101
|
Shafqat A, Omer MH, Albalkhi I, Alabdul Razzak G, Abdulkader H, Abdul Rab S, Sabbah BN, Alkattan K, Yaqinuddin A. Neutrophil extracellular traps and long COVID. Front Immunol 2023; 14:1254310. [PMID: 37828990 PMCID: PMC10565006 DOI: 10.3389/fimmu.2023.1254310] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/06/2023] [Indexed: 10/14/2023] Open
Abstract
Post-acute COVID-19 sequelae, commonly known as long COVID, encompasses a range of systemic symptoms experienced by a significant number of COVID-19 survivors. The underlying pathophysiology of long COVID has become a topic of intense research discussion. While chronic inflammation in long COVID has received considerable attention, the role of neutrophils, which are the most abundant of all immune cells and primary responders to inflammation, has been unfortunately overlooked, perhaps due to their short lifespan. In this review, we discuss the emerging role of neutrophil extracellular traps (NETs) in the persistent inflammatory response observed in long COVID patients. We present early evidence linking the persistence of NETs to pulmonary fibrosis, cardiovascular abnormalities, and neurological dysfunction in long COVID. Several uncertainties require investigation in future studies. These include the mechanisms by which SARS-CoV-2 brings about sustained neutrophil activation phenotypes after infection resolution; whether the heterogeneity of neutrophils seen in acute SARS-CoV-2 infection persists into the chronic phase; whether the presence of autoantibodies in long COVID can induce NETs and protect them from degradation; whether NETs exert differential, organ-specific effects; specifically which NET components contribute to organ-specific pathologies, such as pulmonary fibrosis; and whether senescent cells can drive NET formation through their pro-inflammatory secretome in long COVID. Answering these questions may pave the way for the development of clinically applicable strategies targeting NETs, providing relief for this emerging health crisis.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | | | | | | | | | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | |
Collapse
|
102
|
Ali RA, Minarchick VC, Zahavi M, Rysenga CE, Sturm KA, Hoy CK, Sarosh C, Knight JS, Demoruelle MK. Ginger intake suppresses neutrophil extracellular trap formation in autoimmune mice and healthy humans. JCI Insight 2023; 8:e172011. [PMID: 37737262 PMCID: PMC10561719 DOI: 10.1172/jci.insight.172011] [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: 05/03/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
We previously reported that treatment of mice with 6-gingerol, the most abundant phytochemical in ginger root, leads to phosphodiesterase inhibition that counteracts neutrophil hyperactivity in models of antiphospholipid syndrome (APS) and lupus. Here, we explored the extent to which oral intake of a whole-ginger extract would similarly impact neutrophils in both autoimmune mice and healthy humans. In vitro, a solubilized ginger extract was able to attenuate neutrophil extracellular trap formation (NETosis) by human neutrophils through a mechanism that was dependent upon the cyclic AMP-dependent kinase, protein kinase A. When mice with features of either APS or lupus were administered a ginger extract orally, they demonstrated reduced circulating NETs, as well as the tempering of other disease outcomes, such as large-vein thrombosis (APS) and autoantibody production (lupus). In a pilot clinical trial, which was validated in a second cohort, daily intake of a ginger supplement for 7 days by healthy volunteers boosted neutrophil cAMP, inhibited NETosis in response to disease-relevant stimuli, and reduced circulating plasma NET levels. In summary, this work demonstrates that ginger intake restrains neutrophil hyperactivity in autoimmune mouse models and that ginger consumption by healthy individuals makes their neutrophils more resistant to NETosis.
Collapse
Affiliation(s)
- Ramadan A. Ali
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Valerie C. Minarchick
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Miela Zahavi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christine E. Rysenga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kristin A. Sturm
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Claire K. Hoy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cyrus Sarosh
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - M. Kristen Demoruelle
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
103
|
Hoeter K, Neuberger E, Fischer S, Herbst M, Juškevičiūtė E, Enders K, Rossmann H, Sprinzl MF, Simon P, Bodenstein M, Schaefer M. Evidence for the utility of cfDNA plasma concentrations to predict disease severity in COVID-19: a retrospective pilot study. PeerJ 2023; 11:e16072. [PMID: 37744227 PMCID: PMC10512938 DOI: 10.7717/peerj.16072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 08/20/2023] [Indexed: 09/26/2023] Open
Abstract
Background COVID-19 is a worldwide pandemic caused by the highly infective SARS-CoV-2. There is a need for biomarkers not only for overall prognosis but also for predicting the response to treatments and thus for improvements in the clinical management of patients with COVID-19. Circulating cell-free DNA (cfDNA) has emerged as a promising biomarker in the assessment of various pathological conditions. The aim of this retrospective and observational pilot study was to investigate the range of cfDNA plasma concentrations in hospitalized COVID-19 patients during the first wave of SARS-CoV-2 infection, to relate them to established inflammatory parameters as a correlative biomarker for disease severity, and to compare them with plasma levels in a healthy control group. Methods Lithium-Heparin plasma samples were obtained from COVID-19 patients (n = 21) during hospitalization in the University Medical Centre of Mainz, Germany between March and June 2020, and the cfDNA concentrations were determined by quantitative PCR yielding amplicons of long interspersed nuclear elements (LINE-1). The cfDNA levels were compared with those of an uninfected control group (n = 19). Results Plasma cfDNA levels in COVID-19 patients ranged from 247.5 to 6,346.25 ng/ml and the mean concentration was 1,831 ± 1,388 ng/ml (± standard deviation), which was significantly different from the levels of the uninfected control group (p < 0.001). Regarding clinical complications, the highest correlation was found between cfDNA levels and the myositis (p = 0.049). In addition, cfDNA levels correlated with the "WHO clinical progression scale". D-Dimer and C-reactive protein (CRP) were the clinical laboratory parameters with the highest correlations with cfDNA levels. Conclusion The results of this observational pilot study show a wide range in cfDNA plasma concentrations in patients with COVID-19 during the first wave of infection and confirm that cfDNA plasma concentrations serve as a predictive biomarker of disease severity in COVID-19.
Collapse
Affiliation(s)
- Katharina Hoeter
- Department of Anaesthesiology, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
| | - Elmo Neuberger
- Department of Sports Medicine, Disease Prevention and Rehabilitation, Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Susanne Fischer
- Department of Anaesthesiology, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
| | - Manuel Herbst
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
| | - Ema Juškevičiūtė
- Department of Sports Medicine, Disease Prevention and Rehabilitation, Johannes-Gutenberg Universität Mainz, Mainz, Germany
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Kira Enders
- Department of Sports Medicine, Disease Prevention and Rehabilitation, Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Heidi Rossmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
| | - Martin F. Sprinzl
- Department of Internal Medicine I, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
| | - Perikles Simon
- Department of Sports Medicine, Disease Prevention and Rehabilitation, Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Marc Bodenstein
- Department of Anaesthesiology, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael Schaefer
- Department of Anaesthesiology, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
104
|
Thakur A, Sharma V, Averbek S, Liang L, Pandya N, Kumar G, Cili A, Zhang K. Immune landscape and redox imbalance during neurological disorders in COVID-19. Cell Death Dis 2023; 14:593. [PMID: 37673862 PMCID: PMC10482955 DOI: 10.1038/s41419-023-06102-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
The outbreak of Coronavirus Disease 2019 (COVID-19) has prompted the scientific community to explore potential treatments or vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes the illness. While SARS-CoV-2 is mostly considered a respiratory pathogen, several neurological complications have been reported, raising questions about how it may enter the Central Nervous System (CNS). Receptors such as ACE2, CD147, TMPRSS2, and NRP1 have been identified in brain cells and may be involved in facilitating SARS-CoV-2 entry into the CNS. Moreover, proteins like P2X7 and Panx-1 may contribute to the pathogenesis of COVID-19. Additionally, the role of the immune system in the gravity of COVID-19 has been investigated with respect to both innate and adaptive immune responses caused by SARS-CoV-2 infection, which can lead to a cytokine storm, tissue damage, and neurological manifestations. A redox imbalance has also been linked to the pathogenesis of COVID-19, potentially causing mitochondrial dysfunction, and generating proinflammatory cytokines. This review summarizes different mechanisms of reactive oxygen species and neuro-inflammation that may contribute to the development of severe COVID-19, and recent progress in the study of immunological events and redox imbalance in neurological complications of COVID-19, and the role of bioinformatics in the study of neurological implications of COVID-19.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation-CAS Limited, Hong Kong SAR, Hong Kong.
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sera Averbek
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
- Technische Universität Darmstadt, Darmstadt, Germany
| | - Lifan Liang
- University of Pittsburgh, Pittsburgh, PA, USA
| | - Nirali Pandya
- Department of Chemistry, Faculty of Sciences, National University of Singapore, Singapore, Singapore
| | - Gaurav Kumar
- School of Biosciences and Biomedical Engineering, Department of Clinical Research, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Alma Cili
- Clinic of Hematology, University of Medicine, University Hospital center "Mother Teresa", Tirane, Albania
| | - Kui Zhang
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass sciences, Southwest University, Chongqing, China.
- Cancer Centre, Medical Research Institute, Southwest University, Chongqing, China.
| |
Collapse
|
105
|
Naranjo-Millán JA, Bedoya-Joaqui V, Cañas CA. Systemic lupus erythematosus flare during SARS-CoV-2 infection: Report of 3 cases presented during the fourth wave of the pandemic in Colombia. Int J Rheum Dis 2023; 26:1807-1810. [PMID: 36880687 DOI: 10.1111/1756-185x.14657] [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: 12/04/2022] [Revised: 01/16/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has become a worldwide pandemic since first discovered in 2019. Systemic lupus erythematosus (SLE) flare has been reported in the post-infectious period. In Colombia, the fourth pandemic wave started at the beginning of 2022 when we observed flare of 3 SLE patients during active infection. CASE PRESENTATION We describe 3 patients with inactive SLE, who presented coronavirus disease 2019 and severe flare in early 2022, 2 patients with nephritis and 1 with severe thrombocytopenia. All patients had increase of antinuclear and anti-DNA antibody titers and complement consumption. CONCLUSIONS Three cases with SLE flare concomitant with active SARS-CoV-2 infection were different from others reported earlier in the pandemic with post-infectious flare.
Collapse
Affiliation(s)
- Julián Alberto Naranjo-Millán
- Departamento de Medicina Interna, Facultad de Ciencias de la Salud, Universidad Icesi, Unidad de Reumatología, Cali, Colombia
| | - Vanessa Bedoya-Joaqui
- Departamento de Medicina Interna, Facultad de Ciencias de la Salud, Universidad Icesi, Unidad de Reumatología, Cali, Colombia
| | - Carlos A Cañas
- Departamento de Medicina Interna, Facultad de Ciencias de la Salud, Universidad Icesi, Unidad de Reumatología, Cali, Colombia
- Fundación Valle del Lili, Unidad de Reumatología, Cali, Colombia
- Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional (CIRAT), Universidad Icesi, Cali, Colombia
| |
Collapse
|
106
|
Aghamohamadi N, Shahba F, Zarezadeh Mehrabadi A, Khorramdelazad H, Karimi M, Falak R, Emameh RZ. Age-dependent immune responses in COVID-19-mediated liver injury: focus on cytokines. Front Endocrinol (Lausanne) 2023; 14:1139692. [PMID: 37654571 PMCID: PMC10465349 DOI: 10.3389/fendo.2023.1139692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/21/2023] [Indexed: 09/02/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is potentially pathogenic and causes severe symptoms; in addition to respiratory syndromes, patients might experience other severe conditions such as digestive complications and liver complications injury. The abnormality in the liver is manifested by hepatobiliary dysfunction and enzymatic elevation, which is associated with morbidity and mortality. The direct cytopathic effect, immune dysfunction, cytokine storm, and adverse effects of therapeutic regimens have a crucial role in the severity of liver injury. According to aging and immune system alterations, cytokine patterns may also change in the elderly. Moreover, hyperproduction of cytokines in the inflammatory response to SARS-CoV-2 can lead to multi-organ dysfunction. The mortality rate in elderly patients, particularly those with other comorbidities, is also higher than in adults. Although the pathogenic effect of SARS-CoV-2 on the liver has been widely studied, the impact of age and immune-mediated responses at different ages remain unclear. This review discusses the association between immune system responses in coronavirus disease 2019 (COVID-19) patients of different ages and liver injury, focusing on cytokine alterations.
Collapse
Affiliation(s)
- Nazanin Aghamohamadi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Shahba
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zarezadeh Mehrabadi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Milad Karimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Zolfaghari Emameh
- Department of Energy and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
107
|
Li J, Zhang K, Zhang Y, Gu Z, Huang C. Neutrophils in COVID-19: recent insights and advances. Virol J 2023; 20:169. [PMID: 37533131 PMCID: PMC10398943 DOI: 10.1186/s12985-023-02116-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/06/2023] [Indexed: 08/04/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an acute respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which can lead to acute respiratory distress syndrome (ARDS), multi-organ failure and death, posing significant threat to human health. Studies have found that pathological mechanisms, such as cytokine storms caused by uncontrolled innate immune system activation, release of damage-associated molecular patterns during tissue injury and a high incidence of thrombotic events, are associated with the function and dysfunction of neutrophils. Specifically, the increased formation of low-density neutrophils (LDNs) and neutrophil extracellular traps (NETs) has been shown to be closely linked with the severity and poor prognosis in patients with COVID-19. Our work focuses on understanding the increased number, abnormal activation, lung tissue infiltration, and elevated neutrophil-to-lymphocyte ratio in the pathogenesis of COVID-19. We also explore the involvement of NETs and LDNs in disease progression and thrombosis formation, along with potential therapeutic strategies targeting neutrophil and NETs formation.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Kegong Zhang
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Ye Zhang
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Ziyang Gu
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Changxing Huang
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
108
|
Solomon SD, Lowenstein CJ, Bhatt AS, Peikert A, Vardeny O, Kosiborod MN, Berger JS, Reynolds HR, Mavromichalis S, Barytol A, Althouse AD, Luther JF, Leifer ES, Kindzelski AL, Cushman M, Gong MN, Kornblith LZ, Khatri P, Kim KS, Baumann Kreuziger L, Wahid L, Kirwan BA, Geraci MW, Neal MD, Hochman JS. Effect of the P-Selectin Inhibitor Crizanlizumab on Survival Free of Organ Support in Patients Hospitalized for COVID-19: A Randomized Controlled Trial. Circulation 2023; 148:381-390. [PMID: 37356038 PMCID: PMC10373640 DOI: 10.1161/circulationaha.123.065190] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/12/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND COVID-19 has been associated with endothelial injury, resultant microvascular inflammation and thrombosis. Activated endothelial cells release and express P-selectin and von Willebrand factor, both of which are elevated in severe COVID-19 and may be implicated in the disease pathophysiology. We hypothesized that crizanlizumab, a humanized monoclonal antibody to P-selectin, would reduce morbidity and death in patients hospitalized for COVID-19. METHODS An international, adaptive, randomized controlled platform trial, funded by the National Heart, Lung, and Blood Institute, randomly assigned 422 patients hospitalized with COVID-19 with moderate or severe illness to receive either a single infusion of the P-selectin inhibitor crizanlizumab (at a dose of 5 mg/kg) plus standard of care or standard of care alone in an open-label 1:1 ratio. The primary outcome was organ support-free days, evaluated on an ordinal scale consisting of the number of days alive free of organ support through the first 21 days after trial entry. RESULTS The study was stopped for futility by the data safety monitoring committee. Among 421 randomized patients with known 21-day outcomes, 163 patients (77%) randomized to the crizanlizumab plus standard-of-care arm did not require any respiratory or cardiovascular organ support compared with 169 (80%) in the standard-of-care-alone arm. The adjusted odds ratio for the effect of crizanlizumab on organ support-free days was 0.70 (95% CI, 0.43-1.16), where an odds ratio >1 indicates treatment benefit, yielding a posterior probability of futility (odds ratio <1.2) of 98% and a posterior probability of inferiority (odds ratio <1.0) of 91%. Overall, there were 37 deaths (17.5%) in the crizanlizumab arm and 27 deaths (12.8%) in the standard-of-care arm (hazard ratio, 1.33 [95% CrI, 0.85-2.21]; [probability of hazard ratio>1] = 0.879). CONCLUSIONS Crizanlizumab, a P-selectin inhibitor, did not result in improvement in organ support-free days in patients hospitalized with COVID-19. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT04505774.
Collapse
Affiliation(s)
- Scott D. Solomon
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (S.D.S., A.S.B., A.P., A.B.)
| | | | - Ankeet S. Bhatt
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (S.D.S., A.S.B., A.P., A.B.)
- Kaiser Permanente San Francisco Medical Center, CA (A.S.B.)
| | - Alexander Peikert
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (S.D.S., A.S.B., A.P., A.B.)
| | - Orly Vardeny
- University of Minnesota and the Minneapolis VA Medical Center (O.V.)
| | - Mikhail N. Kosiborod
- Saint Luke’s Mid America Heart Institute, University of Missouri–Kansas City (M.N.K.)
| | - Jeffrey S. Berger
- Cardiovascular Clinical Research Center, NYU Grossman School of Medicine, New York (J.S.B., H.R.R., S.M., J.S.H.)
| | - Harmony R. Reynolds
- Cardiovascular Clinical Research Center, NYU Grossman School of Medicine, New York (J.S.B., H.R.R., S.M., J.S.H.)
| | - Stephanie Mavromichalis
- Cardiovascular Clinical Research Center, NYU Grossman School of Medicine, New York (J.S.B., H.R.R., S.M., J.S.H.)
| | - Anya Barytol
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (S.D.S., A.S.B., A.P., A.B.)
| | | | - James F. Luther
- University of Pittsburgh, PA (A.D.A., J.F.L., M.W.G., M.D.N.)
| | - Eric S. Leifer
- National Heart, Lung, and Blood Institute, Bethesda, MD (E.S.L., A.L.K.)
| | | | - Mary Cushman
- Larner College of Medicine, University of Vermont, Burlington (M.C.)
| | | | | | - Pooja Khatri
- University of Cincinnati Medical Center, OH (P.K.)
| | | | | | | | | | - Mark W. Geraci
- University of Pittsburgh, PA (A.D.A., J.F.L., M.W.G., M.D.N.)
| | - Matthew D. Neal
- University of Pittsburgh, PA (A.D.A., J.F.L., M.W.G., M.D.N.)
| | - Judith S. Hochman
- Cardiovascular Clinical Research Center, NYU Grossman School of Medicine, New York (J.S.B., H.R.R., S.M., J.S.H.)
| |
Collapse
|
109
|
Kangro K, Campbell RA, Tilvawala R, Flick MJ, Wolberg AS. Citrullination of α2-antiplasmin is unlikely to contribute to enhanced plasmin generation in COVID-19 pathophysiology. Res Pract Thromb Haemost 2023; 7:102195. [PMID: 37736033 PMCID: PMC10510058 DOI: 10.1016/j.rpth.2023.102195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/16/2023] [Accepted: 08/07/2023] [Indexed: 09/23/2023] Open
Affiliation(s)
- Kadri Kangro
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Robert A. Campbell
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Ronak Tilvawala
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Matthew J. Flick
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
110
|
Woodruff MC, Bonham KS, Anam FA, Walker TA, Faliti CE, Ishii Y, Kaminski CY, Ruunstrom MC, Cooper KR, Truong AD, Dixit AN, Han JE, Ramonell RP, Haddad NS, Rudolph ME, Yalavarthi S, Betin V, Natoli T, Navaz S, Jenks SA, Zuo Y, Knight JS, Khosroshahi A, Lee FEH, Sanz I. Chronic inflammation, neutrophil activity, and autoreactivity splits long COVID. Nat Commun 2023; 14:4201. [PMID: 37452024 PMCID: PMC10349085 DOI: 10.1038/s41467-023-40012-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023] Open
Abstract
While immunologic correlates of COVID-19 have been widely reported, their associations with post-acute sequelae of COVID-19 (PASC) remain less clear. Due to the wide array of PASC presentations, understanding if specific disease features associate with discrete immune processes and therapeutic opportunities is important. Here we profile patients in the recovery phase of COVID-19 via proteomics screening and machine learning to find signatures of ongoing antiviral B cell development, immune-mediated fibrosis, and markers of cell death in PASC patients but not in controls with uncomplicated recovery. Plasma and immune cell profiling further allow the stratification of PASC into inflammatory and non-inflammatory types. Inflammatory PASC, identifiable through a refined set of 12 blood markers, displays evidence of ongoing neutrophil activity, B cell memory alterations, and building autoreactivity more than a year post COVID-19. Our work thus helps refine PASC categorization to aid in both therapeutic targeting and epidemiological investigation of PASC.
Collapse
Affiliation(s)
- Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Kevin S Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Fabliha A Anam
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Tiffany A Walker
- Department of Medicine, Division of General Internal Medicine, Emory University, Atlanta, GA, USA
| | - Caterina E Faliti
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Yusho Ishii
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | | | - Martin C Ruunstrom
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Kelly Rose Cooper
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Alexander D Truong
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Adviteeya N Dixit
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Jenny E Han
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Richard P Ramonell
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | - Sherwin Navaz
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Scott A Jenks
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Yu Zuo
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Jason S Knight
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Arezou Khosroshahi
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - F Eun-Hyung Lee
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA.
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| |
Collapse
|
111
|
Advani S, Hosseini SMM, Bozorgmehr R, Khameneh-Bagheri A, Mohammadzadeh S, Hasanzadeh T, Jalilian L, Vahidi M, Nofeli AH, Hooshyari Z. Abnormalities of brain imaging in COVID-19 patients with neurological symptoms. CURRENT JOURNAL OF NEUROLOGY 2023; 22:162-169. [PMID: 38011453 PMCID: PMC10626144 DOI: 10.18502/cjn.v22i3.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/09/2023] [Indexed: 11/29/2023]
Abstract
Background: Coronavirus disease 2019 (COVID-19) is a multisystem disease, manifested by several symptoms of various degrees. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) can affect the central nervous system (CNS) through several mechanisms and brain imaging plays an essential role in the diagnosis and evaluation of the neurological involvement of COVID-19. Moreover, brain imaging of patients with COVID-19 would result in a better understanding of SARS-CoV-2 neuro-pathophysiology. In this study, we evaluated the brain imaging findings of patients with COVID-19 in Shohada-e Tajrish Hospital, Tehran, Iran. Methods: This was a single-center, retrospective, and observational study. The hospital records and chest and brain computed tomography (CT) scans of patients with confirmed COVID-19 were reviewed. Results: 161 patients were included in this study (39.1% women, mean age: 60.84). Thirteen patients (8%) had ischemic strokes identified by brain CT. Subdural hematoma, subdural effusion, and subarachnoid hemorrhage were confirmed in three patients. Furthermore, there were four cases of intracranial hemorrhage (ICH) and intraventricular hemorrhage (IVH). Patients with and without abnormal brain CTs had similar average ages. The rate of brain CT abnormalities in both genders did not differ significantly. Moreover, abnormal brain CT was not associated with increased death rate. There was no significant difference in lung involvement (according to lung CT scan) between the two groups. Conclusion: Our experience revealed a wide range of imaging findings in patients with COVID-19 and these findings were not associated with a more severe lung involvement or increased rate of mortality.
Collapse
Affiliation(s)
- Soroor Advani
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyyed Mohammad Mahdi Hosseini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rama Bozorgmehr
- Clinical Research Development Unit, Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khameneh-Bagheri
- Department of Radiology, Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sevda Mohammadzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Taha Hasanzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Laya Jalilian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Vahidi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Hasan Nofeli
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Hooshyari
- Psychiatry and Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
112
|
Fagerhol MK, Schultz NH, Mirlashari MR, Wiedmann MKH, Nissen-Meyer LSH, Søraas AVL, Hetland G. DNase analysed by a novel competitive assay in patients with complications after ChAdOx1 nCoV-19 vaccination and in normal unvaccinated blood donors. Scand J Immunol 2023; 98:e13274. [PMID: 37676118 DOI: 10.1111/sji.13274] [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: 10/25/2022] [Revised: 03/07/2023] [Accepted: 04/18/2023] [Indexed: 09/08/2023]
Abstract
Increased levels of neutrophil extracellular traps (NETs) have been detected in individuals with vaccine complications after the ChAdOx1 nCov vaccine with a correlation between the severity of vaccine side effects and the level of NETosis. DNases may disrupt NETs by degrading their content of DNA, and a balance has been reported between NETs and DNases. Because of this and since the inflammatory marker NETs may be used as a confirmatory test in diagnosing VITT, it is of interest to monitor levels of DNase in patients with increased NETs levels. The current novel rapid DNase ELISA was tested in blood samples of patients with known increased levels of NETs with or without VITT after ChAdOx1 nCoV-19 vaccination. DNase levels in VITT patients were significantly increased compared with normal unvaccinated blood donors and compared with patients with post-vaccination symptoms but not VITT. However, since EDTA was found to inhibit DNase, serum and not EDTA-plasma samples should be applied for DNase testing. The novel DNase assay may serve as a supplementary test to the NETs test when analysing samples from patients with suspected increased NETs levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Geir Hetland
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
113
|
Hocini H, Wiedemann A, Blengio F, Lefebvre C, Cervantes-Gonzalez M, Foucat E, Tisserand P, Surenaud M, Coléon S, Prague M, Guillaumat L, Krief C, Fenwick C, Laouénan C, Bouadma L, Ghosn J, Pantaleo G, Thiébaut R, Lévy Y. Neutrophil Activation and Immune Thrombosis Profiles Persist in Convalescent COVID-19. J Clin Immunol 2023; 43:882-893. [PMID: 36943669 PMCID: PMC10029801 DOI: 10.1007/s10875-023-01459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/24/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE Following a severe COVID-19 infection, a proportion of individuals develop prolonged symptoms. We investigated the immunological dysfunction that underlies the persistence of symptoms months after the resolution of acute COVID-19. METHODS We analyzed cytokines, cell phenotypes, SARS-CoV-2 spike-specific and neutralizing antibodies, and whole blood gene expression profiles in convalescent severe COVID-19 patients 1, 3, and 6 months following hospital discharge. RESULTS We observed persistent abnormalities until month 6 marked by (i) high serum levels of monocyte/macrophage and endothelial activation markers, chemotaxis, and hematopoietic cytokines; (ii) a high frequency of central memory CD4+ and effector CD8+ T cells; (iii) a decrease in anti-SARS-CoV-2 spike and neutralizing antibodies; and (iv) an upregulation of genes related to platelet, neutrophil activation, erythrocytes, myeloid cell differentiation, and RUNX1 signaling. We identified a "core gene signature" associated with a history of thrombotic events, with upregulation of a set of genes involved in neutrophil activation, platelet, hematopoiesis, and blood coagulation. CONCLUSION The lack of restoration of gene expression to a normal profile after up to 6 months of follow-up, even in asymptomatic patients who experienced severe COVID-19, signals the need to carefully extend their clinical follow-up and propose preventive measures.
Collapse
Affiliation(s)
- Hakim Hocini
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Aurélie Wiedemann
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Fabiola Blengio
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Cécile Lefebvre
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Minerva Cervantes-Gonzalez
- Département Épidémiologie Biostatistiques Et Recherche Clinique, AP-HP, Hôpital Bichat, INSERM, Centre d'Investigation Clinique-Epidémiologie Clinique 1425, 75018, Paris, France
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
- APHP- Hôpital Bichat - Médecine Intensive et Réanimation des Maladies Infectieuses, Paris, France
| | - Emile Foucat
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Pascaline Tisserand
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Mathieu Surenaud
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Séverin Coléon
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Mélanie Prague
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
- Department of Public Health, Univ. Bordeaux, Inserm Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France
| | - Lydia Guillaumat
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Corinne Krief
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Cédric Laouénan
- Département Épidémiologie Biostatistiques Et Recherche Clinique, AP-HP, Hôpital Bichat, INSERM, Centre d'Investigation Clinique-Epidémiologie Clinique 1425, 75018, Paris, France
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
| | - Lila Bouadma
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
- APHP- Hôpital Bichat - Médecine Intensive et Réanimation des Maladies Infectieuses, Paris, France
| | - Jade Ghosn
- UMR 1137, Université de Paris, INSERM, IAME, 75018, Paris, France
- AP-HP, Hôpital Bichat, Service de Maladies Infectieuses Et Tropicales, 75018, Paris, France
| | - Giuseppe Pantaleo
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Rodolphe Thiébaut
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
- Department of Public Health, Univ. Bordeaux, Inserm Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France
- CHU de Bordeaux, Pôle de Santé Publique, Service d'Information Médicale, Bordeaux, France
| | - Yves Lévy
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France.
- Assistance Publique-Hôpitaux de Paris, Service Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Créteil, France.
| |
Collapse
|
114
|
Fuzo CA, Fraga-Silva TFC, Maruyama SR, Bastos VAF, Rogerio LA, Takamiya NT, da Silva-Neto PV, Pimentel VE, Toro DM, Pérez MM, de Carvalho JCS, Carmona-Garcia I, Oliveira CNS, Degiovani AM, Ostini FM, Constant LF, de Amorim AP, Vilar FC, Feitosa MR, Parra RS, da Rocha JJR, Feres O, Gaspar GG, Viana AL, Fernandes APM, Santos IKFM, Russo EMS, Cardoso CRB, Sorgi CA, Faccioli LH, Bonato VLD, Dias-Baruffi M. The turning point of COVID-19 severity is associated with a unique circulating neutrophil gene signature. Immunology 2023; 169:323-343. [PMID: 36740582 DOI: 10.1111/imm.13631] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/01/2023] [Indexed: 02/07/2023] Open
Abstract
COVID-19 has a broad spectrum of clinical manifestations associated with the host immune response heterogeneity. Despite the advances in COVID-19 research, it is still crucial to seek a panel of molecular markers that enable accurate stratification of COVID-19 patients. Here, we performed a study that combined analysis of blood transcriptome, demographic data, clinical aspects and laboratory findings from 66 participants classified into different degrees of COVID-19 severity and healthy subjects. We identified a perturbation in blood-leukocyte transcriptional profile associated with COVID-19 aggravation, which was mainly related to processes that disfavoured lymphocyte activation and favoured neutrophil activation. This transcriptional profile stratified patients according to COVID-19 severity. Hence, it enabled identification of a turning point in transcriptional dynamics that distinguished disease outcomes and non-hospitalized from hospitalized moderate patients. Central genes of this unique neutrophil signature were S100A9, ANXA3, CEACAM6, VNN1, OLFM4, IL1R2, TCN1 and CD177. Our study indicates the molecular changes that are linked with the differing clinical aspects presented by humans when suffering from COVID-19, which involve neutrophil activation.
Collapse
Affiliation(s)
- Carlos A Fuzo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thais F C Fraga-Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sandra R Maruyama
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Víctor A F Bastos
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luana A Rogerio
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Nayore T Takamiya
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Pedro V da Silva-Neto
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Vinícius E Pimentel
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Diana M Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Malena M Pérez
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jonatan C S de Carvalho
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ingryd Carmona-Garcia
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Camilla N S Oliveira
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Augusto M Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Fátima M Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Leticia F Constant
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Alessandro P de Amorim
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Fernando C Vilar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marley R Feitosa
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rogerio S Parra
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José J R da Rocha
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Omar Feres
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gilberto G Gaspar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Angelina L Viana
- Departamento de Enfermagem Materno-Infantil e Saúde Pública, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana P M Fernandes
- Departamento de Enfermagem Geral e Especializada, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Isabel K F M Santos
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elisa M S Russo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Cristina R B Cardoso
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos A Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Vânia L D Bonato
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
115
|
Bai X, Schountz T, Buckle AM, Talbert JL, Sandhaus RA, Chan ED. Alpha-1-antitrypsin antagonizes COVID-19: a review of the epidemiology, molecular mechanisms, and clinical evidence. Biochem Soc Trans 2023; 51:1361-1375. [PMID: 37294003 PMCID: PMC10317171 DOI: 10.1042/bst20230078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023]
Abstract
Alpha-1-antitrypsin (AAT), a serine protease inhibitor (serpin), is increasingly recognized to inhibit SARS-CoV-2 infection and counter many of the pathogenic mechanisms of COVID-19. Herein, we reviewed the epidemiologic evidence, the molecular mechanisms, and the clinical evidence that support this paradigm. As background to our discussion, we first examined the basic mechanism of SARS-CoV-2 infection and contend that despite the availability of vaccines and anti-viral agents, COVID-19 remains problematic due to viral evolution. We next underscored that measures to prevent severe COVID-19 currently exists but teeters on a balance and that current treatment for severe COVID-19 remains grossly suboptimal. We then reviewed the epidemiologic and clinical evidence that AAT deficiency increases risk of COVID-19 infection and of more severe disease, and the experimental evidence that AAT inhibits cell surface transmembrane protease 2 (TMPRSS2) - a host serine protease required for SARS-CoV-2 entry into cells - and that this inhibition may be augmented by heparin. We also elaborated on the panoply of other activities of AAT (and heparin) that could mitigate severity of COVID-19. Finally, we evaluated the available clinical evidence for AAT treatment of COVID-19.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, U.S.A
- Department of Academic Affairs, National Jewish Health, Denver, CO, U.S.A
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, U.S.A
| | - Tony Schountz
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, U.S.A
| | - Ashley M. Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- PTNG Bio, Melbourne, Australia
| | - Janet L. Talbert
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, U.S.A
| | | | - Edward D. Chan
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, U.S.A
- Department of Academic Affairs, National Jewish Health, Denver, CO, U.S.A
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, U.S.A
| |
Collapse
|
116
|
Bailey M, Linden D, Guo-Parke H, Earley O, Peto T, McAuley DF, Taggart C, Kidney J. Vascular risk factors for COVID-19 ARDS: endothelium, contact-kinin system. Front Med (Lausanne) 2023; 10:1208866. [PMID: 37448794 PMCID: PMC10336249 DOI: 10.3389/fmed.2023.1208866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
SARS-CoV-2 binds to ACE2 receptors, expressed within the lungs. Risk factors for hospitalization include hypertension, diabetes, ischaemic heart disease and obesity-conditions linked by the presence of endothelial pathology. Viral infection in this setting causes increased conversion of circulating Factor XII to its active form (FXIIa). This is the first step in the contact-kinin pathway, leading to synchronous activation of the intrinsic coagulation cascade and the plasma Kallikrein-Kinin system, resulting in clotting and inflammatory lung disease. Temporal trends are evident from blood results of hospitalized patients. In the first week of symptoms the activated partial thromboplastin time (APTT) is prolonged. This can occur when clotting factors are consumed as part of the contact (intrinsic) pathway. Platelet counts initially fall, reflecting their consumption in coagulation. Lymphopenia occurs after approximately 1 week, reflecting the emergence of a lymphocytic pneumonitis [COVID-19 acute respiratory distress syndrome (ARDS)]. Intrinsic coagulation also induces the contact-kinin pathway of inflammation. A major product of this pathway, bradykinin causes oedema with ground glass opacities (GGO) on imaging in early COVID-19. Bradykinin also causes release of the pleiotrophic cytokine IL-6, which causes lymphocyte recruitment. Thromobosis and lymphocytic pneumonitis are hallmark features of COVID-19 ARDS. In this review we examine the literature with particular reference to the contact-kinin pathway. Measurements of platelets, lymphocytes and APTT should be undertaken in severe infections to stratify for risk of developing ARDS.
Collapse
Affiliation(s)
- Melanie Bailey
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Dermot Linden
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Hong Guo-Parke
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Olivia Earley
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Tunde Peto
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Danny F. McAuley
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Clifford Taggart
- Wellcome - Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Joseph Kidney
- Mater Infirmorum Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
| |
Collapse
|
117
|
Prebensen C, Lefol Y, Myhre PL, Lüders T, Jonassen C, Blomfeldt A, Omland T, Nilsen H, Berdal JE. Longitudinal whole blood transcriptomic analysis characterizes neutrophil activation and interferon signaling in moderate and severe COVID-19. Sci Rep 2023; 13:10368. [PMID: 37365222 PMCID: PMC10293211 DOI: 10.1038/s41598-023-37606-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/24/2023] [Indexed: 06/28/2023] Open
Abstract
A maladaptive inflammatory response has been implicated in the pathogenesis of severe COVID-19. This study aimed to characterize the temporal dynamics of this response and investigate whether severe disease is associated with distinct gene expression patterns. We performed microarray analysis of serial whole blood RNA samples from 17 patients with severe COVID-19, 15 patients with moderate disease and 11 healthy controls. All study subjects were unvaccinated. We assessed whole blood gene expression patterns by differential gene expression analysis, gene set enrichment, two clustering methods and estimated relative leukocyte abundance using CIBERSORT. Neutrophils, platelets, cytokine signaling, and the coagulation system were activated in COVID-19, and this broad immune activation was more pronounced in severe vs. moderate disease. We observed two different trajectories of neutrophil-associated genes, indicating the emergence of a more immature neutrophil phenotype over time. Interferon-associated genes were strongly enriched in early COVID-19 before falling markedly, with modest severity-associated differences in trajectory. In conclusion, COVID-19 necessitating hospitalization is associated with a broad inflammatory response, which is more pronounced in severe disease. Our data suggest a progressively more immature circulating neutrophil phenotype over time. Interferon signaling is enriched in COVID-19 but does not seem to drive severe disease.
Collapse
Affiliation(s)
- Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Kirkeveien 166, 0450, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Yohan Lefol
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Peder L Myhre
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Torben Lüders
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | | | - Anita Blomfeldt
- Department of Microbiology and Infection Control, Akershus University Hospital, Lørenskog, Norway
| | - Torbjørn Omland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Jan-Erik Berdal
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
118
|
Zeng H, Zhuang Y, Li X, Yin Z, Huang X, Peng H. Exploring the potential common denominator pathogenesis of system lupus erythematosus with COVID-19 based on comprehensive bioinformatics analysis. Front Immunol 2023; 14:1179664. [PMID: 37426642 PMCID: PMC10325730 DOI: 10.3389/fimmu.2023.1179664] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Objective Evidences show that there may be a link between SLE and COVID-19. The purpose of this study is to screen out the diagnostic biomarkers of systemic lupus erythematosus (SLE) with COVID-19 and explore the possible related mechanisms by the bioinformatics approach. Methods SLE and COVID-19 datasets were extracted separately from the NCBI Gene Expression Omnibus (GEO) database. The limma package in R was used to obtain the differential genes (DEGs). The protein interaction network information (PPI) and core functional modules were constructed in the STRING database using Cytoscape software. The hub genes were identified by the Cytohubba plugin, and TF-gene together with TF-miRNA regulatory networks were constructed via utilizing the Networkanalyst platform. Subsequently, we generated subject operating characteristic curves (ROC) to verify the diagnostic capabilities of these hub genes to predict the risk of SLE with COVID-19 infection. Finally, a single-sample gene set enrichment (ssGSEA) algorithm was used to analyze immune cell infiltration. Results A total of 6 common hub genes (CDC6, PLCG1, KIF15, LCK, CDC25C, and RASGRP1) were identified with high diagnostic validity. These gene functional enrichments were mainly involved in cell cycle, and inflammation-related pathways. Compared to the healthy controls, abnormal infiltration of immune cells was found in SLE and COVID-19, and the proportion of immune cells linked to the 6 hub genes. Conclusion Our research logically identified 6 candidate hub genes that could predict SLE complicated with COVID-19. This work provides a foothold for further study of potential pathogenesis in SLE and COVID-19.
Collapse
Affiliation(s)
- Huiqiong Zeng
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Futian District, Shenzhen, Guangdong, China
| | - Yu Zhuang
- Department of Rheumatology and Immunology, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
| | - Xiaojuan Li
- Department of Public Health, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Zhihua Yin
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Futian District, Shenzhen, Guangdong, China
| | - Xia Huang
- Department of Xi Yuan Community Health Service Center, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Haiyan Peng
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Futian District, Shenzhen, Guangdong, China
| |
Collapse
|
119
|
Matta B, Battaglia J, Barnes BJ. A New Methodology for the Quantification of Neutrophil Extracellular Traps in Patient Plasma. Bio Protoc 2023; 13:e4701. [PMID: 37397793 PMCID: PMC10308188 DOI: 10.21769/bioprotoc.4701] [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: 12/14/2022] [Revised: 02/21/2023] [Accepted: 04/10/2023] [Indexed: 07/04/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are web-like structures made up of decondensed chromatin fibers along with neutrophil granular proteins that are extruded by neutrophils after activation or in response to foreign microorganisms. NETs have been associated with autoimmune and inflammatory diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis, coronavirus disease 2019 (COVID-19), and others. There are reliable methods available to quantitate NETs from neutrophils, but their accurate quantification in patient plasma or serum remains a challenge. We developed a highly sensitive ELISA to detect NETs in serum/plasma and designed a novel smear immunofluorescence assay to detect NETs in as little as 1 μL of serum/plasma. We further validated our technology on plasma samples from SLE patients and healthy donors that carry interferon regulatory factor 5 genetic risk. The multiplex ELISA combines the use of three antibodies against myeloperoxidase (MPO), citrullinated histone H3 (CitH3), and DNA to detect the NET complexes with higher specificities. The immunofluorescence smear assay can visually detect intact structures of NETs in 1 μL of serum/plasma and provide similar results that correlate with findings from the multiplex ELISA. Furthermore, the smear assay is a relatively simple, inexpensive, and quantifiable method of NET detection for small volumes.
Collapse
Affiliation(s)
- Bharati Matta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Jenna Battaglia
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Betsy J. Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
120
|
Lerkvaleekul B, Charuvanij S, Sukharomana M, Pirojsakul K, Kamolwatwong M, Vilaiyuk S. Outcomes in children with rheumatic diseases following COVID-19 vaccination and infection: data from a large two-center cohort study in Thailand. Front Pediatr 2023; 11:1194821. [PMID: 37360372 PMCID: PMC10285492 DOI: 10.3389/fped.2023.1194821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction Vaccination against coronavirus disease 2019 (COVID-19) is effective in protecting patients from severe COVID-19 infection. Disease flare-up following immunization in children with rheumatic disorders may result in patient reluctance to receive the vaccine. Underlying rheumatic diseases or the use of immunosuppressive drugs may influence the outcomes of COVID-19 vaccination and infection. We aimed to describe outcomes in children with rheumatic diseases following COVID-19 immunization and infection. Methods This retrospective study was performed at two large academic centers in Thailand. During the COVID-19 pandemic, all patients were routinely queried about COVID-19-related conditions. We included patients with rheumatic diseases aged <18 years who received at least one dose of a COVID-19 vaccine or had a history of COVID-19 infection with more than 6 months of recorded follow-up after the last vaccine dose or COVID-19 illness. Demographic information and data on clinical symptoms, disease activity, treatment, outcomes, and COVID-19 vaccination and infection were collected. Results A total of 479 patients were included. Most (229; 47.81%) patients had juvenile idiopathic arthritis, followed by connective tissue diseases (189; 39.46%), vasculitis syndromes (42; 8.76%), and other rheumatic diseases (19; 3.97%). Approximately 90% of patients received at least one dose of COVID-19 vaccination, and half of the patients had COVID-19 infection. Among patients, 10.72% and 3.27% developed a flare after COVID-19 vaccination and COVID-19 illness, respectively. Flare severity after COVID immunization and infection was mainly mild to moderate. The predictor of flare after COVID-19 vaccination was the use of prednisolone ≥10 mg/day before vaccination (hazard ratio: 2.04, 95% confidence interval: 1.05-3.97, p = 0.037). Inactive disease before receiving the COVID-19 vaccination was a predictor of inactive status after a flare (hazard ratio: 2.95, 95% confidence interval: 1.04-8.40; p = 0.043). Overall, 3.36% and 1.61% of patients experienced a new onset of rheumatic disease after receiving the COVID-19 vaccine and after COVID-19 infection, respectively. Conclusion The COVID-19 vaccine is recommended for children with rheumatic disease, particularly those who are in stable condition. After COVID-19 vaccination, patients-especially those with active disease before vaccination or those receiving concurrent prednisolone doses of ≥10 mg/day-should be closely monitored.
Collapse
Affiliation(s)
- Butsabong Lerkvaleekul
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sirirat Charuvanij
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Maynart Sukharomana
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kwanchai Pirojsakul
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Malisa Kamolwatwong
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Soamarat Vilaiyuk
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
121
|
Zhao Y, Han X, Li C, Liu Y, Cheng J, Adhikari BK, Wang Y. COVID-19 and the cardiovascular system: a study of pathophysiology and interpopulation variability. Front Microbiol 2023; 14:1213111. [PMID: 37350790 PMCID: PMC10282193 DOI: 10.3389/fmicb.2023.1213111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/18/2023] [Indexed: 06/24/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans can lead to various degrees of tissue and organ damage, of which cardiovascular system diseases are one of the main manifestations, such as myocarditis, myocardial infarction, and arrhythmia, which threaten the infected population worldwide. These diseases threaten the cardiovascular health of infected populations worldwide. Although the prevalence of coronavirus disease 2019 (COVID-19) has slightly improved with virus mutation and population vaccination, chronic infection, post-infection sequelae, and post-infection severe disease patients still exist, and it is still relevant to study the mechanisms linking COVID-19 to cardiovascular disease (CVD). This article introduces the pathophysiological mechanism of COVID-19-mediated cardiovascular disease and analyzes the mechanism and recent progress of the interaction between SARS-CoV-2 and the cardiovascular system from the roles of angiotensin-converting enzyme 2 (ACE2), cellular and molecular mechanisms, endothelial dysfunction, insulin resistance, iron homeostasis imbalance, and psychosocial factors, respectively. We also discussed the differences and mechanisms involved in cardiovascular system diseases combined with neocoronavirus infection in different populations and provided a theoretical basis for better disease prevention and management.
Collapse
Affiliation(s)
- Yifan Zhao
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaorong Han
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Cheng Li
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Yucheng Liu
- Department of Family and Community Medicine, Feinberg School of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, United States
| | - Jiayu Cheng
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | | | - Yonggang Wang
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
122
|
Moore N, Williams R, Mori M, Bertolusso B, Vernet G, Lynch J, Philipson P, Ledgerwood T, Kidd SP, Thomas C, Garcia-Arias V, Young M, Saeed K, Gordon K, Cortes N. Mid-regional proadrenomedullin (MR-proADM), C-reactive protein (CRP) and other biomarkers in the early identification of disease progression in patients with COVID-19 in the acute NHS setting. J Clin Pathol 2023; 76:400-406. [PMID: 34996755 PMCID: PMC8761594 DOI: 10.1136/jclinpath-2021-207750] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 12/14/2021] [Indexed: 02/02/2023]
Abstract
AIMS There is a lack of biomarkers validated for assessing clinical deterioration in patients with COVID-19 on presentation to secondary or tertiary care. This evaluation looked at the potential clinical application of C reactive protein (CRP), procalcitonin, mid-regional proadrenomedullin (MR-proADM) and white cell count to support prediction of clinical outcomes. METHODS 135 patients presenting to Hampshire Hospitals NHS Foundation Trust between April and June 2020 confirmed to have COVID-19 via reverse-transcription-qPCR were included. Biomarkers from within 24 hours of presentation were used to predict disease progression by Cox regression and area under the receiver operating characteristic curves. The endpoints assessed were 30-day all-cause mortality, intubation and ventilation, critical care admission and non-invasive ventilation (NIV) use. RESULTS Elevated MR-proADM was shown to have the greatest ability to predict 30-day mortality adjusting for age, cardiovascular disease, renal disease and neurological disease. A significant association was also noted between raised MR-proADM and CRP concentrations and the requirement for critical care admission and NIV. CONCLUSIONS The measurement of MR-proADM and CRP in patients with confirmed COVID-19 infection on admission shows significant potential to support clinicians in identifying those at increased risk of disease progression and need for higher level care, subsequently enabling prompt escalation in clinical interventions.
Collapse
Affiliation(s)
- Nathan Moore
- Microbiology Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
| | - Rebecca Williams
- Microbiology Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
| | - Matilde Mori
- Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Gabrielle Vernet
- Emergency Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
| | - Jessica Lynch
- Microbiology Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
| | - Pete Philipson
- University of Newcastle upon Tyne, Newcastle upon Tyne, Tyne and Wear, UK
| | - Thomas Ledgerwood
- Microbiology Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
| | - Stephen P Kidd
- Microbiology Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
| | - Claire Thomas
- Microbiology Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
- Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Michelle Young
- Biochemsitry Department, Whittington Hospital, London, UK
| | - Kordo Saeed
- Faculty of Medicine, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Kirsty Gordon
- Biochemistry Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
| | - Nicholas Cortes
- Microbiology Department, Basingstoke and North Hampshire Hospital, Basingstoke, UK
- Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
123
|
Mohandas S, Jagannathan P, Henrich TJ, Sherif ZA, Bime C, Quinlan E, Portman MA, Gennaro M, Rehman J. Immune mechanisms underlying COVID-19 pathology and post-acute sequelae of SARS-CoV-2 infection (PASC). eLife 2023; 12:e86014. [PMID: 37233729 PMCID: PMC10219649 DOI: 10.7554/elife.86014] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
With a global tally of more than 500 million cases of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections to date, there are growing concerns about the post-acute sequelae of SARS-CoV-2 infection (PASC), also known as long COVID. Recent studies suggest that exaggerated immune responses are key determinants of the severity and outcomes of the initial SARS-CoV-2 infection as well as subsequent PASC. The complexity of the innate and adaptive immune responses in the acute and post-acute period requires in-depth mechanistic analyses to identify specific molecular signals as well as specific immune cell populations which promote PASC pathogenesis. In this review, we examine the current literature on mechanisms of immune dysregulation in severe COVID-19 and the limited emerging data on the immunopathology of PASC. While the acute and post-acute phases may share some parallel mechanisms of immunopathology, it is likely that PASC immunopathology is quite distinct and heterogeneous, thus requiring large-scale longitudinal analyses in patients with and without PASC after an acute SARS-CoV-2 infection. By outlining the knowledge gaps in the immunopathology of PASC, we hope to provide avenues for novel research directions that will ultimately lead to precision therapies which restore healthy immune function in PASC patients.
Collapse
Affiliation(s)
- Sindhu Mohandas
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Prasanna Jagannathan
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford UniversityStanfordUnited States
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Zaki A Sherif
- Department of Biochemistry & Molecular Biology, Howard University College of MedicineWashingtonUnited States
| | - Christian Bime
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, University of Arizona College of MedicineTucsonUnited States
| | - Erin Quinlan
- National Center for Complementary and Integrative Health, National Institutes of HealthBethesdaUnited States
| | - Michael A Portman
- Seattle Children’s Hospital, Division of Pediatric Cardiology, Department of Pediatrics, University of WashingtonSeattleUnited States
| | - Marila Gennaro
- Public Health Research Institute and Department of Medicine, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of MedicineChicagoUnited States
| |
Collapse
|
124
|
Hirsch J, Uzun G, Zlamal J, Singh A, Bakchoul T. Platelet-neutrophil interaction in COVID-19 and vaccine-induced thrombotic thrombocytopenia. Front Immunol 2023; 14:1186000. [PMID: 37275917 PMCID: PMC10237318 DOI: 10.3389/fimmu.2023.1186000] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/04/2023] [Indexed: 06/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is known to commonly induce a thrombotic diathesis, particularly in severely affected individuals. So far, this COVID-19-associated coagulopathy (CAC) has been partially explained by hyperactivated platelets as well as by the prothrombotic effects of neutrophil extracellular traps (NETs) released from neutrophils. However, precise insight into the bidirectional relationship between platelets and neutrophils in the pathophysiology of CAC still lags behind. Vaccine-induced thrombotic thrombocytopenia (VITT) is a rare autoimmune disorder caused by auto-antibody formation in response to immunization with adenoviral vector vaccines. VITT is associated with life-threatening thromboembolic events and thus, high fatality rates. Our concept of the thrombophilia observed in VITT is relatively new, hence a better understanding could help in the management of such patients with the potential to also prevent VITT. In this review we aim to summarize the current knowledge on platelet-neutrophil interplay in COVID-19 and VITT.
Collapse
Affiliation(s)
- Johannes Hirsch
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Günalp Uzun
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Jan Zlamal
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Anurag Singh
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Tamam Bakchoul
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| |
Collapse
|
125
|
Wang M, Yu F, Chang W, Zhang Y, Zhang L, Li P. Inflammasomes: a rising star on the horizon of COVID-19 pathophysiology. Front Immunol 2023; 14:1185233. [PMID: 37251383 PMCID: PMC10213254 DOI: 10.3389/fimmu.2023.1185233] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a contagious respiratory virus that is the cause of the coronavirus disease 2019 (COVID-19) pandemic which has posed a serious threat to public health. COVID-19 is characterized by a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild cold-like symptoms, severe pneumonia or even death. Inflammasomes are supramolecular signaling platforms that assemble in response to danger or microbial signals. Upon activation, inflammasomes mediate innate immune defense by favoring the release of proinflammatory cytokines and triggering pyroptotic cell death. Nevertheless, abnormalities in inflammasome functioning can result in a variety of human diseases such as autoimmune disorders and cancer. A growing body of evidence has showed that SARS-CoV-2 infection can induce inflammasome assembly. Dysregulated inflammasome activation and consequent cytokine burst have been associated with COVID-19 severity, alluding to the implication of inflammasomes in COVID-19 pathophysiology. Accordingly, an improved understanding of inflammasome-mediated inflammatory cascades in COVID-19 is essential to uncover the immunological mechanisms of COVID-19 pathology and identify effective therapeutic approaches for this devastating disease. In this review, we summarize the most recent findings on the interplay between SARS-CoV-2 and inflammasomes and the contribution of activated inflammasomes to COVID-19 progression. We dissect the mechanisms involving the inflammasome machinery in COVID-19 immunopathogenesis. In addition, we provide an overview of inflammasome-targeted therapies or antagonists that have potential clinical utility in COVID-19 treatment.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | | | | | | | | | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
126
|
Ghimire R, Shrestha R, Amaradhi R, Patton T, Whitley C, Chanda D, Liu L, Ganesh T, More S, Channappanavar R. Toll-like receptor 7 (TLR7)-mediated antiviral response protects mice from lethal SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539929. [PMID: 37214943 PMCID: PMC10197544 DOI: 10.1101/2023.05.08.539929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
SARS-CoV-2-induced impaired antiviral and excessive inflammatory responses cause fatal pneumonia. However, the key pattern recognition receptors that elicit effective antiviral and lethal inflammatory responses in-vivo are not well defined. CoVs possess single-stranded RNA (ssRNA) genome that is abundantly produced during infection and stimulates both antiviral interferon (IFN) and inflammatory cytokine/ chemokine responses. Therefore, in this study, using wild-type control and TLR7 deficient BALB/c mice infected with a mouse-adapted SARS-COV-2 (MA-CoV-2), we evaluated the role of TLR7 signaling in MA-CoV-2-induced antiviral and inflammatory responses and disease outcome. We show that TLR7-deficient mice are more susceptible to MA-CoV-2 infection as compared to infected control mice. Further evaluation of MA-CoV-2 infected lungs showed significantly reduced mRNA levels of antiviral type I (IFNα/β) and type III (IFNλ) IFNs, IFN stimulated genes (ISGs, ISG15 and CXCL10), and several pro-inflammatory cytokines/chemokines in TLR7 deficient compared to control mice. Reduced lung IFN/ISG levels and increased morbidity/mortality in TLR7 deficient mice correlated with high lung viral titer. Detailed examination of total cells from MA-CoV-2 infected lungs showed high neutrophil count in TLR7 deficient mice compared to control mice. Additionally, blocking TLR7 activity post-MA-CoV-2 infection using a specific inhibitor also enhanced disease severity. In summary, our results conclusively establish that TLR7 signaling is protective during SARS-CoV-2 infection, and despite robust inflammatory response, TLR7-mediated IFN/ISG responses likely protect the host from lethal disease. Given similar outcomes in control and TLR7 deficient humans and mice, these results show that MA-CoV-2 infected mice serve as excellent model to study COVID-19.
Collapse
|
127
|
Teo A, Chan LLY, Cheung C, Chia PY, Ong SWX, Fong SW, Ng LFP, Renia L, Lye DC, Young BE, Yeo TW. Myeloperoxidase inhibition may protect against endothelial glycocalyx shedding induced by COVID-19 plasma. COMMUNICATIONS MEDICINE 2023; 3:62. [PMID: 37147421 PMCID: PMC10160718 DOI: 10.1038/s43856-023-00293-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/27/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND SARS-CoV-2, the causative agent of COVID-19, is a threat to public health. Evidence suggests increased neutrophil activation and endothelial glycocalyx (EG) damage are independently associated with severe COVID-19. Here, we hypothesised that an increased level of blood neutrophil myeloperoxidase (MPO) is associated with soluble EG breakdown, and inhibiting MPO activity may reduce EG damage. METHODS Analysing a subset of acute and convalescent COVID-19 plasma, 10 from severe and 15 from non-severe COVID-19 cases, and 9 from pre-COVID-19 controls, we determined MPO levels, MPO activity and soluble EG proteins (syndecan-1 and glypican-1) levels by enzyme-linked immunosorbent assay. In vitro primary human aortic endothelial cells were cultured with plasma untreated or treated with specific MPO inhibitors (MPO-IN-28, AZD5904) to determine EG shedding. We then investigated whether inhibiting MPO activity decreased EG degradation. RESULTS In COVID-19 plasma, MPO levels, MPO activity and levels of soluble EG proteins are significantly raised compared to controls, and concentrations increase in proportion to disease severity. Despite clinical recovery, protein concentrations remain significantly elevated. Interestingly, there is a trend of increasing MPO activity in convalescent plasma in both severe and non-severe groups. MPO levels and MPO activity correlate significantly with soluble EG levels and inhibiting MPO activity leads to reduced syndecan-1 shedding, in vitro. CONCLUSIONS Neutrophil MPO may increase EG shedding in COVID-19, and inhibiting MPO activity may protect against EG degradation. Further research is needed to evaluate the utility of MPO inhibitors as potential therapeutics against severe COVID-19.
Collapse
Affiliation(s)
- Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, VIC, Australia.
- National Centre for Infectious Diseases, Singapore, Singapore.
| | - Louisa L Y Chan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science and Technology and Research (A*STAR), Singapore, Singapore
| | - Po Ying Chia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Sean Wei Xiang Ong
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Siew Wai Fong
- A*STAR Infectious Diseases Lab (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Lab (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Singapore
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK
| | - Laurent Renia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- A*STAR Infectious Diseases Lab (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - David Chien Lye
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Barnaby Edward Young
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tsin Wen Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
128
|
Li Y, Hook JS, Ding Q, Xiao X, Chung SS, Mettlen M, Xu L, Moreland JG, Agathocleous M. Neutrophil metabolomics in severe COVID-19 reveal GAPDH as a suppressor of neutrophil extracellular trap formation. Nat Commun 2023; 14:2610. [PMID: 37147288 PMCID: PMC10162006 DOI: 10.1038/s41467-023-37567-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/20/2023] [Indexed: 05/07/2023] Open
Abstract
Severe COVID-19 is characterized by an increase in the number and changes in the function of innate immune cells including neutrophils. However, it is not known how the metabolome of immune cells changes in patients with COVID-19. To address these questions, we analyzed the metabolome of neutrophils from patients with severe or mild COVID-19 and healthy controls. We identified widespread dysregulation of neutrophil metabolism with disease progression including in amino acid, redox, and central carbon metabolism. Metabolic changes in neutrophils from patients with severe COVID-19 were consistent with reduced activity of the glycolytic enzyme GAPDH. Inhibition of GAPDH blocked glycolysis and promoted pentose phosphate pathway activity but blunted the neutrophil respiratory burst. Inhibition of GAPDH was sufficient to cause neutrophil extracellular trap (NET) formation which required neutrophil elastase activity. GAPDH inhibition increased neutrophil pH, and blocking this increase prevented cell death and NET formation. These findings indicate that neutrophils in severe COVID-19 have an aberrant metabolism which can contribute to their dysfunction. Our work also shows that NET formation, a pathogenic feature of many inflammatory diseases, is actively suppressed in neutrophils by a cell-intrinsic mechanism controlled by GAPDH.
Collapse
Affiliation(s)
- Yafeng Li
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica S Hook
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qing Ding
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xue Xiao
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen S Chung
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel Mettlen
- Department of Cell Biology, Quantitative Light Microscopy Core, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica G Moreland
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michalis Agathocleous
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
129
|
Bello S, Lasierra AB, López-Vergara L, de Diego C, Torralba L, de Gopegui PR, Lahoz R, Abadía C, Godino J, Cebollada A, Jimeno B, Bello C, Tejada A, Torres A. IL-6 and cfDNA monitoring throughout COVID-19 hospitalization are accurate markers of its outcomes. Respir Res 2023; 24:125. [PMID: 37147677 PMCID: PMC10161166 DOI: 10.1186/s12931-023-02426-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/18/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Severe COVID-19 entails a dysregulated immune response, most likely inflammation related to a lack of virus control. A better understanding of immune toxicity, immunosuppression balance, and COVID-19 assessments could help determine whether different clinical presentations are driven by specific types of immune responses. The progression of the immune response and tissular damage could predict outcomes and may help in the management of patients. METHODS We collected 201 serum samples from 93 hospitalised patients classified as moderately, severely, and critically ill. We differentiated the viral, early inflammatory, and late inflammatory phases and included 72 patients with 180 samples in separate stages for longitudinal study and 55 controls. We studied selected cytokines, P-selectin, and the tissue damage markers lactate dehydrogenase (LDH) and cell-free DNA (cfDNA). RESULTS TNF-α, IL-6, IL-8, and G-CSF were associated with severity and mortality, but only IL-6 increased since admission in the critical patients and non-survivors, correlating with damage markers. The lack of a significant decrease in IL-6 levels in the critical patients and non-survivors in the early inflammatory phase (a decreased presence in the other patients) suggests that these patients did not achieve viral control on days 10-16. For all patients, lactate dehydrogenase and cfDNA levels increased with severity, and cfDNA levels increased in the non-survivors from the first sample (p = 0.002) to the late inflammatory phase (p = 0.031). In the multivariate study, cfDNA was an independent risk factor for mortality and ICU admission. CONCLUSIONS The distinct progression of IL-6 levels in the course of the disease, especially on days 10-16, was a good marker of progression to critical status and mortality and could guide the start of IL-6 blockade. cfDNA was an accurate marker of severity and mortality from admission and throughout COVID-19 progression.
Collapse
Affiliation(s)
- Salvador Bello
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain.
| | | | - Lucía López-Vergara
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain
| | - Cristina de Diego
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain
| | - Laura Torralba
- Department of Pulmonary Medicine, Miguel Servet University Hospital, CIBERES, Instituto de Investigación Sanitaria (ISS) Aragón, Avenida Isabel La Católica 1-9, 50009, Zaragoza, Spain
| | | | - Raquel Lahoz
- Department of Biochemistry, Miguel Servet University Hospital, Zaragoza, Spain
| | - Claudia Abadía
- Department of Biochemistry, Miguel Servet University Hospital, Zaragoza, Spain
| | - Javier Godino
- Department of Cytometry and Cell Separation, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Alberto Cebollada
- Biocomputing Technical Scientific Service, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Beatriz Jimeno
- Department of Cytometry and Cell Separation, Aragon Institute of Health Sciences (IACS), Zaragoza, Spain
| | - Carlota Bello
- Department of Radiology, Hospital Clínico Lozano Blesa, Zaragoza, Spain
| | - Antonio Tejada
- Intensive Care Unit, Miguel Servet University Hospital, Zaragoza, Spain
| | - Antoni Torres
- Servei de Pneumologia, Hospital Clinic, Universitat de Barcelona, IDIBAPS, ICREA, CIBERESUCICOVID, Barcelona, Spain
| |
Collapse
|
130
|
Herrera VLM, Bosch NA, Lok JJ, Nguyen MQ, Lenae KA, deKay JT, Ryzhov SV, Seder DB, Ruiz-Opazo N, Walkey AJ. Circulating neutrophil extracellular trap (NET)-forming 'rogue' neutrophil subset, immunotype [DEspR + CD11b +], mediate multi-organ failure in COVID-19- an observational study. TRANSLATIONAL MEDICINE COMMUNICATIONS 2023; 8:12. [PMID: 37096233 PMCID: PMC10111078 DOI: 10.1186/s41231-023-00143-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/24/2023] [Indexed: 05/03/2023]
Abstract
Background Cumulative research show association of neutrophils and neutrophil extracellular traps (NETs) with poor outcomes in severe COVID-19. However, to date, there is no curative intent therapy able to block neutrophil/NETs-mediated progression of multi-organ dysfunction. Because of emerging neutrophil heterogeneity, the study of subsets of circulating NET-forming neutrophils [NET + Ns] as mediators of multi-organ failure progression among patients with COVID-19 is critical to identification of therapeutic targets. Methods We conducted a prospective observational study of circulating levels of CD11b + [NET + N] immunotyped for dual endothelin-1/signal peptide receptor (DEspR ±) expression by quantitative immunofluorescence-cytology and causal mediation analysis. In 36 consented adults hospitalized with mod-severe COVID-19, May to September 2020, we measured acute multi-organ failure via SOFA-scores and respiratory failure via SaO2/FiO2 (SF)-ratio at time points t1 (average 5.5 days from ICU/hospital admission) and t2 (the day before ICU-discharge or death), and ICU-free days at day28 (ICUFD). Circulating absolute neutrophil counts (ANC) and [NET + N] subset-specific counts were measured at t1. Spearman correlation and causal mediation analyses were conducted. Results Spearman correlation analyses showed correlations of t1-SOFA with t2-SOFA (rho r S = 0.80) and ICUFD (r S = -0.76); circulating DEspR + [NET + Ns] with t1-SOFA (r S = 0.71), t2-SOFA (r S = 0.62), and ICUFD (r S = -0.63), and ANC with t1-SOFA (r S = 0.71), and t2-SOFA (r S = 0.61).Causal mediation analysis identified DEspR + [NET + Ns] as mediator of 44.1% [95% CI:16.5,110.6] of the causal path between t1-SOFA (exposure) and t2-SOFA (outcome), with 46.9% [15.8,124.6] eliminated when DEspR + [NET + Ns] were theoretically reduced to zero. Concordantly, DEspR + [NET + Ns] mediated 47.1% [22.0,72.3%] of the t1-SOFA to ICUFD causal path, with 51.1% [22.8,80.4%] eliminated if DEspR + [NET + Ns] were reduced to zero. In patients with t1-SOFA > 1, the indirect effect of a hypothetical treatment eliminating DEspR + [NET + Ns] projected a reduction of t2-SOFA by 0.98 [0.29,2.06] points and ICUFD by 3.0 [0.85,7.09] days. In contrast, there was no significant mediation of SF-ratio through DEspR + [NET + Ns], and no significant mediation of SOFA-score through ANC. Conclusions Despite equivalent correlations, DEspR + [NET + Ns], but not ANC, mediated progression of multi-organ failure in acute COVID-19, and its hypothetical reduction is projected to improve ICUFD. These translational findings warrant further studies of DEspR + [NET + Ns] as potential patient-stratifier and actionable therapeutic target for multi-organ failure in COVID-19. Supplementary Information The online version contains supplementary material available at 10.1186/s41231-023-00143-x.
Collapse
Affiliation(s)
- Victoria L. M. Herrera
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts USA
| | - Nicholas A. Bosch
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts USA
| | - Judith J. Lok
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts USA
| | - Mai Q. Nguyen
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts USA
| | - Kaitriona A. Lenae
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts USA
| | | | | | - David B. Seder
- Maine Health Institute for Research, Scarborough, Maine USA
- Department of Critical Care Services, Maine Medical Center, Portland, Maine USA
| | - Nelson Ruiz-Opazo
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts USA
| | - Allan J. Walkey
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts USA
| |
Collapse
|
131
|
Jin X, Wang Y, Xu J, Li Y, Cheng F, Luo Y, Zhou H, Lin S, Xiao F, Zhang L, Lin Y, Zhang Z, Jin Y, Zheng F, Chen W, Zhu A, Tao Y, Zhao J, Kuo T, Li Y, Li L, Wen L, Ou R, Li F, Lin L, Zhang Y, Sun J, Yuan H, Zhuang Z, Sun H, Chen Z, Li J, Zhuo J, Chen D, Zhang S, Sun Y, Wei P, Yuan J, Xu T, Yang H, Wang J, Xu X, Zhong N, Xu Y, Sun K, Zhao J. Plasma cell-free DNA promise monitoring and tissue injury assessment of COVID-19. Mol Genet Genomics 2023; 298:823-836. [PMID: 37059908 PMCID: PMC10104435 DOI: 10.1007/s00438-023-02014-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/25/2023] [Indexed: 04/16/2023]
Abstract
Coronavirus 2019 (COVID-19) is a complex disease that affects billions of people worldwide. Currently, effective etiological treatment of COVID-19 is still lacking; COVID-19 also causes damages to various organs that affects therapeutics and mortality of the patients. Surveillance of the treatment responses and organ injury assessment of COVID-19 patients are of high clinical value. In this study, we investigated the characteristic fragmentation patterns and explored the potential in tissue injury assessment of plasma cell-free DNA in COVID-19 patients. Through recruitment of 37 COVID-19 patients, 32 controls and analysis of 208 blood samples upon diagnosis and during treatment, we report gross abnormalities in cfDNA of COVID-19 patients, including elevated GC content, altered molecule size and end motif patterns. More importantly, such cfDNA fragmentation characteristics reflect patient-specific physiological changes during treatment. Further analysis on cfDNA tissue-of-origin tracing reveals frequent tissue injuries in COVID-19 patients, which is supported by clinical diagnoses. Hence, our work demonstrates and extends the translational merit of cfDNA fragmentation pattern as valuable analyte for effective treatment monitoring, as well as tissue injury assessment in COVID-19.
Collapse
Affiliation(s)
- Xin Jin
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jinjin Xu
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Yimin Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Fanjun Cheng
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yuxue Luo
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Haibo Zhou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511500, Guangdong, China
| | - Shanwen Lin
- Yangjiang People's Hospital, Yangjiang, 529500, Guangdong, China
| | - Fei Xiao
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China
| | - Lu Zhang
- Institute of Infectious Disease, Guangzhou Eighth People's Hospital of Guangzhou Medical University, Guangzhou, 510060, Guangdong, China
| | - Yu Lin
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Yan Jin
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Fang Zheng
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Wei Chen
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Ye Tao
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Tingyou Kuo
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, Guangdong, China
| | - Yuming Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Lingguo Li
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, Guangdong, China
| | - Liyan Wen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Rijing Ou
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Fang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Long Lin
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, Guangdong, China
| | - Yanjun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Hao Yuan
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, Guangdong, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Haixi Sun
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jie Li
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, Guangdong, China
| | - Jianfen Zhuo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | | | - Shengnan Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Yuzhe Sun
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Peilan Wei
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jinwei Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Tian Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- Guangdong Provincial Academician Workstation of BGI Synthetic Genomics, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, 518120, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Yonghao Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
- Institute of Infectious Disease, Guangzhou Eighth People's Hospital of Guangzhou Medical University, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
132
|
Acar A. Pan-Cancer Analysis of the COVID-19 Causal Gene SLC6A20. ACS OMEGA 2023; 8:13153-13161. [PMID: 37041751 PMCID: PMC10081573 DOI: 10.1021/acsomega.3c00407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/22/2023] [Indexed: 06/19/2023]
Abstract
Genome-wide association studies demonstrated that the chromosome 3p31.21 locus was linked to the severity of COVID-19 disease. The SLC6A20 gene was reported to be one of the critical causal genes regulated by this locus. Various studies focused on demonstrating the severity of COVID-19 in cancer patients and reported that elevated SARS-CoV-2-associated gene expression might contribute to increased susceptibility for COVID-19 in cancer patients. Given that pan-cancer association for the COVID-19 causal gene SLC6A20 is lacking, we aimed to perform systematic profiling of SLC6A20 in different malignancies. Human Protein Atlas, UALCAN, and Hepatocellular Carcinoma (HCCDB) databases were used to assess SLC6A20 gene expression changes in The Cancer Genome Atlas samples with respect to their normal counterparts. GEPIA and TIMER2.0 databases were used to determine the correlation between SLC6A20 and COVID-19-associated genes. Different databases were used for identification of the correlation of SCL6A20 with infiltrating immune cells. The canSAR database was utilized to determine the association of SCL6A20 with immune profiling in different malignancies. The STRING database was utilized to determine the protein network interacting with SLC6A20. Here, we showed SLC6A20 mRNA expression in pan-cancer samples and their normal counterparts. Increased SCL6A20 expression was associated with tumor grade, and there was a positive correlation with SARS-CoV-2-associated genes. Furthermore, SLC6A20 expression was positively correlated with infiltrating neutrophils and immune-related signatures. Lastly, SLC6A20 expression was found to be associated with the angiotensin converting enzyme 2 homologue, TMEM27, suggesting a potential link between SLC6A20 and COVID-19. Taken together, these results suggest that elevated SLC6A20 levels might be partly responsible for increased susceptibility of cancer patients to COVID-19 disease. Therapeutic intervention strategies against SLC6A20 in cancer patients, alongside other treatment modalities, might offer a benefit in delaying COVID-19 disease.
Collapse
|
133
|
Nagendra L, Bhattacharya S, Kalra S, Kapoor N. Metformin in COVID-19: Is There a Role Beyond Glycemic Control? Int J Endocrinol Metab 2023; 21:e132965. [PMID: 37654526 PMCID: PMC10467582 DOI: 10.5812/ijem-132965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/26/2023] [Accepted: 04/15/2023] [Indexed: 09/02/2023] Open
Abstract
Context The coronavirus disease 2019 (COVID-19) pandemic is still a cause of worldwide health concern. Diabetes and its associated comorbidities are risk factors for mortality and morbidity in COVID-19. Selecting the right antidiabetic drug to achieve optimal glycemic control might mitigate some of the negative impacts of diabetes. Metformin continues to be the most widely administered antidiabetic agent. There is evidence of its beneficial outcome in COVID-19 independent of its glucose-lowering effect. Evidence Acquisition A thorough literature search was conducted in PubMed, Google Scholar, Scopus, and Web of Science to identify studies investigating metformin in COVID-19. Results Several overlapping mechanisms have been proposed to explain its antiviral properties. It could bring about conformational changes in the angiotensin-converting enzyme-2 receptor and decrease viral entry. The effects on the mammalian target of the rapamycin pathway and cellular pH have been proposed to reduce viral protein synthesis and replication. The immunomodulatory effects of metformin might counter the detrimental effects of hyperinflammation associated with COVID-19. Conclusions These findings call for broader metformin usage to manage hyperglycemia in COVID-19.
Collapse
Affiliation(s)
- Lakshmi Nagendra
- Department of Endocrinology, JSS Medical College and Hospital, JSS Academy of Higher Education & Research (JSS AHER), Mysore, Karnataka, India
| | | | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
| | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India
| |
Collapse
|
134
|
Tahir A, Walia J, Daly T, Gradzka A, Banai R. Rapidly Progressive Glomerulonephritis: A COVID-19 Case Report. Cureus 2023; 15:e37767. [PMID: 37214004 PMCID: PMC10194189 DOI: 10.7759/cureus.37767] [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] [Accepted: 04/18/2023] [Indexed: 05/23/2023] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis is a systemic autoimmune disease that typically presents as a multi-organ manifesting disease of unclear etiology that can predispose to rapidly progressive glomerulonephritis (RPGN). If left untreated, ANCA-associated vasculitis can be fatal, and RPGN can progress to irreversible renal failure. Environmental and genetic factors have been implicated in the pathogenesis of this vasculitis. Coronavirus disease (COVID-19) has been noted to have various physiologic impacts on the body, with literature indicating possible autoimmune effects. We present a rare case of ANCA-associated vasculitis in an elderly male with no known autoimmune history after a recent illness with COVID-19. The patient had been seen as an outpatient with progressively declining renal function until he presented to the hospital with acute renal failure and pericarditis. Workup revealed elevated anti-myeloperoxidase antibody (MPO-AB) and perinuclear ANCA (p-ANCA) antibodies with a biopsy confirming focal cresenteric glomerulonephritis, and the patient was initiated on steroid therapy with notable improvement and a return to baseline kidney function.
Collapse
Affiliation(s)
- Ali Tahir
- Internal Medicine, St. Luke's University Health Network, Bethlehem, USA
| | - Jasmit Walia
- Internal Medicine, St. Luke's University Health Network, Bethlehem, USA
| | - Timothy Daly
- Internal Medicine, St. Luke's University Health Network, Bethlehem, USA
| | - Alexandra Gradzka
- Internal Medicine, St. Luke's University Health Network, Bethlehem, USA
| | - Ruslan Banai
- Internal Medicine, St. Luke's University Health Network, Bethlehem, USA
| |
Collapse
|
135
|
Kim IS, Kim DH, Lee HW, Kim SG, Kim YK, Kim JK. Role of increased neutrophil extracellular trap formation on acute kidney injury in COVID-19 patients. Front Immunol 2023; 14:1122510. [PMID: 37051234 PMCID: PMC10083414 DOI: 10.3389/fimmu.2023.1122510] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
BackgroundA strong association between elevated neutrophil extracellular trap (NET) levels and poor clinical outcomes in patients with coronavirus infection 2019 (COVID-19) has been reported. However, while acute kidney injury (AKI) is a common complication of COVID-19, the role of NETs in COVID-19-associated AKI is unclear. We investigated the association between elevated NETs and AKI and the prognostic role of NETs in COVID-19 patients.MethodsTwo representative markers of NETs, circulating nucleosomes and myeloperoxidase-DNA, were measured in 115 hospitalized patients. Serum levels of interleukin [IL]-6, monocyte chemotactic protein-1 [MCP-1], plasma von Willebrand factor (vWF) and urinary biomarkers of renal tubular damage (β2-microglobulin [β2M] and kidney injury molecule 1 [KIM-1]) were measured.ResultsAKI was found in 43 patients (37.4%), and pre-existing chronic kidney disease (CKD) was a strong risk factor for AKI. Higher circulating NET levels were a significant predictor of increased risk of initial ICU admission, in-hospital mortality (adjusted HR 3.21, 95% CI 1.08–9.19) and AKI (OR 3.67, 95% CI 1.30-10.41), independent of age, diabetes, pre-existing CKD and IL-6 levels. There were strong correlations between circulating nucleosome levels and urinary KIM-1/creatinine (r=0.368, p=0.001) and β2M (r=0.218, p=0.049) levels. NETs were also strongly closely associated with serum vWF (r = 0.356, p<0.001), but not with IL-6 or MCP-1 levels.ConclusionsElevated NETs were closely associated with AKI, which was a strong predictor of mortality. The close association between NETs and vWF may suggest a role for NETs in COVID-19-associated vasculopathy leading to AKI.
Collapse
Affiliation(s)
- In Soo Kim
- Department of Internal Medicine & Kidney Research Institute, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Do Hyun Kim
- Department of Internal Medicine & Kidney Research Institute, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Hoi Woul Lee
- Department of Internal Medicine & Kidney Research Institute, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Sung Gyun Kim
- Department of Internal Medicine & Kidney Research Institute, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Yong Kyun Kim
- Division of Infectious Diseases, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
- *Correspondence: Jwa-Kyung Kim, ; Yong Kyun Kim,
| | - Jwa-Kyung Kim
- Department of Internal Medicine & Kidney Research Institute, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
- *Correspondence: Jwa-Kyung Kim, ; Yong Kyun Kim,
| |
Collapse
|
136
|
Narang J, Jatana S, Ponti AK, Musich R, Gallop J, Wei AH, Seck S, Johnson J, Kokoczka L, Nowacki AS, McBride JD, Mireles-Cabodevila E, Gordon S, Cooper K, Fernandez AP, McDonald C. Abnormal thrombosis and neutrophil activation increase hospital-acquired sacral pressure injuries and morbidity in COVID-19 patients. Front Immunol 2023; 14:1031336. [PMID: 37026002 PMCID: PMC10070761 DOI: 10.3389/fimmu.2023.1031336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023] Open
Abstract
Hospitalized patients have an increased risk of developing hospital-acquired sacral pressure injury (HASPI). However, it is unknown whether SARS-CoV-2 infection affects HASPI development. To explore the role of SARS-CoV-2 infection in HASPI development, we conducted a single institution, multi-hospital, retrospective study of all patients hospitalized for ≥5 days from March 1, 2020 to December 31, 2020. Patient demographics, hospitalization information, ulcer characteristics, and 30-day-related morbidity were collected for all patients with HASPIs, and intact skin was collected from HASPI borders in a patient subset. We determined the incidence, disease course, and short-term morbidity of HASPIs in COVID-19(+) patients, and characterized the skin histopathology and tissue gene signatures associated with HASPIs in COVID-19 disease. COVID-19(+) patients had a 63% increased HASPI incidence rate, HASPIs of more severe ulcer stage (OR 2.0, p<0.001), and HASPIs more likely to require debridement (OR 3.1, p=0.04) compared to COVID-19(-) patients. Furthermore, COVID-19(+) patients with HASPIs had 2.2x increased odds of a more severe hospitalization course compared to COVID-19(+) patients without HASPIs. HASPI skin histology from COVID-19(+) patients predominantly showed thrombotic vasculopathy, with the number of thrombosed vessels being significantly greater than HASPIs from COVID-19(-) patients. Transcriptional signatures of a COVID-19(+) sample subset were enriched for innate immune responses, thrombosis, and neutrophil activation genes. Overall, our results suggest that immunologic dysregulation secondary to SARS-CoV-2 infection, including neutrophil dysfunction and abnormal thrombosis, may play a pathogenic role in development of HASPIs in patients with severe COVID-19.
Collapse
Affiliation(s)
- Jatin Narang
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Samreen Jatana
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - András K. Ponti
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Ryan Musich
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Joshua Gallop
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Angela H. Wei
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sokhna Seck
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jessica Johnson
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Lynne Kokoczka
- Medical Intensive Care Unit, Cleveland Clinic, Cleveland, OH, United States
| | - Amy S. Nowacki
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Jeffrey D. McBride
- Department of Dermatology, The University of Oklahoma College of Medicine, Oklahoma City, OK, United States
| | | | - Steven Gordon
- Department of Infectious Disease, Cleveland Clinic, Cleveland, OH, United States
| | - Kevin Cooper
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Anthony P. Fernandez
- Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Anatomic Pathology, Pathology and Lab Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Christine McDonald
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
137
|
Liu W, Han F, Wan M, Yang XZ. Integrated bioinformatics analysis identifies shared immune changes between ischemic stroke and COVID 19. Front Immunol 2023; 14:1102281. [PMID: 36969251 PMCID: PMC10030956 DOI: 10.3389/fimmu.2023.1102281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
Although COVID-19 is primarily a respiratory disease, its neurological complications, such as ischemic stroke (IS), have aroused growing concerns and reports. However, the molecular mechanisms that underlie IS and COVID-19 are not well understood. Therefore, we implemented transcriptomic analysis from eight GEO datasets consist of 1191 samples to detect common pathways and molecular biomarkers in IS and COVID-19 that help understand the linkage between them. Differentially expressed genes (DEGs) were detected for IS and COVID-19 separately for finding shared mechanisms and we found that immune-related pathways were outlined with statistical significance. JAK2, which was identified as a hub gene, was supposed to be a potential therapeutic gene targets during the immunological process of COVID-19 and IS. Besides, we found a decrease in the proportion of CD8+ T and T helper 2 cells in the peripheral circulation of both COVID and IS patients, and NCR3 expression was significantly correlated with this change. In conclusion, we demonstrated that transcriptomic analyses reported in this study could make a deeper understanding of the common mechanism and might be promising for effective therapeutic for IS and COVID-19.
Collapse
Affiliation(s)
- Wenhao Liu
- Eight-year program of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyao Wan
- Eight-year program of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Zhuang Yang
- Medical Research Center, State Key laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xin-Zhuang Yang,
| |
Collapse
|
138
|
Ferrandis R, Escontrela B, Ferrando C, Hernández M, Herrera J, Hidalgo F, Librero J, Llau JV, Martínez A, Pajares A, Tapia B, Arruti E, Bassas E, Blasi A, Calvo A. Effectiveness of thromboprophylaxis with low molecular weight heparin in critically ill patients with COVID-19. An observational prospective, multicenter study. REVISTA ESPANOLA DE ANESTESIOLOGIA Y REANIMACION 2023; 70:129-139. [PMID: 36842685 PMCID: PMC9957653 DOI: 10.1016/j.redare.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/09/2022] [Indexed: 04/12/2023]
Abstract
INTRODUCTION COVID-19 induces coagulopathy associated with an increase of thromboembolic events. Due to the lack of agreement on recommendations for thromboprophylactic management, the aim of this study was to study the dosages of LMWH used in critically ill COVID-19 patients assessing the effect on their outcome. METHODS We evaluated data of the Reg-COVID19. According to LMWH dose two groups were analyzed: prophylaxis and treatment. Primary outcome was the relationship of LMWH dosage with mortality. Secondary outcomes included the incidence of thrombotic and bleeding events, length of ICU stay, invasive mechanical ventilation, and thrombotic and inflammatory parameters. RESULTS Data of 720 patients were analyzed, 258 in the prophylaxis group and 462 in the treatment group. C Reactive Protein, invasive mechanical ventilation, tocilizumab and corticosteroid treatments were related with the choice of LMWH dose. Hemorrhagic events (66/720, 9.2%) and thrombotic complications (69/720, 9.6%) were similar in both groups (p = .819 and p = .265), as was the time course of the thrombotic events, earlier than hemorrhagic ones (9 [3-18] and 12 [6-19] days respectively). Mortality was lower in prophylaxis group (25.2% versus 35.1%), but once an inverse probability weighting model was applied, we found no effect of LMWH dose. CONCLUSION We found no benefit or harm with the administration of therapeutic or prophylactic LMWH dose in COVID19 critically ill patients. With a similar rate of hemorrhagic or thrombotic events, the LMWH dose had no influence on mortality. More studies are needed to determine the optimal thromboprophylaxis protocol for critically ill patients.
Collapse
Affiliation(s)
- R Ferrandis
- Departamento de Anestesiología y Cuidados Críticos, Hospital Universitari i Politècnic La Fe, Valencia, Spain.
| | - B Escontrela
- Departamento de Anestesiología y Cuidados Críticos, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - C Ferrando
- Departamento de Anestesiología y Cuidados Críticos, Hospital Clínic, Institut d'Investigacions Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, España, CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - M Hernández
- Departamento de Anestesiología y Cuidados Críticos, Hospital Universitario de Cruces, Barakaldo, Vizcaya, Spain
| | - J Herrera
- Departamento de Anestesiología y Cuidados Críticos, Hospital Universitario de Cruces, Barakaldo, Vizcaya, Spain
| | - F Hidalgo
- Departamento de Anestesiología y Cuidados Críticos, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - J Librero
- Navarrabiomed, Complejo Hospitalario de Navarra-Universidad Pública de Navarra, IDISNA, Pamplona, Navarra, Spain
| | - J V Llau
- Departamento de Anestesiología y Cuidados Críticos, Hospital Universitario Doctor Peset, Valencia, Spain
| | - A Martínez
- Jefe de Servicio de Anestesiología y Cuidados Críticos, Hospital Universitario Cruces, Barakaldo, Vizcaya, Spain
| | - A Pajares
- Departamento de Anestesiología y Cuidados Críticos, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - B Tapia
- Departamento de Anestesiología y Cuidados Críticos, Hospital Universitario La Paz, Madrid, Spain
| | - E Arruti
- Innovation and Technology Area, Ubikare SL, Getxo, Vizcaya, Spain
| | - E Bassas
- Departamento de Anestesiología y Cuidados Críticos, Hospital Moisès Broggi, Sant Joan Despí, Barcelona, Spain
| | - A Blasi
- Departamento de Anestesiología y Cuidados Críticos, Hospital Clínic, Institut d'Investigacions Biomèdica Agust Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| | - A Calvo
- Departamento de Anestesiología y Cuidados Críticos, Hospital Clínic, Institut d'Investigacions Biomèdica Agust Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| |
Collapse
|
139
|
Faruqi J, Balasubramanyam A. COVID-19 and diabetes mellitus: a review of the incidence, pathophysiology and management of diabetes during the pandemic. Expert Rev Endocrinol Metab 2023; 18:167-179. [PMID: 36797835 DOI: 10.1080/17446651.2023.2176300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The COVID-19 pandemic has changed the landscape of modern medicine on a global scale. An emerging concern is the recognition of a bidirectional relationship between COVID-19 and diabetes. Diabetes is a risk factor for severe COVID-19 illness. Intriguingly, recent epidemiological and in vitro studies suggest that infection with SARS-CoV-2, the causative viral agent of COVID-19, is associated with new-onset diabetes and worsening diabetes control. These factors have affected the management of diabetes. AREAS COVERED This review provides an overview of our current understanding of the incidence and prevalence of diabetes in relation to the COVID-19 pandemic, highlights studies evaluating SARS-CoV-2's beta cell tropism and its effects on insulin secretion and sensitivity and evaluates the impact of the pandemic on diabetes management and metabolic control. EXPERT OPINION Epidemiological studies have noted an increase in the incidence of new-onset diabetes associated with COVID-19 in patients with phenotypes of type 1 diabetes, type 2 diabetes and Ketosis-Prone Diabetes. Prospective studies are needed to fully elucidate the association between COVID-19 and diabetes and to characterize persons at risk of developing diabetes after SARS-CoV-2 infection, identify those who should be screened for diabetes, and determine the natural histories of different forms of diabetes associated with COVID-19.
Collapse
Affiliation(s)
- Jordana Faruqi
- Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Ashok Balasubramanyam
- Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
140
|
Denorme F, Rustad JL, Portier I, Crandell JL, de Araujo CV, Cody MJ, Campbell RA, Yost CC. Neutrophil extracellular trap inhibition improves survival in neonatal mouse infectious peritonitis. Pediatr Res 2023; 93:862-869. [PMID: 35902703 PMCID: PMC9331023 DOI: 10.1038/s41390-022-02219-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/25/2022] [Accepted: 07/09/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Treatment of neonatal peritonitis and sepsis is challenging. Following infection, neutrophils elaborate neutrophil extracellular traps (NETs)-extracellular lattices of decondensed chromatin decorated with antimicrobial proteins. NETs, however, can augment pathogenic inflammation causing collateral damage. We hypothesized that NET inhibition would improve survival in experimental neonatal infectious peritonitis. METHODS We induced peritonitis in 7 to 10-day-old mice by intraperitoneal injection with cecal slurry. We targeted NETs by treating mice with neonatal NET-Inhibitory Factor (nNIF), an endogenous NET-inhibitor; Cl-amidine, a PAD4 inhibitor; DNase I, a NET degrading enzyme, or meropenem (an antibiotic). We determined peritoneal NET and cytokine levels and circulating platelet-neutrophil aggregates. Survival from peritonitis was followed for 6 days. RESULTS nNIF, Cl-amidine, and DNase I decreased peritoneal NET formation and inflammatory cytokine levels at 24 h compared to controls. nNIF, Cl-amidine, and DNase I decreased circulating platelet-neutrophil aggregates, and NET-targeting treatments significantly increased survival from infectious peritonitis compared to controls. Finally, nNIF administration significantly improved survival in mice treated with sub-optimal doses of meropenem even when treatment was delayed until 2 h after peritonitis induction. CONCLUSIONS NET inhibition improves survival in experimental neonatal infectious peritonitis, suggesting that NETs participate pathogenically in neonatal peritonitis and sepsis. IMPACT 1. Neutrophil extracellular trap formation participates pathogenically in experimental neonatal infectious peritonitis. 2. NET-targeting strategies improve outcomes in a translational model of neonatal infectious peritonitis. 3. NET inhibition represents a potential target for drug development in neonatal sepsis and infectious peritonitis.
Collapse
Affiliation(s)
| | - John L Rustad
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
| | - Irina Portier
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
| | | | - Claudia V de Araujo
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
- Department of Pediatrics/Neonatology, Salt Lake City, UT, 84112, USA
| | - Mark J Cody
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
- Department of Pediatrics/Neonatology, Salt Lake City, UT, 84112, USA
| | - Robert A Campbell
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Christian C Yost
- Molecular Medicine Program, Salt Lake City, UT, 84112, USA.
- Department of Pediatrics/Neonatology, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
141
|
Kapten K, Orczyk K, Smolewska E. Immunity in SARS-CoV-2 Infection: Clarity or Mystery? A Broader Perspective in the Third Year of a Worldwide Pandemic. Arch Immunol Ther Exp (Warsz) 2023; 71:7. [PMID: 36810662 PMCID: PMC9943048 DOI: 10.1007/s00005-023-00673-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/09/2023] [Indexed: 02/23/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its mechanisms have been thoroughly studied by researchers all over the world with the hope of finding answers that may aid the discovery of new treatment options or effective means of prevention. Still, over 2 years into the pandemic that is an immense burden on health care and economic systems, there seem to be more questions than answers. The character and multitude of immune responses elicited in coronavirus disease 2019 (COVID-19) vary from uncontrollable activation of the inflammatory system, causing extensive tissue damage and consequently leading to severe or even fatal disease, to mild or asymptomatic infections in the majority of patients, resulting in the unpredictability of the current pandemic. The aim of the study was to systematize the available data regarding the immune response to SARS-CoV-2, to provide some clarification among the abundance of the knowledge available. The review contains concise and current information on the most significant immune reactions to COVID-19, including components of both innate and adaptive immunity, with an additional focus on utilizing humoral and cellular responses as effective diagnostic tools. Moreover, the authors discussed the present state of knowledge on SARS-CoV-2 vaccines and their efficacy in cases of immunodeficiency.
Collapse
Affiliation(s)
- Katarzyna Kapten
- Department of Pediatric Cardiology and Rheumatology, Central Teaching Hospital of Medical University of Lodz, Lodz, Poland
| | - Krzysztof Orczyk
- Department of Pediatric Cardiology and Rheumatology, Medical University of Lodz, Sporna 36/50, 91-738, Lodz, Poland
| | - Elzbieta Smolewska
- Department of Pediatric Cardiology and Rheumatology, Medical University of Lodz, Sporna 36/50, 91-738, Lodz, Poland.
| |
Collapse
|
142
|
Kosidło JW, Wolszczak-Biedrzycka B, Matowicka-Karna J, Dymicka-Piekarska V, Dorf J. Clinical Significance and Diagnostic Utility of NLR, LMR, PLR and SII in the Course of COVID-19: A Literature Review. J Inflamm Res 2023; 16:539-562. [PMID: 36818192 PMCID: PMC9930576 DOI: 10.2147/jir.s395331] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/18/2023] [Indexed: 02/12/2023] Open
Abstract
Nowadays, society is increasingly struggling with infectious diseases that are characterized by severe course and even death. Recently, the whole world has faced the greatest epidemiological threat, which is COVID-19 caused by SARS CoV-2 virus. SARS CoV-2 infection is often accompanied by severe inflammation, which can lead to the development of different complications. Consequently, clinicians need easily interpreted and effective markers of inflammation that can predict the efficacy of the treatment and patient prognosis. Inflammation is associated with changes in many biochemical and hematological parameters, including leukocyte counts and their populations. In COVID-19, changes in leukocytes count populations such as neutrophils, lymphocytes or monocytes are observed. The numerous research confirm that indicators like neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), platelets-to-lymphocyte ratio (PLR) and systemic inflammatory index (SII) may prove effective in assessment patient prognosis and choosing optimal therapy. Therefore, in this review, we would like to summarize the latest knowledge about the diagnostic utility of systemic inflammatory ratios - NLR, LMR, PLR and SII in patients with COVID-19. We focused on the papers evaluating the diagnostic utility of inflammatory ratios using ROC curve published in the recent 3 years. Identification of biomarkers associated with inflammation would help the selection of patients with severe course of COVID-19 and high risk of death.
Collapse
Affiliation(s)
- Jakub Wiktor Kosidło
- Students’ Scientific Club at the Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Blanka Wolszczak-Biedrzycka
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland,Warmia and Mazury Oncology Center of the Hospital of the Ministry of the Interior and Administration, Olsztyn, Poland
| | - Joanna Matowicka-Karna
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | | | - Justyna Dorf
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland,Correspondence: Justyna Dorf, Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15a St., 15-269, Bialystok, Poland, Tel +48 85 8 31 87 16, Email
| |
Collapse
|
143
|
Artykbaeva GM, Saatov TS. Relationship between severe acute respiratory syndrome coronavirus 2 and diabetes mellitus (review). DIABETES MELLITUS 2023. [DOI: 10.14341/dm12900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Infections caused by SARE-CoV-2 are complicated with the concurrent pathologies, to name hypertension, diabetes mellitus and cardiovascular diseases. High level of glucose in blood weakens the immunity and increase the SARS-CoV-2 replication. Diabetes mellitus aggravates the COVID-19 outcome. The intrusion of SARS-CoV-2 into a host-cell occurs by means of its association with the angiotensin-converting enzyme-2 (ACE 2). Stimulating immune responses the COVID-19 infection causes the cytokine storm, and may result in the lethal outcome in the diabetics.Recent laboratory studies demonstrated that the type1 and type2 diabetes mellitus is the main consequence in 14% of the patients after corona infection. Thus, in 2% of 14% diabetes started progressing due to the corona virus. In the other, diabetes debut occurred as the direct and negative consequence of the disease. Hyperglycemia results in the formation of protein molecules known as the advanced glycation end products (AGEs). The AGEs and their receptors (RAGE) are of high significance in the host-cell’s virus invasion. Consequently, more strict glucose control is necessary for optimal outcome and reduction in mortality. The better control for the COVID-19 course can be provided by the targeted effect on the RAGE axis. The review helps elucidate the molecular mechanism underlying the exacerbation of pathophysiology in the diabetic COVID-19 patients.
Collapse
Affiliation(s)
- G. M. Artykbaeva
- Institute of biophysics and biochemistry, National University of Uzbekistan named after Mirzo Ulugbek
| | - T. S. Saatov
- Institute of biophysics and biochemistry, National University of Uzbekistan named after Mirzo Ulugbek
| |
Collapse
|
144
|
Chandok T, Nasr R, Uday KA. A Case of Antineutrophil Cytoplasmic Antibody Vasculitis-Associated Acute Kidney Injury in a Patient With Asymptomatic COVID-19 Infection. Cureus 2023; 15:e35006. [PMID: 36938153 PMCID: PMC10021030 DOI: 10.7759/cureus.35006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
Vasculitis, or inflammation of blood vessels, is commonly seen with severe acute respiratory syndrome Coronavirus disease 2 (SARS-CoV-2). It is usually triggered by an autoimmune response induced by the virus, infection by the virus itself and trauma to the epithelial vessels caused by the release of cytokines. We present a case of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (pauci-immune crescentic glomerulonephritis [GN]) superimposed on acute kidney injury caused by SARS-CoV-2. Our patient is a 57-year-old Hispanic female who presented with rising creatinine and active urinary sediment in the setting of an asymptomatic COVID-19 infection. A kidney biopsy was done for declining renal function, and positive myeloperoxidase antibodies revealed pauci-immune focal crescentic glomerulonephritis. Normalization of renal function was not achieved with pulse steroids and rituximab. The patient required long-term hemodialysis. Our case here adds to the very few cases of pauci-immune crescentic glomerulonephritis reported in patients with asymptomatic SARS-CoV-2 infection. We recommend keeping this high on the differential in SARS-CoV-2-infected patients presenting with acute kidney injury.
Collapse
Affiliation(s)
| | - Rabih Nasr
- Nephrology, Bronx Care Health System, Bronx, USA
| | | |
Collapse
|
145
|
Herrera VL, Bosch NA, Lok JJ, Nguyen MQ, Lenae KA, deKay JT, Ryzhov SV, Seder DB, Ruiz-Opazo N, Walkey AJ. Circulating neutrophil extracellular trap (NET)-forming 'rogue' neutrophil subset, immunotype [DEspR+CD11b+], mediate multi-organ failure in COVID-19 - an observational study. RESEARCH SQUARE 2023:rs.3.rs-2479844. [PMID: 36778407 PMCID: PMC9915800 DOI: 10.21203/rs.3.rs-2479844/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background: Cumulative research show association of neutrophils and neutrophil extracellular traps (NETs) with poor outcomes in severe COVID-19. However, to date, no curative intent therapy has been identified to block neutrophil/NETs-mediated progression of multi-organ dysfunction. Because of emerging neutrophil heterogeneity, the study of subsets of circulating neutrophil-extracellular trap (NET)-forming neutrophils [NET+Ns] as mediators of multi-organ failure progression among patients with COVID-19 is critical to identification of therapeutic targets. Methods: We conducted a prospective observational study of circulating levels of CD11b+[NET+N] immunotyped for dual endothelin-1/signal peptide receptor, (DEspR±) expression by quantitative immunofluorescence-cytology and causal mediation analysis. In 36 consented adults hospitalized with mod-severe COVID-19, May to September 2020, we measured acute multi-organ failure via SOFA-scores and respiratory failure via SaO2/FiO2 (SF)ratio at time points t1 (average 5.5 days from ICU/hospital admission) and t2 (the day before ICU-discharge or death), and ICU-free days at day28 (ICUFD). Circulating absolute neutrophil counts (ANC) and [NET+N] subset-specific counts were measured at t1. Spearman correlation and causal mediation analyses were conducted. Results: Spearman correlation analyses showed correlations of t1-SOFA with t2-SOFA ( rho r S =0.80) and ICUFD ( r S =-0.76); circulating DEspR+[NET+Ns] with t1-SOFA ( r S = 0.71), t2-SOFA ( r S =0.62), and ICUFD ( r S =-0.63), and ANC with t1-SOFA ( r S =0.71), and t2-SOFA ( r S =0.61). Causal mediation analysis identified DEspR+[NET+Ns] as mediator of 44.1% [95% CI:16.5,110.6] of the causal path between t1-SOFA (exposure) and t2-SOFA (outcome), with 46.9% [15.8,124.6] eliminated when DEspR+[NET+Ns] were theoretically reduced to zero. Concordantly, DEspR+[NET+Ns] mediated 47.1% [22.0,72.3%] of the t1-SOFA to ICUFD causal path, with 51.1% [22.8,80.4%] eliminated if DEspR+[NET+Ns] were reduced to zero. In patients with t1-SOFA >1, the indirect effect of a hypothetical treatment eliminating DEspR+[NET+Ns] projected a reduction of t2-SOFA by 0.98 [0.29,2.06] points and ICUFD by 3.0 [0.85,7.09] days. In contrast, there was no significant mediation of SF-ratio through DEspR+[NET+Ns], and no significant mediation of SOFA-score through ANC. Conclusions: Despite equivalent correlations, DEspR+[NET+Ns], but not ANC, mediated progression of multi-organ failure in acute COVID-19, and its hypothetical reduction is projected to improve ICUFD. These translational findings warrant further studies of DEspR+[NET+Ns] as potential patient-stratifier and actionable therapeutic target for multi-organ failure in COVID-19.
Collapse
Affiliation(s)
- Victoria L.M. Herrera
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine,Corresponding author:
| | - Nicholas A. Bosch
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine
| | - Judith J. Lok
- Department of Mathematics and Statistics, Boston University
| | - Mai Q. Nguyen
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine
| | - Kaitriona A. Lenae
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine
| | | | | | - David B. Seder
- Maine Health Institute for Research,Department of Critical Care Services, Maine Medical Center
| | - Nelson Ruiz-Opazo
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine
| | - Allan J. Walkey
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine
| |
Collapse
|
146
|
Laino ME, Ammirabile A, Motta F, De Santis M, Savevski V, Francone M, Chiti A, Mannelli L, Selmi C, Monti L. Advanced Imaging Supports the Mechanistic Role of Autoimmunity and Plaque Rupture in COVID-19 Heart Involvement. Clin Rev Allergy Immunol 2023; 64:75-89. [PMID: 35089505 PMCID: PMC8796606 DOI: 10.1007/s12016-022-08925-1] [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] [Accepted: 01/11/2022] [Indexed: 01/26/2023]
Abstract
The cardiovascular system is frequently affected by coronavirus disease-19 (COVID-19), particularly in hospitalized cases, and these manifestations are associated with a worse prognosis. Most commonly, heart involvement is represented by myocarditis, myocardial infarction, and pulmonary embolism, while arrhythmias, heart valve damage, and pericarditis are less frequent. While the clinical suspicion is necessary for a prompt disease recognition, imaging allows the early detection of cardiovascular complications in patients with COVID-19. The combination of cardiothoracic approaches has been proposed for advanced imaging techniques, i.e., CT scan and MRI, for a simultaneous evaluation of cardiovascular structures, pulmonary arteries, and lung parenchyma. Several mechanisms have been proposed to explain the cardiovascular injury, and among these, it is established that the host immune system is responsible for the aberrant response characterizing severe COVID-19 and inducing organ-specific injury. We illustrate novel evidence to support the hypothesis that molecular mimicry may be the immunological mechanism for myocarditis in COVID-19. The present article provides a comprehensive review of the available evidence of the immune mechanisms of the COVID-19 cardiovascular injury and the imaging tools to be used in the diagnostic workup. As some of these techniques cannot be implemented for general screening of all cases, we critically discuss the need to maximize the sustainability and the specificity of the proposed tests while illustrating the findings of some paradigmatic cases.
Collapse
Affiliation(s)
- Maria Elena Laino
- Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Angela Ammirabile
- Department of Radiology and Nuclear Medicine, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Francesca Motta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Maria De Santis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Victor Savevski
- Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marco Francone
- Department of Radiology and Nuclear Medicine, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Arturo Chiti
- Department of Radiology and Nuclear Medicine, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Lorenzo Monti
- Department of Radiology and Nuclear Medicine, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
147
|
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: 27] [Impact Index Per Article: 13.5] [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
|
148
|
Tang J, Xu Q, Tang K, Ye X, Cao Z, Zou M, Zeng J, Guan X, Han J, Wang Y, Yang L, Lin Y, Jiang K, Chen X, Zhao Y, Tian D, Li C, Shen W, Du X. Susceptibility identification for seasonal influenza A/H3N2 based on baseline blood transcriptome. Front Immunol 2023; 13:1048774. [PMID: 36713410 PMCID: PMC9878565 DOI: 10.3389/fimmu.2022.1048774] [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: 09/20/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Influenza susceptibility difference is a widely existing trait that has great practical significance for the accurate prevention and control of influenza. Methods Here, we focused on the human susceptibility to the seasonal influenza A/H3N2 of healthy adults at baseline level. Whole blood expression data for influenza A/H3N2 susceptibility from GEO were collected firstly (30 symptomatic and 19 asymptomatic). Then to explore the differences at baseline, a suite of systems biology approaches - the differential expression analysis, co-expression network analysis, and immune cell frequencies analysis were utilized. Results We found the baseline condition, especially immune condition between symptomatic and asymptomatic, was different. Co-expression module that is positively related to asymptomatic is also related to immune cell type of naïve B cell. Function enrichment analysis showed significantly correlation with "B cell receptor signaling pathway", "immune response-activating cell surface receptor signaling pathway" and so on. Also, modules that are positively related to symptomatic are also correlated to immune cell type of neutrophils, with function enrichment analysis showing significantly correlations with "response to bacterium", "inflammatory response", "cAMP-dependent protein kinase complex" and so on. Responses of symptomatic and asymptomatic hosts after virus exposure show differences on resisting the virus, with more effective frontline defense for asymptomatic hosts. A prediction model was also built based on only baseline transcription information to differentiate symptomatic and asymptomatic population with accuracy of 0.79. Discussion The results not only improve our understanding of the immune system and influenza susceptibility, but also provide a new direction for precise and targeted prevention and therapy of influenza.
Collapse
Affiliation(s)
- Jing Tang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Qiumei Xu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Kang Tang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Ye
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zicheng Cao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,School of Public Health, Shantou University, Shantou, China
| | - Min Zou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Jinfeng Zeng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Xinyan Guan
- Department of Chronic Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, China
| | - Jinglin Han
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Yihan Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Lan Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yishan Lin
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Kaiao Jiang
- Palos Verdes Peninsula High School, Rancho Palos Verdes, CA, United States
| | - Xiaoliang Chen
- Department of Chronic Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, China
| | - Yang Zhao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Dechao Tian
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Chunwei Li
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Shen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,*Correspondence: Xiangjun Du, ; Wei Shen,
| | - Xiangjun Du
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China,*Correspondence: Xiangjun Du, ; Wei Shen,
| |
Collapse
|
149
|
De Michele M, Lorenzano S, Piscopo P, Rivabene R, Crestini A, Chistolini A, Stefanini L, Pulcinelli FM, Berto I, Campagna R, Amisano P, Iacobucci M, Cirelli C, Falcou A, Nicolini E, Schiavo OG, Toni D. SARS-CoV-2 infection predicts larger infarct volume in patients with acute ischemic stroke. Front Cardiovasc Med 2023; 9:1097229. [PMID: 36704480 PMCID: PMC9871539 DOI: 10.3389/fcvm.2022.1097229] [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: 11/13/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Background and purpose Acute ischemic stroke (AIS) is a fearful complication of Coronavirus Disease-2019 (COVID-19). Aims of this study were to compare clinical/radiological characteristics, endothelial and coagulation dysfunction between acute ischemic stroke (AIS) patients with and without COVID-19 and to investigate if and how the SARS-CoV-2 spike protein (SP) was implicated in triggering platelet activation. Methods We enrolled AIS patients with COVID-19 within 12 h from onset and compared them with an age- and sex-matched cohort of AIS controls without COVID-19. Neuroimaging studies were performed within 24 h. Blood samples were collected in a subset of 10 patients. Results Of 39 AIS patients, 22 had COVID-19 and 17 did not. Admission levels of Factor VIII and von Willebrand factor antigen were significantly higher in COVID-19 patients and positively correlated with the infarct volume. In multivariate linear regression analyses, COVID-19 was an independent predictor of infarct volume (B 20.318, Beta 0.576, 95%CI 6.077-34.559; p = 0.011). SP was found in serum of 2 of the 10 examined COVID-19 patients. Platelets from healthy donors showed a similar degree of procoagulant activation induced by COVID-19 and non-COVID-19 patients' sera. The anti-SP and anti-FcγRIIA blocking antibodies had no effect in modulating platelet activity in both groups. Conclusions SARS-CoV-2 infection seems to play a major role in endothelium activation and infarct volume extension during AIS.
Collapse
Affiliation(s)
- Manuela De Michele
- Emergency Department, Stroke Unit, Sapienza University of Rome, Rome, Italy
| | - Svetlana Lorenzano
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Roberto Rivabene
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Alessio Crestini
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio Chistolini
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio M. Pulcinelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Irene Berto
- Emergency Department, Stroke Unit, Sapienza University of Rome, Rome, Italy
| | - Roberta Campagna
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Paolo Amisano
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Marta Iacobucci
- Neuroradiology Unit, Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Carlo Cirelli
- Neuroradiology Unit, Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Anne Falcou
- Emergency Department, Stroke Unit, Sapienza University of Rome, Rome, Italy
| | - Ettore Nicolini
- Emergency Department, Stroke Unit, Sapienza University of Rome, Rome, Italy
| | - Oscar G. Schiavo
- Emergency Department, Stroke Unit, Sapienza University of Rome, Rome, Italy
| | - Danilo Toni
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
150
|
Gong HH, Worley MJ, Carver KA, Goldstein DR, Deng JC. Neutrophils drive pulmonary vascular leakage in MHV-1 infection of susceptible A/J mice. Front Immunol 2023; 13:1089064. [PMID: 36685578 PMCID: PMC9853883 DOI: 10.3389/fimmu.2022.1089064] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
Background Lung inflammation, neutrophil infiltration, and pulmonary vascular leakage are pathological hallmarks of acute respiratory distress syndrome (ARDS) which can lethally complicate respiratory viral infections. Despite similar comorbidities, however, infections in some patients may be asymptomatic while others develop ARDS as seen with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections for example. Methods In this study, we infected resistant C57BL/6 and susceptible A/J strains of mice with pulmonary administration of murine hepatitis virus strain 1 (MHV-1) to determine mechanisms underlying susceptibility to pulmonary vascular leakage in a respiratory coronavirus infection model. Results A/J animals displayed increased lung injury parameters, pulmonary neutrophil influx, and deficient recruitment of other leukocytes early in the infection. Moreover, under basal conditions, A/J neutrophils overexpressed primary granule protein genes for myeloperoxidase and multiple serine proteases. During infection, myeloperoxidase and elastase protein were released in the bronchoalveolar spaces at higher concentrations compared to C57BL/6 mice. In contrast, genes from other granule types were not differentially expressed between these 2 strains. We found that depletion of neutrophils led to mitigation of lung injury in infected A/J mice while having no effect in the C57BL/6 mice, demonstrating that an altered neutrophil phenotype and recruitment profile is a major driver of lung immunopathology in susceptible mice. Conclusions These results suggest that host susceptibility to pulmonary coronaviral infections may be governed in part by underlying differences in neutrophil phenotypes, which can vary between mice strains, through mechanisms involving primary granule proteins as mediators of neutrophil-driven lung injury.
Collapse
Affiliation(s)
- Henry H. Gong
- University of Michigan, Ann Arbor, MI, United States
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Matthew J. Worley
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Kyle A. Carver
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Daniel R. Goldstein
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Jane C. Deng
- University of Michigan, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Medicine Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
| |
Collapse
|