101
|
Sudden Hypotension and Increased Serum Interferon-γ and Interleukin-10 Predict Early Macrophage Activation Syndrome in Patients with Systemic Juvenile Idiopathic Arthritis. J Pediatr 2021; 235:203-211.e3. [PMID: 33581106 DOI: 10.1016/j.jpeds.2021.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To identify clinical and laboratory predictors for early macrophage activation syndrome (MAS) associated with systemic juvenile idiopathic arthritis (sJIA). STUDY DESIGN This is a retrospective cohort study of 149 patients with sJIA, of whom 27 had 31 episodes of MAS. We evaluated the clinical and laboratory features of patients with sJIA and MAS and compared them with those without MAS. We focused our analysis on the overall process of MAS development, especially MAS onset. RESULTS As shown in previous studies, we found a high percentage of fever, absence of arthritis, and central nervous system dysfunction at MAS onset in our study cohort. We also found that 35% of patients with MAS had hypotension although not shock, and 22.6% of patients with MAS had gastrointestinal involvement at MAS onset. Compared with patients with MAS without hypotension, patients with MAS and hypotension had greater rates of admission to the intensive care unit; presented with more arthritis, serositis, pneumonia, and gastrointestinal involvement; and had greater white blood cell and absolute neutrophil counts and serum bilirubin levels and lower serum total protein. We confirmed laboratory markers such as platelet counts, lactate dehydrogenase, and aspartate aminotransferase can help to identify early MAS and that ferritin/erythrocyte sedimentation rate ratio of approximately 20.0 had a high diagnostic sensitivity and specificity for MAS. In addition, we discovered that the combination of interferon-γ >17.1 pg/mL and interleukin-10 >7.8 pg/mL appeared to be a good cytokine pattern for the recognition of MAS onset. CONCLUSIONS Sudden hypotension, elevated ferritin/erythrocyte sedimentation rate ratio, and the cytokine pattern of significantly increased interferon-γ and interleukin-10 levels are important markers for early identification of MAS in addition to the traditional characteristics of sJIA-associated MAS.
Collapse
|
102
|
Gao DK, Salomonis N, Henderlight M, Woods C, Thakkar K, Grom AA, Thornton S, Jordan MB, Wikenheiser-Brokamp KA, Schulert GS. IFN-γ is essential for alveolar macrophage driven pulmonary inflammation in macrophage activation syndrome. JCI Insight 2021; 6:e147593. [PMID: 34314387 PMCID: PMC8492332 DOI: 10.1172/jci.insight.147593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Macrophage activation syndrome (MAS) is a life-threatening cytokine storm complicating systemic juvenile idiopathic arthritis (SJIA) driven by IFNγ. SJIA and MAS are associated with an unexplained emerging lung disease (SJIA-LD), with our recent work supporting pulmonary activation of IFNγ pathways pathologically linking SJIA-LD and MAS. Our objective was to mechanistically define the novel observation of pulmonary inflammation in the TLR9 mouse model of MAS. In acute MAS, lungs exhibit mild but diffuse CD4-predominant, perivascular interstitial inflammation with elevated IFNγ, IFN-induced chemokines, and AMΦ expression of IFNγ-induced genes. Single-cell RNA-sequencing confirmed IFN-driven transcriptional changes across lung cell types with myeloid expansion and detection of MAS-specific macrophage populations. Systemic MAS resolution was associated with increased AMΦ and interstitial lymphocytic infiltration. AMΦ transcriptomic analysis confirmed IFNγ-induced proinflammatory polarization during acute MAS, which switches towards an anti-inflammatory phenotype after systemic MAS resolution. Interestingly, recurrent MAS led to increased alveolar inflammation and lung injury, and reset AMΦ polarization towards a proinflammatory state. Furthermore, in mice bearing macrophages insensitive to IFNγ, both systemic feature of MAS and pulmonary inflammation were attenuated. These findings demonstrate that experimental MAS induces IFNγ-driven pulmonary inflammation replicating key features of SJIA-LD, and provides a model system for testing novel treatments directed towards SJIA-LD.
Collapse
Affiliation(s)
- Denny K Gao
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Maggie Henderlight
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Christopher Woods
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Kairavee Thakkar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Alexei A Grom
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Sherry Thornton
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Michael B Jordan
- Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | | | - Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| |
Collapse
|
103
|
Knoll R, Schultze JL, Schulte-Schrepping J. Monocytes and Macrophages in COVID-19. Front Immunol 2021; 12:720109. [PMID: 34367190 PMCID: PMC8335157 DOI: 10.3389/fimmu.2021.720109] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a contagious viral disease caused by SARS-CoV-2 that led to an ongoing pandemic with massive global health and socioeconomic consequences. The disease is characterized primarily, but not exclusively, by respiratory clinical manifestations ranging from mild common cold symptoms, including cough and fever, to severe respiratory distress and multi-organ failure. Macrophages, a heterogeneous group of yolk-sac derived, tissue-resident mononuclear phagocytes of complex ontogeny present in all mammalian organs, play critical roles in developmental, homeostatic and host defense processes with tissue-dependent plasticity. In case of infection, they are responsible for early pathogen recognition, initiation and resolution of inflammation, as well as repair of tissue damage. Monocytes, bone-marrow derived blood-resident phagocytes, are recruited under pathological conditions such as viral infections to the affected tissue to defend the organism against invading pathogens and to aid in efficient resolution of inflammation. Given their pivotal function in host defense and the potential danger posed by their dysregulated hyperinflammation, understanding monocyte and macrophage phenotypes in COVID-19 is key for tackling the disease's pathological mechanisms. Here, we outline current knowledge on monocytes and macrophages in homeostasis and viral infections and summarize concepts and key findings on their role in COVID-19. While monocytes in the blood of patients with moderate COVID-19 present with an inflammatory, interferon-stimulated gene (ISG)-driven phenotype, cellular dysfunction epitomized by loss of HLA-DR expression and induction of S100 alarmin expression is their dominant feature in severe disease. Pulmonary macrophages in COVID-19 derived from infiltrating inflammatory monocytes are in a hyperactivated state resulting in a detrimental loop of pro-inflammatory cytokine release and recruitment of cytotoxic effector cells thereby exacerbating tissue damage at the site of infection.
Collapse
Affiliation(s)
- Rainer Knoll
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L. Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
| | - Jonas Schulte-Schrepping
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
104
|
Tavares J, Silva F, Castro A, Santos J, Rocha G, Almeida M, Martins LS, Dias L, Henriques AC, Cabrita A. Diagnostic challenge of recurrent macrophage activation syndrome before and after kidney transplant. Nefrologia 2021; 41:482-485. [PMID: 36165122 DOI: 10.1016/j.nefroe.2020.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/25/2020] [Indexed: 06/16/2023] Open
Affiliation(s)
- Joana Tavares
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal.
| | - Filipa Silva
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Ana Castro
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Josefina Santos
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Guilherme Rocha
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Manuela Almeida
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - La Salete Martins
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Leonídio Dias
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | | | - António Cabrita
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| |
Collapse
|
105
|
Schulert GS. The IL-18/IFNγ axis in systemic JIA and MAS-new answers, more questions. Rheumatology (Oxford) 2021; 60:3045-3047. [PMID: 33839745 DOI: 10.1093/rheumatology/keab342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center.,Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
106
|
Griffin GD. Does Covera-19 know 'when to hold 'em or 'when to fold 'em? A translational thought experiment. TRANSLATIONAL MEDICINE COMMUNICATIONS 2021; 6:12. [PMID: 34226878 PMCID: PMC8243045 DOI: 10.1186/s41231-021-00090-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/19/2021] [Indexed: 05/09/2023]
Abstract
The function of proteins depends on their structure. The structural integrity of proteins is dynamic and depends on interacting nearby neighboring moieties that influence their properties and induce folding and structural changes. The conformational changes induced by these nearby neighbors in the micro-environmental milieu at that moment are guided by chemical or electrical bonding attractions. There are few literature references that describe the potential for environmental milieu changes to disfavor SARS-CoV-2 attachment to a receptor for survival outside of a host. There are many studies on the effects of pH (acid and base balance) supporting its importance for protein structure and function, but few focus on pH role in extracellular or intracellular protein or actionable requirements of Covera-19. 'Fold 'em or Hold 'em' is seen by the various functions and effects of furin as it seeks an acidic milieu for action or compatible amino acid sequences which is currently aided by its histidine component and the structural changes of proteins as they enter or exit the host. Questions throughout the text are posed to focus on current thoughts as reviewing applicable COVID-19 translational research science in order to understand the complexities of Covid-19. The pH needs of COVID-19 players and its journey through the human host and environment as well as some efficacious readily available repurposed drugs and out-of-the box and easily available treatments are reviewed.
Collapse
Affiliation(s)
- Gerald Dieter Griffin
- Adjunct Faculty, School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA USA
- Adjunct Faculty, School of Pharmacy & Health Sciences, The University of the Pacific, 123 Forest Ave, Pacific Grove, CA 93950 USA
| |
Collapse
|
107
|
Inflammatory biomarkers in pregnant women with COVID-19: a retrospective cohort study. Sci Rep 2021; 11:13350. [PMID: 34172816 PMCID: PMC8233302 DOI: 10.1038/s41598-021-92885-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 06/11/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic viral disease affecting also obstetric patients and uncertainties exist about the prognostic role of inflammatory biomarkers and hemocytometry values in patients with this infection. To clarify that, we have assessed the values of several inflammatory biomarkers and hemocytometry variables in a cohort of obstetric patients hospitalized with COVID-19 and we have correlated the values at admission with the need of oxygen supplementation during the hospitalization. Overall, among 62 (27.3%) pregnant women and 165 (72.7%) postpartum women, 21 (9.2%) patients received oxygen supplementation and 2 (0.9%) required admission to intensive care unit but none died. During hospitalization leukocytes (p < 0.001), neutrophils (p < 0.001), neutrophils to lymphocytes ratio (p < 0.001) and C reactive protein (p < 0.001) decreased significantly, whereas lymphocytes (p < 0.001), platelets (p < 0.001) and ferritin (p = 0.001) increased. Lymphocyte values at admission were correlated with oxygen need, with a 26% higher risk of oxygen supplementation for each 1000 cells decreases. Overall, in obstetric patients hospitalized with COVID-19, C reactive protein is the inflammatory biomarker that better mirrors the course of the disease whereas D-dimer or ferritin are not reliable predictors of poor outcome. Care to the need of oxygen supplementation should be reserved to patients with reduced lymphocyte values at admission.
Collapse
|
108
|
Ahmad N, Parmar A, Kitchen J. A case of macrophage activation syndrome in a patient with anti-synthetase syndrome. Oxf Med Case Reports 2021; 2021:omab045. [PMID: 34158959 PMCID: PMC8212681 DOI: 10.1093/omcr/omab045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/22/2020] [Accepted: 04/11/2021] [Indexed: 11/29/2022] Open
Abstract
Anti-synthetase syndrome (ASS) is an autoimmune disease characterized by autoantibodies against an aminoacyl transfer RNA synthetase with clinical features including interstitial lung disease, non-erosive arthritis, myositis, Raynaud’s phenomenon, unexplained fever and/or mechanic’s hands. Macrophage activation syndrome (MAS) is a potentially fatal hyper- inflammatory syndrome that can occur as a complication of systemic rheumatic diseases. However, the association of MAS and ASS has rarely been reported in the literature. Here, we report this association in a patient with overlap ASS and anti-CCP positive rheumatoid arthritis. First line management with steroids was complicated by diabetic ketoacidosis, hence requiring use of anti-IL1 therapy (anakinra) for disease control.
Collapse
Affiliation(s)
- Nadia Ahmad
- Rheumatology, Royal Berkshire NHS Foundation Trust, Reading RG1 5AN, UK
| | - Aneel Parmar
- Rheumatology, Royal Berkshire NHS Foundation Trust, Reading RG1 5AN, UK
| | - Joanne Kitchen
- Rheumatology, Royal Berkshire NHS Foundation Trust, Reading RG1 5AN, UK
| |
Collapse
|
109
|
Sarkisian DK, Chebotareva NV, McDonnell V, Oganesyan AV, Krasnova TN, Makarov EA. Risk Factors For Kidney Damage In COVID-19 Patients Admitted To The Intensive Care Unit. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background — Acute kidney injury (AKI) reaches 29% in the intensive care unit (ICU). Our study aimed to determine the prevalence, features, and the main AKI factors in critically ill patients with coronavirus disease 2019 (COVID-19). Material and Methods — The study included 37 patients with COVID-19. We analyzed the total blood count test results, biochemical profile panel, coagulation tests, and urine samples. We finally estimated the markers of kidney damage and mortality. Result — All patients in ICU had proteinuria, and 80.5% of patients had hematuria. AKI was observed in 45.9% of patients. Independent risk factors were age more than 60 years, increased C-reactive protein (CRP) level, and decreased platelet count. Conclusion — Kidney damage was observed in most critically ill patients with COVID-19. The independent risk factors for AKI in critically ill patients were elderly age, a cytokine response with a high CRP level.
Collapse
|
110
|
Huang LW, Wei JCC, Chen DY, Chen YJ, Tang KT, Ko TM, Chen HH. Bidirectional association between systemic lupus erythematosus and macrophage activation syndrome: a nationwide population-based study. Rheumatology (Oxford) 2021; 61:1123-1132. [PMID: 34146089 DOI: 10.1093/rheumatology/keab502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/09/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES To determine the bidirectional relationship between macrophage activation syndrome (MAS) and systemic lupus erythematosus (SLE). METHODS Using the 1997-2013 Taiwan's National Health Insurance Research Database, we identified patients with newly diagnosed SLE from 2001 to 2013 from the whole beneficiaries and selected individuals without SLE from a one-million representative population. Propensity score (PS) matching was performed to balance incident SLE patients and individuals without SLE according to age, sex, comorbidities and medical utilization. The association between a history of MAS and SLE was studied using conditional logistic regression analysis shown as adjusted odds ratio (aOR). The risk of MAS associated with SLE was analyzed using Cox proportional regression analysis shown as hazard ratio (aHR), and we conducted a sensitivity analysis using various definitions of MAS. RESULTS We finally included 10,481 SLE patients and 20,962 PS-matched (1:2) non-SLE individuals. The correlation between a history of MAS and SLE did not reach statistical significance after adjustment for potential confounders (aOR, 1.18; 95% confidence interval, 0.80-1.75) in the age/sex-matched populations. In the 1:2 PS-matched populations, the risk of MAS markedly increased in patients with SLE (aHR, 7.18; 95% confidence interval, 4.97-10.36). Other risk factors for MAS included female, age ≥ 65 years, low income, a history of inflammatory bowel disease, and MAS history. CONCLUSION This nationwide, population-based study revealed that a history of MAS was not significantly associated with SLE risk. However, the risk of MAS was markedly associated with SLE and history of MAS.
Collapse
Affiliation(s)
- Lu-Wei Huang
- Department of Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - James Cheng-Chung Wei
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, China Medical University, Taichung, Taiwan.,Translational Medicine Laboratory, Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan
| | - Yen-Ju Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kuo-Tung Tang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tai-Ming Ko
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University (NYCU), Tainan, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsin-Hua Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Biomedical Science and Rong-Hsing Research Center for Translational Medicine, Chung-Hsing University, Taichung, Taiwan
| |
Collapse
|
111
|
COVID-19 and Malignancy: Exploration of the possible genetic and epigenetic interlinks and overview of the vaccination scenario. Cancer Treat Res Commun 2021; 28:100425. [PMID: 34171559 PMCID: PMC8213508 DOI: 10.1016/j.ctarc.2021.100425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Malignancy is one of the prime global causes of mortality. Cancer Patients suffering from SARS-CoV-2 have demonstrated higher rates of severe complications exacerbating towards death. Possible genetic and epigenetic alterations may exist in cancer patients which have the potential to contribute towards their increased vulnerability towards COVID-19. METHOD An exhaustive literature search using 'COVID-19', 'SARS-CoV-2', 'Cancer', 'Malignancy', 'Relationships', Interlinks', 'Genetic', 'Epigenetic', 'Epidemiological studies', 'Clinical Studies', 'Vaccination', 'Vaccine scenario' were conducted in PubMed and EMBASE till 2nd June 2021. RESULT In this narrative review, 17 epidemiological studies were listed which focused on clinical parameters of several malignancy patient cohorts who contracted COVID-19. Besides, genetic and epigenetic alterations seen among cancer patients are also discussed which may plausibly increase the vulnerability of cancer patients to SARS-CoV-2 infection. Also, global vaccination scenario among malignant patients along with the necessity to prioritize them in the vaccination campaigns are also elaborated. CONCLUSION Genetic and epigenetic modifications present in ACE2, TMPRSS2, IL-6 and several cytokines require more in-depth research to elucidate the shared mechanisms of malignancy and SARS-CoV-2.
Collapse
|
112
|
Lipopolysaccharide stimulation test on cultured PBMCs assists the discrimination of cryopyrin-associated periodic syndrome from systemic juvenile idiopathic arthritis. Sci Rep 2021; 11:11903. [PMID: 34099791 PMCID: PMC8185076 DOI: 10.1038/s41598-021-91354-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Systemic juvenile idiopathic arthritis (sJIA) and cryopyrin-associated periodic syndrome (CAPS) share many common manifestations. We aim to identify an applicable method to assist disease discrimination. Inflammatory cytokines were measured in the plasma of patients with CAPS, sJIA with persistent disease course and healthy controls. Supernatants collected from non-stimulated peripheral blood mononuclear cells (PBMCs) and those undergone inflammasome stimulation tests utilizing lipopolysaccharide (LPS) with and without adenosine triphosphate (ATP) were investigated. Inflammatory cytokines in patient plasma fail to differentiate sJIA from CAPS. PBMCs from sJIA secrets higher amount of IL-1β and IL-18 while CAPS PBMCs produces more caspase-1 without stimulation. IL-1β, IL-18, and caspase-1 were significantly elevated among CAPS PBMCs (all p < 0.05) upon LPS stimulation, but not when additional ATPs were provided. Levels of cytokines and PBMC responses to the stimulation assays were similar among all sJIA patients regardless of their history of macrophage activation syndrome. Unstimulated PBMC activities and the LPS inflammasome stimulation assay without exogenic ATPs can assist the differentiation of CAPS from sJIA with persistent disease course.
Collapse
|
113
|
Ombrello MJ, Schulert GS. COVID-19 and cytokine storm syndrome: are there lessons from macrophage activation syndrome? Transl Res 2021; 232:1-12. [PMID: 33684592 PMCID: PMC7934701 DOI: 10.1016/j.trsl.2021.03.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022]
Abstract
Although interest in "cytokine storms" has surged over the past decade, it was massively amplified in 2020 when it was suggested that a subset of patients with COVID-19 developed a form of cytokine storm. The concept of cytokine storm syndromes (CSS) encompasses diverse conditions or circumstances that coalesce around potentially lethal hyperinflammation with hemodynamic compromise and multiple organ dysfunction syndrome. Macrophage activation syndrome (MAS) is a prototypic form of CSS that develops in the context of rheumatic diseases, particularly systemic juvenile idiopathic arthritis. The treatment of MAS relies heavily upon corticosteroids and cytokine inhibitors, which have proven to be lifesaving therapies in MAS, as well as in other forms of CSS. Within months of the recognition of SARS-CoV2 as a human pathogen, descriptions of COVID-19 patients with hyperinflammation emerged. Physicians immediately grappled with identifying optimal therapeutic strategies for these patients, and despite clinical distinctions such as marked coagulopathy with endothelial injury associated with COVID-19, borrowed from the experiences with MAS and other CSS. Initial reports of patients treated with anti-cytokine agents in COVID-19 were promising, but recent large, better-controlled studies of these agents have had mixed results suggesting a more complex pathophysiology. Here, we discuss how the comparison of clinical features, immunologic parameters and therapeutic response data between MAS and hyperinflammation in COVID-19 can provide new insight into the pathophysiology of CSS.
Collapse
Affiliation(s)
- Michael J Ombrello
- Translational Genetics and Genomics Unit, Pediatric Translational Research Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland.
| | - Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
114
|
Oh EJ, Yoon JH, Park KH, Bae HJ, Yun SJ, Min GJ, Park SS, Park S, Lee SE, Cho BS, Eom KS, Kim YJ, Lee S, Kim HJ, Min CK, Cho SG, Han K, Lee JW. Natural-killer cell cytotoxicity as a diagnostic and prognostic marker for adult patients with secondary hemophagocytic lymphohistiocytosis: a prospective phase II observational study. Ther Adv Hematol 2021; 12:20406207211020544. [PMID: 34104375 PMCID: PMC8172956 DOI: 10.1177/20406207211020544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/04/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Hemophagocytic lymphohistiocytosis (HLH) can be life-threatening if not detected and treated appropriately. The diagnosis of HLH can be confusing due to other similar febrile diseases that present with cytopenia. Natural-killer cell (NK)-cytotoxicity is an important diagnostic parameter for primary HLH; however, its role in secondary HLH in adults has not been well-elucidated. METHODS We prospectively enrolled 123 adult patients with febrile conditions accompanied by cytopenia or marrow hemophagocytosis. A diagnosis of HLH was based on HLH-2004 criteria and treated based on HLH-94 protocol. NK-cytotoxicity was calculated at the time of diagnosis by K562-cell direct lysis using flow-cytometry. RESULTS HLH (n = 60) was determined to be caused by Epstein-Barr virus (EBV) (n = 11), infection other than EBV (n = 16), malignancies (n = 19), and unknown (n = 14). Febrile diseases other than HLH (n = 63) were diagnosed as autoimmune disease (n = 22), malignancies (n = 21), infection (n = 12), non-malignant hematological diseases (n = 6), and unknown (n = 2). A lower NK-cytotoxicity level was observed at diagnosis in patients with HLH, compared with other causes of febrile disease (12.1% versus 26.2%, p < 0.001). However, NK-cytotoxicity had a borderline effect on diagnosis of HLH, with an area under receiver operation characteristic curve of 0.689. It also showed no significant role for the prediction of survival outcome. Multivariate analysis revealed that malignant disease and high ferritin level were related with poor survival outcome. In non-malignant disease subgroups, old age, EBV-association, and low NK-cytotoxicity were related with poor survival. CONCLUSIONS Febrile disease with cytopenia was associated with decreased NK-cytotoxicity, especially in adults with HLH; however, its diagnostic role for adult HLH is still arguable. The diagnostic criteria for adult HLH should be further discussed. TRIAL REGISTRATION Clinical Research Information Service [Internet]; Osong (Chungcheongbuk-do), Korea, Centers for Disease Control and Prevention, Ministry of Health and Welfare (Republic of Korea); https://cris.nih.go.kr/cris/index.jsp; Feb, 16th 2016; KCT0001886 (KC15TISE0936).
Collapse
Affiliation(s)
- Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae-Ho Yoon
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Ki Hyun Park
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun Joo Bae
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - So Jeong Yun
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gi June Min
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Soo Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Silvia Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung-Sik Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki-Seong Eom
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoo-Jin Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang-Ki Min
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok-Goo Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyungja Han
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Wook Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
115
|
Feng Y, Dong H, Sun B, Hu Y, Yang Y, Jia Y, Jia L, Zhong X, Zhao R. METTL3/METTL14 Transactivation and m 6A-Dependent TGF-β1 Translation in Activated Kupffer Cells. Cell Mol Gastroenterol Hepatol 2021; 12:839-856. [PMID: 33992834 PMCID: PMC8340128 DOI: 10.1016/j.jcmgh.2021.05.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Transforming growth factor β1 (TGF-β1) secreted from activated Kupffer cells (KC) promotes the progression of nonalcoholic steatohepatitis (NASH) to liver fibrosis. N6-methyladenosine (m6A) RNA modification participates in various cell stress responses, yet it remains unknown whether it plays a role in TGF-β1 upregulation in activated KCs. METHODS Western blot, dot blot, and liquid chromatography with tandem mass spectrometry were used to determine the expression of m6A methyltransferase, METTL3, and METTL14, as well as global m6A modification. RNA-sequencing and m6A-seq were employed to screen differentially expressed genes and responsive m6A peaks. Nuclear factor κB (NF-κB)-mediated METTL3/METTL14 transactivation were validated with chromatin immunoprecipitation polymerase chain reaction and dual-luciferase reporter system, and the role of m6A in TGF-β1 upregulation was further verified in METTL3/METTL14-deficient KCs and myeloid lineage cell-specific METTL14 knockout mice. RESULTS Serum lipopolysaccharide (LPS) concentration is increased in high-fat diet-induced NASH rats. TGF-β1 upregulation is closely associated with METTL3/METTL14 upregulation and global m6A hypermethylation, in both NASH rat liver and LPS-activated KCs. LPS-responsive m6A peaks are identified on the 5' untranslated region (UTR) of TGF-β1 messenger RNA (mRNA). NF-κB directly transactivates METTL3 and METTL14 genes. LPS-stimulated TGF-β1 expression is abolished in METTL3/METTL14-deficient KCs and myeloid lineage cell-specific METTL14 knockout mice. Mutation of m6A sites on the 5'UTR of TGF-β1 mRNA blocks LPS-induced increase of luciferase reporter activity. CONCLUSIONS NF-κB acts as transcription factor to transactivate METTL3/METTL14 genes upon LPS challenge, leading to global RNA m6A hypermethylation. Increased m6A on the 5'UTR of TGF-β1 mRNA results in m6A-dependent translation of TGF-β1 mRNA in a cap-independent manner. We identify a novel role of m6A modification in TGF-β1 upregulation, which helps to shed light on the molecular mechanism of NASH progression.
Collapse
Affiliation(s)
- Yue Feng
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China; Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Haibo Dong
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China; Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Bo Sun
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China; Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Yun Hu
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China; Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Yang Yang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China; Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China; Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Longfei Jia
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China; Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China.
| |
Collapse
|
116
|
Lu M, Xie Y, Guan X, Wang M, Zhu L, Zhang S, Ning Q, Han M. Clinical analysis and a novel risk predictive nomogram for 155 adult patients with hemophagocytic lymphohistiocytosis. Ann Hematol 2021; 100:2181-2193. [PMID: 33977332 DOI: 10.1007/s00277-021-04551-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/02/2021] [Indexed: 12/16/2022]
Abstract
Recently, more and more attention has been paid on adult hemophagocytic lymphohistiocytosis (HLH), a disease with complicated symptoms and high mortality. In order to analyze the clinical characteristics and prediction risk factors of mortality, we designed a retrospective study with 1-year follow-up and included 155 patients admitted to Tongji Hospital diagnosed as HLH. One hundred seven patients formed the training cohort for nomogram development, and 48 patients formed the validation cohort to confirm the model's performance. All patients' clinical characteristics, laboratory results, medical records, and prognosis were analyzed. Among all the 107 patients in the training cohort, 46 were male and 61 were female, with the median age of 49.0 (IQR 31.0-63.0). The 1-year mortality rate was 43.9% (47/107) and 45.8% (22/48) in the training and validation cohort, respectively. And further multivariate logistic regression analysis in the training cohort showed that male (odds ratio 5.534, 95% CI 1.507-20.318, p = 0.010), altered mental status (11.876, 1.882-74.947, p = 0.008), serum ferritin ≥ 31,381 μg/L (8.273, 1.855-36.883, p = 0.006), and IL-6 ≥ 18.59 pg/mL (19.446, 1.527-247.642, p = 0.022) were independent risk factor of mortality. A nomogram included the four prediction factors mentioned above was also tabled to help clinicians evaluate the probability of poor outcome. Area under the receiver operating characteristic curve (AUROC) analysis, calibration curves, and decision curve analysis (DCA) certify the accuracy and the clinical usefulness of the nomogram. Our research reveals that male, altered mental status, serum ferritin ≥ 31,381 µg/L, and IL-6 ≥ 18.59 pg/mL are four independent predictors for poor prognosis. Doctors should pay more attention to patients with altered mental status, high serum ferritin, and IL-6 level, who have a higher risk of death.
Collapse
Affiliation(s)
- Mengxin Lu
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanghao Xie
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoxu Guan
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Wang
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin Zhu
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shen Zhang
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin Ning
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Meifang Han
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
117
|
Iwata J, Yamamura H, Takano A, Horiuchi Y. A case of corticosteroid‐refractory adult‐onset Still's disease successfully controlled with tocilizumab despite transient neutropenia and thrombocytopenia. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2021. [DOI: 10.1002/cia2.12168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Jun‐ichi Iwata
- Department of Dermatology International University of Health and Welfare Atami Hospital Atami Japan
| | - Hitomi Yamamura
- Department of Dermatology International University of Health and Welfare Atami Hospital Atami Japan
| | - Aiko Takano
- Department of Dermatology International University of Health and Welfare Atami Hospital Atami Japan
| | - Yoshihito Horiuchi
- Department of Dermatology International University of Health and Welfare Atami Hospital Atami Japan
| |
Collapse
|
118
|
Chen G, Liao Q, Ai J, Yang B, Bai H, Chen J, Liu F, Cao Y, Liu H, Li K. Immune Response to COVID-19 During Pregnancy. Front Immunol 2021; 12:675476. [PMID: 34012458 PMCID: PMC8126657 DOI: 10.3389/fimmu.2021.675476] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/13/2021] [Indexed: 12/16/2022] Open
Abstract
Pregnant women are generally more susceptible to viral infection. Although the impact of SARS-CoV-2 on pregnant women remains to be determined, evidence indicates that risks of adverse clinical outcomes are similar in pregnancy to the general population. Here we analyzed clinical symptoms and outcomes of 20 pregnant and 299 reproductive-aged non-pregnant female COVID-19 patients who were hospitalized during the same period. Laboratory measurements were compared among mild cases and healthy pregnant women. Our study found that pregnant patients showed enhanced innate immune response evident by higher neutrophils and C-reactive protein. Cytokines, chemokines, and growth factors (CCGFs) profiles from 11 pregnant and 4 non-pregnant COVID-19 patients and 10 healthy pregnant female patients, and lymphocyte subsets analysis of 7 pregnant patients and 19 non-pregnant patients, indicate suppressed cytokine storm and potential enhanced CD8+ T cell and NK cell activity in pregnant patients with COVID-19, which may be essential in contributing to the unique anti-SARS-CoV-2 response in pregnancy.
Collapse
Affiliation(s)
- Ge Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyue Liao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jihui Ai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hualin Bai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengyuan Liu
- Biological Sciences Greenhouse, College of Art & Sciences, The Ohio State University, Columbus, OH, United States
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiyi Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kezhen Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
119
|
Rabaan AA, Al-Ahmed SH, Muhammad J, Khan A, Sule AA, Tirupathi R, Mutair AA, Alhumaid S, Al-Omari A, Dhawan M, Tiwari R, Sharun K, Mohapatra RK, Mitra S, Bilal M, Alyami SA, Emran TB, Moni MA, Dhama K. Role of Inflammatory Cytokines in COVID-19 Patients: A Review on Molecular Mechanisms, Immune Functions, Immunopathology and Immunomodulatory Drugs to Counter Cytokine Storm. Vaccines (Basel) 2021; 9:436. [PMID: 33946736 PMCID: PMC8145892 DOI: 10.3390/vaccines9050436] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a severe pandemic of the current century. The vicious tentacles of the disease have been disseminated worldwide with unknown complications and repercussions. Advanced COVID-19 syndrome is characterized by the uncontrolled and elevated release of pro-inflammatory cytokines and suppressed immunity, leading to the cytokine storm. The uncontrolled and dysregulated secretion of inflammatory and pro-inflammatory cytokines is positively associated with the severity of the viral infection and mortality rate. The secretion of various pro-inflammatory cytokines such as TNF-α, IL-1, and IL-6 leads to a hyperinflammatory response by recruiting macrophages, T and B cells in the lung alveolar cells. Moreover, it has been hypothesized that immune cells such as macrophages recruit inflammatory monocytes in the alveolar cells and allow the production of large amounts of cytokines in the alveoli, leading to a hyperinflammatory response in severely ill patients with COVID-19. This cascade of events may lead to multiple organ failure, acute respiratory distress, or pneumonia. Although the disease has a higher survival rate than other chronic diseases, the incidence of complications in the geriatric population are considerably high, with more systemic complications. This review sheds light on the pivotal roles played by various inflammatory markers in COVID-19-related complications. Different molecular pathways, such as the activation of JAK and JAK/STAT signaling are crucial in the progression of cytokine storm; hence, various mechanisms, immunological pathways, and functions of cytokines and other inflammatory markers have been discussed. A thorough understanding of cytokines' molecular pathways and their activation procedures will add more insight into understanding immunopathology and designing appropriate drugs, therapies, and control measures to counter COVID-19. Recently, anti-inflammatory drugs and several antiviral drugs have been reported as effective therapeutic drug candidates to control hypercytokinemia or cytokine storm. Hence, the present review also discussed prospective anti-inflammatory and relevant immunomodulatory drugs currently in various trial phases and their possible implications.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia;
| | - Shamsah H. Al-Ahmed
- Specialty Paediatric Medicine, Qatif Central Hospital, Qatif 32654, Saudi Arabia;
| | - Javed Muhammad
- Department of Microbiology, The University of Haripur, Khyber Pakhtunkhwa 22620, Pakistan;
| | - Amjad Khan
- Department of Public Health/Nutrition, The University of Haripur, Khyber Pakhtunkhwa 22620, Pakistan;
| | - Anupam A Sule
- Medical Director of Informatics and Outcomes, St Joseph Mercy Oakland, Pontiac, MI 48341, USA;
| | - Raghavendra Tirupathi
- Department of Medicine Keystone Health, Penn State University School of Medicine, Hershey, PA 16801, USA;
- Department of Medicine, Wellspan Chambersburg and Waynesboro (Pa.) Hospitals, Chambersburg, PA 16801, USA
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Alahsa 36342, Saudi Arabia;
- College of Nursing, Prince Nora University, Riyadh 11564, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, NSW 2522, Australia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Ministry of Health, Alahsa 31982, Saudi Arabia;
| | - Awad Al-Omari
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
- Dr. Sulaiman Al-Habib Medical Group, Critical Care and Infection Control Department, Research Centre, Riyadh 11372, Saudi Arabia
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141027, Punjab, India;
- The Trafford Group of Colleges, Manchester WA14 5PQ, UK
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh; Pandit DeenDayal Upadhyaya PashuChikitsa Vigyan Vishwavidyalaya Evam Go AnusandhaSansthan (DUVASU), Mathura 281001, Uttar Pradesh, India;
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Mathura 281001, Uttar Pradesh, India;
| | - Ranjan K. Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar 758002, Odisha, India;
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; or
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Salem A. Alyami
- Department of Mathematics and Statistics, Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh;
| | - Mohammad Ali Moni
- WHO Collaborating Centre on eHealth, UNSW Digital Health, School of Public Health and Community Medicine, Faculty of Medicine, UNSW Sydney, NSW 2052, Australia
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| |
Collapse
|
120
|
Valdez-Cruz NA, García-Hernández E, Espitia C, Cobos-Marín L, Altamirano C, Bando-Campos CG, Cofas-Vargas LF, Coronado-Aceves EW, González-Hernández RA, Hernández-Peralta P, Juárez-López D, Ortega-Portilla PA, Restrepo-Pineda S, Zelada-Cordero P, Trujillo-Roldán MA. Integrative overview of antibodies against SARS-CoV-2 and their possible applications in COVID-19 prophylaxis and treatment. Microb Cell Fact 2021; 20:88. [PMID: 33888152 PMCID: PMC8061467 DOI: 10.1186/s12934-021-01576-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/03/2021] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 is a novel β-coronavirus that caused the COVID-19 pandemic disease, which spread rapidly, infecting more than 134 million people, and killing almost 2.9 million thus far. Based on the urgent need for therapeutic and prophylactic strategies, the identification and characterization of antibodies has been accelerated, since they have been fundamental in treating other viral diseases. Here, we summarized in an integrative manner the present understanding of the immune response and physiopathology caused by SARS-CoV-2, including the activation of the humoral immune response in SARS-CoV-2 infection and therefore, the synthesis of antibodies. Furthermore, we also discussed about the antibodies that can be generated in COVID-19 convalescent sera and their associated clinical studies, including a detailed characterization of a variety of human antibodies and identification of antibodies from other sources, which have powerful neutralizing capacities. Accordingly, the development of effective treatments to mitigate COVID-19 is expected. Finally, we reviewed the challenges faced in producing potential therapeutic antibodies and nanobodies by cell factories at an industrial level while ensuring their quality, efficacy, and safety.
Collapse
Affiliation(s)
- Norma A Valdez-Cruz
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México.
| | - Enrique García-Hernández
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Clara Espitia
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Laura Cobos-Marín
- Facultad de Medicina Veterinaria Y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Av. Brasil N° 2950, Valparaíso, Chile
| | - Carlos G Bando-Campos
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Luis F Cofas-Vargas
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Enrique W Coronado-Aceves
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Ricardo A González-Hernández
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Pablo Hernández-Peralta
- Facultad de Medicina Veterinaria Y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Daniel Juárez-López
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Paola A Ortega-Portilla
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Sara Restrepo-Pineda
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Patricio Zelada-Cordero
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Mauricio A Trujillo-Roldán
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Ciudad de México, México.
| |
Collapse
|
121
|
Rowaiye AB, Okpalefe OA, Onuh Adejoke O, Ogidigo JO, Hannah Oladipo O, Ogu AC, Oli AN, Olofinase S, Onyekwere O, Rabiu Abubakar A, Jahan D, Islam S, Dutta S, Haque M. Attenuating the Effects of Novel COVID-19 (SARS-CoV-2) Infection-Induced Cytokine Storm and the Implications. J Inflamm Res 2021; 14:1487-1510. [PMID: 33889008 PMCID: PMC8057798 DOI: 10.2147/jir.s301784] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/16/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic constitutes an arduous global health challenge, and the increasing number of fatalities calls for the speedy pursuit of a remedy. This review emphasizes the changing aspects of the COVID-19 disease, featuring the cytokine storm's pathological processes. Furthermore, we briefly reviewed potential therapeutic agents that may modulate and alleviate cytokine storms. The literature exploration was made using PubMed, Embase, MEDLINE, Google scholar, and China National Knowledge Infrastructure databases to retrieve the most recent literature on the etiology, diagnostic markers, and the possible prophylactic and therapeutic options for the management of cytokine storm in patients hospitalized with COVID-19 disease. The causative agent, severe acute respiratory coronavirus-2 (SARS-CoV-2), continually threatens the efficiency of the immune system of the infected individuals. As the first responder, the innate immune system provides primary protection against COVID-19, affecting the disease's progression, clinical outcome, and prognosis. Evidence suggests that the fatalities associated with COVID-19 are primarily due to hyper-inflammation and an aberrant immune function. Accordingly, the magnitude of the release of pro-inflammatory cytokines such as interleukin (IL)-1, (IL-6), and tumor necrosis alpha (TNF-α) significantly differentiate between mild and severe cases of COVID-19. The early prediction of a cytokine storm is made possible by several serum chemistry and hematological markers. The prompt use of these markers for diagnosis and the aggressive prevention and management of a cytokine release syndrome is critical in determining the level of morbidity and fatality associated with COVID-19. The prophylaxis and the rapid treatment of cytokine storm by clinicians will significantly enhance the fight against the dreaded COVID-19 disease.
Collapse
Affiliation(s)
- Adekunle Babajide Rowaiye
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | | | - Olukemi Onuh Adejoke
- Department of Medical Biotechnology, National Biotechnology Development Agency, Abuja, Nigeria
| | - Joyce Oloaigbe Ogidigo
- Bioresources Development Centre, Abuja, National Biotechnology Development Agency, Abuja, Nigeria
| | - Oluwakemi Hannah Oladipo
- Bioresources Development Centre, Ilorin, National Biotechnology Development Agency, Kwara State, Nigeria
| | - Amoge Chidinma Ogu
- Department of Medical Biotechnology, National Biotechnology Development Agency, Abuja, Nigeria
| | - Angus Nnamdi Oli
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Samson Olofinase
- Department of Genetics, Genomics, Bioinformatics, National Biotechnology Development Agency, Abuja, Nigeria
| | - Onyekachi Onyekwere
- Bioresources Development Centre, Ubulu-Uku, National Biotechnology Development Agency, Delta State, Nigeria
| | - Abdullahi Rabiu Abubakar
- Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Bayero University, Kano, 700233, Nigeria
| | - Dilshad Jahan
- Department of Hematology, Asgar Ali Hospital, Gandaria, Dhaka, 1204, Bangladesh
| | - Salequl Islam
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Siddhartha Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur, 57000, Malaysia
| |
Collapse
|
122
|
Amere Subbarao S. Cancer vs. SARS-CoV-2 induced inflammation, overlapping functions, and pharmacological targeting. Inflammopharmacology 2021; 29:343-366. [PMID: 33723711 PMCID: PMC7959277 DOI: 10.1007/s10787-021-00796-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/27/2021] [Indexed: 12/15/2022]
Abstract
Inflammation is an intrinsic defence mechanism triggered by the immune system against infection or injury. Chronic inflammation allows the host to recover or adapt through cellular and humoral responses, whereas acute inflammation leads to cytokine storms resulting in tissue damage. In this review, we present the overlapping outcomes of cancer inflammation with virus-induced inflammation. The study emphasises how anti-inflammatory drugs that work against cancer inflammation may work against the inflammation caused by the viral infection. It is established that the cytokine storm induced in response to SARS-CoV-2 infection contributes to disease-associated mortality. While cancer remains the second among the diseases associated with mortality worldwide, cancer patients' mortality rates are often observed upon extended periods after illness, usually ranging from months to years. However, the mortality rates associated with COVID-19 disease are robust. The cytokine storm induced by SARS-CoV-2 infection appeared to be responsible for the multi-organ failure and increased mortality rates. Since both cancer and COVID-19 disease share overlapping inflammatory mechanisms, repurposing some anticancer and anti-inflammatory drugs for COVID-19 may lower mortality rates. Here, we review some of these inflammatory mechanisms and propose some potential chemotherapeutic agents to intervene in them. We also discuss the repercussions of anti-inflammatory drugs such as glucocorticoids and hydroxychloroquine with zinc or antiviral drugs such as ivermectin and remdesivir against SARS-CoV-2 induced cytokine storm. In this review, we emphasise on various possibilities to reduce SARS-CoV-2 induced cytokine storm.
Collapse
|
123
|
Chizinga M, Kalra SS, Innabi A, Rackauskas M, Ataya A, Emtiazjoo A. Macrophage activating syndrome causing decompensated right heart failure. Respir Med Case Rep 2021; 33:101409. [PMID: 34401257 PMCID: PMC8348922 DOI: 10.1016/j.rmcr.2021.101409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/24/2021] [Indexed: 12/03/2022] Open
Abstract
Background Macrophage activating syndrome (MAS) is a form of hemophagocytic lymphohistiocytosis (HLH), a rare complication of autoimmune disease that is characterized by cytokine storm and multiorgan failure. Case summary A 32-year-old male presented with acutely decompensated pulmonary arterial hypertension and right heart failure secondary to MAS. The patient was immediately started on inhaled and intravenous epoprostenol, vasopressors and dexamethasone and anakinra were administered. Despite the therapies given, the patient's condition continued to decline, and he was placed on veno-arterial extracorporeal membrane oxygenation (VA-ECMO) support. Over a few days, his clinical condition improved, and he was decannulated from VA-ECMO and later transitioned oral treprositinil and was discharged home. Due to its non-specific clinical manifestations, the diagnosis of MAS depends on high clinical suspicion and initial laboratory work up such as thrombocytopenia, transaminitis, hyperferritinemia, hypertriglyceridemia, hypofibrinogenemia, etc. In our patient, MAS led to decompensated Pulmonary Arterial Hypertension (PAH) leading to right heart failure that was refractory to inhaled and intravenous epoprostenol and vasopressors and required VA-ECMO as a bridge to recovery while his MAS was managed by anakinra and dexamethasone. Conclusion MAS can result in acute decompensation of PAH and right heart failure. Besides RV failure management, immunosuppressants such as anakinra, etoposide, etc. should be utilized early in the management of MAS. In refractory right heart failure, VA-ECMO can be considered as a bridge to recovery. There is a paucity of literature supporting the utilization of VA-ECMO in the management of refractory right heart failure caused by MAS in adults and much of the data stems from pediatric studies. This case serves as a fine example of successful use of VA-ECMO in adult population.
Collapse
Affiliation(s)
- Mwelwa Chizinga
- University of Florida, Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Gainesville, FL, USA
| | - Saminder Singh Kalra
- University of Florida, Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Gainesville, FL, USA
| | - Ayoub Innabi
- University of Florida, Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Gainesville, FL, USA
| | - Mindaugas Rackauskas
- University of Florida, Department of Surgery, Division of Thoracic Surgery, Gainesville, FL, USA
| | - Ali Ataya
- University of Florida, Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Gainesville, FL, USA
| | - Amir Emtiazjoo
- University of Florida, Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Gainesville, FL, USA
| |
Collapse
|
124
|
Chen M, Zhao Q, Diao L, Xue K, Ruan Y, Xue F, Li J, Shi R, Pan M, Zheng J, Cao H. Distribution of anti-melanoma differentiation associated gene 5 (MDA5) IgG subclasses in MDA5+ dermatomyositis. Rheumatology (Oxford) 2021; 61:430-439. [PMID: 33742662 DOI: 10.1093/rheumatology/keab268] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES The anti-melanoma differentiation-associated gene 5 (MDA5) antibody is the main predictor of interstitial lung disease (ILD) in dermatomyositis (DM) and clinically amyopathic dermatomyositis (CADM). Nevertheless, a subset of MDA5+ patients have a favorable prognosis. We aimed to determine the possibility of using anti-MDA5 antibody isotypes and IgG subclasses for evaluating ILD risk. METHODS The isotypes (IgG, IgA and IgM) of anti-MDA5 were detected in serum samples of 36 anti-MDA5+ patients with DM/CADM using enzyme-linked immunosorbent assay (ELISA). IgG subclasses of anti-MDA5 antibodies were further investigated. Laboratory findings and cumulative survival were analyzed based on the isotypes of anti-MDA5 and subclasses of anti-MDA5 IgG. RESULTS Among the MDA5+ patients with DM/CADM, the positive rates of anti-MDA5 IgG, IgA, IgM were 100%, 97%, and 6%, respectively. The positive rates of anti-MDA5 IgG1, IgG2, IgG3, and IgG4 were 72%, 25%, 0%, and 28%, respectively. The incidence of acute interstitial pneumonia, mortality rate, and serum ferritin were significantly higher in anti-MDA5 IgG1+ patients than those in anti-MDA5 IgG1- patients with DM/CADM (P = 0.0027, 0.015, 0.0011, respectively). The sensitivity and specificity of anti-MDA5 IgG1 for predicting mortality were 100% and 41.7%, respectively. A combination of anti-MDA5 IgG1 and IgG4 for predicting mortality, yielded better specificity (87.5%). CONCLUSION IgA and IgG are the primary anti-MDA5 antibody isotypes. Anti-MDA5 IgG1 is the primary component of MDA5 IgG subclasses and anti-MDA5 IgG1 and IgG4 might serve as useful biomarkers for predicting mortality in DM-ILD.
Collapse
Affiliation(s)
- Mengya Chen
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Qian Zhao
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Licheng Diao
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Ke Xue
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Yeping Ruan
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Feng Xue
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Jian Li
- Department of Clinical Research Center, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Ruofei Shi
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Meng Pan
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Jie Zheng
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Hua Cao
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| |
Collapse
|
125
|
Li C, Liu M, Liu K, Li M, Liu Y, Li T, Wei Y, Long Y, He W, Shi X, Li Y, Zhang H. BATF2 balances the T cell-mediated immune response of CADM with an anti-MDA5 autoantibody. Biochem Biophys Res Commun 2021; 551:155-160. [PMID: 33740622 DOI: 10.1016/j.bbrc.2021.02.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Clinically amyopathic dermatomyositis (CADM) is a subtype of dermatomyositis (DM) characterized by low-grade or absent muscle inflammation but frequent and rapidly progressive interstitial lung disease (RP-ILD) and skin ulcers with anti-melanoma differentiation-associated gene 5 (anti-MDA5) autoantibodies. Basic leucine zipper transcription factor ATF-like 2 (BATF2) is thought to function as an inhibitor of tumours and inflammation. Here, we aimed to investigate the roles of BATF2 in Th cell differentiation of CADM with an anti-MDA5 autoantibody (anti-MDA5+ CADM). METHODS Naive CD4+ T cells from human peripheral blood mononuclear cells (PBMCs) of healthy controls (HCs) were isolated and then cultured with IL-12, TGF-β or TGF-β plus IL-6 following anti-CD3 and anti-CD28 stimulations. The expression of BATF2 was measured by real-time PCR. The percentages of Th1, Th17 and Treg CD4+ T cells were detected by flow cytometry. BATF2 knockdown of CD4+ T cells was performed using small interfering RNAs (siRNAs). RESULTS The expression of BATF2 in PBMCs was higher in anti-MDA5+ CADM patients than in healthy controls. The BATF2 mRNA expression was increased under Th1 and Treg polarization but decreased under Th17 polarization. Th17 cell activation-associated genes were possibly increased while Th1 and Treg cell differentiation-associated genes were inhibited by posttranscriptional gene silencing of BATF2 in CD4+ T cells. CONCLUSIONS BATF2 promoted Th1 and Treg cell differentiation but suppressed Th17 cell activation in anti-MDA5+ CADM.
Collapse
Affiliation(s)
- Caiyan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Meidong Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Muyuan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Yanjuan Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Tao Li
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Yu Wei
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China.
| | - Ying Long
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China.
| | - Weijia He
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China.
| | - Xueyan Shi
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Yisha Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China.
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
126
|
Chen X, Kang Y, Luo J, Pang K, Xu X, Wu J, Li X, Jin S. Next-Generation Sequencing Reveals the Progression of COVID-19. Front Cell Infect Microbiol 2021; 11:632490. [PMID: 33777844 PMCID: PMC7991797 DOI: 10.3389/fcimb.2021.632490] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/10/2021] [Indexed: 01/08/2023] Open
Abstract
The novel coronavirus SARS-CoV-2 (causing the disease COVID-19) has caused a highly transmissible and ongoing pandemic worldwide. Due to its rapid development, next-generation sequencing plays vital roles in many aspects. Here, we summarize the current knowledge on the origin and human transmission of SARS-CoV-2 based on NGS analysis. The ACE2 expression levels in various human tissues and relevant cells were compared to provide insights into the mechanism of SAS-CoV-2 infection. Gut microbiota dysbiosis observed by metagenome sequencing and the immunogenetics of COVID-19 patients according to single-cell sequencing analysis were also highlighted. Overall, the application of these sequencing techniques could be meaningful for finding novel intermediate SARS-CoV-2 hosts to block interspecies transmission. This information will further benefit SARS-CoV-2 diagnostic development and new therapeutic target discovery. The extensive application of NGS will provide powerful support for our fight against future public health emergencies.
Collapse
Affiliation(s)
- Xiaomin Chen
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yutong Kang
- Wenzhou Key Laboratory of Sanitary Microbiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jing Luo
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kun Pang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xin Xu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jinyu Wu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
127
|
Chen S, Lai SWT, Brown CE, Feng M. Harnessing and Enhancing Macrophage Phagocytosis for Cancer Therapy. Front Immunol 2021; 12:635173. [PMID: 33790906 PMCID: PMC8006289 DOI: 10.3389/fimmu.2021.635173] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy has revolutionized the paradigm for the clinical management of cancer. While FDA-approved cancer immunotherapies thus far mainly exploit the adaptive immunity for therapeutic efficacy, there is a growing appreciation for the importance of innate immunity in tumor cell surveillance and eradication. The past decade has witnessed macrophages being thrust into the spotlight as critical effectors of an innate anti-tumor response. Promising evidence from preclinical and clinical studies have established targeting macrophage phagocytosis as an effective therapeutic strategy, either alone or in combination with other therapeutic moieties. Here, we review the recent translational advances in harnessing macrophage phagocytosis as a pivotal therapeutic effort in cancer treatment. In addition, this review emphasizes phagocytosis checkpoint blockade and the use of nanoparticles as effective strategies to potentiate macrophages for phagocytosis. We also highlight chimeric antigen receptor macrophages as a next-generation therapeutic modality linking the closely intertwined innate and adaptive immunity to induce efficacious anti-tumor immune responses.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Seigmund W. T. Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Christine E. Brown
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| |
Collapse
|
128
|
McGonagle D, Ramanan AV, Bridgewood C. Immune cartography of macrophage activation syndrome in the COVID-19 era. Nat Rev Rheumatol 2021; 17:145-157. [PMID: 33547426 PMCID: PMC7863615 DOI: 10.1038/s41584-020-00571-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2020] [Indexed: 02/06/2023]
Abstract
A hyperinflammatory 'cytokine storm' state termed macrophage activation syndrome (MAS), culminating from a complex interplay of genetics, immunodeficiency, infectious triggers and dominant innate immune effector responses, can develop across disparate entities including systemic juvenile idiopathic arthritis (sJIA) and its counterpart adult-onset Still disease (AOSD), connective tissue diseases, sepsis, infection, cancers and cancer immunotherapy. Classifying MAS using the immunological disease continuum model, with strict boundaries that define the limits of innate and adaptive immunity, at one boundary is MAS with loss of immune function, as occurs in the 'perforinopathies' and some cases of sJIA-AOSD. Conversely, at the other boundary, immune hypersensitivity with gain of immune function in MHC class II-associated sJIA-AOSD and with chimeric antigen receptor (CAR) T cell therapy also triggers MAS. This provides a benchmark for evaluating severe inflammation in some patients with COVID-19 pneumonia, which cripples primary type I interferon immunity and usually culminates in a lung-centric 'second wave' cytokine-driven alveolitis with associated immunothrombosis; this phenomenon is generally distinct from MAS but can share features with the proposed 'loss of immune function' MAS variant. This loss and gain of function MAS model offers immune cartography for a novel mechanistic classification of MAS with therapeutic implications.
Collapse
Affiliation(s)
- Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, UK.
| | - Athimalaipet V Ramanan
- University Hospitals Bristol NHS Foundation Trust & Translational Health Sciences, University of Bristol, Bristol, UK
| | - Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
129
|
A fine line between macrophage activation syndrome and multisystem inflammatory syndrome in children - literature review based on two case reports. Reumatologia 2021; 59:47-57. [PMID: 33707796 PMCID: PMC7944956 DOI: 10.5114/reum.2021.102871] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/14/2021] [Indexed: 12/24/2022] Open
Abstract
Multisystem inflammatory syndrome in children is a newly described condition temporarily associated with severe acute respiratory syndrome coronavirus 2. The primary observations indicated coronavirus disease 2019 infection in children to be mild. However, recent reports have revealed a correlation between multisystem inflammatory symptoms and coronavirus infection. The manifestation of the disease is similar to Kawasaki disease, toxic shock syndrome and macrophage activation syndrome. Treatment includes intravenous corticosteroids, immunoglobulin and biological therapy (anakinra, tocilizumab, infliximab). In this article we compare world reports of multisystem inflammatory syndrome in children to case reports of two patients who were hospitalized recently at the Department of Paediatric Cardiology and Rheumatology. In the course of diagnostics they presented numerous symptoms which required differentiation from multisystem inflammatory syndrome in children.
Collapse
|
130
|
Kang YW, Park S, Lee KJ, Moon D, Kim YM, Lee SW. Understanding the Host Innate Immune Responses against SARS-CoV-2 Infection and COVID-19 Pathogenesis. Immune Netw 2021; 21:e1. [PMID: 33728094 PMCID: PMC7937512 DOI: 10.4110/in.2021.21.e1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 12/26/2022] Open
Abstract
The emergence of a new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has become a significant health concern worldwide. Undoubtedly, a better understanding of the innate and adaptive immune responses against SARS-CoV-2 and its relationship with the coronavirus disease 2019 (COVID-19) pathogenesis will be the sole basis for developing and applying therapeutics. This review will summarize the published results that relate to innate immune responses against infections with human coronaviruses including SARS-CoV-1 and SARS-CoV-2 in both humans and animal models. The topics encompass the innate immune sensing of the virus to the dysregulation of various innate immune cells during infection and disease progression.
Collapse
Affiliation(s)
- Yeon-Woo Kang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Subin Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Kun-Joo Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Dain Moon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Young-Min Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| |
Collapse
|
131
|
Barberis E, Vanella VV, Falasca M, Caneapero V, Cappellano G, Raineri D, Ghirimoldi M, De Giorgis V, Puricelli C, Vaschetto R, Sainaghi PP, Bruno S, Sica A, Dianzani U, Rolla R, Chiocchetti A, Cantaluppi V, Baldanzi G, Marengo E, Manfredi M. Circulating Exosomes Are Strongly Involved in SARS-CoV-2 Infection. Front Mol Biosci 2021; 8:632290. [PMID: 33693030 PMCID: PMC7937875 DOI: 10.3389/fmolb.2021.632290] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Knowledge of the host response to the novel coronavirus SARS-CoV-2 remains limited, hindering the understanding of COVID-19 pathogenesis and the development of therapeutic strategies. During the course of a viral infection, host cells release exosomes and other extracellular vesicles carrying viral and host components that can modulate the immune response. The present study used a shotgun proteomic approach to map the host circulating exosomes’ response to SARS-CoV-2 infection. We investigated how SARS-CoV-2 infection modulates exosome content, exosomes’ involvement in disease progression, and the potential use of plasma exosomes as biomarkers of disease severity. A proteomic analysis of patient-derived exosomes identified several molecules involved in the immune response, inflammation, and activation of the coagulation and complement pathways, which are the main mechanisms of COVID-19–associated tissue damage and multiple organ dysfunctions. In addition, several potential biomarkers—such as fibrinogen, fibronectin, complement C1r subcomponent and serum amyloid P-component—were shown to have a diagnostic feature presenting an area under the curve (AUC) of almost 1. Proteins correlating with disease severity were also detected. Moreover, for the first time, we identified the presence of SARS-CoV-2 RNA in the exosomal cargo, which suggests that the virus might use the endocytosis route to spread infection. Our findings indicate circulating exosomes’ significant contribution to several processes—such as inflammation, coagulation, and immunomodulation—during SARS-CoV-2 infection. The study’s data are available via ProteomeXchange with the identifier PXD021144.
Collapse
Affiliation(s)
- Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy.,ISALIT, Novara, Italy
| | - Virginia V Vanella
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Marco Falasca
- Metabolic Signalling Group, Curtin Medical School, Curtin University, Perth, WA, Australia
| | - Valeria Caneapero
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Giuseppe Cappellano
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy.,Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Davide Raineri
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy.,Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Marco Ghirimoldi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Chiara Puricelli
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Rosanna Vaschetto
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Pier Paolo Sainaghi
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Azienda Ospedaliero-Universitaria "Maggiore della Carità", Novara, Italy
| | - Stefania Bruno
- Città della Salute e della Scienza and Molecular Biotechnology Center, Torino, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.,Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Umberto Dianzani
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Roberta Rolla
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Vincenzo Cantaluppi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Gianluca Baldanzi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy.,Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy.,ISALIT, Novara, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy.,ISALIT, Novara, Italy
| |
Collapse
|
132
|
Gruenbacher G, Gander H, Dobler G, Rahm A, Klaver D, Thurnher M. The human G protein-coupled ATP receptor P2Y 11 is a target for anti-inflammatory strategies. Br J Pharmacol 2021; 178:1541-1555. [PMID: 33463722 PMCID: PMC9328440 DOI: 10.1111/bph.15379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background and Purpose The ATP receptor P2Y11, which couples to Gq and Gs proteins, senses cell stress and promotes cytoprotective responses. P2Y11 receptors are upregulated during differentiation of M2 macrophages. However, it is unclear whether and how P2Y11 receptors contribute to the anti‐inflammatory properties of M2 macrophages. Experimental Approach Transcriptome and secretome profiling of ectopic P2Y11 receptors was used to analyse their signalling and function. Findings were validated in human monocyte‐derived M2 macrophages. The suramin analogue NF340 and P2Y11 receptor‐knockout cells confirmed that agonist‐mediated responses were specific to P2Y11 receptor stimulation. Key Results Temporal transcriptome profiling of P2Y11 receptor stimulation showed a strong and tightly controlled response of IL‐1 receptors, including activation of the IL‐1 receptor target genes, IL6 and IL8. Secretome profiling confirmed the presence of IL‐6 and IL‐8 proteins and additionally identified soluble tumour necrosis factor receptor 1 and 2 (sTNFR1 and sTNFR2) as targets of P2Y11 receptor activation. Raised levels of intracellular cAMP in M2 macrophages, after inhibition of phosphodiesterases (PDE), especially PDE4, strongly increased P2Y11 receptor‐induced release of sTNFR2 through ectodomain shedding mediated by TNF‐α converting enzyme (TACE/ADAM17). Both IL‐1α and IL‐1ß synergistically enhanced P2Y11 receptor‐ induced IL‐6 and IL‐8 secretion and release of sTNFR2. During lipopolysaccharide‐induced activation of TLR4, which shares the downstream signalling pathway with IL‐1 receptors, P2Y11 receptors specifically prevented secretion of TNF‐α. Conclusions and Implications Targeting P2Y11 receptors activates IL‐1 receptor signalling to promote sTNFR2 release and suppress TLR4 signalling to prevent TNF‐α secretion, thus facilitating resolution of inflammation.
Collapse
Affiliation(s)
- Georg Gruenbacher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Gander
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Dobler
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Rahm
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Klaver
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
133
|
Macrophage activation syndrome in a newborn: report of a case associated with neonatal lupus erythematosus and a summary of the literature. Pediatr Rheumatol Online J 2021; 19:13. [PMID: 33568193 PMCID: PMC7877111 DOI: 10.1186/s12969-021-00500-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Macrophage activation syndrome (MAS) is a life-threatening hyperinflammatory syndrome and is caused by a severely dysregulated immune response. It has rarely been associated with neonatal lupus. CASE PRESENTATION We present a female neonate with MAS born to a mother who had cutaneous lupus erythematosus with circulating anti-nuclear antibodies (ANA), anti-SSA, anti-SSB and anti-extractable nuclear antigen (anti-ENA) antibodies. Because of neonatal lupus (NLE) with a total atrioventricular block, epicardial pacemaker implantation was required on the sixth day of life. Following surgery she developed non-remitting fever and disseminated erythematous skin lesions. A diagnosis of MAS was made based on these symptoms, with hyperferritinemia, elevated transaminases, hypertriglyceridemia, and a skin biopsy that showed hemophagocytosis. Our patient was treated with steroids for 3 months with good effect. No relapse has occurred. CONCLUSIONS MAS is a rare complication of neonatal lupus that may be difficult to diagnose, but needs to be treated promptly. In this article, pathogenesis and overlap of MAS and hemophagocytic lymphohistiocytosis (HLH) has been described. Diagnosis of MAS can be difficult. Different diagnostic criteria are used in both diagnosing MAS and HLH. Validated criteria for diagnosis of MAS in other disease than systemic onset JIA have not been validated yet. In NLE, diagnosing MAS is even more difficult, since skin lesions are already common in NLE. We show the potential additional value of skin biopsy in diagnosing MAS.
Collapse
|
134
|
Han ZB, Wu J, Liu J, Li HM, Guo K, Sun T. Adult-onset Still's disease evolving with multiple organ failure and death: A case report and review of the literature. World J Clin Cases 2021; 9:886-897. [PMID: 33585636 PMCID: PMC7852636 DOI: 10.12998/wjcc.v9.i4.886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/28/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adult-onset Still’s disease (AOSD) is a rare systemic inflammatory disease, which is characterized by daily fever and arthritis, with an evanescent rash and neutrophilic leukocytosis. To date, there has been no definite laboratory or imaging test available for diagnosing AOSD; the diagnosis is one of exclusion, which can be very challenging. In particular, AOSD patients may experience different complications affecting their clinical picture, management, and prognosis. The treatment of AOSD remains largely empirical and involves therapeutic agents.
CASE SUMMARY We report the case of a 36-year-old woman who presented with fever, red rash, arthralgia, and sore throat. Her serum ferritin level and white blood cell count were markedly elevated, and the first diagnosis 22 years prior was "juvenile rheumatoid arthritis of systemic type". The patient was treated with prednisone, sulfasalazine, methotrexate, and leflunomide. After remission of her symptoms, the patient stopped taking the medications, and the disease recurred. Ultimately, the patient was diagnosed with adult-onset Still's disease. Relapse occurred several times due to self-medication withdrawal, and an interleukin-6 antagonist (tocilizumab/Actemra) was administered to control the disease. Recently, she was hospitalized because an incision did not heal, and the patient suddenly developed high fever and diarrhea during hospitalization. The patient's disease progressed violently and quickly developed into macrophage activation syndrome, disseminated intravascular coagulation, shock, and multiple organ failure. The patient had sudden cardiac arrest, and she died despite emergency rescue efforts.
CONCLUSION AOSD patients need regular follow-up in the long-term treatment process, and must press formulary standard medication, and do not voluntarily withdraw or reduce the dose. Otherwise it may cause disease back-and-forth or serious life-threatening complications. Meanwhile, strict management of trauma, infections, tumors, and other diseases may contribute to improved outcomes in patients with complications.
Collapse
Affiliation(s)
- Zhong-Bin Han
- Department of Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Ju Wu
- Department of Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Jing Liu
- Department of ICU, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - He-Ming Li
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Kai Guo
- Department of Surgery, Second People's Hospital of Jinzhong City, Jinzhong 030600, Shanxin Province, China
| | - Tong Sun
- Department of ICU, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| |
Collapse
|
135
|
Bashir DA, Da Q, Pradhan S, Sekhar N, Valladolid C, Lam F, Guffey D, Goldman J, Desai MS, Cruz MA, Allen C, Nguyen TC, Vijayan KV. Secretion of von Willebrand Factor and Suppression of ADAMTS-13 Activity by Markedly High Concentration of Ferritin. Clin Appl Thromb Hemost 2021; 27:1076029621992128. [PMID: 33539188 PMCID: PMC7868463 DOI: 10.1177/1076029621992128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hyperferritinemia is associated with poor outcomes in critically ill patients with sepsis, hemophagocytic lymphohistiocytosis (HLH), macrophage activation syndromes (MAS) and coronavirus disease 19 (COVID-19). Autopsies of hyperferritinemic patients that succumbed to either sepsis, HLH, MAS or COVID-19 have revealed disseminated microvascular thromboses with von Willebrand factor (VWF)-, platelets-, and/or fibrin-rich microthrombi. It is unknown whether high plasma ferritin concentration actively promotes microvascular thrombosis, or merely serves as a prognostic biomarker in these patients. Here, we show that secretion of VWF from human umbilical vein endothelial cells (HUVEC) is significantly enhanced by 100,000 ng/ml of recombinant ferritin heavy chain protein (FHC). Ferritin fraction that was isolated by size exclusion chromatography from the plasma of critically ill HLH patients promoted VWF secretion from HUVEC, compared to similar fraction from non-critically ill control plasma. Furthermore, recombinant FHC moderately suppressed the activity of VWF cleaving metalloprotease ADAMTS-13. These observations suggest that a state of marked hyperferritinemia could promote thrombosis and organ injury by inducing endothelial VWF secretion and reducing the ADAMTS-13 activity.
Collapse
Affiliation(s)
- Dalia A Bashir
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA
| | - Qi Da
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Subhashree Pradhan
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Nitin Sekhar
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christian Valladolid
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Fong Lam
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA
| | - Danielle Guffey
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Jordana Goldman
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA
| | - Moreshwar S Desai
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA
| | - Miguel A Cruz
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Carl Allen
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA
| | - Trung C Nguyen
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA
| | - K Vinod Vijayan
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
136
|
Gao YM, Xu G, Wang B, Liu BC. Cytokine storm syndrome in coronavirus disease 2019: A narrative review. J Intern Med 2021; 289:147-161. [PMID: 32696489 PMCID: PMC7404514 DOI: 10.1111/joim.13144] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022]
Abstract
Cytokine storm syndrome (CSS) is a critical clinical condition induced by a cascade of cytokine activation, characterized by overwhelming systemic inflammation, hyperferritinaemia, haemodynamic instability and multiple organ failure (MOF). At the end of 2019, the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in Wuhan, China, and rapidly developed into a global pandemic. More and more evidence shows that there is a dramatic increase of inflammatory cytokines in patients with COVID-19, suggesting the existence of cytokine storm in some critical illness patients. Here, we summarize the pathogenesis, clinical manifestation of CSS, and highlight the current understanding about the recognition and potential therapeutic options of CSS in COVID-19.
Collapse
Affiliation(s)
- Y-M Gao
- From the, Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - G Xu
- Department of Nephrology, Tongji Hospital, University of HuaZhong Science and Technology, Wuhan, China
| | - B Wang
- From the, Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - B-C Liu
- From the, Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
137
|
Choudhary S, Sharma K, Silakari O. The interplay between inflammatory pathways and COVID-19: A critical review on pathogenesis and therapeutic options. Microb Pathog 2021; 150:104673. [PMID: 33278517 PMCID: PMC7709793 DOI: 10.1016/j.micpath.2020.104673] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/28/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023]
Abstract
COVID-19, caused by SARS-CoV-2, emerged as the deadliest outbreak that has now become a serious health issue to mankind. Activation of inflammatory signaling pathways and cytokine storm are crucial factors that lead to acute respiratory distress syndrome (ARDS) in COVID-19 patients. Excessive secretion of pro-inflammatory cytokines and chemokines leads to the dysregulation of the innate immune system. The cytokine storm attracts many inflammatory cells that infiltrate into the lung tissues and ultimately cause immune damage. In addition to the dysregulation of the immune system, dysfunction of the renin-angiotensin system (RAS) due to the downregulation of ACE2 is also associated with the mortality of COVID-19 patients. Both the mechanisms are directly or indirectly associated with cytokine storm that promotes vascular hyperpermeability, vascular edema leading to hypercoagulation and hence multiorgan damage. As of now, there is no specific treatment available for COVID-19, but scientists have purposed several treatment options including cytokine inhibitors, JAK inhibitors, immunomodulators, plasma therapy, etc. In this article, we have provided the detailed mechanism of occurrence of SARS-CoV-2 induced inflammatory storm and its connection with the pre-existing inflammatory conditions. Possible treatment options to cope up with the severe clinical manifestations of COVID-19 are also discussed.
Collapse
Affiliation(s)
- Shalki Choudhary
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Kajal Sharma
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Om Silakari
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
138
|
Aigner L, Pietrantonio F, Bessa de Sousa DM, Michael J, Schuster D, Reitsamer HA, Zerbe H, Studnicka M. The Leukotriene Receptor Antagonist Montelukast as a Potential COVID-19 Therapeutic. Front Mol Biosci 2020; 7:610132. [PMID: 33392263 PMCID: PMC7773944 DOI: 10.3389/fmolb.2020.610132] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
The emergence and global impact of COVID-19 has focused the scientific and medical community on the pivotal influential role of respiratory viruses as causes of severe pneumonia, on the understanding of the underlying pathomechanisms, and on potential treatment for COVID-19. The latter concentrates on four different strategies: (i) antiviral treatments to limit the entry of the virus into the cell and its propagation, (ii) anti-inflammatory treatment to reduce the impact of COVID-19 associated inflammation and cytokine storm, (iii) treatment using cardiovascular medication to reduce COVID-19 associated thrombosis and vascular damage, and (iv) treatment to reduce the COVID-19 associated lung injury. Ideally, effective COVID-19 treatment should target as many of these mechanisms as possible arguing for the search of common denominators as potential drug targets. Leukotrienes and their receptors qualify as such targets: they are lipid mediators of inflammation and tissue damage and well-established targets in respiratory diseases like asthma. Besides their role in inflammation, they are involved in various other aspects of lung pathologies like vascular damage, thrombosis, and fibrotic response, in brain and retinal damages, and in cardiovascular disease. In consequence, leukotriene receptor antagonists might be potential candidates for COVID-19 therapeutics. This review summarizes the current knowledge on the potential involvement of leukotrienes in COVID-19, and the rational for the use of the leukotriene receptor antagonist montelukast as a COVID-19 therapeutic.
Collapse
Affiliation(s)
- Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | | | - Diana Marisa Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Johanna Michael
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Daniela Schuster
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Herbert Anton Reitsamer
- Department of Ophthalmology and Optometry, University Clinic Salzburg, Paracelsus Medical University, Salzburg, Austria.,Research Program of Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University Salzburg, Salzburg, Austria
| | | | - Michael Studnicka
- Department of Pulmonary Medicine, University Clinic Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| |
Collapse
|
139
|
Lariccia V, Magi S, Serfilippi T, Toujani M, Gratteri S, Amoroso S. Challenges and Opportunities from Targeting Inflammatory Responses to SARS-CoV-2 Infection: A Narrative Review. J Clin Med 2020; 9:E4021. [PMID: 33322733 PMCID: PMC7763517 DOI: 10.3390/jcm9124021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/05/2020] [Accepted: 12/09/2020] [Indexed: 02/08/2023] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) is a global pandemic that continues to sweep across the world, posing an urgent need for effective therapies and prevention of the spread of the severe acute respiratory syndrome related to coronavirus-2 (SARS-CoV-2). A major hypothesis that is currently guiding research and clinical care posits that an excessive and uncontrolled surge of pro-inflammatory cytokines (the so-called "cytokine storm") drives morbidity and mortality in the most severe cases. In the overall efforts made to develop effective and safe therapies (including vaccines) for COVID-19, clinicians are thus repurposing ready-to-use drugs with direct or indirect anti-inflammatory and immunomodulatory activities. Speculatively, there are many opportunities and challenges in targeting immune/inflammatory processes in the evolving settings of COVID-19 disease because of the need to safely balance the fight against virus and aggressive inflammation versus the suppression of host immune defenses and the risk of additional harms in already compromised patients. To this end, many studies are globally underway to weigh the pros and cons of tailoring drugs used for inflammatory-driven conditions to COVID-19 patient care, and the next step will be to summarize the growing clinical trial experience into clean clinical practice. Based on the current evidence, anti-inflammatory drugs should be considered as complementary approaches to anti-viral drugs that need to be timely introduced in the management of COVID-19 according to disease severity. While drugs that target SARS-CoV-2 entry or replication are expected to confer the greatest benefits at the early stage of the infection, anti-inflammatory drugs would be more effective in limiting the inflammatory processes that drive the worsening of the disease.
Collapse
Affiliation(s)
- Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (S.M.); (T.S.); (M.T.)
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (S.M.); (T.S.); (M.T.)
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (S.M.); (T.S.); (M.T.)
| | - Marwa Toujani
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (S.M.); (T.S.); (M.T.)
| | - Santo Gratteri
- Institute of Legal Medicine, University “Magna Graecia”, 88100 Catanzaro, Italy;
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (S.M.); (T.S.); (M.T.)
| |
Collapse
|
140
|
Kooistra EJ, Waalders NJB, Grondman I, Janssen NAF, de Nooijer AH, Netea MG, van de Veerdonk FL, Ewalds E, van der Hoeven JG, Kox M, Pickkers P. Anakinra treatment in critically ill COVID-19 patients: a prospective cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:688. [PMID: 33302991 PMCID: PMC7726611 DOI: 10.1186/s13054-020-03364-w] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/29/2020] [Indexed: 01/29/2023]
Abstract
Background A subset of critically ill COVID-19 patients develop a hyperinflammatory state. Anakinra, a recombinant interleukin-1 receptor antagonist, is known to be effective in several hyperinflammatory diseases. We investigated the effects of anakinra on inflammatory parameters and clinical outcomes in critically ill, mechanically ventilated COVID-19 patients with clinical features of hyperinflammation. Methods In this prospective cohort study, 21 critically ill COVID-19 patients treated with anakinra were compared to a group of standard care. Serial data of clinical inflammatory parameters and concentrations of multiple circulating cytokines were determined and aligned on start day of anakinra in the treatment group, and median start day of anakinra in the control group. Analysis was performed for day − 10 to + 10 relative to alignment day. Clinical outcomes were analyzed during 28 days. Additionally, three sensitivity analyses were performed: (1) using propensity score-matched groups, (2) selecting patients who did not receive corticosteroids, and (3) using a subset of the control group aimed to match the criteria (fever, elevated ferritin) for starting anakinra treatment. Results Baseline patient characteristics and clinical parameters on ICU admission were similar between groups. As a consequence of bias by indication, plasma levels of aspartate aminotransferase (ASAT) (p = 0.0002), ferritin (p = 0.009), and temperature (p = 0.001) were significantly higher in the anakinra group on alignment day. Following treatment, no relevant differences in kinetics of circulating cytokines were observed between both groups. Decreases of clinical parameters, including temperature (p = 0.03), white blood cell counts (p = 0.02), and plasma levels of ferritin (p = 0.003), procalcitonin (p = 0.001), creatinine (p = 0.01), and bilirubin (p = 0.007), were more pronounced in the anakinra group. No differences in duration of mechanical ventilation or ICU length of stay were observed between groups. Sensitivity analyses confirmed these results. Conclusions Anakinra is effective in reducing clinical signs of hyperinflammation in critically ill COVID-19 patients. A randomized controlled trial is warranted to draw conclusion about the effects of anakinra on clinical outcomes.
Collapse
Affiliation(s)
- Emma J Kooistra
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Nicole J B Waalders
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Inge Grondman
- Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Nico A F Janssen
- Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Aline H de Nooijer
- Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Mihai G Netea
- Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Department of Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Frank L van de Veerdonk
- Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Esther Ewalds
- Department of Intensive Care Medicine, Bernhoven Hospital, 5406PT, Uden, The Netherlands
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands. .,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500HB, Nijmegen, The Netherlands.
| | | |
Collapse
|
141
|
Lombardi A, Trombetta E, Cattaneo A, Castelli V, Palomba E, Tirone M, Mangioni D, Lamorte G, Manunta M, Prati D, Ceriotti F, Gualtierotti R, Costantino G, Aliberti S, Scaravilli V, Grasselli G, Gori A, Porretti L, Bandera A. Early Phases of COVID-19 Are Characterized by a Reduction in Lymphocyte Populations and the Presence of Atypical Monocytes. Front Immunol 2020; 11:560330. [PMID: 33362757 PMCID: PMC7756112 DOI: 10.3389/fimmu.2020.560330] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/13/2020] [Indexed: 12/26/2022] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 is a recently discovered pathogen responsible of coronavirus disease 2019 (COVID-19). The immunological changes associated with this infection are largely unknown. Methods We evaluated the peripheral blood mononuclear cells profile of 63 patients with COVID-19 at diagnosis. We also assessed the presence of association with inflammatory biomarkers and the 28-day mortality. Results Lymphocytopenia was present in 51 of 63 (80.9%) patients, with a median value of 720 lymphocytes/µl (IQR 520-1,135). This reduction was mirrored also on CD8+ (128 cells/µl, IQR 55-215), natural killer (67 cells/µl, IQR 35-158) and natural killer T (31 cells/µl, IQR 11-78) cells. Monocytes were preserved in total number but displayed among them a subpopulation with a higher forward and side scatter properties, composed mainly of cells with a reduced expression of both CD14 and HLA-DR. Patients who died in the 28 days from admission (N=10, 15.9%), when compared to those who did not, displayed lower mean values of CD3+ (337.4 cells/µl vs 585.9 cells/µl; p=0.028) and CD4+ cells (232.2 cells/µl vs 381.1 cells/µl; p=0.042) and an higher percentage of CD8+/CD38+/HLA-DR+ lymphocytes (13.5% vs 7.6%; p=0.026). Discussion The early phases of COVID-19 are characterized by lymphocytopenia, predominance of Th2-like lymphocytes and monocytes with altered immune profile, which include atypical mononuclear cells.
Collapse
Affiliation(s)
- Andrea Lombardi
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Elena Trombetta
- Flow Cytometry and Cell Sorting Laboratory, Clinical Laboratory, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Alessandra Cattaneo
- Flow Cytometry and Cell Sorting Laboratory, Clinical Laboratory, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Valeria Castelli
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Emanuele Palomba
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Mario Tirone
- Flow Cytometry and Cell Sorting Laboratory, Clinical Laboratory, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Davide Mangioni
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milano, Italy
| | - Giuseppe Lamorte
- Translational Medicine and Biobank Unit, Department of Transfusion Medicine and Hematology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Maria Manunta
- Translational Medicine and Biobank Unit, Department of Transfusion Medicine and Hematology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Ferruccio Ceriotti
- Clinical Laboratory, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Roberta Gualtierotti
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Giorgio Costantino
- Emergency Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Stefano Aliberti
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
- Respiratory Unit and Cystic Fibrosis Adult Center, Internal Medicine Department, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Vittorio Scaravilli
- Departement of Anaesthesia, Critical care and Emergency, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Giacomo Grasselli
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
- Departement of Anaesthesia, Critical care and Emergency, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milano, Italy
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
| | - Laura Porretti
- Flow Cytometry and Cell Sorting Laboratory, Clinical Laboratory, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milano, Italy
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
| |
Collapse
|
142
|
"Cytokine storm", not only in COVID-19 patients. Mini-review. Immunol Lett 2020; 228:38-44. [PMID: 33007369 PMCID: PMC7524442 DOI: 10.1016/j.imlet.2020.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/10/2020] [Accepted: 09/22/2020] [Indexed: 01/09/2023]
Abstract
Cytokine storm is a form of uncontrolled systemic inflammatory reaction activated by a variety of factors and leading to a harmful homeostatic process, even to patient's death. Triggers that start the reaction are infection, systemic diseases and rarely anaphylaxis. Cytokine storm is frequently mentioned in connection to medical interventions such as transplantation or administration of drugs. Presented mini-review would like to show current possibilities how to fight or even stop such a life-threatening, immune-mediated process in order to save lives, not only in COVID-19 patients. Early identification of rising state and multilevel course of treatment is imperative. The most widely used molecule for systemic treatment remains tocilizumab. Except for anti IL-6 treatment, contemporary research opens the possibilities for combination of pharmaceutical, non-pharmaceutical and adjunctive treatment in a successful fight with consequences of cytokine storm. Further work is needed to discover the exact signaling pathways that lead to cytokine storm and to determine how these effector molecules and/or combination of processes can help to resolve this frequently fatal episode of inflammation. It is a huge need for all scientists and clinicians to establish a physiological rational for new therapeutic targets that might lead to more personalized medicine approaches.
Collapse
|
143
|
Kappert K, Jahić A, Tauber R. Assessment of serum ferritin as a biomarker in COVID-19: bystander or participant? Insights by comparison with other infectious and non-infectious diseases. Biomarkers 2020; 25:616-625. [PMID: 32700561 DOI: 10.1080/1354750x.2020.1797880] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The 2019 coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus has an impact on all aspects of patient care. Serum ferritin generally represents a biomarker of choice when iron deficiency is suspected. However, ferritin is also an acute-phase-protein exhibiting elevated serum concentration in various inflammatory diseases. Here we focus on the role of serum ferritin for diagnostic and clinical management of patients with COVID-19 in comparison with other infectious and non-infectious diseases. METHODS We examined scientific articles listed in PubMed reporting on ferritin in various infectious and non-infectious diseases. We then compared these results with nine current COVID-19 ferritin reports published in 2020. RESULTS Several non-infectious, as well as non-COVID-19 infectious diseases, are characterised by a partly dramatic elevation of serum ferritin levels. All COVID-19 studies published between February and May 2020, which documented laboratory serum ferritin, indicate ferritin as a biomarker of COVID-19 severity in hospitalised patients. CONCLUSIONS Serum ferritin may be considered both a prognostic and stratifying biomarker that can also contribute to therapeutic decision-making concerning patients with COVID-19. It should be emphasised, however, that most scientific reports refer to cohorts in the Asian region. Further validation in other cohorts is urgently required.
Collapse
Affiliation(s)
- Kai Kappert
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Amir Jahić
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Rudolf Tauber
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| |
Collapse
|
144
|
Vega MA, Simón-Fuentes M, González de la Aleja A, Nieto C, Colmenares M, Herrero C, Domínguez-Soto Á, Corbí ÁL. MAFB and MAF Transcription Factors as Macrophage Checkpoints for COVID-19 Severity. Front Immunol 2020; 11:603507. [PMID: 33312178 PMCID: PMC7708330 DOI: 10.3389/fimmu.2020.603507] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/19/2020] [Indexed: 01/10/2023] Open
Abstract
Defective IFN production and exacerbated inflammatory and pro-fibrotic responses are hallmarks of SARS-CoV-2 infection in severe COVID-19. Based on these hallmarks, and considering the pivotal role of macrophages in COVID-19 pathogenesis, we hypothesize that the transcription factors MAFB and MAF critically contribute to COVID-19 progression by shaping the response of macrophages to SARS-CoV-2. Our proposal stems from the recent identification of pathogenic lung macrophage subsets in severe COVID-19, and takes into consideration the previously reported ability of MAFB to dampen IFN type I production, as well as the critical role of MAFB and MAF in the acquisition and maintenance of the transcriptional signature of M-CSF-conditioned human macrophages. Solid evidences are presented that link overexpression of MAFB and silencing of MAF expression with clinical and biological features of severe COVID-19. As a whole, we propose that a high MAFB/MAF expression ratio in lung macrophages could serve as an accurate diagnostic tool for COVID-19 progression. Indeed, reversing the macrophage MAFB/MAF expression ratio might impair the exacerbated inflammatory and profibrotic responses, and restore the defective IFN type I production, thus becoming a potential strategy to limit severity of COVID-19.
Collapse
Affiliation(s)
- Miguel A. Vega
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | | | | | | | | | | | | | - Ángel L. Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| |
Collapse
|
145
|
Atemnkeng Ntam V, Klein A, Horneff G. Safety and efficacy of anakinra as first-line or second-line therapy for systemic onset juvenile idiopathic arthritis - data from the German BIKER registry. Expert Opin Drug Saf 2020; 20:93-100. [PMID: 33148061 DOI: 10.1080/14740338.2021.1843631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Background: The IL-1 receptor-antagonist anakinra is recommended for the treatment of systemic juvenile idiopathic arthritis (sJIA) and was recently approved for first-line treatment. Long-term data from clinical practise are scarce. Methods: SJIA patients from the German biologics in pediatric rheumatology (BIKER) registry starting anakinra were grouped into two cohorts: Patients in the first-line cohort received no prior sJIA treatment except NSAID and a maximum of 3 days of steroids. Second-line cohort patients were pre-treated with steroids; DMARDs or biologics. Patient characteristics, disease-activity parameters, efficacy, and safety-parameters were compared. Results: Until December 2018, 51 anakinra patients were documented, representing 117.96 patient-years. Mean disease duration was 3.5 (± 3.8) years. At baseline, all anakinra first-line users had active systemic disease compared to 82% in the second-line users. Significant JADAS-10 improvement at last follow-up was observed in both cohorts (p = 0.02, p = 0.0014). Substantial numbers of patients in both groups reached JADAS-MDA/JADAS-remission/inactive disease (66.7%50%50% in first-liners and 60%45%70% in second-liners). Rates of serious adverse events were comparable and consistent with the overall AE profile of anakinra in patients. Conclusion: This analysis adds to the established safety profile of anakinra and demonstrates that anakinra is effective as first-line or second-line treatment.
Collapse
Affiliation(s)
- V Atemnkeng Ntam
- Asklepios Clinic Sankt Augustin, Centre of Pediatric Rheumatology , Sankt Augustin, Germany
| | - A Klein
- Asklepios Clinic Sankt Augustin, Centre of Pediatric Rheumatology , Sankt Augustin, Germany.,Department of Pediatrics, University Clinic, University of Cologne , Germany
| | - G Horneff
- Asklepios Clinic Sankt Augustin, Centre of Pediatric Rheumatology , Sankt Augustin, Germany.,Department of Pediatrics, University Clinic, University of Cologne , Germany
| |
Collapse
|
146
|
Tavares J, Silva F, Castro A, Santos J, Rocha G, Almeida M, Martins LS, Dias L, Henriques AC, Cabrita A. Diagnostic challenge of recurrent macrophage activation syndrome before and after kidney transplant. Nefrologia 2020. [PMID: 33168264 DOI: 10.1016/j.nefro.2020.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Joana Tavares
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal.
| | - Filipa Silva
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Ana Castro
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Josefina Santos
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Guilherme Rocha
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Manuela Almeida
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - La Salete Martins
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Leonídio Dias
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| | | | - António Cabrita
- Serviço de Nefrologia do Centro Hospitalar Universitário do Porto, Porto, Portugal
| |
Collapse
|
147
|
Histopathologic and Autopsy Findings in Patients Diagnosed With Coronavirus Disease 2019 (COVID-19): What We Know So Far Based on Correlation With Clinical, Morphologic and Pathobiological Aspects. Adv Anat Pathol 2020; 27:363-370. [PMID: 32732586 DOI: 10.1097/pap.0000000000000276] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In response to the current outbreak of Coronavirus Disease-2019 (COVID-19), a fast body of literature emerged providing insights into the clinical and radiologic aspects of the novel disease, while the pathologic manifestations on tissue are yet to be sufficiently characterized mainly due to paucity of autopsy and biopsy of these cases. It is essential for both the clinicians and pathologists to maintain up-to-date knowledge of this continuously evolving topic in the midst of the current pandemic. Besides, understanding the impact of any disease in tissue pathology is crucial for better analysis of the pathogenesis and speculation of potential therapeutic targets. This review aimed to highlight the potential implication of COVID-19 in the pathology of various organ systems. We discuss the pathologic findings of the lungs, gastrointestinal tract, liver, brain, kidneys, heart, and the reproductive and immune systems that are associated with COVID-19. It seems that the respiratory, immune, and the digestive systems are the major targets of the disease. Mild mononuclear inflammatory cell infiltration is the most frequent histologic finding in general. Besides organ-specific changes, microthrombi, especially noticed in lungs, kidneys, and prostates, are the most significant observation microscopically. In addition, the possible mechanisms of organ injury were also reviewed.
Collapse
|
148
|
Samavedam S. Immunomodulation and COVID-19: Is There a Winning Combination? Indian J Crit Care Med 2020; 24:1015-1017. [PMID: 33384504 PMCID: PMC7751030 DOI: 10.5005/jp-journals-10071-23691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) has been testing the boundaries of science on several fronts. Pharmacotherapy has seen the highs of several drugs being tried as the best option as well as the lows of no drug proven to be effective. In this edition of the Indian Journal of Critical Care Medicine, Mahale et al. retrospectively evaluated a combination of drugs targeted at immunomodulation. HOW TO CITE THIS ARTICLE Samavedam S. Immunomodulation and COVID-19: Is There a Winning Combination? Indian J Crit Care Med 2020;24(11):1015-1017.
Collapse
Affiliation(s)
- Srinivas Samavedam
- Department of Critical Care, Virinchi Hospital, Hyderabad, Telangana, India
| |
Collapse
|
149
|
Nasonov E, Samsonov M. The role of Interleukin 6 inhibitors in therapy of severe COVID-19. Biomed Pharmacother 2020; 131:110698. [PMID: 32920514 PMCID: PMC7455113 DOI: 10.1016/j.biopha.2020.110698] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Cytokine storm syndrome (CSS) is a severe complication of inflammatory immune diseases or treatment of malignancies; it may also appear during the progression of COVID-19. CSS is caused by dysregulation of the synthesis of cytokines, including pro-inflammatory, regulatory, and anti-inflammatory cytokines and chemokines, leading to pathologic activation of innate and adaptive (Th1 and Th17 mediated) immunity. Interleukin-6 (IL-6) plays an important role in the pathogenesis of CSS. The significant role of IL-6 in pathogenesis of COVID-19 was confirmed in a range of studies, which showed that the plasma concentration of IL-6 was increased in patients with severe COVID-19. Currently, IL-6 inhibitor therapeutics are not yet approved for the treatment of COVID-19; however, these medicines, including tocilizumab (TCZ) are used off-label for the treatment of patients with severe COVID-19, including life-threatening conditions. The role of IL-6 in the pathogenesis of CSS during COVID-19 is important however, a number of related issues are not yet clear. These issues include the indications for treatment with IL-6 inhibitors, as well as the estimation of risk associated with the disease, outcomes, treatment options, and adverse drug reactions. The development of personalized immunomodulatory therapy, with respect to the role of cytokines in pathogenesis, requires the studies that aimed to find other relevant therapeutic targets for the treatment of CSS in patients with COVID-19. These therapeutic targets include inhibition of IL-1, IL-6, TNFα, GM-CSF, IFNγ, IL-17, IL-18, and also activation of the complement system. The challenge of CSS in patients with COVID-19 is identifying the correct scientific targets and developing clinical trials aimed to evaluate the pathogenesis and treat immune-mediated inflammatory diseases (IMIDs). Hopefully, the significant efforts of scientists and physicians across the globe will improve the prognosis in COVID-19 patients and provide useful information on IMIDs required to support the struggle for treating potential viral outbreaks, and treatment of well-known IMIDs.
Collapse
Affiliation(s)
- E Nasonov
- V.A. Nasonova Research Institute of Rheumatology, Moscow, Russia; I.M. Sechenov First Moscow State Medical University, MOH, Moscow, Russia; Kashirskoye roadway, 34А, 1115522, Moscow, Russia; Trubetskaya str, 8, bdg. 2, 2119991, Moscow, Russia.
| | - M Samsonov
- RPharm JSC, Leninsky prospect 111, 11942, Moscow, Russia.
| |
Collapse
|
150
|
Spihlman AP, Gadi N, Wu SC, Moulton VR. COVID-19 and Systemic Lupus Erythematosus: Focus on Immune Response and Therapeutics. Front Immunol 2020; 11:589474. [PMID: 33193418 PMCID: PMC7661632 DOI: 10.3389/fimmu.2020.589474] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/12/2020] [Indexed: 01/08/2023] Open
Abstract
The SARS-CoV-2 novel coronavirus has caused the COVID-19 pandemic with over 35 million cases and over a million deaths worldwide as of early October 2020. The populations most affected are the elderly and especially those with underlying comorbidities. In terms of race and ethnicity, black and hispanic populations are affected at disproportionately higher rates. Individuals with underlying conditions that cause an immune-compromised state are considered vulnerable to this infection. The immune response is an important determinant in viral infections including coronaviruses, not only in the antiviral defense but also in the disease progression, severity, and clinical outcomes of COVID-19. Systemic lupus erythematosus is a chronic autoimmune disease which also disproportionately afflicts black and hispanic populations. In lupus patients, an aberrant immune response is characterized by the presence of circulating autoantibodies, lymphopenia, aberrant T cells, and proinflammatory cytokines along with defective regulatory mechanisms, leading to immune-mediated damage to tissues. Lupus patients are often treated with immune-suppressants and therefore are immune-compromised and more susceptible to infections and may be vulnerable to coronavirus infection. While the anti-viral immune response is important to protect from coronavirus infection, an uncontrolled proinflammatory cytokine response can lead to cytokine storm which causes damage to the lungs and other organs, causing significant morbidity and mortality. Better understanding of the underlying immune response and therapeutic strategies in lupus and COVID-19 is important to guide management of this deadly infectious disease in the context of lupus and vice-versa.
Collapse
Affiliation(s)
- Allison P Spihlman
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Boston University School of Medicine, Boston, MA, United States
| | - Nirupa Gadi
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Boston University School of Medicine, Boston, MA, United States
| | - Samantha C Wu
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Boston University School of Medicine, Boston, MA, United States
| | - Vaishali R Moulton
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|