had lower hemoglobin levels and lower hematocrit, lymphocyte, and eosinophil counts but higher WBC and neutrophil counts compared to survivors. PCA on the CBC results of survivors yielded a significantly different profile than non-survivors, indicating the possibility of its use in the context of COVID-19. The diagnostic value of CBC in the clinical management of COVID-19 should be utilized in clinical guidelines for managing COVID-19 cases.
Collapse
Affiliation(s)
- Anwar A Sayed
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
10
|
Budylowski P, Chau SLL, Banerjee A, Guvenc F, Samson R, Hu Q, Fiddes L, Seifried L, Chao G, Buchholz M, Estacio A, Cheatley PL, Pavenski K, Patriquin CJ, Liu Y, Sheikh-Mohamed S, Crasta K, Yue F, Pasic MD, Mossman K, Gingras AC, Gommerman JL, Ehrhardt GRA, Mubareka S, Ostrowski M. A Significant Contribution of the Classical Pathway of Complement in SARS-CoV-2 Neutralization of Convalescent and Vaccinee Sera. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1922-1931. [PMID: 38683124 DOI: 10.4049/jimmunol.2300320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/09/2024] [Indexed: 05/01/2024]
Abstract
Although high titers of neutralizing Abs in human serum are associated with protection from reinfection by SARS-CoV-2, there is considerable heterogeneity in human serum-neutralizing Abs against SARS-CoV-2 during convalescence between individuals. Standard human serum live virus neutralization assays require inactivation of serum/plasma prior to testing. In this study, we report that the SARS-CoV-2 neutralization titers of human convalescent sera were relatively consistent across all disease states except for severe COVID-19, which yielded significantly higher neutralization titers. Furthermore, we show that heat inactivation of human serum significantly lowered neutralization activity in a live virus SARS-CoV-2 neutralization assay. Heat inactivation of human convalescent serum was shown to inactivate complement proteins, and the contribution of complement in SARS-CoV-2 neutralization was often >50% of the neutralizing activity of human sera without heat inactivation and could account for neutralizing activity when standard titers were zero after heat inactivation. This effect was also observed in COVID-19 vaccinees and could be abolished in individuals who were undergoing treatment with therapeutic anti-complement Abs. Complement activity was mainly dependent on the classical pathway with little contributions from mannose-binding lectin and alternative pathways. Our study demonstrates the importance of the complement pathway in significantly increasing viral neutralization activity against SARS-CoV-2 in spike seropositive individuals.
Collapse
Affiliation(s)
- Patrick Budylowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Serena L L Chau
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Arinjay Banerjee
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Furkan Guvenc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Reuben Samson
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Queenie Hu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Lindsey Fiddes
- Microscopy Imaging Lab, University of Toronto, Toronto, Ontario, Canada
| | - Laurie Seifried
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Gary Chao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Megan Buchholz
- Apheresis Unit, Kidney and Metabolism Program, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Antonio Estacio
- Keenan Research Centre for Biomedical Science of St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Patti Lou Cheatley
- Apheresis Unit, Kidney and Metabolism Program, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Katerina Pavenski
- Apheresis Unit, Kidney and Metabolism Program, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Christopher J Patriquin
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Yanling Liu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Kimberly Crasta
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - FengYun Yue
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Maria D Pasic
- Department of Immunology, Unity Health Toronto, Toronto, Ontario, Canada
| | - Karen Mossman
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | | | - Götz R A Ehrhardt
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Samira Mubareka
- Sunnybrook Research Institute, Sunnybrook Hospital, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science of St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Lee JD, Woodruff TM. Complement(ing) long-COVID thromboinflammation and pathogenesis. Trends Immunol 2024; 45:397-399. [PMID: 38637200 DOI: 10.1016/j.it.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
The persistence or recurrence of symptoms after acute SARS-CoV-2 infection, termed 'long COVID', presents a formidable challenge to global healthcare systems. Recent research by Cervia-Hasler and colleagues delves into the intricate immunological landscape in patients with long COVID, demonstrating an interplay between complement and coagulation, driven by antiviral antibodies and tissue damage.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia; Queensland Brain Institute, the University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
12
|
Kircheis R. In Silico Analyses Indicate a Lower Potency for Dimerization of TLR4/MD-2 as the Reason for the Lower Pathogenicity of Omicron Compared to Wild-Type Virus and Earlier SARS-CoV-2 Variants. Int J Mol Sci 2024; 25:5451. [PMID: 38791489 PMCID: PMC11121871 DOI: 10.3390/ijms25105451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The SARS-CoV-2 Omicron variants have replaced all earlier variants, due to increased infectivity and effective evasion from infection- and vaccination-induced neutralizing antibodies. Compared to earlier variants of concern (VoCs), the Omicron variants show high TMPRSS2-independent replication in the upper airway organs, but lower replication in the lungs and lower mortality rates. The shift in cellular tropism and towards lower pathogenicity of Omicron was hypothesized to correlate with a lower toll-like receptor (TLR) activation, although the underlying molecular mechanisms remained undefined. In silico analyses presented here indicate that the Omicron spike protein has a lower potency to induce dimerization of TLR4/MD-2 compared to wild type virus despite a comparable binding activity to TLR4. A model illustrating the molecular consequences of the different potencies of the Omicron spike protein vs. wild-type spike protein for TLR4 activation is presented. Further analyses indicate a clear tendency for decreasing TLR4 dimerization potential during SARS-CoV-2 evolution via Alpha to Gamma to Delta to Omicron variants.
Collapse
|
13
|
Dos Ramos Almeida CJL, Veras FP, Paiva IM, Schneider AH, da Costa Silva J, Gomes GF, Costa VF, Silva BMS, Caetite DB, Silva CMS, Salina ACG, Martins R, Bonilha CS, Cunha LD, Jamur MC, da Silva LLP, Arruda E, Zamboni DS, Louzada-Junior P, de Oliveira RDR, Alves-Filho JC, Cunha TM, de Queiroz Cunha F. Neutrophil Virucidal Activity Against SARS-CoV-2 Is Mediated by Neutrophil Extracellular Traps. J Infect Dis 2024; 229:1352-1365. [PMID: 38015657 DOI: 10.1093/infdis/jiad526] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Inflammation in the lungs and other vital organs in COVID-19 is characterized by the presence of neutrophils and a high concentration of neutrophil extracellular traps (NETs), which seems to mediate host tissue damage. However, it is not known whether NETs could have virucidal activity against SARS-CoV-2. METHODS We investigated whether NETs could prevent SARS-CoV-2 replication in neutrophils and epithelial cells and what the consequence of NETs degradation would be in K18-humanized ACE2 transgenic mice infected with SARS-CoV-2. RESULTS Here, by immunofluorescence microscopy, we observed that viral particles colocalize with NETs in neutrophils isolated from patients with COVID-19 or healthy individuals and infected in vitro. The inhibition of NETs production increased virus replication in neutrophils. In parallel, we observed that NETs inhibited virus abilities to infect and replicate in epithelial cells after 24 hours of infection. Degradation of NETs with DNase I prevented their virucidal effect in vitro. Using K18-humanized ACE2 transgenic mice, we observed a higher viral load in animals treated with DNase I. However, the virucidal effect of NETs was not dependent on neutrophil elastase or myeloperoxidase activity. CONCLUSIONS Our results provide evidence of the role of NETosis as a mechanism of SARS-CoV-2 viral capture and inhibition.
Collapse
Affiliation(s)
| | - Flávio Protásio Veras
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto
| | - Isadora Marques Paiva
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Ayda Henriques Schneider
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Juliana da Costa Silva
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Giovanni Freitas Gomes
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Victor Ferreira Costa
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | - Diego Brito Caetite
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | | | - Ronaldo Martins
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Caio Santos Bonilha
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | - Maria Célia Jamur
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
| | - Luís Lamberti Pinto da Silva
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Eurico Arruda
- Department of Cellular and Molecular Biology and Pathogenic Bioagents
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | | | - Paulo Louzada-Junior
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | | | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| | - Fernando de Queiroz Cunha
- Center for Research in Inflammatory Diseases
- Department of Pharmacology, Ribeirão Preto Medical School
| |
Collapse
|
14
|
Jiao Y, Zhou L, Huo J, Li H, Zhu H, Chen D, Lu Y. Flavonoid substituted polysaccharides from Tamarix chinensis Lour. alleviate H1N1-induced acute lung injury via inhibiting complement system. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117651. [PMID: 38135232 DOI: 10.1016/j.jep.2023.117651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Viral pneumonia is a highly pathogenic respiratory infectious disease associated with excessive activation of the complement system. Our previous studies found that the anticomplement polysaccharides from some medicinal plants could significantly alleviate H1N1-induced acute lung injury (H1N1-ALI). The leaves and twigs of Tamarix chinensis Lour. are traditionally used as a Chinese medicine Xiheliu for treating inflammatory disorders. Interestingly, its crude polysaccharides (MBAP90) showed potent anticomplement activity in vitro. AIM OF THE STUDY To evaluate the therapeutic effects and possible mechanism of MBAP90 on viral pneumonia and further isolate and characterize the key active substance of MBAP90. MATERIALS AND METHODS The protective effects of MBAP90 were evaluated by survival tests and pharmacodynamic experiments on H1N1-ALI mice. Histopathological changes, viral load, inflammatory markers, and complement deposition in lungs were analyzed by H&E staining, enzyme-linked immunosorbent assay (ELISA), and immunohistochemistry (IHC), respectively. An anticomplement homogenous polysaccharide (MBAP-3) was obtained from MBAP90 by bio-guided separation, and its structure was further characterized by methylation analysis and NMR spectroscopy. RESULTS Oral administration of MBAP90 at a dose of 400 mg/kg significantly increased the survival rate of mice infected with the lethal H1N1 virus. In H1N1-induced ALI, mice treated with MBAP90 (200 and 400 mg/kg) could decrease the lung index, lung pathological injury, the levels of excessive proinflammatory cytokines (IL-6, TNF-α, MCP-1, IL-18, and IL-1β), and complement levels (C3c and C5b-9). In addition, MBAP-3 was characterized as a novel homogenous polysaccharide with potent in vitro anticomplement activity (CH50: 0.126 ± 0.002 mg/mL), containing 10.51% uronic acids and 9.67% flavonoids, which were similar to the composition of MBAP90. The backbone of MBAP-3 consisted of →4)-α-D-Glcp-(1→, →3,4,6)-α-D-Glcp-(1→, and →3,4)-α-D-Glcp-(1→, with branches comprising α-L-Araf-(1→, α-D-GlcpA-(1→, →4,6)-α-D-Manp-(1→ and →4)-β-D-Galp-(1 → . Particularly, O-6 of →4)-β-D-Galp-(1→ was conjugated with a flavonoid, myricetin. CONCLUSIONS MBAP90 could ameliorate H1N1-ALI by inhibiting inflammation and over-activation of the complement system. These polysaccharides (MBAP90 and MBAP-3) with relative high contents of uronic acid and flavonoid substituent might be vital components of T. chinensis for treating viral pneumonia.
Collapse
Affiliation(s)
- Yukun Jiao
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, China.
| | - Lishuang Zhou
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, China.
| | - Jiangyan Huo
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, China.
| | - Hong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| | - Haiyan Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai, China.
| | - Daofeng Chen
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, China; Institutes of Integrative Medicine, School of Pharmacy, Fudan University, Shanghai, China.
| | - Yan Lu
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Radke J, Meinhardt J, Aschman T, Chua RL, Farztdinov V, Lukassen S, Ten FW, Friebel E, Ishaque N, Franz J, Huhle VH, Mothes R, Peters K, Thomas C, Schneeberger S, Schumann E, Kawelke L, Jünger J, Horst V, Streit S, von Manitius R, Körtvélyessy P, Vielhaber S, Reinhold D, Hauser AE, Osterloh A, Enghard P, Ihlow J, Elezkurtaj S, Horst D, Kurth F, Müller MA, Gassen NC, Melchert J, Jechow K, Timmermann B, Fernandez-Zapata C, Böttcher C, Stenzel W, Krüger E, Landthaler M, Wyler E, Corman V, Stadelmann C, Ralser M, Eils R, Heppner FL, Mülleder M, Conrad C, Radbruch H. Proteomic and transcriptomic profiling of brainstem, cerebellum and olfactory tissues in early- and late-phase COVID-19. Nat Neurosci 2024; 27:409-420. [PMID: 38366144 DOI: 10.1038/s41593-024-01573-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/08/2024] [Indexed: 02/18/2024]
Abstract
Neurological symptoms, including cognitive impairment and fatigue, can occur in both the acute infection phase of coronavirus disease 2019 (COVID-19) and at later stages, yet the mechanisms that contribute to this remain unclear. Here we profiled single-nucleus transcriptomes and proteomes of brainstem tissue from deceased individuals at various stages of COVID-19. We detected an inflammatory type I interferon response in acute COVID-19 cases, which resolves in the late disease phase. Integrating single-nucleus RNA sequencing and spatial transcriptomics, we could localize two patterns of reaction to severe systemic inflammation, one neuronal with a direct focus on cranial nerve nuclei and a separate diffuse pattern affecting the whole brainstem. The latter reflects a bystander effect of the respiratory infection that spreads throughout the vascular unit and alters the transcriptional state of mainly oligodendrocytes, microglia and astrocytes, while alterations of the brainstem nuclei could reflect the connection of the immune system and the central nervous system via, for example, the vagus nerve. Our results indicate that even without persistence of severe acute respiratory syndrome coronavirus 2 in the central nervous system, local immune reactions are prevailing, potentially causing functional disturbances that contribute to neurological complications of COVID-19.
Collapse
Affiliation(s)
- Josefine Radke
- Institute of Pathology, Universitätsmedizin Greifswald, Greifswald, Germany.
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Jenny Meinhardt
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tom Aschman
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Lorenz Chua
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Vadim Farztdinov
- Core Facility High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sören Lukassen
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Foo Wei Ten
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ekaterina Friebel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Naveed Ishaque
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jonas Franz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Valerie Helena Huhle
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ronja Mothes
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kristin Peters
- Institute of Pathology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Carolina Thomas
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Shirin Schneeberger
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elisa Schumann
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Leona Kawelke
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Jünger
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Viktor Horst
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Streit
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Regina von Manitius
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Péter Körtvélyessy
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto von Guerike University Magdeburg, Magdeburg, Germany
| | - Dirk Reinhold
- Institute of Molecular and Clinical Immunology, Otto von Guerike University Magdeburg, Magdeburg, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Berlin, Germany
| | - Anja Osterloh
- Department of Pathology, University Medical Center Ulm, Ulm, Germany
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jana Ihlow
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sefer Elezkurtaj
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David Horst
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel A Müller
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils C Gassen
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Julia Melchert
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Jechow
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Camila Fernandez-Zapata
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Chotima Böttcher
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Markus Landthaler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institut für Biologie, Humboldt Universität, Berlin, Germany
| | - Emanuel Wyler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Victor Corman
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Centre for Infection Research (DZIF), associated partner, Berlin, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Ralser
- Core Facility High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Eils
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank L Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Cluster of Excellence NeuroCure, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Conrad
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Helena Radbruch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Pushalkar S, Wu S, Maity S, Pressler M, Rendleman J, Vitrinel B, Jeffery L, Abdelhadi R, Chen M, Ross T, Carlock M, Choi H, Vogel C. Complex changes in serum protein levels in COVID-19 convalescents. Sci Rep 2024; 14:4479. [PMID: 38396092 PMCID: PMC10891133 DOI: 10.1038/s41598-024-54534-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The COVID-19 pandemic, triggered by severe acute respiratory syndrome coronavirus 2, has affected millions of people worldwide. Much research has been dedicated to our understanding of COVID-19 disease heterogeneity and severity, but less is known about recovery associated changes. To address this gap in knowledge, we quantified the proteome from serum samples from 29 COVID-19 convalescents and 29 age-, race-, and sex-matched healthy controls. Samples were acquired within the first months of the pandemic. Many proteins from pathways known to change during acute COVID-19 illness, such as from the complement cascade, coagulation system, inflammation and adaptive immune system, had returned to levels seen in healthy controls. In comparison, we identified 22 and 15 proteins with significantly elevated and lowered levels, respectively, amongst COVID-19 convalescents compared to healthy controls. Some of the changes were similar to those observed for the acute phase of the disease, i.e. elevated levels of proteins from hemolysis, the adaptive immune systems, and inflammation. In contrast, some alterations opposed those in the acute phase, e.g. elevated levels of CETP and APOA1 which function in lipid/cholesterol metabolism, and decreased levels of proteins from the complement cascade (e.g. C1R, C1S, and VWF), the coagulation system (e.g. THBS1 and VWF), and the regulation of the actin cytoskeleton (e.g. PFN1 and CFL1) amongst COVID-19 convalescents. We speculate that some of these shifts might originate from a transient decrease in platelet counts upon recovery from the disease. Finally, we observed race-specific changes, e.g. with respect to immunoglobulins and proteins related to cholesterol metabolism.
Collapse
Affiliation(s)
- Smruti Pushalkar
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| | - Shaohuan Wu
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Shuvadeep Maity
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
- Birla Institute of Technology and Science-Pilani (BITS Pilani), Hyderabad, India
| | - Matthew Pressler
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Justin Rendleman
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Burcu Vitrinel
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Lauren Jeffery
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Ryah Abdelhadi
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Mechi Chen
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Ted Ross
- Cleveland Clinic Florida Research & Innovation Center, Port St. Lucie, FL, USA
| | - Michael Carlock
- Cleveland Clinic Florida Research & Innovation Center, Port St. Lucie, FL, USA
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christine Vogel
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
17
|
He X, Tan T, Yang S, Feng Y, Wen Q. Characterisation of an anticomplement polysaccharide BP-S1 from seeds of Brucea javanica. Nat Prod Res 2024:1-13. [PMID: 38189427 DOI: 10.1080/14786419.2023.2300399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/24/2023] [Indexed: 01/09/2024]
Abstract
Bioactivity-guided purification obtained polysaccharide BP-S1 from seeds of Brucea javanica. The results showed that BP-S1 was a homogenous polysaccharide with molecular weight of 45.7 kDa, mainly composed of arabinose and glucose in the ratio of 1.0:1.0 and the backbone of BP-S1 was deduced to be →3,4)-α-Glup-(1→ with branches of →2)-α-Arap-(1→and α-Arap-(1→, and the possible repetitive units were speculated according to result of methylation and 2D-NMR. Moreover, BP-S1 is a periodic rope-like structure. Functional analysis revealed that BP-S1 inhibited complement activation on the classic and alternative pathways with values of CH50 0.073 ± 0.012 mg/mL and AP50 0.097 ± 0.004 mg/mL, respectively. In mechanism study, using complement component depleted-sera methods indicated that BP-S1 selectively interacted with C3 and C4 components.
Collapse
Affiliation(s)
- Xiao He
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, P. R. China
| | - Ting Tan
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, P. R. China
| | - Shilin Yang
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, P. R. China
- State Key Laboratory of Innovative Drug and Efficient Energy- Saving Pharmaceutical Equipment, Nanchang, Jiangxi, China
| | - Yulin Feng
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, P. R. China
- State Key Laboratory of Innovative Drug and Efficient Energy- Saving Pharmaceutical Equipment, Nanchang, Jiangxi, China
| | - Quan Wen
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, P. R. China
- State Key Laboratory of Innovative Drug and Efficient Energy- Saving Pharmaceutical Equipment, Nanchang, Jiangxi, China
| |
Collapse
|
18
|
Abakar MAA, Hamad DHA, Faisal E, Omer HMFA, Faki MTM, Idris AEM, Omer R, Osman Z, Elhassan EAG, Abrahim-Holie MA, Hakami MA, Alghamdi A, Alfahed A, Aloraini GS, Alharthi NS, Waggiallah HA. Comparative analysis of immunological biomarkers in COVID-19 and bacterial pneumonia. J Med Life 2023; 16:1844-1851. [PMID: 38585537 PMCID: PMC10994609 DOI: 10.25122/jml-2023-0273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/19/2023] [Indexed: 04/09/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a severe and infectious respiratory condition caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This case-control study aimed to evaluate serum levels of various immunological markers in patients with COVID-19 compared to those with bacterial pneumonia and a healthy control group. Serum samples were collected from adult participants across various COVID-19 isolation centers, including Kassala State and Ahmed Gasim Hospital, between April and June 2021. The study included 70 patients diagnosed with COVID-19, 30 with bacterial pneumonia, and 50 healthy controls. Serum levels of C-reactive protein (CRP), complement components C3 and C4, and cytokines IL-8, IL-10, IL-12, TNF-α, and IFN-γ were measured using standard reagent kits. Serum level of CRP was significantly elevated in both bacterial pneumonia and COVID-19 but significantly higher among patients with bacterial pneumonia. C3 and C4 were also increased in both patient groups, with C3 significantly higher in bacterial pneumonia. IL-8, IL-10, IL-12, TNF-α, and IFN-γ were significantly increased in bacterial pneumonia and SARS-Cov-2 compared to healthy controls. However, IFN-γ was significantly increased among patients with COVID-19 than patients with bacterial pneumonia. This study highlights the potential significant impact of COVID-19 on the immunological biomarkers investigated.
Collapse
Affiliation(s)
| | | | - Eman Faisal
- Department of Microbiology, Faculty of Medical Laboratory, University of Kassala, Kassala, Kassala State, Sudan
| | | | | | | | - Rouwida Omer
- Ministry of Health, Kassala, Kassala State, Sudan
| | - Zeinab Osman
- Medical Laboratory, Khartoum, Khartoum State, Sudan
| | | | - Mohamed Ahmed Abrahim-Holie
- Department of Medical Microbiology and Immunology, Faculty of Medical Laboratory Sciences, Alzaiem Alazhari University, Khartoum, Khartoum State, Sudan
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Abdullah Alghamdi
- Deparment of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Abdulaziz Alfahed
- Deparment of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Ghfren Suliman Aloraini
- Deparment of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Nahed Sail Alharthi
- Deparment of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Hisham Ali Waggiallah
- Deparment of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| |
Collapse
|
19
|
Albalawi M, Alwasaidi T, Almohammadi M, Alzarief AA, Elyamany G. Paroxysmal nocturnal hemoglobinuria in a patient post COVID-19 virus infection: A case report with literature review. Clin Case Rep 2023; 11:e8011. [PMID: 37822485 PMCID: PMC10562656 DOI: 10.1002/ccr3.8011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/21/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023] Open
Abstract
COVID 19 is a serious infection that originated in Wuhan, China and has resulted in worldwide morbidity and mortality. It continues to be a major health concern in 2022, being associated with multiorgan failure. Although the pathophysiology of the disease and its complications are not well understood, it is believed that a cytokine storm, triggered by complement activation may be responsible for the severity and complications of the disease. As of now, there is no definitive treatment available. Hematological changes associated with COVID-19 include lymphopenia, anemia, thrombocytopenia, disseminated intravascular coagulopathy, and thrombosis. Paroxysmal nocturnal hemoglobinuria (PNH), on the other hand, is an acquired clonal hematopoietic stem cell disorder that occurs due to an acquired PIG-A mutation affecting the hematopoietic stem cells. Interestingly, PNH exhibits some clinical and laboratory manifestations like those seen in COVID-19. In this report, we present a rare case of PNH that developed following a COVID-19 infection.
Collapse
Affiliation(s)
- Mohammed Albalawi
- Department of Internal Medicine, College of MedicineTaibah UniversityMadinahSaudi Arabia
| | - Turki Alwasaidi
- Department of Internal Medicine, College of MedicineTaibah UniversityMadinahSaudi Arabia
- Medicine DepartmentPrince Mohammed Bin Abdul‐Aziz Hospital‐National Guard HospitalMadinahSaudi Arabia
| | - Mohammed Almohammadi
- King Abdullah International Medical Research CenterKing Saud bin Abdulaziz University for Health Sciences, Pathology and Laboratory Medicine, King Abdulaziz Medical CityJeddahSaudi Arabia
| | | | - Ghaleb Elyamany
- Central Military Laboratory and Blood BankPrince Sultan Military Medical CityRiyadhSaudi Arabia
| |
Collapse
|
20
|
Musat O, Sorop VB, Sorop MI, Lazar V, Marti DT, Susan M, Avram CR, Oprisoni A, Vulcanescu DD, Horhat FG, Bagiu IC, Horhat DI, Diaconu MM. COVID-19 and Laboratory Markers from Romanian Patients-A Narrative Review. Life (Basel) 2023; 13:1837. [PMID: 37763241 PMCID: PMC10532991 DOI: 10.3390/life13091837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/09/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
COVID-19 has significantly impacted the whole world, and Romania was no exception. Biomarkers play a crucial role in understanding and managing the disease. However, research regarding laboratory analyses for patients with COVID-19 is fairly limited. For detection, PCR testing is still considered the golden standard, while antibodies are still useful for monitoring both patients and their vaccination status. In our country, biomarkers such as CRP, LDH, transaminases, cardiac, and iron markers have been used to assess the status of patients and even predict illness outcome. CRP, IL-6, LDH, FER, fibrinogen, creatinine, and vitamin D levels have been associated with increased severity, risk of ICU admission, and death. Cardiac markers and D-dimers are also good predictors, but their role seems more important in patients with complications. HDL cholesterol and BUN levels were also suggested as potential biomarkers. Hematological issues in SARS-CoV-2 infections include neutrophilia, lymphopenia and their ratio, while PCT, which is a marker of bacterial infections, is better to be used in patients with co- or supra-infections. The current research is a narrative review that focuses on the laboratory results of Romanian COVID-19 patients. The goal of this article is to provide an update on the research on biomarkers and other laboratory tests conducted inside the borders of Romania and identify gaps in this regard. Secondly, options for further research are discussed and encouraged.
Collapse
Affiliation(s)
- Ovidiu Musat
- Department of Ophthalmology, “Carol Davila” University of Medicine and Pharmacy, Dionisie Lupu Street, No. 37, Sector 2, 020021 Bucharest, Romania;
- Department of Ophthalmology, “Dr Carol Davila” Central Military Emergency University Hospital, Mircea Vulcanescu Street, No. 88, 010825 Bucharest, Romania
| | - Virgiliu Bogdan Sorop
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (V.B.S.); (M.M.D.)
| | - Madalina Ioana Sorop
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (M.I.S.); (D.D.V.)
| | - Viorica Lazar
- Department of General Medicine, “Vasile Goldis” University of Medicine, Liviu Rebreanu Street, No. 86, 310048 Arad, Romania;
- Pediatric Clinic II, Clinical Hospital Emergency of Arad County, Andrényi Károly Street, No. 2-4, 310037 Arad, Romania
| | - Daniela Teodora Marti
- Department of Biology and Life Sciences, “Vasile Goldis” University of Medicine, Liviu Rebreanu Street, No. 86, 310048 Arad, Romania;
- Clinical Analysis Laboratory Clinical Hospital Emergency of Arad County, Andrényi Károly Street, No. 2-4, 310037 Arad, Romania
| | - Monica Susan
- Department of Internal Medicine, Centre for Preventive Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Cecilia Roberta Avram
- Department of Residential Training and Post-University Courses, “Vasile Goldis” Western University, Liviu Rebreanu Street 86, 310414 Arad, Romania;
| | - Andrada Oprisoni
- Department of Pediatrics, Discipline of Pediatric Oncology and Hematology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Dan Dumitru Vulcanescu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (M.I.S.); (D.D.V.)
- Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Clinical Analysis Laboratory, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Florin George Horhat
- Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Clinical Analysis Laboratory, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Iulia Cristina Bagiu
- Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Clinical Analysis Laboratory, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Delia Ioana Horhat
- Department of ENT, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Mircea Mihai Diaconu
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (V.B.S.); (M.M.D.)
| |
Collapse
|
21
|
Chen P, Wu M, He Y, Jiang B, He ML. Metabolic alterations upon SARS-CoV-2 infection and potential therapeutic targets against coronavirus infection. Signal Transduct Target Ther 2023; 8:237. [PMID: 37286535 DOI: 10.1038/s41392-023-01510-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/18/2023] [Accepted: 05/19/2023] [Indexed: 06/09/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by coronavirus SARS-CoV-2 infection has become a global pandemic due to the high viral transmissibility and pathogenesis, bringing enormous burden to our society. Most patients infected by SARS-CoV-2 are asymptomatic or have mild symptoms. Although only a small proportion of patients progressed to severe COVID-19 with symptoms including acute respiratory distress syndrome (ARDS), disseminated coagulopathy, and cardiovascular disorders, severe COVID-19 is accompanied by high mortality rates with near 7 million deaths. Nowadays, effective therapeutic patterns for severe COVID-19 are still lacking. It has been extensively reported that host metabolism plays essential roles in various physiological processes during virus infection. Many viruses manipulate host metabolism to avoid immunity, facilitate their own replication, or to initiate pathological response. Targeting the interaction between SARS-CoV-2 and host metabolism holds promise for developing therapeutic strategies. In this review, we summarize and discuss recent studies dedicated to uncovering the role of host metabolism during the life cycle of SARS-CoV-2 in aspects of entry, replication, assembly, and pathogenesis with an emphasis on glucose metabolism and lipid metabolism. Microbiota and long COVID-19 are also discussed. Ultimately, we recapitulate metabolism-modulating drugs repurposed for COVID-19 including statins, ASM inhibitors, NSAIDs, Montelukast, omega-3 fatty acids, 2-DG, and metformin.
Collapse
Affiliation(s)
- Peiran Chen
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Mandi Wu
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Yaqing He
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China
| | - Binghua Jiang
- Cell Signaling and Proteomic Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China.
| |
Collapse
|
22
|
Nayak SS, Naidu A, Sudhakaran SL, Vino S, Selvaraj G. Prospects of Novel and Repurposed Immunomodulatory Drugs against Acute Respiratory Distress Syndrome (ARDS) Associated with COVID-19 Disease. J Pers Med 2023; 13:664. [PMID: 37109050 PMCID: PMC10142859 DOI: 10.3390/jpm13040664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is intricately linked with SARS-CoV-2-associated disease severity and mortality, especially in patients with co-morbidities. Lung tissue injury caused as a consequence of ARDS leads to fluid build-up in the alveolar sacs, which in turn affects oxygen supply from the capillaries. ARDS is a result of a hyperinflammatory, non-specific local immune response (cytokine storm), which is aggravated as the virus evades and meddles with protective anti-viral innate immune responses. Treatment and management of ARDS remain a major challenge, first, because the condition develops as the virus keeps replicating and, therefore, immunomodulatory drugs are required to be used with caution. Second, the hyperinflammatory responses observed during ARDS are quite heterogeneous and dependent on the stage of the disease and the clinical history of the patients. In this review, we present different anti-rheumatic drugs, natural compounds, monoclonal antibodies, and RNA therapeutics and discuss their application in the management of ARDS. We also discuss on the suitability of each of these drug classes at different stages of the disease. In the last section, we discuss the potential applications of advanced computational approaches in identifying reliable drug targets and in screening out credible lead compounds against ARDS.
Collapse
Affiliation(s)
- Smruti Sudha Nayak
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Akshayata Naidu
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Sajitha Lulu Sudhakaran
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Sundararajan Vino
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Gurudeeban Selvaraj
- Centre for Research in Molecular Modeling, Department of Chemistry and Biochemistry, Concordia University-Loyola Campus, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
23
|
Askari H, Rabiei F, Lohrasbi F, Ghadir S, Ghasemi-Kasman M. The Latest Cellular and Molecular Mechanisms of COVID-19 on Non-Lung Organs. Brain Sci 2023; 13:brainsci13030415. [PMID: 36979225 PMCID: PMC10046222 DOI: 10.3390/brainsci13030415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Understanding the transmission pathways of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will aid in developing effective therapies directed at the virus’s life cycle or its side effects. While severe respiratory distress is the most common symptom of a coronavirus 2019 (COVID-19) infection, the virus is also known to cause damage to almost every major organ and system in the body. However, it is not obvious whether pathological changes in extra-respiratory organs are caused by direct infection, indirect, or combination of these effects. In this narrative review, we first elaborate on the characteristics of SARS-CoV-2, followed by the mechanisms of this virus on various organs such as brain, eye, and olfactory nerve and different systems such as the endocrine and gastrointestinal systems.
Collapse
Affiliation(s)
- Hamid Askari
- Student Research Committee, Babol University of Medical Sciences, Babol 47176-47745, Iran
| | - Fatemeh Rabiei
- Student Research Committee, Babol University of Medical Sciences, Babol 47176-47745, Iran
| | - Fatemeh Lohrasbi
- Student Research Committee, Babol University of Medical Sciences, Babol 47176-47745, Iran
| | - Sara Ghadir
- Student Research Committee, Babol University of Medical Sciences, Babol 47176-47745, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol 47176-47745, Iran
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol 47176-47745, Iran
- Correspondence: ; Tel./Fax: +98-11-32190557
| |
Collapse
|
24
|
Huo Y, Li C, Li Y, Li X, Xu P, Bao Z, Liu W. Detecting early-warning signals for influenza by dysregulated dynamic network biomarkers. Brief Funct Genomics 2023:7036269. [PMID: 36787234 DOI: 10.1093/bfgp/elad006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/15/2023] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
As a dynamical system, complex disease always has a sudden state transition at the tipping point, which is the result of the long-term accumulation of abnormal regulations. This paper proposes a novel approach to detect the early-warning signals of influenza A (H3N2 and H1N1) outbreaks by dysregulated dynamic network biomarkers (dysregulated DNBs) for individuals. The results of cross-validation show that our approach can detect early-warning signals before the symptom appears successfully. Unlike the traditional DNBs, our dysregulated DNBs are anchored and very few, which is essential for disease early diagnosis in clinical practice. Moreover, the genes of dysregulated DNBs are significantly enriched in the influenza-related pathways. The source code of this paper can be freely downloaded from https://github.com/YanhaoHuo/dysregulated-DNBs.git.
Collapse
Affiliation(s)
- Yanhao Huo
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China
| | - Chuchu Li
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China
| | - Yujie Li
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China
| | - Xianbin Li
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China.,School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Peng Xu
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China.,School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Zhenshen Bao
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China.,School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Wenbin Liu
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
25
|
Zerangian N, Erabi G, Poudineh M, Monajjem K, Diyanati M, Khanlari M, Khalaji A, Allafi D, Faridzadeh A, Amali A, Alizadeh N, Salimi Y, Ghane Ezabadi S, Abdi A, Hasanabadi Z, ShojaeiBaghini M, Deravi N. Venous thromboembolism in viral diseases: A comprehensive literature review. Health Sci Rep 2023; 6:e1085. [PMID: 36778773 PMCID: PMC9900357 DOI: 10.1002/hsr2.1085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/25/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
Venous thromboembolism (VTE) is known to be a common respiratory and/or cardiovascular complication in hospitalized patients with viral infections. Numerous studies have proven human immunodeficiency virus infection to be a prothrombotic condition. An elevated VTE risk has been observed in critically ill H1N1 influenza patients. VTE risk is remarkably higher in patients infected with the Hepatitis C virus in contrast to uninfected subjects. The elevation of D-dimer levels supported the association between Chikungunya and the Zika virus and the rise of clinical VTE risk. Varicella-zoster virus is a risk factor for both cellulitis and the consequent invasive bacterial disease which may take part in thrombotic initiation. Eventually, hospitalized patients infected with the coronavirus disease of 2019 (COVID-19), the cause of the ongoing worldwide pandemic, could mainly suffer from an anomalous risk of coagulation activation with enhanced venous thrombosis events and poor quality clinical course. Although the risk of VTE in nonhospitalized COVID-19 patients is not known yet, there are a large number of guidelines and studies on thromboprophylaxis administration for COVID-19 cases. This study aims to take a detailed look at the effect of viral diseases on VTE, the epidemiology of VTE in viral diseases, and the diagnosis and treatment of VTE.
Collapse
Affiliation(s)
- Nasibeh Zerangian
- Health Education and Health Promotion, Department of Health Education and Health Promotion, School of HealthMashhad University of Medical SciencesMashhadIran
| | - Gisou Erabi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | | | - Kosar Monajjem
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Maryam Diyanati
- Student Research CommitteeRafsanjan University of Medical SciencesRafsanjanIran
| | - Maryam Khanlari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | | | - Diba Allafi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
| | - Arian Amali
- Student Research Committee, Paramedical DepartmentIslamic Azad University, Mashhad BranchMashhadIran
| | - Nilufar Alizadeh
- Doctor of Medicine (MD), School of MedicineIran University of Medical SciencesTehranIran
| | - Yasaman Salimi
- Student Research CommitteeKermanshah University of Medical SciencesKermanshahIran
| | - Sajjad Ghane Ezabadi
- Student's Scientific Research Center, School of MedicineTehran University of Medical SciencesTehranIran
| | - Amir Abdi
- Student Research Committee, School of Medicine, Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Zahra Hasanabadi
- Doctor of Medicine (MD), School of MedicineQazvin University of Medical ScienceQazvinIran
| | - Mahdie ShojaeiBaghini
- Medical Informatics Research Center, Institute for Futures Studies in HealthKerman University of Medical SciencesKermanIran
| | - Niloofar Deravi
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
26
|
Nasrollahi H, Talepoor AG, Saleh Z, Eshkevar Vakili M, Heydarinezhad P, Karami N, Noroozi M, Meri S, Kalantar K. Immune responses in mildly versus critically ill COVID-19 patients. Front Immunol 2023; 14:1077236. [PMID: 36793739 PMCID: PMC9923185 DOI: 10.3389/fimmu.2023.1077236] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
The current coronavirus pandemic (COVID-19), caused by SARS-CoV-2, has had devastating effects on the global health and economic system. The cellular and molecular mediators of both the innate and adaptive immune systems are critical in controlling SARS-CoV-2 infections. However, dysregulated inflammatory responses and imbalanced adaptive immunity may contribute to tissue destruction and pathogenesis of the disease. Important mechanisms in severe forms of COVID-19 include overproduction of inflammatory cytokines, impairment of type I IFN response, overactivation of neutrophils and macrophages, decreased frequencies of DC cells, NK cells and ILCs, complement activation, lymphopenia, Th1 and Treg hypoactivation, Th2 and Th17 hyperactivation, as well as decreased clonal diversity and dysregulated B lymphocyte function. Given the relationship between disease severity and an imbalanced immune system, scientists have been led to manipulate the immune system as a therapeutic approach. For example, anti-cytokine, cell, and IVIG therapies have received attention in the treatment of severe COVID-19. In this review, the role of immunity in the development and progression of COVID-19 is discussed, focusing on molecular and cellular aspects of the immune system in mild vs. severe forms of the disease. Moreover, some immune- based therapeutic approaches to COVID-19 are being investigated. Understanding key processes involved in the disease progression is critical in developing therapeutic agents and optimizing related strategies.
Collapse
Affiliation(s)
- Hamid Nasrollahi
- Radio-Oncology Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Saleh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Eshkevar Vakili
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paria Heydarinezhad
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karami
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Noroozi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seppo Meri
- Department of Bacteriology and Immunology, University of Helsinki and Diagnostic Center of the Helsinki University Hospital, Helsinki, Finland
| | - Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
27
|
Kemper C, Ferreira VP, Paz JT, Holers VM, Lionakis MS, Alexander JJ. Complement: The Road Less Traveled. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:119-125. [PMID: 36596217 PMCID: PMC10038130 DOI: 10.4049/jimmunol.2200540] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/11/2022] [Indexed: 01/04/2023]
Abstract
The complement field has recently experienced a strong resurgence of interest because of the unexpected discovery of new complement functions extending complement's role beyond immunity and pathogen clearance, a growing list of diseases in which complement plays a role, and the proliferation of complement therapeutics. Importantly, although the majority of complement components in the circulation are generated by the liver and activated extracellularly, complement activation unexpectedly also occurs intracellularly across a broad range of cells. Such cell-autonomous complement activation can engage intracellular complement receptors, which then drive noncanonical cell-specific effector functions. Thus, much remains to be discovered about complement biology. In this brief review, we focus on novel noncanonical activities of complement in its "classic areas of operation" (kidney and brain biology, infection, and autoimmunity), with an outlook on the next generation of complement-targeted therapeutics.
Collapse
Affiliation(s)
- Claudia Kemper
- Complement and Inflammation Research Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, OH
| | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco CA
- Department of Neurology, University of California, San Francisco, San Francisco, CA
- Neurosciences Graduate Program, University of California, San Francisco, San Francisco, CA
| | - V Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD; and
| | | |
Collapse
|
28
|
Kumari B, Hajela K, Ali A, Sharma AK, Yadav RK, Ranjan A, Nair R, Bharti S, Dipankar S, Singh PK, Sharma S. Evaluation of C4b as an adjunct marker in symptomatic RT-PCR negative Covid-19 cases. Indian J Clin Biochem 2023; 38:102-109. [PMID: 35756690 PMCID: PMC9207170 DOI: 10.1007/s12291-022-01033-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/04/2022] [Indexed: 01/24/2023]
Abstract
Introduction Detecting low viral load has been a challenge in this pandemic, which has led to its escalated transmission. Complement activation has been implicated in pathogenesis of Covid-19 infection. Thus, evaluation of complement activation in suspected Covid-19 infection may help to detect infection and limit false negative cases thus limiting transmission of infection. We speculate that measuring C4b, produced from an activated complement system due to the presence of Covid-19 may help in its detection, even when the viral titers are low. Methods Plasma C4b levels of symptomatic RT-PCR positive patients (cases, n = 40); symptomatic RT-PCR negative patients (n = 35) and asymptomatic RT-PCR negative controls (n = 40) were evaluated. Plasma C5b-9, IL-6, D-dimer and C1-Inhibitor (C1-INH) were also measured in cases and controls. ELISA kits were used for all measurements. Statistical analyses were carried out using Stata, version 12 (Stata Corp., Texas, USA). Results C4b levels were found to be significantly increased in RT-PCR positive patients as compared to asymptomatic RT-PCR negative controls. RT-PCR negative but symptomatic patients still showed increased C4b levels. The significantly higher levels of C4b in cases with a cut-off value of ≥ 116 ng/ml with optimum sensitivity and specificity of 80% and 52% respectively is indicative of its possible use as an adjunct marker. Increased levels of D-dimer, IL6, along with decreased levels of C1-INH were found in cases compared to controls. Whereas, C5b-9 levels were not significantly raised in cases. Conclusions The results of our study suggests that plasma C4b may help to detect infection in false negative cases of RT-PCR that escape detection owing to low viral load. However, to confirm it a large-scale study is needed. Supplementary Information The online version contains supplementary material available at 10.1007/s12291-022-01033-z.
Collapse
Affiliation(s)
- Bandana Kumari
- Department of Biochemistry, All India Institute of Medical Sciences Patna, Patna, Bihar India
| | - Krishnan Hajela
- Schools of Life Sciences, Devi Ahilya Vishwavidyalaya, Indore, MP India
| | - Asgar Ali
- Department of Biochemistry, All India Institute of Medical Sciences Patna, Patna, Bihar India
| | - Abhay Kumar Sharma
- Department of Biochemistry, All India Institute of Medical Sciences Patna, Patna, Bihar India
| | - Rajesh Kumar Yadav
- Department of Biochemistry, All India Institute of Medical Sciences Patna, Patna, Bihar India
| | - Alok Ranjan
- Department of Community and Family Medicine, All India Institute of Medical Sciences Patna, Patna, Bihar India
| | - Rathish Nair
- College of Nursing, All India Institute of Medical Sciences Patna, Patna, Bihar India
| | - Shreekant Bharti
- Department of Pathology, All India Institute of Medical Sciences Patna, Patna, Bihar India
| | - Satish Dipankar
- Department of Physiology, All India Institute of Medical Sciences Mangalagiri, Mangalagiri, AP India
| | | | - Sadhana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences Patna, Patna, Bihar India
- Dept. of Biochemistry, Faculty In-charge, Medical Education Cell AIIMS, Patna, Bihar India
| |
Collapse
|
29
|
HIF-1α-Dependent Metabolic Reprogramming, Oxidative Stress, and Bioenergetic Dysfunction in SARS-CoV-2-Infected Hamsters. Int J Mol Sci 2022; 24:ijms24010558. [PMID: 36614003 PMCID: PMC9820273 DOI: 10.3390/ijms24010558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
The mechanistic interplay between SARS-CoV-2 infection, inflammation, and oxygen homeostasis is not well defined. Here, we show that the hypoxia-inducible factor (HIF-1α) transcriptional pathway is activated, perhaps due to a lack of oxygen or an accumulation of mitochondrial reactive oxygen species (ROS) in the lungs of adult Syrian hamsters infected with SARS-CoV-2. Prominent nuclear localization of HIF-1α and increased expression of HIF-1α target proteins, including glucose transporter 1 (Glut1), lactate dehydrogenase (LDH), and pyruvate dehydrogenase kinase-1 (PDK1), were observed in areas of lung consolidation filled with infiltrating monocytes/macrophages. Upregulation of these HIF-1α target proteins was accompanied by a rise in glycolysis as measured by extracellular acidification rate (ECAR) in lung homogenates. A concomitant reduction in mitochondrial respiration was also observed as indicated by a partial loss of oxygen consumption rates (OCR) in isolated mitochondrial fractions of SARS-CoV-2-infected hamster lungs. Proteomic analysis further revealed specific deficits in the mitochondrial ATP synthase (Atp5a1) within complex V and in the ATP/ADP translocase (Slc25a4). The activation of HIF-1α in inflammatory macrophages may also drive proinflammatory cytokine production and complement activation and oxidative stress in infected lungs. Together, these findings support a role for HIF-1α as a central mediator of the metabolic reprogramming, inflammation, and bioenergetic dysfunction associated with SARS-CoV-2 infection.
Collapse
|
30
|
Chung J, Vig V, Sun X, Han X, O’Connor GT, Chen X, DeAngelis MM, Farrer LA, Subramanian ML. Genome-Wide Pleiotropy Study Identifies Association of PDGFB with Age-Related Macular Degeneration and COVID-19 Infection Outcomes. J Clin Med 2022; 12:jcm12010109. [PMID: 36614910 PMCID: PMC9821609 DOI: 10.3390/jcm12010109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022] Open
Abstract
Age-related macular degeneration (AMD) has been implicated as a risk factor for severe consequences from COVID-19. We evaluated the genetic architecture shared between AMD and COVID-19 (critical illness, hospitalization, and infections) using analyses of genetic correlations and pleiotropy (i.e., cross-phenotype meta-analysis) of AMD (n = 33,976) and COVID-19 (n ≥ 1,388,342) and subsequent analyses including expression quantitative trait locus (eQTL), differential gene expression, and Mendelian randomization (MR). We observed a significant genetic correlation between AMD and COVID-19 infection (rG = 0.10, p = 0.02) and identified novel genome-wide significant associations near PDGFB (best SNP: rs130651; p = 2.4 × 10-8) in the pleiotropy analysis of the two diseases. The disease-risk allele of rs130651 was significantly associated with increased gene expression levels of PDGFB in multiple tissues (best eQTL p = 1.8 × 10-11 in whole blood) and immune cells (best eQTL p = 7.1 × 10-20 in T-cells). PDGFB expression was observed to be higher in AMD cases than AMD controls {fold change (FC) = 1.02; p = 0.067}, as well as in the peak COVID-19 symptom stage (11-20 days after the symptom onset) compared to early/progressive stage (0-10 days) among COVID-19 patients over age 40 (FC = 2.17; p = 0.03) and age 50 (FC = 2.15; p = 0.04). Our MR analysis found that the liability of AMD risk derived from complement system dysfunction {OR (95% CI); hospitalization = 1.02 (1.01-1.03), infection = 1.02 (1.01-1.03) and increased levels of serum cytokine PDGF-BB {β (95% CI); critical illness = 0.07 (0.02-0.11)} are significantly associated with COVID-19 outcomes. Our study demonstrated that the liability of AMD is associated with an increased risk of COVID-19, and PDGFB may be responsible for the severe COVID-19 outcomes among AMD patients.
Collapse
Affiliation(s)
- Jaeyoon Chung
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Viha Vig
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Xinyu Sun
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Xudong Han
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - George T. O’Connor
- Department of Medicine (Pulmonary & Critical Care), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Xuejing Chen
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Margaret M. DeAngelis
- Department of Population Health Sciences and Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo and VA Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, NY 14203, USA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Departments of Epidemiology and Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- Correspondence: (L.A.F.); (M.L.S.); Tel.: +1-617-358-3550 (L.A.F.); +1-617-414-2020 (M.L.S.)
| | - Manju L. Subramanian
- Department of Medicine (Pulmonary & Critical Care), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Correspondence: (L.A.F.); (M.L.S.); Tel.: +1-617-358-3550 (L.A.F.); +1-617-414-2020 (M.L.S.)
| |
Collapse
|
31
|
Trivedi VS, Magnusen AF, Rani R, Marsili L, Slavotinek AM, Prows DR, Hopkin RJ, McKay MA, Pandey MK. Targeting the Complement-Sphingolipid System in COVID-19 and Gaucher Diseases: Evidence for a New Treatment Strategy. Int J Mol Sci 2022; 23:14340. [PMID: 36430817 PMCID: PMC9695449 DOI: 10.3390/ijms232214340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2)-induced disease (COVID-19) and Gaucher disease (GD) exhibit upregulation of complement 5a (C5a) and its C5aR1 receptor, and excess synthesis of glycosphingolipids that lead to increased infiltration and activation of innate and adaptive immune cells, resulting in massive generation of pro-inflammatory cytokines, chemokines and growth factors. This C5a-C5aR1-glycosphingolipid pathway- induced pro-inflammatory environment causes the tissue damage in COVID-19 and GD. Strikingly, pharmaceutically targeting the C5a-C5aR1 axis or the glycosphingolipid synthesis pathway led to a reduction in glycosphingolipid synthesis and innate and adaptive immune inflammation, and protection from the tissue destruction in both COVID-19 and GD. These results reveal a common involvement of the complement and glycosphingolipid systems driving immune inflammation and tissue damage in COVID-19 and GD, respectively. It is therefore expected that combined targeting of the complement and sphingolipid pathways could ameliorate the tissue destruction, organ failure, and death in patients at high-risk of developing severe cases of COVID-19.
Collapse
Affiliation(s)
- Vyoma Snehal Trivedi
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Albert Frank Magnusen
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Reena Rani
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Luca Marsili
- Department of Neurology, James J. and Joan A. Gardner Center for Parkinson’s Disease and Movement Disorders, University of Cincinnati, 3113 Bellevue Ave, Cincinnati, OH 45219, USA
| | - Anne Michele Slavotinek
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Daniel Ray Prows
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Robert James Hopkin
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Mary Ashley McKay
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| |
Collapse
|
32
|
Qin R, Kurz E, Chen S, Zeck B, Chiribogas L, Jackson D, Herchen A, Attia T, Carlock M, Rapkiewicz A, Bar-Sagi D, Ritchie B, Ross TM, Mahal LK. α2,6-Sialylation Is Upregulated in Severe COVID-19, Implicating the Complement Cascade. ACS Infect Dis 2022; 8:2348-2361. [PMID: 36219583 PMCID: PMC9578644 DOI: 10.1021/acsinfecdis.2c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Indexed: 01/29/2023]
Abstract
Better understanding of the molecular mechanisms underlying COVID-19 severity is desperately needed in current times. Although hyper-inflammation drives severe COVID-19, precise mechanisms triggering this cascade and what role glycosylation might play therein are unknown. Here we report the first high-throughput glycomic analysis of COVID-19 plasma samples and autopsy tissues. We find that α2,6-sialylation is upregulated in the plasma of patients with severe COVID-19 and in autopsied lung tissue. This glycan motif is enriched on members of the complement cascade (e.g., C5, C9), which show higher levels of sialylation in severe COVID-19. In the lung tissue, we observe increased complement deposition, associated with elevated α2,6-sialylation levels, corresponding to elevated markers of poor prognosis (IL-6) and fibrotic response. We also observe upregulation of the α2,6-sialylation enzyme ST6GAL1 in patients who succumbed to COVID-19. Our work identifies a heretofore undescribed relationship between sialylation and complement in severe COVID-19, potentially informing future therapeutic development.
Collapse
Affiliation(s)
- Rui Qin
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Emma Kurz
- Department
of Cell Biology, NYU Grossman School of
Medicine, 550 First Avenue, New York, New York 10016, United
States
| | - Shuhui Chen
- Department
of Chemistry, Biomedical Research Institute, New York University, New York, New York10003, United States
| | - Briana Zeck
- Center
for Biospecimen Research and Development, NYU Langone, New York, New York 10016, United
States
| | - Luis Chiribogas
- Center
for Biospecimen Research and Development, NYU Langone, New York, New York 10016, United
States
| | - Dana Jackson
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Alex Herchen
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Tyson Attia
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Michael Carlock
- Center for
Vaccines and Immunology, University of Georgia, Athens, Georgia 30605, United States
| | - Amy Rapkiewicz
- Department
of Pathology, NYU Long Island School of
Medicine, Mineola, New York 11501, United
States
| | - Dafna Bar-Sagi
- Department
of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Bruce Ritchie
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Ted M. Ross
- Center for
Vaccines and Immunology, University of Georgia, Athens, Georgia 30605, United States
| | - Lara K. Mahal
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
33
|
Dang Q, Li S. Exploring Public Discussions Regarding COVID-19 Vaccinations on Microblogs in China: Findings from Machine Learning Algorithms. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13476. [PMID: 36294061 PMCID: PMC9603472 DOI: 10.3390/ijerph192013476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 06/16/2023]
Abstract
Large-scale, widespread COVID-19 vaccination is the most effective means of cutting off the spread of the novel coronavirus and establishing an immune barrier. Due to the large population base in China, it has been a very difficult task to establish such an immune barrier. Therefore, this study aims to explore the public's discussions related to COVID-19 vaccinations on microblogs and to detect their sentiments toward COVID-19 vaccination so as to improve the vaccination rate in China. This study employed machine learning methods in the field of artificial intelligence to analyze mass data obtained from SinaWeibo. A total of 1,478,875 valid microblog texts were collected between December 2020 and June 2022, the results of which indicated that: (1) overall, negative texts (38.7%) slightly outweighed positive texts (36.1%); "Good" (63%) dominated positive texts, while "disgust" (44.6%) and "fear" (35.8%) dominated negative texts; (2) six overarching themes related to COVID-19 vaccination were identified: public trust in the Chinese government, changes in daily work and study, vaccine economy, international COVID-19 vaccination, the COVID-19 vaccine's R&D, and COVID-19 vaccination for special groups. These themes and sentiments can clarify the public's reactions to COVID-19 vaccination and help Chinese officials' response to vaccine hesitancy. Furthermore, this study seeks to make up for the lack of focus on big data in public health and epidemiology research, and to provide novel insights for future studies.
Collapse
Affiliation(s)
- Qiong Dang
- Climate and Health Communication Center, School of Journalism and Communication, Guangxi University, Nanning 530004, China
| | | |
Collapse
|
34
|
Hamanaka RB, Mutlu GM. Our Compliments to the Authors: Decay Accelerating Factor and the Complement System in Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2022; 67:415-416. [PMID: 35901283 PMCID: PMC9564930 DOI: 10.1165/rcmb.2022-0268ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Robert B Hamanaka
- Section of Pulmonary and Critical Care Medicine The University of Chicago Chicago, Illinois
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care Medicine The University of Chicago Chicago, Illinois
| |
Collapse
|
35
|
Laurent P, Yang C, Rendeiro AF, Nilsson-Payant BE, Carrau L, Chandar V, Bram Y, tenOever BR, Elemento O, Ivashkiv LB, Schwartz RE, Barrat FJ. Sensing of SARS-CoV-2 by pDCs and their subsequent production of IFN-I contribute to macrophage-induced cytokine storm during COVID-19. Sci Immunol 2022; 7:eadd4906. [PMID: 36083891 PMCID: PMC9853436 DOI: 10.1126/sciimmunol.add4906] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Lung-infiltrating macrophages create a marked inflammatory milieu in a subset of patients with COVID-19 by producing a cytokine storm, which correlates with increased lethality. However, these macrophages are largely not infected by SARS-CoV-2, so the mechanism underlying their activation in the lung is unclear. Type I interferons (IFN-I) contribute to protecting the host against SARS-CoV-2 but may also have some deleterious effect, and the source of IFN-I in the lungs of infected patients is not well defined. Plasmacytoid dendritic cells (pDCs), a key cell type involved in antiviral responses, can produce IFN-I in response to SARS-CoV-2. We observed the infiltration of pDCs in the lungs of SARS-CoV-2-infected patients, which correlated with strong IFN-I signaling in lung macrophages. In patients with severe COVID-19, lung macrophages expressed a robust inflammatory signature, which correlated with persistent IFN-I signaling at the single-cell level. Hence, we observed the uncoupling in the kinetics of the infiltration of pDCs in the lungs and the associated IFN-I signature, with the cytokine storm in macrophages. We observed that pDCs were the dominant IFN-α-producing cells in response to the virus in the blood, whereas macrophages produced IFN-α only when in physical contact with infected epithelial cells. We also showed that IFN-α produced by pDCs, after the sensing of SARS-CoV-2 by TLR7, mediated changes in macrophages at both transcriptional and epigenetic levels, which favored their hyperactivation by environmental stimuli. Together, these data indicate that the priming of macrophages can result from the response by pDCs to SARS-CoV-2, leading to macrophage activation in patients with severe COVID-19.
Collapse
Affiliation(s)
- Paôline Laurent
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Chao Yang
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - André F. Rendeiro
- Institute for Computational Biomedicine and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Benjamin E. Nilsson-Payant
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave., New York, NY 10029, USA
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Lucia Carrau
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave., New York, NY 10029, USA
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Vasuretha Chandar
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Benjamin R. tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave., New York, NY 10029, USA
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- WorldQuant Initiative for Quantitative Prediction and Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY 10029, USA
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
| | - Lionel B. Ivashkiv
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
| | - Robert E. Schwartz
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| |
Collapse
|
36
|
Lo MW, Amarilla AA, Lee JD, Albornoz EA, Modhiran N, Clark RJ, Ferro V, Chhabra M, Khromykh AA, Watterson D, Woodruff TM. SARS-CoV-2 triggers complement activation through interactions with heparan sulfate. Clin Transl Immunology 2022; 11:e1413. [PMID: 35999893 PMCID: PMC9387400 DOI: 10.1002/cti2.1413] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 12/30/2022] Open
Abstract
Objectives To determine whether SARS‐CoV‐2 can trigger complement activation, the pathways that are involved and the functional significance of the resultant effect. Methods SARS‐CoV‐2 was inoculated into a human lepirudin‐anticoagulated whole blood model, which contains a full repertoire of complement factors and leukocytes that express complement receptors. Complement activation was determined by measuring C5a production with an ELISA, and pretreatment with specific inhibitors was used to identify the pathways involved. The functional significance of this was then assessed by measuring markers of C5a signalling including leukocyte C5aR1 internalisation and CD11b upregulation with flow cytometry. Results SARS‐CoV‐2 inoculation in this whole blood model caused progressive C5a production over 24 h, which was significantly reduced by inhibitors for factor B, C3, C5 and heparan sulfate. However, this phenomenon could not be replicated in cell‐free plasma, highlighting the requirement for cell surface interactions with heparan sulfate. Functional analysis of this phenomenon revealed that C5aR1 signalling and CD11b upregulation in granulocytes and monocytes was delayed and only occurred after 24 h. Conclusion SARS‐CoV‐2 is a noncanonical alternative pathway activator that progressively triggers complement activation through interactions with heparan sulfate.
Collapse
Affiliation(s)
- Martin W Lo
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia
| | - John D Lee
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Eduardo A Albornoz
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia
| | - Richard J Clark
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| | - Mohit Chhabra
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| |
Collapse
|
37
|
De Leeuw E, Van Damme KFA, Declercq J, Bosteels C, Maes B, Tavernier SJ, Detalle L, Smart T, Glatt S, Debeuf N, Deckers J, Lameire S, Vandecasteele SJ, De Neve N, Demedts IK, Govaerts E, Knoop C, Vanhove K, Moutschen M, Terryn W, Depuydt P, Van Braeckel E, Haerynck F, Hendrickx TCJ, Parrein V, Lalla M, Brittain C, Lambrecht BN. Efficacy and safety of the investigational complement C5 inhibitor zilucoplan in patients hospitalized with COVID-19: an open-label randomized controlled trial. Respir Res 2022; 23:202. [PMID: 35945604 PMCID: PMC9361275 DOI: 10.1186/s12931-022-02126-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/01/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The efficacy and safety of complement inhibition in COVID-19 patients is unclear. METHODS A multicenter randomized controlled, open-label trial. Hospitalized COVID-19 patients with signs of systemic inflammation and hypoxemia (PaO2/FiO2 below 350 mmHg) were randomized (2:1 ratio) to receive standard of care with or without the C5 inhibitor zilucoplan daily for 14 days, under antibiotic prophylaxis. The primary outcome was improvement in oxygenation at day 6 and 15. RESULTS 81 patients were randomly assigned to zilucoplan (n = 55) or the control group (n = 26). 78 patients were included in the safety and primary analysis. Most were men (87%) and the median age was 63 years. The mean improvement in PaO2/FiO2 from baseline to day 6 was 56.4 mmHg in the zilucoplan group and 20.6 mmHg in the control group (mean difference + 35.8; 95% confidence interval (CI) - 9.4 to 80.9; p = 0.12), an effect also observed at day 15. Day 28 mortality was 9% in the zilucoplan and 21% in the control group (odds ratio 0.4; 95% CI 0.1 to 1.5). At long-term follow up, the distance walked in a 6-min test was 539.7 m in zilucoplan and 490.6 m in the control group (p = 0.18). Zilucoplan lowered serum C5b-9 (p < 0.001) and interleukin-8 (p = 0.03) concentration compared with control. No relevant safety differences between the zilucoplan and control group were identified. CONCLUSION Administration of zilucoplan to COVID-19 patients in this proof-of-concept randomized trial was well tolerated under antibiotic prophylaxis. While not reaching statistical significance, indicators of respiratory function (PaO2/FiO2) and clinical outcome (mortality and 6-min walk test) suggest that C5 inhibition might be beneficial, although this requires further research in larger randomized studies.
Collapse
Affiliation(s)
- Elisabeth De Leeuw
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Karel F A Van Damme
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jozefien Declercq
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Cedric Bosteels
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bastiaan Maes
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Simon J Tavernier
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | | | | | | | - Nincy Debeuf
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Julie Deckers
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Sahine Lameire
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | | | - Nikolaas De Neve
- Department of Anesthesiology and Intensive Care Medicine, OLV Hospital, Aalst, Belgium
| | - Ingel K Demedts
- Department of Respiratory Medicine, AZ Delta Roeselare-Menen, Roeselare, Belgium
| | - Elke Govaerts
- Department of Pulmonary Medicine, AZ Sint-Lucas Gent, Ghent, Belgium
| | - Christiane Knoop
- Department of Pulmonary Medicine, CHU Erasme Université Libre de Bruxelles, Brussels, Belgium
| | - Karolien Vanhove
- Department of Pneumology and Respiratory Oncology, AZ Vesalius, Tongeren, Belgium
| | - Michel Moutschen
- Department of Infectious Diseases and General Internal Medicine, CHU Sart-Tilman, Université de Liège, Liège, Belgium
| | - Wim Terryn
- Department of General Internal Medicine and Nephrology, Jan Yperman Hospital, Ieper, Belgium
| | - Pieter Depuydt
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Intensive Care Unit, Ghent University Hospital, Ghent, Belgium
| | - Eva Van Braeckel
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Tine C J Hendrickx
- Clinical Trial Center, Pharmacy Department, AZ Sint-Lucas Gent, Ghent, Belgium
| | | | | | | | - Bart N Lambrecht
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
38
|
Lamerton RE, Marcial-Juarez E, Faustini SE, Perez-Toledo M, Goodall M, Jossi SE, Newby ML, Chapple I, Dietrich T, Veenith T, Shields AM, Harper L, Henderson IR, Rayes J, Wraith DC, Watson SP, Crispin M, Drayson MT, Richter AG, Cunningham AF. SARS-CoV-2 Spike- and Nucleoprotein-Specific Antibodies Induced After Vaccination or Infection Promote Classical Complement Activation. Front Immunol 2022; 13:838780. [PMID: 35860286 PMCID: PMC9289266 DOI: 10.3389/fimmu.2022.838780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 06/07/2022] [Indexed: 12/19/2022] Open
Abstract
Antibodies specific for the spike glycoprotein (S) and nucleocapsid (N) SARS-CoV-2 proteins are typically present during severe COVID-19, and induced to S after vaccination. The binding of viral antigens by antibody can initiate the classical complement pathway. Since complement could play pathological or protective roles at distinct times during SARS-CoV-2 infection we determined levels of antibody-dependent complement activation along the complement cascade. Here, we used an ELISA assay to assess complement protein binding (C1q) and the deposition of C4b, C3b, and C5b to S and N antigens in the presence of antibodies to SARS-CoV-2 from different test groups: non-infected, single and double vaccinees, non-hospitalised convalescent (NHC) COVID-19 patients and convalescent hospitalised (ITU-CONV) COVID-19 patients. C1q binding correlates strongly with antibody responses, especially IgG1 levels. However, detection of downstream complement components, C4b, C3b and C5b shows some variability associated with the subject group from whom the sera were obtained. In the ITU-CONV, detection of C3b-C5b to S was observed consistently, but this was not the case in the NHC group. This is in contrast to responses to N, where median levels of complement deposition did not differ between the NHC and ITU-CONV groups. Moreover, for S but not N, downstream complement components were only detected in sera with higher IgG1 levels. Therefore, the classical pathway is activated by antibodies to multiple SARS-CoV-2 antigens, but the downstream effects of this activation may differ depending the disease status of the subject and on the specific antigen targeted.
Collapse
Affiliation(s)
- Rachel E. Lamerton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Edith Marcial-Juarez
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Sian E. Faustini
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Marisol Perez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Margaret Goodall
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Siân E. Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Maddy L. Newby
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Iain Chapple
- Periodontal Research Group, School of Dentistry, Institute of Clinical Sciences, University of Birmingham, and Birmingham Community Healthcare National Health Service Trust, Birmingham, United Kingdom
| | - Thomas Dietrich
- Periodontal Research Group, School of Dentistry, Institute of Clinical Sciences, University of Birmingham, and Birmingham Community Healthcare National Health Service Trust, Birmingham, United Kingdom
| | - Tonny Veenith
- Department of Critical Care Medicine, University Hospitals Birmingham National Health Service (NHS) Trust, Birmingham, United Kingdom
| | - Adrian M. Shields
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Lorraine Harper
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Julie Rayes
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - David C. Wraith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Mark T. Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Alex G. Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
39
|
Stefanile A, Cellerino M, Koudriavtseva T. Elevated risk of thrombotic manifestations of SARS-CoV-2 infection in cancer patients: A literature review. EXCLI JOURNAL 2022; 21:906-920. [PMID: 36172074 PMCID: PMC9489888 DOI: 10.17179/excli2022-5073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 11/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) results in higher risks of hospitalization or death in older patients and those with multiple comorbidities, including malignancies. Patients with cancer have greater risks of COVID-19 onset and worse prognosis. This excess is mainly explained by thrombotic complications. Indeed, an imbalance in the equilibrium between clot formation and bleeding, increased activation of coagulation, and endothelial dysfunction characterize both COVID-19 patients and those with cancer. With this review, we provide a summary of the pathological mechanisms of coagulation and thrombotic manifestations in these patients and discuss the possible therapeutic implications of these phenomena.
Collapse
Affiliation(s)
- Annunziata Stefanile
- Department Clinical Pathology and Cancer Biobank, IRCCS Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), 00144 Rome, Italy
| | - Maria Cellerino
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, IFO, Via Elio Chianesi 53, 00144, Rome, Italy,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Genoa, Italy,*To whom correspondence should be addressed: Maria Cellerino, Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, IFO, Via Elio Chianesi 53, 00144, Rome, Italy and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Genoa, Italy, E-mail:
| | - Tatiana Koudriavtseva
- Medical Direction, IRCCS Regina Elena National Cancer Institute, Istituti Fisioterapici Ospitalieri (IFO), 00144 Rome, Italy
| |
Collapse
|
40
|
Bruiners N, Guerrini V, Ukey R, Dikdan RJ, Yang JH, Mishra PK, Onyuka A, Handler D, Vieth J, Carayannopoulos M, Guo S, Pollen M, Pinter A, Tyagi S, Feingold D, Philipp C, Libutti SK, Gennaro ML. Longitudinal Analysis of Biologic Correlates of COVID-19 Resolution: Case Report. Front Med (Lausanne) 2022; 9:915367. [PMID: 35783607 PMCID: PMC9240225 DOI: 10.3389/fmed.2022.915367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
While the biomarkers of COVID-19 severity have been thoroughly investigated, the key biological dynamics associated with COVID-19 resolution are still insufficiently understood. We report a case of full resolution of severe COVID-19 due to convalescent plasma transfusion. Following transfusion, the patient showed fever remission, improved respiratory status, and rapidly decreased viral burden in respiratory fluids and SARS-CoV-2 RNAemia. Longitudinal unbiased proteomic analysis of plasma and single-cell transcriptomics of peripheral blood cells conducted prior to and at multiple times after convalescent plasma transfusion identified the key biological processes associated with the transition from severe disease to disease-free state. These included (i) temporally ordered upward and downward changes in plasma proteins reestablishing homeostasis and (ii) post-transfusion disappearance of a subset of monocytes characterized by hyperactivated Interferon responses and decreased TNF-α signaling. Monitoring specific dysfunctional myeloid cell subsets in peripheral blood may provide prognostic keys in COVID-19.
Collapse
Affiliation(s)
- Natalie Bruiners
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Valentina Guerrini
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Rahul Ukey
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
| | - Ryan J. Dikdan
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
| | - Jason H. Yang
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Pankaj Kumar Mishra
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
| | - Alberta Onyuka
- Global Tuberculosis Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Deborah Handler
- Global Tuberculosis Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Joshua Vieth
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, United States
| | - Mary Carayannopoulos
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Shuang Guo
- Division of Hematology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Maressa Pollen
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Abraham Pinter
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Sanjay Tyagi
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Daniel Feingold
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Claire Philipp
- Division of Hematology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Steven K. Libutti
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, United States
- *Correspondence: Steven K. Libutti
| | - Maria Laura Gennaro
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, United States
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
- Maria Laura Gennaro
| |
Collapse
|
41
|
Qin R, Kurz E, Chen S, Zeck B, Chiribogas L, Jackson D, Herchen A, Attia T, Carlock M, Rapkiewicz A, Bar-Sagi D, Ritchie B, Ross TM, Mahal LK. α2,6-Sialylation is Upregulated in Severe COVID-19 Implicating the Complement Cascade. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.06.06.22275981. [PMID: 35702159 PMCID: PMC9196116 DOI: 10.1101/2022.06.06.22275981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Better understanding of the mechanisms of COVID-19 severity is desperately needed in current times. Although hyper-inflammation drives severe COVID-19, precise mechanisms triggering this cascade and what role glycosylation might play therein is unknown. Here we report the first high-throughput glycomic analysis of COVID-19 plasma samples and autopsy tissues. We find α2,6-sialylation is upregulated in plasma of patients with severe COVID-19 and in the lung. This glycan motif is enriched on members of the complement cascade, which show higher levels of sialylation in severe COVID-19. In the lung tissue, we observe increased complement deposition, associated with elevated α2,6-sialylation levels, corresponding to elevated markers of poor prognosis (IL-6) and fibrotic response. We also observe upregulation of the α2,6-sialylation enzyme ST6GAL1 in patients who succumbed to COVID-19. Our work identifies a heretofore undescribed relationship between sialylation and complement in severe COVID-19, potentially informing future therapeutic development.
Collapse
Affiliation(s)
- Rui Qin
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Emma Kurz
- Department of Cell Biology, NYU Grossman School of Medicine, 550 1st Avenue, New York, New York, USA
| | - Shuhui Chen
- Department of Chemistry, Biomedical Research Institute, New York University, New York, New York, USA
| | - Briana Zeck
- Center for Biospecimen Research and Development, NYU Langone, New York, New York, USA
| | - Luis Chiribogas
- Center for Biospecimen Research and Development, NYU Langone, New York, New York, USA
| | - Dana Jackson
- University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Alex Herchen
- University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Tyson Attia
- University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Michael Carlock
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Amy Rapkiewicz
- Department of Pathology, NYU Long Island School of Medicine, Mineola, NY, USA
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York, USA
| | - Bruce Ritchie
- University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Lara K. Mahal
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
42
|
Govender S, Naicker T. The Contribution of Complement Protein C1q in COVID-19 and HIV Infection Comorbid with Preeclampsia: A Review. Int Arch Allergy Immunol 2022; 183:1114-1126. [PMID: 35661665 PMCID: PMC9393774 DOI: 10.1159/000524976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 11/19/2022] Open
Abstract
Dysregulation in component 1q (C1q) levels is associated with weak placental development in preeclampsia (PE). Human immunodeficiency virus infection (HIV-1) triggers the C1q complex, resulting in opsonization of healthy host cells, contributing to their removal, and augmented progression of HIV disease. In coronavirus disease 2019 (COVID-19)-infected patients, the deposition of C1q activates the complement. Considering the paucity of data, this review highlights a significant gap in the potential of C1q in the immunocompromised state of preeclamptic HIV-infected women and COVID-19 infection. In PE, C1q is downregulated; while in antiretroviral treatment-treated HIV/COVID-19 infected patients, C1q is upregulated. It is plausible that C1q is augmented in the triad and may exacerbate severity of disease. This thereby provides a foundation for future intended research which involves the investigation of single nucleotide polymorphism expression of the C1q gene, specifically in these diseases.
Collapse
Affiliation(s)
- Sumeshree Govender
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
43
|
Kircheis R, Planz O. Could a Lower Toll-like Receptor (TLR) and NF-κB Activation Due to a Changed Charge Distribution in the Spike Protein Be the Reason for the Lower Pathogenicity of Omicron? Int J Mol Sci 2022; 23:ijms23115966. [PMID: 35682644 PMCID: PMC9180620 DOI: 10.3390/ijms23115966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023] Open
Abstract
The novel SARS-CoV-2 Omicron variant B.1.1.529, which emerged in late 2021, is currently active worldwide, replacing other variants, including the Delta variant, due to an enormously increased infectivity. Multiple substitutions and deletions in the N-terminal domain (NTD) and the receptor binding domain (RBD) in the spike protein collaborate with the observed increased infectivity and evasion from therapeutic monoclonal antibodies and vaccine-induced neutralizing antibodies after primary/secondary immunization. In contrast, although three mutations near the S1/S2 furin cleavage site were predicted to favor cleavage, observed cleavage efficacy is substantially lower than in the Delta variant and also lower compared to the wild-type virus correlating with significantly lower TMPRSS2-dependent replication in the lungs, and lower cellular syncytium formation. In contrast, the Omicron variant shows high TMPRSS2-independent replication in the upper airway organs, but lower pathogenicity in animal studies and clinics. Based on recent data, we present here a hypothesis proposing that the changed charge distribution in the Omicron’s spike protein could lead to lower activation of Toll-like receptors (TLRs) in innate immune cells, resulting in lower NF-κB activation, furin expression, and viral replication in the lungs, and lower immune hyper-activation.
Collapse
Affiliation(s)
- Ralf Kircheis
- Syntacoll GmbH, 93342 Saal an der Donau, Germany
- Correspondence: ; Tel.: +49-151-167-90606
| | - Oliver Planz
- Interfaculty Institute for Cell Biology, Department of Immunology, Eberhard Karls University Tuebingen, 72076 Tübingen, Germany;
| |
Collapse
|
44
|
Burwick RM, Dellapiana G, Newman RA, Smithson SD, Naqvi M, Williams J, Wong MS, Bautista M, Gaden A, Kazani SD, Dunn DA, Ma MH, Mitter S, Monteleone JPR, Ortiz SR, Ghandehari S, Merin N, Zakowski MI, Karumanchi SA. Complement blockade with eculizumab for treatment of severe Coronavirus Disease 2019 in pregnancy: A case series. Am J Reprod Immunol 2022; 88:e13559. [PMID: 35514201 PMCID: PMC9347938 DOI: 10.1111/aji.13559] [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: 12/04/2021] [Revised: 03/03/2022] [Accepted: 04/25/2022] [Indexed: 12/16/2022] Open
Abstract
Problem We evaluated eculizumab, a complement protein C5 inhibitor, for treatment of severe COVID‐19 in pregnant and postpartum individuals. Method of Study Protocol ECU‐COV‐401 (clinicaltrials.gov NCT04355494) is an open label, multicenter, Expanded Access Program (EAP), evaluating eculizumab for treatment of severe COVID‐19. Participants enrolled at our center from August 2020 to February 2021. Hospitalized patients were eligible if they had severe COVID‐19 with bilateral pulmonary infiltrates and oxygen requirement. Eculizumab was administered on day 1 (1200 mg IV) with additional doses if still hospitalized (1200 mg IV on Days 4 and 8; 900 mg IV on Days 15 and 22; optional doses on Days 12 and 18). The primary outcome was survival at Day 15. Secondary outcomes included survival at Day 29, need for mechanical ventilation, and duration of hospital stay. We evaluated pharmacokinetic and pharmacodynamic data, safety, and adverse outcomes. Results Eight participants were enrolled at the Cedars‐Sinai Medical Center, six during pregnancy (mean 30 ± 4.0 weeks) and two in the postpartum period. Baseline oxygen requirement ranged from 2 L/min nasal cannula to 12 L/min by non‐rebreather mask. The median number of doses of eculizumab was 2 (range 1–3); the median time to hospital discharge was 5.5 days (range 3–12). All participants met the primary outcome of survival at Day 15, and all were alive and free of mechanical ventilation at Day 29. In three participants we demonstrated that free C5 and soluble C5b‐9 levels decreased following treatment. There were no serious adverse maternal or neonatal events attributed to eculizumab at 3 months. Conclusion We describe use of eculizumab to treat severe COVID‐19 in a small series of pregnant and postpartum adults. A larger, controlled study in pregnancy is indicated.
Collapse
Affiliation(s)
- Richard M Burwick
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gabriela Dellapiana
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Rachel A Newman
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sarah D Smithson
- Department of Obstetrics and Gynecology, Loma Linda Medical Center, Los Angeles, California, USA
| | - Mariam Naqvi
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - John Williams
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Melissa S Wong
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Martha Bautista
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Anna Gaden
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shamsah D Kazani
- Clinical Development & Translational Sciences, Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Derek A Dunn
- Clinical Development & Translational Sciences, Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Mark H Ma
- Bioanalytical and Biomarker Development, Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Sanjay Mitter
- Biostatistics - Quantitative Sciences, Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Jonathan P R Monteleone
- Clinical Development & Translational Sciences, Pharmacometrics, Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Stephan R Ortiz
- Clinical Development & Translational Sciences, Clinical Pharmacology, Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Sara Ghandehari
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Noah Merin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mark I Zakowski
- Department of Anesthesiology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - S Ananth Karumanchi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
45
|
Gaikwad H, Li Y, Wang G, Li R, Dai S, Rester C, Kedl R, Saba L, Banda NK, Scheinman RI, Patrick C, Mallela KM, Moein Moghimi S, Simberg D. Antibody-Dependent Complement Responses toward SARS-CoV-2 Receptor-Binding Domain Immobilized on "Pseudovirus-like" Nanoparticles. ACS NANO 2022; 16:acsnano.2c02794. [PMID: 35507641 PMCID: PMC9092195 DOI: 10.1021/acsnano.2c02794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/28/2022] [Indexed: 05/09/2023]
Abstract
Many aspects of innate immune responses to SARS viruses remain unclear. Of particular interest is the role of emerging neutralizing antibodies against the receptor-binding domain (RBD) of SARS-CoV-2 in complement activation and opsonization. To overcome challenges with purified virions, here we introduce "pseudovirus-like" nanoparticles with ∼70 copies of functional recombinant RBD to map complement responses. Nanoparticles fix complement in an RBD-dependent manner in sera of all vaccinated, convalescent, and naı̈ve donors, but vaccinated and convalescent donors with the highest levels of anti-RBD antibodies show significantly higher IgG binding and higher deposition of the third complement protein (C3). The opsonization via anti-RBD antibodies is not an efficient process: on average, each bound antibody promotes binding of less than one C3 molecule. C3 deposition is exclusively through the alternative pathway. C3 molecules bind to protein deposits, but not IgG, on the nanoparticle surface. Lastly, "pseudovirus-like" nanoparticles promote complement-dependent uptake by granulocytes and monocytes in the blood of vaccinated donors with high anti-RBD titers. Using nanoparticles displaying SARS-CoV-2 proteins, we demonstrate subject-dependent differences in complement opsonization and immune recognition.
Collapse
Affiliation(s)
- Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Yue Li
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ronghui Li
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Shaodong Dai
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Cody Rester
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ross Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Laura Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Nirmal K. Banda
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, 1775 Aurora Court, Aurora, CO, 80045, USA
| | - Robert I. Scheinman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Casey Patrick
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Krishna M.G. Mallela
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - S. Moein Moghimi
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- School of Pharmacy, King George VI Building, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Translational and Clinical Research Institute, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
46
|
Vitkov L, Knopf J, Krunić J, Schauer C, Schoen J, Minnich B, Hannig M, Herrmann M. Periodontitis-Derived Dark-NETs in Severe Covid-19. Front Immunol 2022; 13:872695. [PMID: 35493525 PMCID: PMC9039207 DOI: 10.3389/fimmu.2022.872695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/23/2022] [Indexed: 11/15/2022] Open
Abstract
The frequent severe COVID-19 course in patients with periodontitis suggests a link of the aetiopathogenesis of both diseases. The formation of intravascular neutrophil extracellular traps (NETs) is crucial to the pathogenesis of severe COVID-19. Periodontitis is characterised by an increased level of circulating NETs, a propensity for increased NET formation, delayed NET clearance and low-grade endotoxemia (LGE). The latter has an enormous impact on innate immunity and susceptibility to infection with SARS-CoV-2. LPS binds the SARS-CoV-2 spike protein and this complex, which is more active than unbound LPS, precipitates massive NET formation. Thus, circulating NET formation is the common denominator in both COVID-19 and periodontitis and other diseases with low-grade endotoxemia like diabetes, obesity and cardiovascular diseases (CVD) also increase the risk to develop severe COVID-19. Here we discuss the role of propensity for increased NET formation, DNase I deficiency and low-grade endotoxaemia in periodontitis as aggravating factors for the severe course of COVID-19 and possible strategies for the diminution of increased levels of circulating periodontitis-derived NETs in COVID-19 with periodontitis comorbidity.
Collapse
Affiliation(s)
- Ljubomir Vitkov
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University, Homburg, Germany.,Department of Environment & Biodiversity, University of Salzburg, Salzburg, Austria.,Department of Dental Pathology, University of East Sarajevo, East Sarajevo, Bosnia and Herzegovina
| | - Jasmin Knopf
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jelena Krunić
- Department of Dental Pathology, University of East Sarajevo, East Sarajevo, Bosnia and Herzegovina
| | - Christine Schauer
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Janina Schoen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Bernd Minnich
- Department of Environment & Biodiversity, University of Salzburg, Salzburg, Austria
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University, Homburg, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
47
|
Parker SE, Bellingham MC, Woodruff TM. Complement drives circuit modulation in the adult brain. Prog Neurobiol 2022; 214:102282. [DOI: 10.1016/j.pneurobio.2022.102282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/24/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022]
|
48
|
Nilsson B, Persson B, Eriksson O, Fromell K, Hultström M, Frithiof R, Lipcsey M, Huber-Lang M, Ekdahl KN. How the Innate Immune System of the Blood Contributes to Systemic Pathology in COVID-19-Induced ARDS and Provides Potential Targets for Treatment. Front Immunol 2022; 13:840137. [PMID: 35350780 PMCID: PMC8957861 DOI: 10.3389/fimmu.2022.840137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
Most SARS-CoV-2 infected patients experience influenza-like symptoms of low or moderate severity. But, already in 2020 early during the pandemic it became obvious that many patients had a high incidence of thrombotic complications, which prompted treatment with high doses of low-molecular-weight heparin (LMWH; typically 150-300IU/kg) to prevent thrombosis. In some patients, the disease aggravated after approximately 10 days and turned into a full-blown acute respiratory distress syndrome (ARDS)-like pulmonary inflammation with endothelialitis, thrombosis and vascular angiogenesis, which often lead to intensive care treatment with ventilator support. This stage of the disease is characterized by dysregulation of cytokines and chemokines, in particular with high IL-6 levels, and also by reduced oxygen saturation, high risk of thrombosis, and signs of severe pulmonary damage with ground glass opacities. The direct link between SARS-CoV-2 and the COVID-19-associated lung injury is not clear. Indirect evidence speaks in favor of a thromboinflammatory reaction, which may be initiated by the virus itself and by infected damaged and/or apoptotic cells. We and others have demonstrated that life-threatening COVID-19 ARDS is associated with a strong activation of the intravascular innate immune system (IIIS). In support of this notion is that activation of the complement and kallikrein/kinin (KK) systems predict survival, the necessity for usage of mechanical ventilation, acute kidney injury and, in the case of MBL, also coagulation system activation with thromboembolism. The general properties of the IIIS can easily be translated into mechanisms of COVID-19 pathophysiology. The prognostic value of complement and KKsystem biomarkers demonstrate that pharmaceuticals, which are licensed or have passed the phase I trial stage are promising candidate drugs for treatment of COVID-19. Examples of such compounds include complement inhibitors AMY-101 and eculizumab (targeting C3 and C5, respectively) as well as kallikrein inhibitors ecallantide and lanadelumab and the bradykinin receptor (BKR) 2 antagonist icatibant. In this conceptual review we discuss the activation, crosstalk and the therapeutic options that are available for regulation of the IIIS.
Collapse
Affiliation(s)
- Bo Nilsson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Barbro Persson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Eriksson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Karin Fromell
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.,Unit for Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Miklos Lipcsey
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.,Hedenstierna Laboratory, Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Kristina N Ekdahl
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
49
|
Aripova T, Muratkhodjaev J. A novel concept of human antiviral protection: It's all about RNA (Review). Biomed Rep 2022; 16:29. [PMID: 35251616 PMCID: PMC8889527 DOI: 10.3892/br.2022.1512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/10/2022] [Indexed: 11/06/2022] Open
Abstract
The comparative analysis of the antiviral protective mechanisms, including protozoa and RNA interference in multicellular organisms, has revealed their similarity and provided a basic understanding of adaptive immunity. The present article summarizes the latest studies on RNA-guided gene regulation in human antiviral protection, and its importance. Additionally, the role of both neutralizing antibodies and the interferon system in viral invasion is considered. The interferon system is an additional mechanism for suppressing viral infections in humans, which shifts cells into an 'alarm' mode to attempt to prevent further contagion. The primary task of the human central immune system is to maintain integrity and to protect against foreign organisms. In this review, a novel concept is proposed: Antiviral protection in all organisms can be achieved through an intracellular RNA-guided mechanism. A simple and effective defence against viruses is incorporation of a part of a virus's DNA (spacer) into the hosts chromosomes. Following reinfection, RNA transcripts of this spacer are created to direct nuclease enzymes to destroy the viral genome. This is an example of real-time adaptive immunity potentially possessed by every cell with a full complement of chromosomes, and an indicator that antiviral immunity is not only mediated by the presence of neutralizing antibodies and memory B- and T-cells, but also by the presence of specific spacers in the DNA of individuals who have recovered from a viral infection.
Collapse
Affiliation(s)
- Tamara Aripova
- Institute of Immunology and Human Genomics, Academy of Sciences of Uzbekistan, Tashkent 100060, Uzbekistan
| | - Javdat Muratkhodjaev
- Institute of Immunology and Human Genomics, Academy of Sciences of Uzbekistan, Tashkent 100060, Uzbekistan
- GENEX LLC Pharmaceutical Company, Tashkent 100052, Uzbekistan
| |
Collapse
|
50
|
Maccio U, Zinkernagel AS, Schuepbach R, Probst-Mueller E, Frontzek K, Brugger SD, Hofmaenner DA, Moch H, Varga Z. Long-Term Persisting SARS-CoV-2 RNA and Pathological Findings: Lessons Learnt From a Series of 35 COVID-19 Autopsies. Front Med (Lausanne) 2022; 9:778489. [PMID: 35223894 PMCID: PMC8865372 DOI: 10.3389/fmed.2022.778489] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/04/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Long-term sequelae of coronavirus disease 2019 (COVID-19), including the interaction between persisting viral-RNA and specific tissue involvement, pose a challenging issue. In this study, we addressed the chronological correlation (after first clinical diagnosis and postmortem) between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA and organ involvement. METHODS The presence of postmortem SARS-CoV-2 RNA from 35 complete COVID-19 autopsies was correlated with the time interval between the first diagnosis of COVID-19 and death and with its relationship to morphologic findings. RESULTS Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA can be evident up to 40 days after the first diagnosis and can persist to 94 hours after death. Postmortem SARS-CoV-2 RNA was mostly positive in lungs (70%) and trachea (69%), but all investigated organs were positive with variable frequency. Late-stage tissue damage was evident up to 65 days after initial diagnosis in several organs. Positivity for SARS-CoV-2 RNA in pulmonary swabs correlated with diffuse alveolar damage (p = 0.0009). No correlation between positive swabs and other morphologic findings was present. Cerebral (p = 0.0003) and systemic hemorrhages (p = 0.009), cardiac thrombi (p = 0.04), and ischemic events (p = 0.03) were more frequent in the first wave, whereas bacterial pneumonia (p = 0.03) was more prevalent in the second wave. No differences in biometric data, clinical comorbidities, and other autopsy findings were found. CONCLUSIONS Our data provide evidence not only of long-term postmortem persisting SARS-CoV-2 RNA but also of tissue damage several weeks after the first diagnosis of SARS-CoV-2 infection. Additional conditions, such as concomitant bacterial pulmonary superinfection, lung aspergillosis, thromboembolic phenomena, and hemorrhages can further worsen tissue damage.
Collapse
Affiliation(s)
- Umberto Maccio
- Department of Pathology and Molecular Pathology, University Hospital of Zürich, University of Zurich, Zurich, Switzerland
| | - Annelies S Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, University of Zurich, Zurich, Switzerland
| | - Reto Schuepbach
- Institute of Intensive Care, University Hospital Zurich, University Hospital of Zürich, Zurich, Switzerland
| | | | - Karl Frontzek
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Silvio D Brugger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, University of Zurich, Zurich, Switzerland
| | - Daniel Andrea Hofmaenner
- Institute of Intensive Care, University Hospital Zurich, University Hospital of Zürich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital of Zürich, University of Zurich, Zurich, Switzerland
| | - Zsuzsanna Varga
- Department of Pathology and Molecular Pathology, University Hospital of Zürich, University of Zurich, Zurich, Switzerland
| |
Collapse
|