1
|
Søe-Jensen P, Clausen NE, Bestle MH, Andersen LPK, Lange T, Johansson PI, Stensballe J. Efficacy and safety of a 72-h infusion of prostacyclin (1 ng/kg/min) in mechanically ventilated patients with pulmonary infection and endotheliopathy-protocol for the multicenter randomized, placebo-controlled, blinded, investigator-initiated COMBAT-ARF trial. Acta Anaesthesiol Scand 2025; 69:e14565. [PMID: 39704260 DOI: 10.1111/aas.14565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Acute respiratory failure (ARF) is common in critically ill patients, and 50% of patients in intensive care units require mechanical ventilation [3, 4]. The COVID-19 pandemic revealed that COVID-19 infection induced ARF caused by damage to the microvascular pulmonary endothelium. In a randomized clinical trial, mechanically ventilated COVID-19 patients with severe endotheliopathy, as defined by soluble thrombomodulin (sTM) ≥ 4 ng/mL, were randomized to evaluate the effect of a 72-h infusion of low-dose prostacyclin 1 ng/kg/min or placebo. Twenty-eight-day mortality was 21.9% versus 43.6% in the prostacyclin and the placebo groups, respectively (RR 0.50; CI 0.24 to 0.96 p = .06). The aim of the current trial is to investigate if this beneficial effect and safety of prostacyclin also are present in any patient with suspected pulmonary infection requiring mechanical ventilation and concomitant severe endotheliopathy. MATERIALS AND METHODS This is a multi-center, randomized, blinded, clinical investigator-initiated phase 3 trial in mechanically ventilated patients with suspected pulmonary infection and severe endotheliopathy, as defined by sTM ≥4 ng/mL. Patients are randomized 1:1 to a 72-h infusion of low-dose prostacyclin (iloprost) 1 ng/kg/min or placebo (an equal volume of saline). Four-hundred fifty patients will be included. The primary endpoint is 28-day all-cause mortality. Secondary endpoints include 90-day mortality, days alive without vasopressor, mechanical ventilation, and renal replacement therapy in the ICU within 28 and 90 days, and the number of serious adverse reactions or serious adverse events within the first 7 days. DISCUSSION This trial will investigate the efficacy and safety of prostacyclin vs. placebo for 72-hours in mechanically ventilated patients with any suspected pulmonary infection and severe endotheliopathy, as defined by sTM ≥4 ng/mL. Trial endpoints focus on the potential effect of prostacyclin to reduce 28-day all-cause mortality.
Collapse
Affiliation(s)
- Peter Søe-Jensen
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
| | - Niels E Clausen
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Morten H Bestle
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital-North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lars P K Andersen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Anaesthesia and Intensive Care, Zealand University Hospital-Køge, Køge, Denmark
| | - Theis Lange
- Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Pär I Johansson
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- CAG Center for Endotheliomics, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Jakob Stensballe
- CAG Center for Endotheliomics, Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Anesthesiology, Surgery and Trauma Center, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
2
|
Culiat C, Soni D, Malkes W, Wienhold M, Zhang LH, Henry E, Dragan M, Kar S, Angeles DM, Eaker S, Biswas R. NELL1 variant protein (NV1) modulates hyper-inflammation, Th-1 mediated immune response, and the HIF-1α hypoxia pathway to promote healing in viral-induced lung injury. Biochem Biophys Res Commun 2025; 744:151198. [PMID: 39706056 DOI: 10.1016/j.bbrc.2024.151198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Research underscores the urgent need for technological innovations to treat lung tissue damage from viral infections and the lasting impact of COVID-19. Our study demonstrates the effectiveness of recombinant human NV1 protein in promoting a pro-healing extracellular matrix that regulates homeostasis in response to excessive tissue reactions caused by infection and injury. NV1 achieves this by calibrating multiple biological mechanisms, including reducing hyperinflammatory cytokine levels (e.g., IFN-γ, TNF-α, IL-10, and IP-10), enhancing the production of proteins involved in viral inactivation and clearance through endocytosis and phagocytosis (e.g., IL-9, IL-1α), regulating pro-clotting and thrombolytic pathways (e.g., downregulates SERPINE 1 and I-TAC during Th1-mediated inflammation), maintaining cell survival under hypoxic conditions via HIF-1α regulation through the M3K5-JNK-AP-1 and TSC2-mTOR pathways, and promoting blood vessel formation. Our findings reveal NV1 as a potential therapeutic candidate for treating severe lung injuries caused by inflammatory and hypoxic conditions from viral infections and related diseases.
Collapse
Affiliation(s)
| | - Dharmendra Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | | | - Mark Wienhold
- NellOne Therapeutics Inc., Knoxville, TN, 37931, USA
| | | | | | | | | | | | - Shannon Eaker
- NellOne Therapeutics Inc., Knoxville, TN, 37931, USA
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
3
|
Ayako RM, Patel K, Ndede I, Nordgren J, Larrson M, Mining SK. Inflammatory, Hematological, and Biochemical Biomarkers in COVID-19 Patients. Immun Inflamm Dis 2024; 12:e70078. [PMID: 39641395 PMCID: PMC11621974 DOI: 10.1002/iid3.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
INTRODUCTION There are few accurate prognostic indications of the illness's development and severity for COVID-19, despite certain biomarkers having been investigated. The unexpected nature of COVID-19's course, which can quickly progress from asymptomatic to life-threatening symptoms, lies at the heart of the disease's intricacy. Predicting SARS-CoV-2 pathogenicity through laboratory biomarkers and as such, identifying the patients' illness severity at the time of their initial admission would be crucial in improving patient care. In this study, we sought to evaluate the potential of hematological, biochemical, and inflammatory biomarkers in predicting the course of COVID-19 at a tertiary hospital in western Kenya. METHODS This cross-sectional study involved 48 COVID-19 patients (16 asymptomatic; 16 moderate symptomatic; and 16 severe symptomatic) and 48 age-sex-matched COVID-19-negative clients attending the Moi Teaching and Referral Hospital, Kenya. Demographic information, self-reported chronic illnesses, symptoms, and laboratory results were collected at recruitment. RESULTS Significantly, the severity of COVID-19 was associated with; hemoglobin (p < 0.0001), white blood cells (p = 0.0022), hematocrit (p < 0.0001), blood urea nitrogen (p = 0.01), blood sodium (p = 0.0002), potassium (p = 0.0483), C-reactive protein (p = 0.0002), and Lactate Dehydrogenase (p < 0.0001). Regression analysis of CRP revealed a strong positive correlation (p = 0.0006) whereas LDH revealed a weak positive correlation (p < 0.0001) with COVID-19 disease severity. Discriminative accuracy was highest when asymptomatic was compared to severe COVID-19 for CRP and LDH (AUC: 0.8867, 95% CI: 0.7532-1.000) and (AUC: 1.000, 95% CI: 1.000-1.000) respectively. CONCLUSION The hematological indices, inflammatory and biochemical biomarkers studied have the potential to predict the course of COVID-19. These parameters may be useful in helping design appropriate care for COVID-19 patients.
Collapse
Affiliation(s)
- Rebeccah M. Ayako
- Department of PathologyMoi UniversityEldoretKenya
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and VirologyLinköping UniversityLinköpingSweden
| | | | - Isaac Ndede
- Department of PathologyMoi UniversityEldoretKenya
| | - Johan Nordgren
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and VirologyLinköping UniversityLinköpingSweden
| | - Marie Larrson
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and VirologyLinköping UniversityLinköpingSweden
| | | |
Collapse
|
4
|
Cîrjaliu RE, Gurrala SV, Nallapati B, Krishna V, Oancea C, Tudorache E, Marc M, Bratosin F, Bogdan I, Rosca O, Barata PI, Hangan LT, Chirilă SI, Fildan AP. Prognostic Implications of Initial Radiological Findings of Pulmonary Fibrosis in Patients with Acute SARS-CoV-2 Infection: A Prospective Multicentric Study. Diseases 2024; 12:285. [PMID: 39589960 PMCID: PMC11592577 DOI: 10.3390/diseases12110285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/02/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Pulmonary fibrosis detected during the acute phase of SARS-CoV-2 infection may significantly influence patient prognosis. This study aimed to evaluate the prognostic value of initial high-resolution computed tomography (HRCT) findings of pulmonary fibrosis in hospitalized COVID-19 patients and to examine how these findings relate to disease severity and clinical outcomes, with a particular focus on the development and validation of predictive scoring systems. In this multicentric prospective cohort study from January 2023 to January 2024, 120 adult patients with confirmed SARS-CoV-2 infection requiring hospitalization were enrolled from two Romanian university hospitals. Patients were categorized based on the presence (n = 60) or absence (n = 60) of pulmonary fibrosis signs on admission HRCT scans, identified by reticular opacities, traction bronchiectasis, honeycombing, and architectural distortion. Biochemical analyses, severity scores (SOFA, APACHE II, NEWS 2), and novel compound scores combining clinical and radiological data were assessed. Patients with HRCT evidence of pulmonary fibrosis had significantly higher severity scores and worse clinical outcomes. The HRCT score alone was a strong predictor of severe COVID-19 (area under the ROC curve [AUC] = 0.885), with a best cutoff value of 9.72, yielding 85.7% sensitivity and 79.8% specificity. Compound Score 1, integrating SOFA, APACHE II, and HRCT scores, demonstrated excellent predictive performance with an AUC of 0.947, sensitivity of 92.5%, and specificity of 88.9%. Compound Score 2, combining systemic inflammation markers (SIRI, SII) and NEWS 2, also showed a strong predictive capability (AUC = 0.913), with 89.2% sensitivity and 85.7% specificity at the optimal cutoff. Regression analysis revealed that Compound Score 1 had the highest hazard ratio for severe COVID-19 outcomes (HR = 4.89; 95% CI: 3.40-7.05), indicating its superior prognostic value over individual markers and traditional severity scores. Initial HRCT findings of pulmonary fibrosis are significantly associated with increased disease severity in hospitalized COVID-19 patients. The HRCT score is a valuable prognostic tool, and, when combined with clinical severity scores into Compound Score 1, it enhances the prediction of severe COVID-19 outcomes with high sensitivity and specificity. These compound scores facilitate the early identification of high-risk patients, guiding clinical decision-making and optimizing patient management to improve outcomes.
Collapse
Affiliation(s)
- Roxana-Elena Cîrjaliu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (R.-E.C.); (L.T.H.); (S.I.C.); (A.-P.F.)
| | - Sri Vidhya Gurrala
- NRI Academy of Medical Sciences, NTR Health University, Mangalagiri Road, Chinakakani, Guntur, Andhra Pradesh 522503, India;
| | - Balaji Nallapati
- Department of General Medicine, Katuri Medical College and Hospital, Katuri City 522019, India;
| | - Vamsi Krishna
- Sri Devaraj Urs Medical College, Kolar 563101, India;
| | - Cristian Oancea
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.O.); (M.M.); (P.I.B.)
| | - Emanuela Tudorache
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.O.); (M.M.); (P.I.B.)
| | - Monica Marc
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.O.); (M.M.); (P.I.B.)
| | - Felix Bratosin
- Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (F.B.); (I.B.); (O.R.)
| | - Iulia Bogdan
- Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (F.B.); (I.B.); (O.R.)
| | - Ovidiu Rosca
- Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (F.B.); (I.B.); (O.R.)
| | - Paula Irina Barata
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.O.); (M.M.); (P.I.B.)
| | - Laurentiu Tony Hangan
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (R.-E.C.); (L.T.H.); (S.I.C.); (A.-P.F.)
| | - Sergiu Ioachim Chirilă
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (R.-E.C.); (L.T.H.); (S.I.C.); (A.-P.F.)
| | - Ariadna-Petronela Fildan
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (R.-E.C.); (L.T.H.); (S.I.C.); (A.-P.F.)
| |
Collapse
|
5
|
Ullah S, Noureddine Z, Sathian B, Narayanankutty K, Asirvatham T, Abubacker M, Omar M, Awadh MN, Al-Kuwari F, Saad R. Rehabilitation and functional outcomes of COVID-19 patients in a rehabilitation hospital in Qatar. Qatar Med J 2024; 2024:45. [PMID: 39372687 PMCID: PMC11450274 DOI: 10.5339/qmj.2024.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/23/2024] [Indexed: 10/08/2024] Open
Abstract
Background Patients recovering from severe COVID-19 infections have experienced prolonged cognitive, physical, and psychological sequelae, including cardiorespiratory and motor deconditioning, neurological deterioration, anxiety, and depression. The impact of rehabilitation post-acute COVID-19 infection was recognized in the literature, but studies assessing and quantifying specific functional outcomes were lacking. This study aims to describe the characteristics and quantify the changes in functional outcomes of patients admitted to Qatar Rehabilitation Institute (QRI) for inpatient rehabilitation (IPR) post-COVID-19 infection during a 10-month period in 2021. Methods This is a retrospective observational cohort study, which included individuals over 18 years of age with a documented COVID-19-positive diagnosis who were admitted to QRI for IPR due to COVID-19 complications. Data was collected by the investigators from January 1, 2021, until October 30, 2021. A total of 243 patients were included in this study. The changes in functional rehabilitation outcomes were assessed and quantified at both the patient's baseline (on admission to QRI) and after completion of IPR (on discharge). The duration of the IPR program varied based on each patient's baseline assessment. Patients were given a total of 8-12 weeks to achieve their rehabilitation goals and were discharged once those goals were met. Several validated tools were utilized in this study including Functional Independence Measure (FIM), Berg Balance Scale (BBS), Dynamic Gait Index (DGI), Modified Medical Research Council (mMRC) Dyspnea Scale, Mini-Mental State Examination (MMSE), and Right- and left-Hand Grip Strength. In addition, patients' diet, the need for respiratory support, and the presence of a tracheostomy tube before and after IPR were also recorded. Results In total, 84.4% of the included patients were males (n = 205); with a mean age of 52.44 ± 12.99 years. The most commonly reported comorbidities were type 2 diabetes (62.1%) and hypertension (49.8%) with 83.5% of patients experiencing critical illness neuromyopathy. The average patients' length of stay in QRI was 33.92 ± 27.72 days. A statistically significant improvement in all functional outcome scales was noted following the completion of the IPR program (p = 0.001). The number of patients requiring modification to their diet or feeding via nasogastric tube (NGT) significantly decreased by 35% and 93%, respectively (p = 0.001). Patients requiring respiratory support decreased by 98% (p = 0.001) and the need for a tracheostomy tube among patients was reduced by 95% (p = 0.001). Conclusion IPR following COVID-19 infection was associated with significant functional, motor, and cardiorespiratory improvement. Dedicating clinics for post-COVID-19 rehabilitation would ensure improved patient outcomes and enhanced recovery.
Collapse
Affiliation(s)
- Sami Ullah
- Department of Physical Medicine and Rehabilitation, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
- MetroHealth Rehabilitation Institute of Ohio-Case Western Reserve University, Cleveland, OH, United States
| | - Zahra Noureddine
- Clinical Pharmacy Department, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar *
| | - Brijesh Sathian
- Geriatrics and Long-Term Care Department, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Krishnaprasad Narayanankutty
- Department of Physical Medicine and Rehabilitation, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
| | - Thajus Asirvatham
- Occupational Therapy Department, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
| | - Muhaiadeen Abubacker
- Physiotherapy Department, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
| | - Moustafa Omar
- Nursing Department, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
| | - Mohammed Nassir Awadh
- Department of Physical Medicine and Rehabilitation, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
| | - Fatima Al-Kuwari
- Department of Physical Medicine and Rehabilitation, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
| | - Rafat Saad
- Department of Physical Medicine and Rehabilitation, Qatar Rehabilitation Institute, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
6
|
Karim ZA, Reese RA, Smith AN, Blackadar ME, Arora V, Moore NM, Johnson EA. Positive impact of nutrition in the prevention of peripheral vascular disease and severe acute respiratory syndrome coronavirus 2: review. Front Nutr 2024; 11:1418028. [PMID: 39364158 PMCID: PMC11448360 DOI: 10.3389/fnut.2024.1418028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
Recent research has shown that there is a link between the trend of cardiovascular disease (CVD), chronic symptoms of SARS-CoV-2 (COVID-19), and medical nutrition therapy. Making positive changes to an individual's lifestyle can help to reduce the symptoms that follow exposure to CVD and COVID-19. Sustainable nutrition and lifestyle changes can positively impact an individual's health. Studies have considered the risk factors associated with the disease, medical history, the link between nutrition and peripheral vascular disease (PVD), symptom management, and the interrelationship between nutrition, COVID-19, and PVD. One study has demonstrated that Western Dietary intake can boost the innate immune system while suppressing humoral response, causing chronic inflammation and poor host defense against viruses. However, further investigation is needed to confirm. Patients with PVD and COVID-19 have experienced a reduction in side effects when prescribed a regimen of medical nutrition therapy, heart-healthy diets, and adequate physical activity before and after symptoms of both diseases appear. This approach has proven to be a protective factor during the combination of both illnesses. Our findings indicate that balanced diet and lifestyle are essential in supporting an optimal immune system that can reduce the risk of virus load in individuals at risk of infection and symptoms from COVID-19 and PVD.
Collapse
Affiliation(s)
- Zubair A Karim
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Rebecca A Reese
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Adrianne N Smith
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Madeline E Blackadar
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Vishal Arora
- Department of Medicine: Cardiology, Wellstar MCG Health, Augusta University, Augusta, GA, United States
| | - Nicole M Moore
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Emily A Johnson
- Department of Nutrition and Dietetics, College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| |
Collapse
|
7
|
Buchynskyi M, Oksenych V, Kamyshna I, Budarna O, Halabitska I, Petakh P, Kamyshnyi O. Genomic insight into COVID-19 severity in MAFLD patients: a single-center prospective cohort study. Front Genet 2024; 15:1460318. [PMID: 39296547 PMCID: PMC11408174 DOI: 10.3389/fgene.2024.1460318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
This study investigated the influence of single nucleotide polymorphisms (SNPs) in genes associated with the interferon pathway (IFNAR2 rs2236757), antiviral response (OAS1 rs10774671, OAS3 rs10735079), and viral entry (ACE2 rs2074192) on COVID-19 severity and their association with nonalcoholic fatty liver disease (MAFLD). We did not observe a significant association between the investigated SNPs and COVID-19 severity. While the IFNAR2 rs2236757 A allele was correlated with higher creatinine levels upon admission and the G allele was correlated with lower band neutrophils upon discharge, these findings require further investigation. The distribution of OAS gene polymorphisms (rs10774671 and rs10735079) did not differ between MAFLD patients and non-MAFLD patients. Our study population's distribution of ACE2 rs2074192 genotypes and alleles differed from that of the European reference population. Overall, our findings suggest that these specific SNPs may not be major contributors to COVID-19 severity in our patient population, highlighting the potential role of other genetic factors and environmental influences.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Olena Budarna
- Department of Neurology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
8
|
Navas Boada P, Chamorro K, Ballaz S. Survival analysis of COVID-19 versus non-COVID-19 patients requiring intensive care for acute respiratory distress syndrome: An observational retrospective study. CANADIAN JOURNAL OF RESPIRATORY THERAPY : CJRT = REVUE CANADIENNE DE LA THERAPIE RESPIRATOIRE : RCTR 2024; 60:112-121. [PMID: 39268526 PMCID: PMC11392457 DOI: 10.29390/001c.122402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/07/2024] [Indexed: 09/15/2024]
Abstract
Background/Aim This study analyzed clinical factors impacting the survival of COVID-19 patients with acute respiratory distress síndrome, or ARDS (CARDS) to ICU compared to non-COVID-19 ARDS patients. Methods Clinical variables from 1,008 CARDS cases and 332 ARDS cases were computed using learning algorithms. The multivariable Cox proportional hazards regression models with the enter method evaluated risk factors and ICU mortality relationships. The survival analysis was completed with Kaplan-Meier and the log-rank tests. Results A Random Forest model revealed that mechanical ventilation-related factors, oxygenation, blood pH, superinfection, shock, and ICU length of stay have the greatest effects on ICU survival. According to a multivariate Cox model, reintubation and a high-flow nasal cannula were essential for survival in CARDS patients during the ICU stay. The length of stay in the ICU diminishes in patients older than 45 years, regardless of the source of ARDS. Conclusion This study gives recommendations for the respiratory care of ARDS in COVID-19 patients.
Collapse
Affiliation(s)
- Paulo Navas Boada
- School of Biological Sciences and Engineering Universidad Yachay Tech
| | - Kevin Chamorro
- School of Mathematics and Computational Sciences Universidad Yachay Tech
| | | |
Collapse
|
9
|
Yu Q, Zhou X, Kapini R, Arsecularatne A, Song W, Li C, Liu Y, Ren J, Münch G, Liu J, Chang D. Cytokine Storm in COVID-19: Insight into Pathological Mechanisms and Therapeutic Benefits of Chinese Herbal Medicines. MEDICINES (BASEL, SWITZERLAND) 2024; 11:14. [PMID: 39051370 PMCID: PMC11270433 DOI: 10.3390/medicines11070014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/20/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Cytokine storm (CS) is the main driver of SARS-CoV-2-induced acute respiratory distress syndrome (ARDS) in severe coronavirus disease-19 (COVID-19). The pathological mechanisms of CS are quite complex and involve multiple critical molecular targets that turn self-limited and mild COVID-19 into a severe and life-threatening concern. At present, vaccines are strongly recommended as safe and effective treatments for preventing serious illness or death from COVID-19. However, effective treatment options are still lacking for people who are at the most risk or hospitalized with severe disease. Chinese herbal medicines have been shown to improve the clinical outcomes of mild to severe COVID-19 as an adjunct therapy, particular preventing the development of mild to severe ARDS. This review illustrates in detail the pathogenesis of CS-involved ARDS and its associated key molecular targets, cytokines and signalling pathways. The therapeutic targets were identified particularly in relation to the turning points of the development of COVID-19, from mild symptoms to severe ARDS. Preclinical and clinical studies were reviewed for the effects of Chinese herbal medicines together with conventional therapies in reducing ARDS symptoms and addressing critical therapeutic targets associated with CS. Multiple herbal formulations, herbal extracts and single bioactive phytochemicals with or without conventional therapies demonstrated strong anti-CS effects through multiple mechanisms. However, evidence from larger, well-designed clinical trials is lacking and their detailed mechanisms of action are yet to be well elucidated. More research is warranted to further evaluate the therapeutic value of Chinese herbal medicine for CS in COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Qingyuan Yu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
- Xiyuan Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Rotina Kapini
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Anthony Arsecularatne
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Wenting Song
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Chunguang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Yang Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Junguo Ren
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Gerald Münch
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| |
Collapse
|
10
|
Anwar HM, Salem GEM, Abd El-Latief HM, Osman AAE, Ghanem SK, Khan H, Chavanich S, Darwish A. Therapeutic potential of proteases in acute lung injury and respiratory distress syndrome via TLR4/Nrf2/NF-kB signaling modulation. Int J Biol Macromol 2024; 267:131153. [PMID: 38574930 DOI: 10.1016/j.ijbiomac.2024.131153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/02/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
The COVID-19 pandemic has drawn attention to acute lung injury and respiratory distress syndrome as major causes of death, underscoring the urgent need for effective treatments. Protease enzymes possess a wide range of beneficial effects, including antioxidant, anti-inflammatory, antifibrotic, and fibrinolytic effects. This study aimed to evaluate the potential therapeutic effects of bacterial protease and chymotrypsin in rats in mitigating acute lung injury induced by lipopolysaccharide. Molecular docking was employed to investigate the inhibitory effect of bacterial protease and chymotrypsin on TLR-4, the receptor for lipopolysaccharide. Bacterial protease restored TLR-4, Nrf2, p38 MAPK, NF-kB, and IKK-β levels to normal levels, while chymotrypsin normalized TLR-4, IKK-β, IL-6, and IL-17 levels. The expression of TGF-β, caspase-3, and VEGF in the bacterial protease- and chymotrypsin-treated groups was markedly reduced. Our results suggest that both therapies ameliorate LPS-induced acute lung injury and modulate the TLR4/Nrf2/NF-k signaling pathway. Each protease exhibited distinct mechanisms, with bacterial protease showing a better response to oxidative stress, edema, and fibrosis, whereas chymotrypsin provided a better response in the acute phase and innate immunity. These findings highlight the potential of each protease as a promising therapeutic option for acute lung injury and respiratory distress syndrome.
Collapse
Affiliation(s)
- Hend Mohamed Anwar
- Department of Biochemistry, Egyptian Drug Authority (EDA), Former National Organization for Drug Control and Research (NODCAR), Giza 11221, Egypt
| | - Gad Elsayed Mohamed Salem
- Reef Biology Research Group, Department of Marine Science, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Hanan M Abd El-Latief
- Zoology Department, Women's College for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| | - Amany Abd Elhameid Osman
- Zoology Department, Women's College for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| | - Sahar K Ghanem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Sohag University, Egypt
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| | - Suchana Chavanich
- Reef Biology Research Group, Department of Marine Science, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Aquatic Resources Research Institute, Chulalongkorn University, Bangkok, Thailand.
| | - Alshaymaa Darwish
- Biochemistry Department, Faculty of Pharmacy, Sohag University, Sohag, Egypt.
| |
Collapse
|
11
|
Trinh CD, Le VN, Le VNB, Pham NT, Le VD. Lung abnormalities on computed tomography of Vietnamese patients with COVID-19 and the association with medical variables. IJID REGIONS 2024; 10:183-190. [PMID: 38351902 PMCID: PMC10862005 DOI: 10.1016/j.ijregi.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
Objectives Patients with COVID-19 may experience a lung injury without presenting clinical symptoms. Early detection of lung injury in patients with COVID-19 is required to enhance prediction and prevent severe progression. Methods Lung lesions in patients with COVID-19 were defined using the Fleischner Society terminology. Chest computed tomography lesions and their correlation with demographic characteristics and medical variables were identified. Results Patients with mild and moderate COVID-19 had up to 45% lung injuries, whereas critical patients had 55%. However, patients with mild and moderate COVID-19 typically had low-level lung injuries. Ground-glass (68.1%), consolidation (48.8%), opacity (36.3%), and nodular (6.9%) lung lesions were the most prevalent in patients with COVID-19. Patients with COVID-19 infected with the Delta variant had worse lung injury than those infected with the Alpha and Omicron. People vaccinated with ≥2 doses showed a lower risk of lung injury than those vaccinated with <1 dose. Patients <18 years old were less likely to have a lung injury than patients >18 years old. The treatment outcomes were unaffected by the severity of the lung injury. Conclusion Patients with mild COVID-19 had a similar risk of lung injury as patients with severe COVID-19. Thus, using chest computed tomography to detect lung injury can enhance the treatment outcomes and reduce the patient's risk of pulmonary complications.
Collapse
Affiliation(s)
- Cong Dien Trinh
- Departments of Infectious Disease, Military Hospital 103, Hanoi, Vietnam
| | - Van Nam Le
- Departments of Infectious Disease, Military Hospital 103, Hanoi, Vietnam
| | | | - Ngoc Thach Pham
- Micobiology and Molecular Biology Department, National Hospital for Tropical Diseases, Hanoi, Vietnam
| | - Van Duyet Le
- Micobiology and Molecular Biology Department, National Hospital for Tropical Diseases, Hanoi, Vietnam
| |
Collapse
|
12
|
Edalat F, Khakpour N, Heli H, Letafati A, Ramezani A, Hosseini SY, Moattari A. Immunological mechanisms of the nucleocapsid protein in COVID-19. Sci Rep 2024; 14:3711. [PMID: 38355695 PMCID: PMC10867304 DOI: 10.1038/s41598-024-53906-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 02/06/2024] [Indexed: 02/16/2024] Open
Abstract
The emergence of corona virus disease 2019 (COVID-19), resulting from Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has left an indelible mark on a global scale, causing countless infections and fatalities. This investigation delves into the role of the SARS-CoV-2 nucleocapsid (N) protein within the HEK293 cells, shedding light on its influence over apoptosis, interferon signaling, and cytokines production. The N gene was amplified, inserted into the pAdTrack-CMV vector, and then transfected to the HEK293 cells. Changes in the expression of IRF3, IRF7, IFN-β, BAK, BAX, and BCL-2 genes were evaluated. The levels of proinflammatory cytokines of IL-6, IL-12, IL-1β, and TNF-α were also determined. The N protein exhibited an anti-apoptotic effect by modulating critical genes associated with apoptosis, including BAK, BAX, and BCL-2. This effect potentially prolonged the survival of infected cells. The N protein also played a role in immune evasion by suppressing the interferon pathway, evidenced by the downregulation of essential interferon regulatory factors of IRF3 and IRF7, and IFN-β expression. The N protein expression led to a substantial increase in the production of proinflammatory cytokines of IL-6, IL-12, IL-1β, and TNF-α. The N protein emerged as a versatile factor and was exerted over apoptosis, interferon signaling, and cytokine production. These findings carry potential implications for the development of targeted therapies to combat COVID-19 and mitigate its global health impact.
Collapse
Affiliation(s)
- Fahime Edalat
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Niloofar Khakpour
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Heli
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afagh Moattari
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
13
|
Eldesouki RE, Kishk RM, Abd El-Fadeal NM, Mahran RI, Kamel N, Riad E, Nemr N, Kishk SM, Mohammed EAM. Association of IL-10-592 C > A /-1082 A > G and the TNFα -308 G > A with susceptibility to COVID-19 and clinical outcomes. BMC Med Genomics 2024; 17:40. [PMID: 38287362 PMCID: PMC10826193 DOI: 10.1186/s12920-023-01793-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/31/2023] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Variation in host immune responses to SARS-CoV-2 is regulated by multiple genes involved in innate viral response and cytokine storm emergence like IL-10 and TNFa gene polymorphisms. We hypothesize that IL-10; -592 C > A and - 1082 A > G and TNFa-308 G > A are associated with the risk of SARS-COV2 infections and clinical outcome. METHODS Genotyping, laboratory and radiological investigations were done to 110 COVID-19 patients and 110 healthy subjects, in Ismailia, Egypt. RESULTS A significant association between the - 592 A allele, A containing genotypes under all models (p < 0.0001), and TNFa A allele with risk to infection was observed but not with the G allele of the - 1082. The - 592 /-1082 CG and the - 592 /-1082/ -308 CGG haplotypes showed higher odds in COVID-19 patients. Severe lung affection was negatively associated with - 592, while positive association was observed with - 1082. Higher D-dimer levels were strongly associated with the - 1082 GG genotype. Survival outcomes were strongly associated with the GA genotype of TNFa. -308 as well as AGG and AAA haplotypes. CONCLUSION IL-10 and TNFa polymorphisms should be considered for clinical and epidemiological evaluation of COVID-19 patients.
Collapse
Affiliation(s)
- Raghda E Eldesouki
- Genetics Unit, Histology Department, Faculty of Medicine, Suez Canal University, 41522, Ismailia, Egypt.
| | - Rania M Kishk
- Microbiology and immunology Department, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
| | - Noha M Abd El-Fadeal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
- Biochemistry Department, Ibn Sina National College for Medical Studies, Jeddah, Kingdom of Saudi Arabia
| | - Rama I Mahran
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
| | - Noha Kamel
- Clinical Pathology Department, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
| | - Eman Riad
- Pulmonology Unit, Internal Medicine Department, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
| | - Nader Nemr
- Endemic and Infectious Diseases Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Safaa M Kishk
- Pharmaceutical Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | | |
Collapse
|
14
|
Sapra L, Saini C, Das S, Mishra PK, Singh A, Mridha AR, Yadav PK, Srivastava RK. Lactobacillus rhamnosus (LR) ameliorates pulmonary and extrapulmonary acute respiratory distress syndrome (ARDS) via targeting neutrophils. Clin Immunol 2024; 258:109872. [PMID: 38113963 DOI: 10.1016/j.clim.2023.109872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/25/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Pulmonary and extrapulmonary acute respiratory distress syndrome (ARDS) is a life-threatening respiratory failure associated with high mortality. Despite progress in our understanding of the pathological mechanism causing the crippling illness, there are currently no targeted pharmaceutical treatments available for it. Recent discoveries have emphasized the existence of a potential nexus between gut and lung health fueling novel approaches including probiotics for the treatment of ARDS. We thus investigated the prophylactic-potential of Lactobacillus rhamnosus-(LR) in lipopolysaccharide (LPS)-induced pulmonary and cecal ligation puncture (CLP) induced extrapulmonary ARDS mice. Our in-vivo findings revealed that pretreatment with LR significantly ameliorated vascular-permeability (edema) of the lungs via modulating the neutrophils along with significantly reducing the expression of inflammatory-cytokines in the BALF, lungs and serum in both pulmonary and extrapulmonary mice-models. Interestingly, our ex-vivo immunofluorescence and flow cytometric data suggested that mechanistically LR via short chain fatty acids (butyrate being the most potent and efficient in ameliorating the pathophysiology of both pulmonary and extra-pulmonary ARDS) targets the phagocytic and neutrophils extracellular traps (NETs) releasing potential of neutrophils. Moreover, our in-vivo data further corroborated our ex-vivo findings and suggested that butyrate exhibits enhanced potential in ameliorating the pathophysiology of ARDS via reducing the infiltration of neutrophils into the lungs. Altogether, our study establishes the prophylactic role of LR and its associated metabolites in the prevention and management of both pulmonary and extrapulmonary ARDS via targeting neutrophils.
Collapse
Affiliation(s)
- Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Chaman Saini
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sneha Das
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pradyumna K Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, MP 462001, India
| | - Anurag Singh
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Asit R Mridha
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pardeep K Yadav
- Central Animal Facility, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
15
|
Fares K, El-Deeb MK, Elsammak O, Ouf A, Saeed HMS, Baess A, Elsammak M, El-Attar E. SNP (A > G - rs13057211) but not GT(n) polymorphism in HMOX-1 promotor gene is associated with COVID-19 mortality. BMC Pulm Med 2023; 23:514. [PMID: 38129860 PMCID: PMC10734135 DOI: 10.1186/s12890-023-02785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
INTRODUCTION COVID-19 causes severe inflammatory respiratory distress syndrome. The global pandemic caused millions of cases of morbidity and mortality worldwide. Patients may present with variable symptoms including dyspnea, fever, and GIT manifestations. The HMOX-1 gene is located on the long (q) arm of chromosome 22 at position 12.3. HMOX-1 is expressed in all mammalian tissues at basal levels and is considered as a stress response enzyme. HMOX-1 has a specific polymorphic site with variable GT(n) repeats at the promotor region. Several authors evaluated the HMOX-1 GT(n) promoter polymorphism in different inflammatory conditions. We evaluated HMOX-1 promoter polymorphism in relation to serum Hemoxygenase level and inflammatory makers (CRP, Ferritin, PCT, IL-6 and D-dimer) in patients affected by SARS-COV-2 disease. SUBJECTS AND METHODS Ninety patients confirmed to be infected with COVID-19 were followed up till the study end point (recovery and discharge or death). HMOX-1 promotor GT(n) polymorphism was evaluated using Sanger sequencing. HMOX-1 enzyme serum level was measured by ELISA and the level of different inflammatory markers was assessed by available commercial kits. RESULTS A novel Single nucleotide polymorphism (SNP) (A > G) - rs13057211 in the GT(n) region of HMOX-1 promoter gene was found in 40 (61.5%) COVID-19 patients out of the studied 65 patients. This (A > G) SNP was associated with higher mortality rate in COVID-19 as it was detected in 27 patients (75% of the patients who succumbed to the disease) (p = 0.021, Odds ratio = 3.7; 95% CI:1.29-10.56). Serum IL-6 (Interleuken-6) was positively correlated the length of Hospital Stay (LOHS) and procalcitonin (PCT); (p = 0.014, r: 0.651 and p < 0.001, r:0.997) respectively while negatively correlated with levels of HMOX-1 enzyme serum level (p = 0.013, r: -0.61). CRP correlated positively with LOHS (p = 0.021, r = 0.4), PCT (p = 0.044, r = 0.425) and age (p < 0.001, r = 0.685). Higher levels of D-Dimer and PCT were observed in patients with the long repeat. There was no significant difference between patients who recovered and those who died from COVID-19 as regards HMOX-1 level and GT(n) polymorphism. CONCLUSION We report a novel SNP (A > G, rs13057211) in the GT(n) region of HMOX-1 promoter gene that was associated with mortality in COVID-19 patients, however no significant difference was found in HMOX-1 serum level or HMOX-1 (GT)n repeats within the studied groups.
Collapse
Affiliation(s)
- Kerolos Fares
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Mona K El-Deeb
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al Jouf University, Sakakah, Saudi Arabia
| | - Omar Elsammak
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amged Ouf
- Department of Biology and Biotechnology Graduate Program, School of Sciences and Engineering (SSE), The American University in Cairo (AUC), New Cairo, Egypt
| | - Hesham Mahmoud Sayd Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research Alexandria University, Alexandria, Egypt
| | - Ayman Baess
- Department of Chest Diseases, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed Elsammak
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| | - Eman El-Attar
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
16
|
Santos TA, Oliveira JED, Fonseca CDD, Barbosa DA, Belasco AGDS, Miura CRM. Sepsis and COVID-19: outcomes in young adults in intensive care. Rev Bras Enferm 2023; 76:e20230037. [PMID: 38055486 DOI: 10.1590/0034-7167-2023-0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/23/2023] [Indexed: 12/08/2023] Open
Abstract
OBJECTIVE to verify sepsis incidence among young adults admitted to intensive care due to COVID-19 and to analyze its association with demographic, clinical and outcome variables. METHODS a quantitative, longitudinal, retrospective and analytical study, consisting of 58 adults aged 20 to 40 years in intensive care for SARS-CoV-2. It was carried out in a university hospital, from March 2020 to December 2021, with data collected from electronic medical records. RESULTS sepsis incidence was 65%. Sepsis was associated with acute kidney injury, use of vasoactive drugs and mechanical ventilation, being admitted to the emergency room, severity according to the Simplified Acute Physiology Score III and bacterial pulmonary co-infection, the latter being the most frequent etiology for sepsis. CONCLUSIONS there was a high sepsis incidence, with 42% of deaths, which points to the importance of investing in preventive measures, especially in relation to bacterial pulmonary coinfections.
Collapse
|
17
|
Angelidis CD, Georgalas I, Giachos I, Symeonidis C, Mani A, Rotsos T. A 30-Year-Old Man with a Recent History of COVID-19 Requiring Treatment with Corticosteroids Who Developed Bilateral Central Serous Chorioretinopathy During 7-Month Follow-Up. AMERICAN JOURNAL OF CASE REPORTS 2023; 24:e940241. [PMID: 38007612 PMCID: PMC10687741 DOI: 10.12659/ajcr.940241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 10/24/2023] [Accepted: 08/23/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Central serous chorioretinopathy (CSCR) involves a localized serous macular detachment, secondary to retinal pigment epithelial and choroidal vascular changes, which can be an adverse effect of corticosteroid use. Most CSCR cases resolve spontaneously, and normal vision returns, while some chronic cases can result in blindness. This report is of a 30-year-old man with a recent history of Corona virus disease (COVID)-19 requiring corticosteroid treatment who developed bilateral CSCR with unilateral fibrin and a 7-month follow-up. CASE REPORT A 30-year-old male patient presented with malaise and high fever. The patient tested positive for COVID-19, caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus and was admitted. During hospitalization, he received intravenous (IV) corticosteroids for 1 week (6 mg dexamethasone IV once daily). Following hospitalization, the patient received per os methylprednisolone 16 mg (16 mg once daily for 3 days, 8 mg once daily for 3 days, 4 mg once daily for 3 days, and 2 mg once daily for 3 days). One month later, the patient presented with bilateral visual acuity (VA) deterioration and acute CSCR. The diagnosis and follow-up were performed by optical coherence tomography (OCT) and fundus fluorescein angiography (FFA). The patient was followed-up for a period of 7 months, during which, although the VA improved and remained stable, the OCT findings were changing. CONCLUSIONS This report highlights the importance of timely ophthalmological examination in patients with sudden vision loss and identification of the association between corticosteroid use and CSCR, as well as the importance of a longer follow-up period.
Collapse
Affiliation(s)
| | - Ilias Georgalas
- 1 Department of Ophthalmology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Giachos
- 1 Department of Ophthalmology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Aikaterini Mani
- 1 Department of Ophthalmology, National and Kapodistrian University of Athens, Athens, Greece
| | - Tryfon Rotsos
- 1 Department of Ophthalmology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
18
|
Atefi N, Goodarzi A, Riahi T, Khodabandehloo N, Talebi Taher M, Najar Nobari N, Seirafianpour F, Mahdi Z, Baghestani A, Valizadeh R. Evaluation of the efficacy and safety of oral N-acetylcysteine in patients with COVID-19 receiving the routine antiviral and hydroxychloroquine protocol: A randomized controlled clinical trial. Immun Inflamm Dis 2023; 11:e1083. [PMID: 38018602 PMCID: PMC10659758 DOI: 10.1002/iid3.1083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND The current absence of gold-standard or all-aspect favorable therapies for COVID-19 renders a focus on multipotential drugs proposed to prevent or treat this infection or ameliorate its signs and symptoms vitally important. The present well-designed randomized controlled trial (RCT) sought to evaluate the efficacy and safety of N-acetylcysteine (NAC) as adjuvant therapy for 60 hospitalized Iranian patients with COVID-19. METHODS Two 30-person diets, comprising 15 single diets of Kaletra (lopinavir/ritonavir) + hydroxychloroquine (HCQ) with/without NAC (600 mg TDS) and atazanavir/ritonavir + HCQ with/without NAC (600 mg TDS), were administered in the study. RESULTS At the end of the study, a further decrease in C-reactive protein was observed in the NAC group (P = 0.008), and no death occurred in the atazanavir/ritonavir + HCQ + NAC group, showing that the combination of these drugs may reduce mortality. The atazanavir/ritonavir + HCQ and atazanavir/ritonavir + NAC groups exhibited the highest O2 saturation at the end of the study and a significant rise in O2 saturation following intervention commencement, including NAC (P > 0.05). Accordingly, oral or intravenous NAC, if indicated, may enhance O2 saturation, blunt the inflammation trend (by reducing C-reactive protein), and lower mortality in hospitalized patients with COVID-19. CONCLUSION The NAC could be more effective as prophylactic or adjuvant therapy in stable non-severe cases of COVID-19 with a particularly positive role in the augmentation of O2 saturation and faster reduction of the CRP level and inflammation or could be effective for better controlling of COVID-19 or its therapy-related side effects.
Collapse
Affiliation(s)
- Najmolsadat Atefi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of MedicineIran University of Medical SciencesTehranIran
| | - Azadeh Goodarzi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of MedicineIran University of Medical SciencesTehranIran
| | - Taghi Riahi
- Department of Internal Medicine, School of MedicineIran University of Medical SciencesTehranIran
| | - Niloofar Khodabandehloo
- Department of Geriatric Medicine, School of MedicineIran University of Medical SciencesTehranIran
| | - Mahshid Talebi Taher
- Department of Infectious Disease, School of Medicine, Antimicrobial Resistance Research Center, Immunology and Infectious Disease Research InstituteIran University of Medical SciencesTehranIran
| | - Niloufar Najar Nobari
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of MedicineIran University of Medical SciencesTehranIran
| | | | - Zeinab Mahdi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of MedicineIran University of Medical SciencesTehranIran
| | - Amir Baghestani
- Department of General Medicine, Rasool Akram Medical Complex, School of MedicineIran University of Medical SciencesTehranIran
| | | |
Collapse
|
19
|
Chujan S, Nakareangrit W, Suriyo T, Satayavivad J. Integrated Transcriptomics and Network Analysis of Potential Mechanisms and Health Effects of Convalescent COVID-19 Patients. Bioinform Biol Insights 2023; 17:11779322231206684. [PMID: 37881207 PMCID: PMC10594973 DOI: 10.1177/11779322231206684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/22/2023] [Indexed: 10/27/2023] Open
Abstract
Coronaviral disease 2019 (COVID-19) is a recent pandemic disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Currently, there are still cases of COVID-19 around the world that can develop into persistent symptoms after discharge. The constellation of symptoms, termed long COVID, persists for months and can lead to various diseases such as lung inflammation and cardiovascular disease, which may lead to considerable financial burden and possible risk to human health. Moreover, the molecular mechanisms underlying the post-pandemic syndrome of COVID-19 remain unclear. In this study, we aimed to explore the molecular mechanism, disease association, and possible health risks in convalescent COVID-19 patients. Gene expression data from a human convalescent COVID-19 data set was compared with a data set from healthy normal individuals in order to identify differentially expressed genes (DEGs). To determine biological function and potential pathway alterations, the GO and KEGG databases were used to analyze the DEGs. Disease association, tissue, and organ-specific analyses were used to identify possible health effects. A total of 250 DEGs were identified between healthy and convalescent COVID-19 subjects. The biological function alterations identified revealed cytokine interactions and increased inflammation through NF-κB1, RELA, JUN, STAT3, and SP1. Interestingly, the most significant pathways were cytokine-cytokine receptor interaction, altered lipid metabolism, and atherosclerosis that play a crucial role in convalescent COVID-19. In addition, we also found pneumonitis, dermatitis, and autoimmune diseases. Based on our study, convalescent COVID-19 is associated with inflammation in a variety of organs that could lead to autoimmune and inflammatory diseases, as well as atherosclerosis. These findings are a first step toward fully exploring the disease mechanisms in depth to understand the relationship between post-COVID-19 infection and potential health risks. This is necessary for the development of appropriate strategies for the prevention and treatment of long COVID.
Collapse
Affiliation(s)
- Suthipong Chujan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | | | - Tawit Suriyo
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| |
Collapse
|
20
|
Taher I, El-Masry E, Abouelkheir M, Taha AE. Anti‑inflammatory effect of metformin against an experimental model of LPS‑induced cytokine storm. Exp Ther Med 2023; 26:415. [PMID: 37559933 PMCID: PMC10407980 DOI: 10.3892/etm.2023.12114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/23/2023] [Indexed: 08/11/2023] Open
Abstract
Cytokine storm is one of the leading causes of death in patients with COVID-19. Metformin has been shown to inhibit the action of a wide range of proinflammatory cytokines such as IL-6, and TNF-α which may ultimately affect cytokine storm due to Covid-19. The present study analyzed the anti-inflammatory effect of oral and intraperitoneal (IP) metformin administration routes in a mouse model of lipopolysaccharide (LPS)-induced cytokine storm. A total of 60 female BALB/c mice were randomly assigned to one of six groups: i) Control; ii) LPS model; iii) oral saline + LPS; iv) oral metformin + LPS; v) IP saline + LPS; and vi) IP metformin + LPS. Metformin or saline were administered to the mice for 30 days, after which an IP injection of 0.5 mg/kg LPS induced a cytokine storm in the five treatment groups. Mice were sacrificed and serum cytokine levels were measured. Pretreatment of mice with either oral or IP metformin significantly reduced the increase in IL-1, IL-6 and TNF-α following LPS injection. Both metformin administration routes significantly reduced IL-1 and TNF-α levels, although IP metformin appeared to be significantly more effective at reducing IL-6 levels compared with oral metformin. Neither the oral or IP route of administration of metformin demonstrated a significant effect on IL-17 levels. Based on its ability to suppress the proinflammatory LPS-induced cytokine storm, metformin may have future potential benefits in ameliorating human diseases caused by elevated cytokine levels.
Collapse
Affiliation(s)
- Ibrahim Taher
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Eman El-Masry
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
- Department of Medical Microbiology and Immunology, College of Medicine, Menoufia University, Shebin El Koum 32511, Egypt
| | - Mohamed Abouelkheir
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed E. Taha
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
21
|
Song Y, Gou Y, Gao J, Chen D, Zhang H, Zhao W, Qian F, Xu A, Shen Y. Lomerizine attenuates LPS-induced acute lung injury by inhibiting the macrophage activation through reducing Ca 2+ influx. Front Pharmacol 2023; 14:1236469. [PMID: 37693893 PMCID: PMC10484514 DOI: 10.3389/fphar.2023.1236469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening lung diseases with high mortality rates, predominantly attributable to acute and severe pulmonary inflammation. Lomerizine (LMZ) is a calcium channel blocker previously used in preventing and treating migraine. Here, we found that LMZ inhibited inflammatory responses and lung pathological injury by reducing pulmonary edema, neutrophil infiltration and pro-inflammatory cytokine production in lipopolysaccharide (LPS)-induced ALI mice. In vitro experiments, upon treating with LMZ, the expression of interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α was attenuated in macrophages. The phosphorylation of p38 MAPK, ERK1/2, JNK, and NF-κB p65 was inhibited after LMZ treatment. Furthermore, LPS-induced Ca2+ influx was reduced by treating with LMZ, which correlated with inhibition of pro-inflammatory cytokine production. And L-type Ca2+ channel agonist Bay K8644 (BK) could restore cytokine generation. In conclusion, our study demonstrated that LMZ alleviates LPS-induced ALI and is a potential agent for treating ALI/ARDS.
Collapse
Affiliation(s)
- Yunduan Song
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Clinical Laboratory, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yusen Gou
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jiameng Gao
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Dongxin Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Haibo Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjuan Zhao
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Qian
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ajing Xu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Shen
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
22
|
Brogna B, Bignardi E, Megliola A, Laporta A, La Rocca A, Volpe M, Musto LA. A Pictorial Essay Describing the CT Imaging Features of COVID-19 Cases throughout the Pandemic with a Special Focus on Lung Manifestations and Extrapulmonary Vascular Abdominal Complications. Biomedicines 2023; 11:2113. [PMID: 37626610 PMCID: PMC10452395 DOI: 10.3390/biomedicines11082113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
With the Omicron wave, SARS-CoV-2 infections improved, with less lung involvement and few cases of severe manifestations. In this pictorial review, there is a summary of the pathogenesis with particular focus on the interaction of the immune system and gut and lung axis in both pulmonary and extrapulmonary manifestations of COVID-19 and the computed tomography (CT) imaging features of COVID-19 pneumonia from the beginning of the pandemic, describing the typical features of COVID-19 pneumonia following the Delta variant and the atypical features appearing during the Omicron wave. There is also an outline of the typical features of COVID-19 pneumonia in cases of breakthrough infection, including secondary lung complications such as acute respiratory distress disease (ARDS), pneumomediastinum, pneumothorax, and lung pulmonary thromboembolism, which were more frequent during the first waves of the pandemic. Finally, there is a description of vascular extrapulmonary complications, including both ischemic and hemorrhagic abdominal complications.
Collapse
Affiliation(s)
- Barbara Brogna
- Department of Interventional and Emergency Radiology, San Giuseppe Moscati Hospital, 83100 Avellino, Italy; (A.L.); (A.L.R.); (L.A.M.)
| | - Elio Bignardi
- Department of Radiology, Francesco Ferrari Hospital, ASL Lecce, 73042 Casarano, Italy;
| | - Antonia Megliola
- Radiology Unit, “Frangipane” Hospital, ASL Avellino, 83031 Ariano Irpino, Italy; (A.M.); (M.V.)
| | - Antonietta Laporta
- Department of Interventional and Emergency Radiology, San Giuseppe Moscati Hospital, 83100 Avellino, Italy; (A.L.); (A.L.R.); (L.A.M.)
| | - Andrea La Rocca
- Department of Interventional and Emergency Radiology, San Giuseppe Moscati Hospital, 83100 Avellino, Italy; (A.L.); (A.L.R.); (L.A.M.)
| | - Mena Volpe
- Radiology Unit, “Frangipane” Hospital, ASL Avellino, 83031 Ariano Irpino, Italy; (A.M.); (M.V.)
| | - Lanfranco Aquilino Musto
- Department of Interventional and Emergency Radiology, San Giuseppe Moscati Hospital, 83100 Avellino, Italy; (A.L.); (A.L.R.); (L.A.M.)
| |
Collapse
|
23
|
Schmidt F, Abdesselem HB, Suhre K, Vaikath NN, Sohail MU, Al-Nesf M, Bensmail I, Mashod F, Sarwath H, Bernhardt J, Schaefer-Ramadan S, Tan TM, Morris PE, Schenck EJ, Price D, Mohamed-Ali V, Al-Maadheed M, Arredouani A, Decock J, Blackburn JM, Choi AMK, El-Agnaf OM. Auto-immunoproteomics analysis of COVID-19 ICU patients revealed increased levels of autoantibodies related to the male reproductive system. Front Physiol 2023; 14:1203723. [PMID: 37520825 PMCID: PMC10374950 DOI: 10.3389/fphys.2023.1203723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Background: Coronavirus disease (COVID-19) manifests many clinical symptoms, including an exacerbated immune response and cytokine storm. Autoantibodies in COVID-19 may have severe prodromal effects that are poorly understood. The interaction between these autoantibodies and self-antigens can result in systemic inflammation and organ dysfunction. However, the role of autoantibodies in COVID-19 complications has yet to be fully understood. Methods: The current investigation screened two independent cohorts of 97 COVID-19 patients [discovery (Disc) cohort from Qatar (case = 49 vs. control = 48) and replication (Rep) cohort from New York (case = 48 vs. control = 28)] utilizing high-throughput KoRectly Expressed (KREX) Immunome protein-array technology. Total IgG autoantibody responses were evaluated against 1,318 correctly folded and full-length human proteins. Samples were randomly applied on the precoated microarray slides for 2 h. Cy3-labeled secondary antibodies were used to detect IgG autoantibody response. Slides were scanned at a fixed gain setting using the Agilent fluorescence microarray scanner, generating a 16-bit TIFF file. Group comparisons were performed using a linear model and Fisher's exact test. Differentially expressed proteins were used for KEGG and WIKIpathway annotation to determine pathways in which the proteins of interest were significantly over-represented. Results and conclusion: Autoantibody responses to 57 proteins were significantly altered in the COVID-19 Disc cohort compared to healthy controls (p ≤ 0.05). The Rep cohort had altered autoantibody responses against 26 proteins compared to non-COVID-19 ICU patients who served as controls. Both cohorts showed substantial similarities (r 2 = 0.73) and exhibited higher autoantibody responses to numerous transcription factors, immunomodulatory proteins, and human disease markers. Analysis of the combined cohorts revealed elevated autoantibody responses against SPANXN4, STK25, ATF4, PRKD2, and CHMP3 proteins in COVID-19 patients. The sequences for SPANXN4 and STK25 were cross-validated using sequence alignment tools. ELISA and Western blot further verified the autoantigen-autoantibody response of SPANXN4. SPANXN4 is essential for spermiogenesis and male fertility, which may predict a potential role for this protein in COVID-19-associated male reproductive tract complications, and warrants further research.
Collapse
Affiliation(s)
- Frank Schmidt
- Proteomics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Houari B. Abdesselem
- Proteomics Core Facility, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Hamad Bin Khalifa University (HBKU), Doha, Qatar
- Neurological Disorders Research Center, QBRI, HBKU, Qatar Foundation, Doha, Qatar
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Nishant N. Vaikath
- Neurological Disorders Research Center, QBRI, HBKU, Qatar Foundation, Doha, Qatar
| | | | - Maryam Al-Nesf
- Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Center of Metabolism and Inflammation, Division of Medicine, University College London, London, United Kingdom
| | - Ilham Bensmail
- Proteomics Core Facility, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Fathima Mashod
- Proteomics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Hina Sarwath
- Proteomics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Joerg Bernhardt
- Institute for Microbiology, University of Greifswald, Greifswald, Germany
| | | | - Ti-Myen Tan
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Sengenics Corporation, Damansara Heights, Kuala Lumpur, Malaysia
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Priscilla E. Morris
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Sengenics Corporation, Damansara Heights, Kuala Lumpur, Malaysia
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Edward J. Schenck
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York Presbyterian Hospital—Weill Cornell Medical Center, Weill Cornell Medicine, New York, NY, United States
| | - David Price
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York Presbyterian Hospital—Weill Cornell Medical Center, Weill Cornell Medicine, New York, NY, United States
| | - Vidya Mohamed-Ali
- Center of Metabolism and Inflammation, Division of Medicine, University College London, London, United Kingdom
- Anti-Doping Laboratory Qatar, Doha, Qatar
| | - Mohammed Al-Maadheed
- Center of Metabolism and Inflammation, Division of Medicine, University College London, London, United Kingdom
- Anti-Doping Laboratory Qatar, Doha, Qatar
| | - Abdelilah Arredouani
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Julie Decock
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Jonathan M. Blackburn
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Sengenics Corporation, Damansara Heights, Kuala Lumpur, Malaysia
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Augustine M. K. Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York Presbyterian Hospital—Weill Cornell Medical Center, Weill Cornell Medicine, New York, NY, United States
| | - Omar M. El-Agnaf
- Neurological Disorders Research Center, QBRI, HBKU, Qatar Foundation, Doha, Qatar
| |
Collapse
|
24
|
Dymicka-Piekarska V, Dorf J, Milewska A, Łukaszyk M, Kosidło JW, Kamińska J, Wolszczak-Biedrzycka B, Naumnik W. Neutrophil/Lymphocyte Ratio (NLR) and Lymphocyte/Monocyte Ratio (LMR) - Risk of Death Inflammatory Biomarkers in Patients with COVID-19. J Inflamm Res 2023; 16:2209-2222. [PMID: 37250103 PMCID: PMC10224725 DOI: 10.2147/jir.s409871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Aim The aim of our retrospective study was search for new prognostic parameters, which can help quickly and cheaply identify patients with risk for severe course of SARS-CoV-2 infection. Materials and Methods The following peripheral blood combination biomarkers were calculated: NLR (neutrophil/lymphocytes ratio), LMR (lymphocyte/monocyte ratio), PLR (platelet/lymphocyte ratio), dNLR (neutrophils/(white blood cells - neutrophils)), NLPR (neutrophil/(lymphocyte × platelet ratio)) in 374 patients who were admitted to the Temporary Hospital no 2 of Clinical Hospital in Bialystok (Poland) with COVID-19. The patients were divided into four groups depending on the severity of the course of COVID-19 using MEWS classification. Results The NLR and dNLR were significantly increased with the severity of COVID-19, according to MEWS score. The AUC for the assessed parameters was higher in predicting death in patients with COVID-19: NLR (0.656, p=0.0018, cut-off=6.22), dNLR (0.615, p=0.02, cut-off=3.52) and LMR (0.609, p=0.03, cut-off=2.06). Multivariate COX regression analysis showed that NLR median above 5.56 (OR: 1.050, P=0.002), LMR median below 2.23 (OR: 1.021, P=0.011), and age >75 years old (OR: 1.072, P=0.000) had a significant association with high risk of death during COVID-19. Conclusion Our results indicate that NLR, dNLR, and LMR calculated on admission to the hospital can quickly and easy identify patients with risk of a more severe course of COVID-19. Increase NLR and decrease LMR have a significant predictive value in COVID-19 patient's mortality and might be a potential biomarker for predicting death in COVID-19 patients.
Collapse
Affiliation(s)
| | - Justyna Dorf
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Białystok, Poland
| | - Anna Milewska
- Department of Biostatistics and Medical Informatics, Medical University of Bialystok, Białystok, Poland
| | - Mateusz Łukaszyk
- Temporary Hospital No 2 of Clinical Hospital in Bialystok, 1st Department of Lung Diseases and Tuberculosis, Medical University of Bialystok, Białystok, Poland
| | - Jakub Wiktor Kosidło
- Students Scientific Club at the Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Białystok, Poland
| | - Blanka Wolszczak-Biedrzycka
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Wojciech Naumnik
- Temporary Hospital No 2 of Clinical Hospital in Bialystok, 1st Department of Lung Diseases and Tuberculosis, Medical University of Bialystok, Białystok, Poland
| |
Collapse
|
25
|
Yudhawati R, Shimizu K. PGE2 Produced by Exogenous MSCs Promotes Immunoregulation in ARDS Induced by Highly Pathogenic Influenza A through Activation of the Wnt-β-Catenin Signaling Pathway. Int J Mol Sci 2023; 24:ijms24087299. [PMID: 37108459 PMCID: PMC10138595 DOI: 10.3390/ijms24087299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Acute respiratory distress syndrome is an acute respiratory failure caused by cytokine storms; highly pathogenic influenza A virus infection can induce cytokine storms. The innate immune response is vital in this cytokine storm, acting by activating the transcription factor NF-κB. Tissue injury releases a danger-associated molecular pattern that provides positive feedback for NF-κB activation. Exogenous mesenchymal stem cells can also modulate immune responses by producing potent immunosuppressive substances, such as prostaglandin E2. Prostaglandin E2 is a critical mediator that regulates various physiological and pathological processes through autocrine or paracrine mechanisms. Activation of prostaglandin E2 results in the accumulation of unphosphorylated β-catenin in the cytoplasm, which subsequently reaches the nucleus to inhibit the transcription factor NF-κB. The inhibition of NF-κB by β-catenin is a mechanism that reduces inflammation.
Collapse
Affiliation(s)
- Resti Yudhawati
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Indonesia-Japan Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60286, Indonesia
| | - Kazufumi Shimizu
- Indonesia-Japan Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60286, Indonesia
- Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
26
|
Gómez Duque M, Medina R, Enciso C, Beltran E, Hernandez K, Molano Franco D, Masclans JR. Usefulness of Inhaled Sedation in Patients With Severe ARDS Due to COVID-19. Respir Care 2023; 68:293-299. [PMID: 36414277 PMCID: PMC10027142 DOI: 10.4187/respcare.10371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Sedation in intensive care is fundamental for optimizing clinical outcomes. For many years the world has been facing high rates of opioid use, and to combat the increasing opioid addiction plans at both national and international level have been implemented.1 The COVID-19 pandemic posed a major challenge for health systems and also increased the use of sedatives and opioid analgesia for prolonged periods of time, and at high doses, in a significant proportion of patients. In our institutions, the shortage of many drugs for intravenous (IV) analgosedation forces us to alternatives to replace out-of-stock drugs or to seek sedation goals, which are difficult to obtain with traditional drugs at high doses.2 METHODS: This was an analytical retrospective cohort study evaluating the follow-up of subjects with inclusion criteria from ICU admission to discharge (alive or dead). Five end points were measured: need for high-dose opioids (≥ 200 µg/h), comparison of inhaled versus IV sedation of opioid analgesic doses, midazolam dose, need for muscle relaxant, and risk of delirium. RESULTS A total of 283 subjects were included in the study, of whom 230 were administered IV sedation and 53 inhaled sedation. In the inhaled sedation group, the relative risks (RRs) were 0.5 (95% CI 0.4-0.8, P = .045) for need of high-dose fentanyl, 0.3 (95% CI 0.20-0.45, P < .001) for need of muscle relaxant, and 0.8 (95% CI 0.61-1.15, P = .25) for risk of delirium. The median difference of fentanyl dose between the inhaled sedation and IV sedation groups was 61 µg/h or 1,200 µg/d (2.2 ampules/d, P < .001), and that of midazolam dose was 5.7 mg/h. CONCLUSIONS Inhaled sedation was associated with lower doses of opioids, benzodiazepines, and muscle relaxants compared to IV sedation. This therapy should be considered as an alternative in critically ill patients requiring prolonged ventilatory support and where IV sedation is not possible, always under adequate supervision of ICU staff.
Collapse
Affiliation(s)
- Mario Gómez Duque
- Service of Intensive Care Medicine, Hospital de San José, Fundación Universitaria de Ciencias de la Salud, Research Group CIMCA, Bogota, Colombia
| | - Ronald Medina
- Service of Intensive Care Medicine, Hospital de San José, Fundación Universitaria de Ciencias de la Salud, Research Group CIMCA, Bogota, Colombia
| | - Cesar Enciso
- Service of Intensive Care Medicine, Hospital de San José, Fundación Universitaria de Ciencias de la Salud, Research Group CIMCA, Bogota, Colombia
| | - Edgar Beltran
- Service of Intensive Care Medicine, Hospital de San José, Fundación Universitaria de Ciencias de la Salud, Research Group CIMCA, Bogota, Colombia
| | - Kevin Hernandez
- Service of Intensive Care Medicine, Hospital de San José, Fundación Universitaria de Ciencias de la Salud, Research Group CIMCA, Bogota, Colombia
| | - Daniel Molano Franco
- Service of Intensive Care Medicine, Hospital de San José, Los Cobos Medical Center, Research Group GRIBOS, Bogotá, Colombia.
| | - Joan R Masclans
- Service of Intensive Care Medicine, Hospital del Mar de Barcelona, IMIM (GREPAC), Department of Medicine (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
27
|
McCarthy SD, Rohde CB, Angel M, Masterson CH, MacLoughlin R, Fandiño J, González HE, Byrnes D, Laffey JG, O'Toole D. Aerosolized Pulmonary Delivery of mRNA Constructs Attenuates Severity of Escherichia coli Pneumonia in the Rat. Nucleic Acid Ther 2023; 33:148-158. [PMID: 36811461 PMCID: PMC10066785 DOI: 10.1089/nat.2022.0049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS), a rapid onset inflammatory lung disease with no effective specific therapy, typically has pathogenic etiology termed pneumonia. In previous studies nuclear factor-κB (NF-κB) inhibitor α super-repressor (IκBα-SR) and extracellular superoxide dismutase 3 (SOD3) reduced pneumonia severity when prophylactically delivered by viral vector. In this study, mRNA coding for green fluorescent protein, IκBα-SR, or SOD3 was complexed with cationic lipid, passed through a vibrating mesh nebulizer, and delivered to cell culture or directly to rats undergoing Escherichia coli pneumonia. Injury level was then assessed at 48 h. In vitro, expression was observed as early as 4 h in lung epithelial cells. IκBα-SR and wild-type IκBα mRNAs attenuated inflammatory markers, while SOD3 mRNA induced protective and antioxidant effects. In rat E. coli pneumonia, IκBα-SR mRNA reduced arterial carbon dioxide (pCO2) and reduced lung wet/dry ratio. SOD3 mRNA improved static lung compliance and alveolar-arterial oxygen gradient (AaDO2) and decreased bronchoalveolar lavage (BAL) bacteria load. White cell infiltration and inflammatory cytokine concentrations in BAL and serum were reduced by both mRNA treatments compared to scrambled mRNA controls. These findings indicate nebulized mRNA therapeutics are a promising approach to ARDS therapy, with rapid expression of protein and observable amelioration of pneumonia symptoms.
Collapse
Affiliation(s)
- Sean D McCarthy
- CÚRAM and Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | | | - Matt Angel
- Factor Bioscience Ltd., Cambridge, Massachusetts, USA
| | - Claire H Masterson
- CÚRAM and Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | | | - Juan Fandiño
- CÚRAM and Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | - Héctor E González
- CÚRAM and Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | - Declan Byrnes
- CÚRAM and Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | - John G Laffey
- CÚRAM and Regenerative Medicine Institute, University of Galway, Galway, Ireland
| | - Daniel O'Toole
- CÚRAM and Regenerative Medicine Institute, University of Galway, Galway, Ireland
| |
Collapse
|
28
|
Jarvas G, Szerenyi D, Jankovics H, Vonderviszt F, Tovari J, Takacs L, Foldes F, Somogyi B, Jakab F, Guttman A. Microbead-based extracorporeal immuno-affinity virus capture: a feasibility study to address the SARS-CoV-2 pandemic. Mikrochim Acta 2023; 190:95. [PMID: 36808576 PMCID: PMC9937867 DOI: 10.1007/s00604-023-05671-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/22/2023] [Indexed: 02/20/2023]
Abstract
In this paper, we report on the utilization of micro-technology based tools to fight viral infections. Inspired by various hemoperfusion and immune-affinity capture systems, a blood virus depletion device has been developed that offers highly efficient capture and removal of the targeted virus from the circulation, thus decreasing virus load. Single-domain antibodies against the Wuhan (VHH-72) virus strain produced by recombinant DNA technology were immobilized on the surface of glass micro-beads, which were then utilized as stationary phase. For feasibility testing, the virus suspension was flown through the prototype immune-affinity device that captured the viruses and the filtered media left the column. The feasibility test of the proposed technology was performed in a Biosafety Level 4 classified laboratory using the Wuhan SARS-CoV-2 strain. The laboratory scale device actually captured 120,000 virus particles from the culture media circulation proving the feasibility of the suggested technology. This performance has an estimated capture ability of 15 million virus particles by using the therapeutic size column design, representing three times over-engineering with the assumption of 5 million genomic virus copies in an average viremic patient. Our results suggested that this new therapeutic virus capture device could significantly lower virus load thus preventing the development of more severe COVID-19 cases and consequently reducing mortality rate.
Collapse
Affiliation(s)
- Gabor Jarvas
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, Veszprem, Hungary
| | - Dora Szerenyi
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, Veszprem, Hungary
| | - Hajnalka Jankovics
- Bio-Nanosystems Laboratory, Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, Veszprem, Hungary
| | - Ferenc Vonderviszt
- Bio-Nanosystems Laboratory, Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, Veszprem, Hungary
| | - Jozsef Tovari
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Laszlo Takacs
- Laboratory of Monoclonal Antibody Proteomics, Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Fanni Foldes
- National Virology Laboratory, BSL-4 Laboratory, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pecs, Pecs, Hungary
| | - Balazs Somogyi
- National Virology Laboratory, BSL-4 Laboratory, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pecs, Pecs, Hungary
| | - Ferenc Jakab
- National Virology Laboratory, BSL-4 Laboratory, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pecs, Pecs, Hungary
| | - Andras Guttman
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, Veszprem, Hungary.
| |
Collapse
|
29
|
Molecular imaging of chemokine-like receptor 1 (CMKLR1) in experimental acute lung injury. Proc Natl Acad Sci U S A 2023; 120:e2216458120. [PMID: 36626557 PMCID: PMC9934297 DOI: 10.1073/pnas.2216458120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The lack of techniques for noninvasive imaging of inflammation has challenged precision medicine management of acute respiratory distress syndrome (ARDS). Here, we determined the potential of positron emission tomography (PET) of chemokine-like receptor-1 (CMKLR1) to monitor lung inflammation in a murine model of lipopolysaccharide-induced injury. Lung uptake of a CMKLR1-targeting radiotracer, [64Cu]NODAGA-CG34, was significantly increased in lipopolysaccharide-induced injury, correlated with the expression of multiple inflammatory markers, and reduced by dexamethasone treatment. Monocyte-derived macrophages, followed by interstitial macrophages and monocytes were the major CMKLR1-expressing leukocytes contributing to the increased tracer uptake throughout the first week of lipopolysaccharide-induced injury. The clinical relevance of CMKLR1 as a biomarker of lung inflammation in ARDS was confirmed using single-nuclei RNA-sequencing datasets which showed significant increases in CMKLR1 expression among transcriptionally distinct subsets of lung monocytes and macrophages in COVID-19 patients vs. controls. CMKLR1-targeted PET is a promising strategy to monitor the dynamics of lung inflammation and response to anti-inflammatory treatment in ARDS.
Collapse
|
30
|
Al‐Kuraishy HM, Al‐Gareeb AI, Mohammed AA, Alexiou A, Papadakis M, Batiha GE. The potential link between Covid-19 and multiple myeloma: A new saga. Immun Inflamm Dis 2022; 10:e701. [PMID: 36444620 PMCID: PMC9673426 DOI: 10.1002/iid3.701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Covid-19 is considered a primary respiratory disease-causing viral pneumonia and, in severe cases, leads to acute lung injury and acute respiratory distress syndrome (ARDS). In addition, though, extra-pulmonary manifestations of Covid-19 have been shown. Furthermore, severe acute respiratory distress syndrome coronavirus type 2 (SARS-CoV-2) infection may coexist with several malignancies, including multiple myeloma (MM). METHODS This critical literature review aimed to find the potential association between SARS-CoV-2 infection and MM in Covid-19 patients with underlying MM. Narrative literature and databases search revealed that ARDS is developed in both MM and Covid-19 due to hypercalcemia and proteasome dysfunction. RESULTS Notably, the expression of angiogenic factors and glutamine deficiency could link Covid-19 severity and MM in the pathogenesis of cardiovascular complications. MM and Covid-19 share thrombosis as a typical complication; unlike thrombosis in Covid-19, which reflects disease severity, thrombosis does not reflect disease severity in MM. In both conditions, thromboprophylaxis is essential to prevent pulmonary thrombosis and other thromboembolic disorders. Moreover, Covid-19 may exacerbate the development of acute kidney injury and neurological complications in MM patients. CONCLUSION These findings highlighted that MM patients might be a risk group for Covid-19 severity due to underlying immunosuppression and most of those patients need specific management in the Covid-19 era.
Collapse
Affiliation(s)
- Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali A Mohammed
- The Chest Clinic, Barts Health NHS TrustWhipps Cross University HospitalLondonUK
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
31
|
Robert G, Bernhard JC, Capon G, Alezra E, Estrade V, Blanc P, Bladou F, Bensadoun H. Consequences of SARS-CoV-2 pandemic on urological surgery in France: a nationwide analysis of the healthcare system database. BMJ Open 2022; 12:e066220. [PMID: 36375970 PMCID: PMC9664026 DOI: 10.1136/bmjopen-2022-066220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
The SARS-CoV-2 outbreak overwhelmed the healthcare systems worldwide. Saturation of hospitals and the risk of contagion led to a reduction in the care of other diseases. OBJECTIVE To determine the impact of SARS-CoV-2 pandemic on urological surgery in France during the year 2020. DESIGN, SETTING AND PARTICIPANTS An observational descriptive study was conducted on anonymised data collected from the national healthcare database established each year as part of the Program for the Medicalization of Information Systems in Medicine, Surgery, Obstetrics and Odontology. INTERVENTION None. PRIMARY AND SECONDARY OUTCOME MEASURES We gathered the number of urology surgical procedures carried out between 2010 and 2019, and we observed the difference between the forecast and actual number of urological surgeries performed in 2020. RESULTS Urological surgeries decreased by 11.4%, non-oncological surgeries being more affected (-13.1%) than oncological ones (-4.1%). Among the most relevant surgeries, female urinary incontinence (-44.7%) and benign prostatic hyperplasia (-20.8%) were the most impacted ones, followed by kidney cancer (-9%), urolithiasis (-8.7%), radical cystectomy for bladder cancer (-6.1%), prostate cancer (-3.6%) and transurethral resection of bladder tumour (-2%). Public hospitals had a more reduced activity (-17.7%) than private ones (-9.1%). Finally, the distribution of the reduction in urological activities by region did not correspond to the regional burden of SARS-CoV-2. CONCLUSIONS Urological care was severely affected during SARS-CoV-2 pandemic. Even if oncological surgeries were prioritised, the longer it takes to receive appropriate care, the greater the risk on survival impact. TRIAL REGISTRATION The data collection and analysis was authorised by the French Data Protection Authority (CNIL) under the number1 861 282v2.
Collapse
Affiliation(s)
- Gregoire Robert
- Department of Urology, CHU de Bordeaux, Bordeaux, France
- Université de Bordeaux, Talence, France
| | | | - Grégoire Capon
- Department of Urology, CHU de Bordeaux, Bordeaux, France
| | - Eric Alezra
- Department of Urology, CHU de Bordeaux, Bordeaux, France
| | | | - Peggy Blanc
- Department of Urology, CHU de Bordeaux, Bordeaux, France
| | - Franck Bladou
- Department of Urology, CHU de Bordeaux, Bordeaux, France
- Université de Bordeaux, Talence, France
| | | |
Collapse
|
32
|
Todor SB, Bîrluțiu V, Topîrcean D, Mihăilă RG. Role of biological markers and CT severity score in predicting mortality in patients with COVID‑19: An observational retrospective study. Exp Ther Med 2022; 24:698. [PMID: 36277141 PMCID: PMC9535394 DOI: 10.3892/etm.2022.11634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/05/2022] [Indexed: 11/05/2022] Open
Abstract
COVID-19 pandemic is a continuing ongoing emergency of public concern. Early identification of markers associated with disease severity and mortality can lead to a prompter therapeutic approach. The present study conducted a multivariate analysis of different markers associated with mortality in order to establish their predictive role. Confirmed cases of 697 patients were examined. Demographic data, clinical symptoms and comorbidities were evaluated. Laboratory and imaging severity scores were reviewed. A total of 133 (19.1%) out of 697 patients succumbed during hospitalization. Obesity was the most common comorbidity, followed by hypertension, diabetes, coronary heart disease and chronic kidney disease. Compared with the survivor patients, non-survivors had a higher prevalence of diabetes, chronic kidney disease and coronary heart disease, as well as higher values of laboratory markers such as neutrophil-lymphocyte ratio (NLR), D-dimer, procalcitonin, IL-6 and C Reactive protein (CRP) and respectively high values of imaging severity scores. Multivariate regression analysis showed that high values of the proposed markers and chest computerized tomography (CT) severity imaging score were predictive for in hospital death: NLR [hazard ratio (HR): 3.127 confidence interval (CI) 95: 2.137-4.576]; D-dimer [HR: 6.223 (CI 95:3.809-10.167)]; procalcitonin [HR: 4.414 (CI 95:2.804-6.948)]; IL-6 [HR: 3.344 (CI 95:1.423-7.855)]; CRP [HR:2.997 (CI 95:1.940-4.630)]; and CT severity score [HR: 3.068 (CI 95:1.777-5.299)]. Laboratory markers and imaging severity scores could be used to stratify mortality risk in COVID-19 patients.
Collapse
Affiliation(s)
- Samuel-Bogdan Todor
- Pneumology Department, Pneumophtisiology Hospital Sibiu, Sibiu 550196, Romania
| | - Victoria Bîrluțiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Sibiu 550169, Romania
| | - Diana Topîrcean
- Hematology Department, Emergency County Clinical Hospital Sibiu, Sibiu 550245, Romania
| | | |
Collapse
|
33
|
Madurka I, Vishnevsky A, Soriano JB, Gans SJ, Ore DJS, Rendon A, Ulrik CS, Bhatnagar S, Krishnamurthy S, Mc Harry K, Welte T, Fernandez AA, Mehes B, Meiser K, Gatlik E, Sommer U, Junge G, Rezende E, Fernandez AA, Bagu AM, Amido FH, Costa MB, Brigante JA, Franco G, Ahmed NJ, Zerega N, Bacci MR, Fernandes CC, Ragognete HG, de Carvalho Rezende EA, Jaoude CVG, de Olivera EP, Malacize VQ, Stadnik CMB, Ramos EA, Kist GR, Barbosa GR, Filik H, Nalin S, Ulrik CS, Tidemandsen C, Hakansson K, Benfield T, Pedersen KBH, Welte T, Bachman M, Stoll M, Olzik I, Scharf N, Shearman N, Pink I, Frey A, Schulze P, Sayehli CM, Weismann D, Klinker H, Goebeler ME, Maier L, Geismann F, Hanses F, Zeller J, Hupf J, Lubnow M, Sag S, Ripfel S, Pabel S, Bauernfeind S, Leisner U, Hitzenbichler F, Madurka I, Iharos D, Toth KK, Hejja M, Esze T, Bhatnagar S, Mohan A, Pandit A, Kumar B, Ratre BK, Tiwari P, Singh R, Vig S, Bhopale S, Bhan S, Budhraja A, Agrawal A, Krishnamurthy S, Srikanth A, Kaneesan K, Unnithan MRJ, Srinivasan N, Velayuthaswamy N, Gounder SKM, Vaidyanathan V, Saha A, Bhattacharjee A, Datta A, Rendon A, Ortiz AC, Moncivais BS, Rodriguez BNL, Ramirez EJR, Perez ROF, Perez DLC, Osornio JS, Ortega MLM, Medina MAJ, Gans SJ, VanDen Berg JW, Boom L, Panhuis E, Lancee G, Lammens M, Boeve-Epping N, Ore DJS, Bustios ERM, Flores EMZ, Farronay MIM, Orihuela BG, del Pino RM, Vishnevsky AY, Morozov E, Repnikov I, Kiseleva M, Kotov ME, Terskikh MM, Zykov VA, Smolyarchuk EA, Kurguzova D, Garkavi DA, Messnikov O, Kharlamova S, Bondareva YA, Sementsov KV, Katagarov DN, Belekhov GA, Alferov SP, Martynenko TI, Vasileva E, Lazarenko IV, Gatalsky KK, Rudikh OV, Ganova OS, Paraeva OS, Pashkevich VV, Vishneva EM, Martynov AV, Isakova AP, Egorova EA, Gaygolnik TV, Pinzhina VN, Hinovker VV, Abramov VG, Ignatova GL, Blinova EV, Grebneva IV, Rodionova OV, Antonov VN, Trufanov KV, Krylov AA, Radchenko EN, McHarry K, Snyman E, Soriano J, Serrano DR, Vergara AM, Marcos MC, Viladomiu AS, Cardozo C, Garcia F. DFV890: a new oral NLRP3 inhibitor—tested in an early phase 2a randomised clinical trial in patients with COVID-19 pneumonia and impaired respiratory function. Infection 2022; 51:641-654. [PMID: 36104613 PMCID: PMC9473473 DOI: 10.1007/s15010-022-01904-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/27/2022] [Indexed: 01/08/2023]
Abstract
Background Coronavirus-associated acute respiratory distress syndrome (CARDS) has limited effective therapy to date. NLRP3 inflammasome activation induced by SARS-CoV-2 in COVID-19 contributes to cytokine storm. Methods This randomised, multinational study enrolled hospitalised patients (18–80 years) with COVID-19-associated pneumonia and impaired respiratory function. Eligible patients were randomised (1:1) via Interactive Response Technology to DFV890 + standard-of-care (SoC) or SoC alone for 14 days. Primary endpoint was APACHE II score at Day 14 or on day-of-discharge (whichever-came-first) with worst-case imputation for death. Other key assessments included clinical status, CRP levels, SARS-CoV-2 detection, other inflammatory markers, in-hospital outcomes, and safety. Findings Between May 27, 2020 and December 24, 2020, 143 patients (31 clinical sites, 12 countries) were randomly assigned to DFV890 + SoC (n = 71) or SoC alone (n = 72). Primary endpoint to establish clinical efficacy of DFV890 vs. SoC, based on combined APACHE II score, was not met; LSM (SE), 8·7 (1.06) vs. 8·6 (1.05); p = 0.467. More patients treated with DFV890 vs. SoC showed ≥ 1-level improvement in clinical status (84.3% vs. 73.6% at Day 14), earlier clearance of SARS-CoV-2 (76.4% vs. 57.4% at Day 7), and mechanical ventilation-free survival (85.7% vs. 80.6% through Day 28), and there were fewer fatal events in DFV890 group (8.6% vs. 11.1% through Day 28). DFV890 was well tolerated with no unexpected safety signals. Interpretation DFV890 did not meet statistical significance for superiority vs. SoC in primary endpoint of combined APACHE II score at Day 14. However, early SARS-CoV-2 clearance, improved clinical status and in-hospital outcomes, and fewer fatal events occurred with DFV890 vs. SoC, and it may be considered as a protective therapy for CARDS. Trial registration ClinicalTrials.gov, NCT04382053. Supplementary Information The online version contains supplementary material available at 10.1007/s15010-022-01904-w.
Collapse
|
34
|
Castillejos-López M, Torres-Espíndola LM, Huerta-Cruz JC, Flores-Soto E, Romero-Martinez BS, Velázquez-Cruz R, Higuera-Iglesias A, Camarena Á, Torres-Soria AK, Salinas-Lara C, Fernández-Plata R, Alvarado-Vásquez N, Solís-Chagoyán H, Ruiz V, Aquino-Gálvez A. Ivermectin: A Controversial Focal Point during the COVID-19 Pandemic. Life (Basel) 2022; 12:1384. [PMID: 36143420 PMCID: PMC9502658 DOI: 10.3390/life12091384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 01/08/2023] Open
Abstract
The SARS-CoV-2 pandemic has confirmed the apocalyptic predictions that virologists have been making for several decades. The challenge the world is facing is that of trying to find a possible treatment, and a viable and expedient option for addressing this challenge is the repurposing of drugs. However, in some cases, although these drugs are approved for use in humans, the mechanisms of action involved are unknown. In this sense, to justify its therapeutic application to a new disease, it is ideal, but not necessary, to know the basic mechanisms of action involved in a drug's biological effects. This review compiled the available information regarding the various effects attributed to Ivermectin. The controversy over its use for the treatment of COVID-19 is demonstrated by this report that considers the proposal unfeasible because the therapeutic doses proposed to achieve this effect cannot be achieved. However, due to the urgent need to find a treatment, an exhaustive and impartial review is necessary in order to integrate the knowledge that exists, to date, of the possible mechanisms through which the treatment may be helpful in defining safe doses and schedules of Ivermectin.
Collapse
Affiliation(s)
- Manuel Castillejos-López
- Departamento de Epidemiología y Estadística, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | | | - Juan Carlos Huerta-Cruz
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Bianca S. Romero-Martinez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | - Anjarath Higuera-Iglesias
- Departamento de Investigación en Epidemiología Clínica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Ángel Camarena
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Ana Karen Torres-Soria
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala UNAM, Mexico City 54090, Mexico
| | - Citlaltepetl Salinas-Lara
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala UNAM, Mexico City 54090, Mexico
| | - Rosario Fernández-Plata
- Departamento de Epidemiología y Estadística, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Noé Alvarado-Vásquez
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - Héctor Solís-Chagoyán
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Víctor Ruiz
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| |
Collapse
|
35
|
Robinson J, Santarelli A, Wilks N, Latu L, Charran O, Lalitsasivimol D, Dietrich T, Ashurst J. A Comparison of Inhaled Epoprostenol in Patients With Acute Respiratory Distress Syndrome and COVID-19-Associated Acute Respiratory Distress Syndrome. Cureus 2022; 14:e28274. [PMID: 36158384 PMCID: PMC9492349 DOI: 10.7759/cureus.28274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Acute respiratory distress syndrome (ARDS) and coronavirus disease 2019 (COVID-19)-associated acute respiratory distress syndrome (CARDS) are both characterized by non-cardiogenic pulmonary edema and severe hypoxemia that leads to a high percentage of patients suffering in-hospital mortality. Mechanistically, inhaled epoprostenol (iEPO) has shown a role in the treatment of ARDS and CARDS but little data are available directly comparing the two disease processes. Due to the lack of evidence of iEPO in ARDS and CARDS, the authors sought to compare the pulmonary effects of iEPO for mechanically ventilated patients with CARDS against a case match control of those with ARDS. Methods A retrospective cohort of all patients receiving iEPO between January 1, 2020, and February 22, 2022, was reviewed. Patients with ARDS were case-matched in a 2:1 allocation ratio of CARDS to ARDS by the number of medical comorbidities and age +/- 5 years. Clinical data collected included patient demographics, laboratory values, ventilator settings, length of hospitalization, and 28-day mortality. Comparisons of the effectiveness of iEPO between ARDS and CARDS were conducted using the chi-squared statistic for categorical variables and the Mann-Whitney statistic for continuous variables. Results A total of 72 patients were included in the final analysis, with 24 having ARDS and 48 CARDS. The number of medical comorbidities was no different for patients with ARDS or CARDs (p = 0.18), though the frequency of patients diagnosed with coronary artery disease (p=0.007), congestive heart failure (p=0.003), chronic obstructive pulmonary disease (p=0.004), and pulmonary hypertension (p=0.004) did vary between the two groups. A moderate but non-significant difference in pre-iEPO partial pressure of oxygen (PaO2)/fraction of inspired oxygen (FiO2) ratio was noted between the groups (0.74 vs 0.65; p=0.33). Following iEPO treatment, patients with ARDS showed a greater PaO2/FiO2 ratio than those with CARDS (0.87 vs 0.70; p=0.02). CARDS patients who received iEPO had a longer length of stay as compared to those with ARDS (17.5 vs 12.5 days; p=0.01). However, no difference was noted in 28-day mortality between the two groups (14 vs 34; p=0.29). Conclusion In this small sample from a single community hospital, a statistically significant improvement in the PaO2/FiO2 ratio was noted for both those with ARDS and CARDS. However, those with CARDS who were given iEPO had a longer length of stay without a significant difference in mortality as compared to those with traditional ARDS.
Collapse
|
36
|
Akbil B, Meyer T, Stubbemann P, Thibeault C, Staudacher O, Niemeyer D, Jansen J, Mühlemann B, Doehn J, Tabeling C, Nusshag C, Hirzel C, Sanchez DS, Nieters A, Lother A, Duerschmied D, Schallner N, Lieberum JN, August D, Rieg S, Falcone V, Hengel H, Kölsch U, Unterwalder N, Hübner RH, Jones TC, Suttorp N, Drosten C, Warnatz K, Spinetti T, Schefold JC, Dörner T, Sander LE, Corman VM, Merle U, Kurth F, von Bernuth H, Meisel C, Goffinet C. Early and Rapid Identification of COVID-19 Patients with Neutralizing Type I Interferon Auto-antibodies. J Clin Immunol 2022; 42:1111-1129. [PMID: 35511314 PMCID: PMC9069123 DOI: 10.1007/s10875-022-01252-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/14/2022] [Indexed: 12/16/2022]
Abstract
PURPOSE Six to 19% of critically ill COVID-19 patients display circulating auto-antibodies against type I interferons (IFN-AABs). Here, we establish a clinically applicable strategy for early identification of IFN-AAB-positive patients for potential subsequent clinical interventions. METHODS We analyzed sera of 430 COVID-19 patients from four hospitals for presence of IFN-AABs by ELISA. Binding specificity and neutralizing activity were evaluated via competition assay and virus-infection-based neutralization assay. We defined clinical parameters associated with IFN-AAB positivity. In a subgroup of critically ill patients, we analyzed effects of therapeutic plasma exchange (TPE) on the levels of IFN-AABs, SARS-CoV-2 antibodies and clinical outcome. RESULTS The prevalence of neutralizing AABs to IFN-α and IFN-ω in COVID-19 patients from all cohorts was 4.2% (18/430), while being undetectable in an uninfected control cohort. Neutralizing IFN-AABs were detectable exclusively in critically affected (max. WHO score 6-8), predominantly male (83%) patients (7.6%, 18/237 for IFN-α-AABs and 4.6%, 11/237 for IFN-ω-AABs in 237 patients with critical COVID-19). IFN-AABs were present early post-symptom onset and at the peak of disease. Fever and oxygen requirement at hospital admission co-presented with neutralizing IFN-AAB positivity. IFN-AABs were associated with lower probability of survival (7.7% versus 80.9% in patients without IFN-AABs). TPE reduced levels of IFN-AABs in three of five patients and may increase survival of IFN-AAB-positive patients compared to those not undergoing TPE. CONCLUSION IFN-AABs may serve as early biomarker for the development of severe COVID-19. We propose to implement routine screening of hospitalized COVID-19 patients for rapid identification of patients with IFN-AABs who most likely benefit from specific therapies.
Collapse
Affiliation(s)
- Bengisu Akbil
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tim Meyer
- Labor Berlin GmbH, Department of Immunology, Charité - Universitätsmedizin Berlin, Sylter Str. 2, 13353, Berlin, Germany
| | - Paula Stubbemann
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Charlotte Thibeault
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Olga Staudacher
- Labor Berlin GmbH, Department of Immunology, Charité - Universitätsmedizin Berlin, Sylter Str. 2, 13353, Berlin, Germany
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Daniela Niemeyer
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZIF German Centre for Infection Research (DZIF), Partner Site Charité, 10117, Berlin, Germany
| | - Jenny Jansen
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara Mühlemann
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZIF German Centre for Infection Research (DZIF), Partner Site Charité, 10117, Berlin, Germany
| | - Jan Doehn
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christoph Tabeling
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christian Nusshag
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Cédric Hirzel
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David Sökler Sanchez
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexandra Nieters
- University Medical Center Freiburg and Faculty of Medicine, University of Freiburg, Center for Biobanking, FREEZE-Biobank, Freiburg, Germany
| | - Achim Lother
- Cardiology and Medical Intensive Care, Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Duerschmied
- Cardiology and Medical Intensive Care, Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jan Nikolaus Lieberum
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dietrich August
- Division of Infectious Diseases, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Siegbert Rieg
- Division of Infectious Diseases, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Valeria Falcone
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Hartmut Hengel
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Uwe Kölsch
- Labor Berlin GmbH, Department of Immunology, Charité - Universitätsmedizin Berlin, Sylter Str. 2, 13353, Berlin, Germany
| | - Nadine Unterwalder
- Labor Berlin GmbH, Department of Immunology, Charité - Universitätsmedizin Berlin, Sylter Str. 2, 13353, Berlin, Germany
| | - Ralf-Harto Hübner
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Terry C Jones
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZIF German Centre for Infection Research (DZIF), Partner Site Charité, 10117, Berlin, Germany
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Downing St, Cambridge, CB2 3EJ, UK
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZIF German Centre for Infection Research (DZIF), Partner Site Charité, 10117, Berlin, Germany
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thibaud Spinetti
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse, CH-3010, Bern, Switzerland
| | - Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse, CH-3010, Bern, Switzerland
| | - Thomas Dörner
- Department of Medicine/Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- DRFZ, Berlin, Germany
| | - Leif Erik Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZIF German Centre for Infection Research (DZIF), Partner Site Charité, 10117, Berlin, Germany
- Labor Berlin GmbH, Berlin, Germany
| | - Uta Merle
- Department of Gastroenterology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and Department of Medicine I, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| | - Horst von Bernuth
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Labor Berlin GmbH, Department of Immunology, Charité - Universitätsmedizin Berlin, Sylter Str. 2, 13353, Berlin, Germany.
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany.
| | - Christian Meisel
- Labor Berlin GmbH, Department of Immunology, Charité - Universitätsmedizin Berlin, Sylter Str. 2, 13353, Berlin, Germany.
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany.
| | - Christine Goffinet
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
37
|
Raimondi F, Cazzaniga S, Annibali S, Novelli L, Brivio M, Pappacena S, Malandrino L, Bonaffini PA, Bianco I, Liggeri N, Gritti P, Lorini FL, Sironi S, Di Marco F. Extent and Distribution of Parenchymal Abnormalities in Baseline CT-Scans Do Not Predict Awake Prone Positioning Response in COVID-19 Related ARDS. Diagnostics (Basel) 2022; 12:diagnostics12081848. [PMID: 36010199 PMCID: PMC9406535 DOI: 10.3390/diagnostics12081848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/24/2022] [Accepted: 07/24/2022] [Indexed: 12/15/2022] Open
Abstract
Prone positioning is frequently used for non-intubated hypoxemic patients with COVID-19, although conclusive evidence is still lacking. The aim of the present study was to investigate whether baseline CT-scans could predict the improvement in oxygenation in COVID-19 related Acute respira-tory syndrome (ARDS) patients when pronated. Methods: A retrospective study of COVID-19 patients who underwent non-invasive ventilation (NIV) and prone positioning was conducted. Results: Forty-five patients were included. On average, 50% of the overall lung volume was affected by the disease, as observed in the CT-scans, with ground glass opacities (GGOs) and consolidations accounting for 44% and 4%, respectively. The abnormalities were mainly posterior, as demonstrated by posterior/anterior distribution ratios of 1.5 and 4.4 for GGO and consolidation, respectively. The median PaO2/FiO2 ratio during NIV in a supine position (SP1) was 140 [IQR 108–169], which improved by 67% (+98) during prone positioning, on average. Once supine positioning was resumed (SP2), the improvement in oxygenation was maintained in 28 patients (62% of the overall population, categorized as “responders”). We found no significant differences between responders and non-responders in terms of the extent (p = 0.92) and the distribution of parenchymal abnormalities seen in the baseline CT (p = 0.526). Conclusion: Despite the lack of a priori estimation of the sample size, considering the absence of any trends in the differences and correlations, we can reasonably conclude that the baseline chest CT-scan does not predict a gas-exchange response in awake prone-positioned patients with COVID-19 related ARDS. Physicians dealing with this category of patients should not rely on the imaging at presentation when evaluating whether to pronate patients.
Collapse
Affiliation(s)
- Federico Raimondi
- Pulmonary Medicine Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (F.R.); (L.N.); (S.P.); (L.M.)
| | - Sara Cazzaniga
- Department of Intensive Critical Care, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.C.); (M.B.); (P.G.); (F.L.L.)
| | - Simona Annibali
- Department of Diagnostic Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.A.); (P.A.B.); (I.B.); (N.L.); (S.S.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Luca Novelli
- Pulmonary Medicine Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (F.R.); (L.N.); (S.P.); (L.M.)
| | - Matteo Brivio
- Department of Intensive Critical Care, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.C.); (M.B.); (P.G.); (F.L.L.)
| | - Simone Pappacena
- Pulmonary Medicine Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (F.R.); (L.N.); (S.P.); (L.M.)
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
| | - Luca Malandrino
- Pulmonary Medicine Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (F.R.); (L.N.); (S.P.); (L.M.)
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
| | - Pietro Andrea Bonaffini
- Department of Diagnostic Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.A.); (P.A.B.); (I.B.); (N.L.); (S.S.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Ilaria Bianco
- Department of Diagnostic Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.A.); (P.A.B.); (I.B.); (N.L.); (S.S.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Noemi Liggeri
- Department of Diagnostic Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.A.); (P.A.B.); (I.B.); (N.L.); (S.S.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Paolo Gritti
- Department of Intensive Critical Care, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.C.); (M.B.); (P.G.); (F.L.L.)
| | - Ferdinando Luca Lorini
- Department of Intensive Critical Care, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.C.); (M.B.); (P.G.); (F.L.L.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Sandro Sironi
- Department of Diagnostic Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (S.A.); (P.A.B.); (I.B.); (N.L.); (S.S.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Fabiano Di Marco
- Pulmonary Medicine Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (F.R.); (L.N.); (S.P.); (L.M.)
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-035-2673456
| |
Collapse
|
38
|
Orendáčová M, Kvašňák E. Effects of vaccination, new SARS-CoV-2 variants and reinfections on post-COVID-19 complications. Front Public Health 2022; 10:903568. [PMID: 35968477 PMCID: PMC9372538 DOI: 10.3389/fpubh.2022.903568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Post-COVID-19 complications involve a variety of long-lasting health complications emerging in various body systems. Since the prevalence of post-COVID-19 complications ranges from 8-47% in COVID-19 survivors, it represents a formidable challenge to COVID-19 survivors and the health care system. Post-COVID-19 complications have already been studied in the connection to risk factors linked to their higher probability of occurrence and higher severity, potential mechanisms underlying the pathogenesis of post-COVID-19 complications, and their functional and structural correlates. Vaccination status has been recently revealed to represent efficient prevention from long-term and severe post-COVID-19 complications. However, the exact mechanisms responsible for vaccine-induced protection against severe and long-lasting post-COVID-19 complications remain elusive. Also, to the best of our knowledge, the effects of new SARS-CoV-2 variants and SARS-CoV-2 reinfections on post-COVID-19 complications and their underlying pathogenesis remain to be investigated. This hypothesis article will be dedicated to the potential effects of vaccination status, SARS-CoV-2 reinfections, and new SARS-CoV-2 variants on post-COVID-19 complications and their underlying mechanisms Also, potential prevention strategies against post-COVID complications will be discussed.
Collapse
Affiliation(s)
- Mária Orendáčová
- Department of Medical Biophysics and Medical Informatics, Third Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | | |
Collapse
|
39
|
Dysphagia incidence in intensive care unit patients with coronavirus disease 2019: retrospective analysis following systematic dysphagia screening. J Laryngol Otol 2022; 136:1278-1283. [DOI: 10.1017/s0022215122001517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Abstract
Objective
Post-extubation dysphagia in critically ill patients is known to affect about 18 per cent of mixed medical-surgical intensive care unit patients. This study investigated the incidence of post-extubation dysphagia in adult intensive care unit patients with coronavirus disease 2019.
Method
This study was a retrospective analysis of consecutive intensive care unit patients prospectively screened for dysphagia. Systematic screening of all extubated intensive care unit patients at our tertiary centre was performed using the Bernese intensive care unit dysphagia algorithm. The primary outcome measure was the incidence of post-extubation dysphagia.
Results
A total of 231 critically ill adult coronavirus disease 2019 positive patients were included, and 81 patients remained in the final analysis after exclusion criteria were applied (e.g. patients transferred). Dysphagia screening positivity was 25 of 81 (30.9 per cent), with 28.2 per cent (22 of 78) having confirmed dysphagia by specialist examination within 24 hours (n = 3 lost to follow up).
Conclusion
In this observational study, it was observed that the incidence of dysphagia in adult critically ill coronavirus disease 2019 patients was about 31 per cent (i.e. increased when compared with a historical pre-pandemic non-coronavirus disease 2019 intensive care unit cohort).
Collapse
|
40
|
Polastri M, Campanello S. Strokes of a pen: seeing far beyond COVID-19. Eur Respir J 2022; 59:59/6/2102264. [PMID: 35654452 DOI: 10.1183/13993003.02264-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 02/28/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Massimiliano Polastri
- Dept of Continuity of Care and Disability, Physical Medicine and Rehabilitation, St Orsola University Hospital, Bologna, Italy
| | - Salvatore Campanello
- Dept of Cardiac-Thoracic-Vascular Diseases, Unit of Pneumonology, St Orsola University Hospital, Bologna, Italy
| |
Collapse
|
41
|
Sagulkoo P, Suratanee A, Plaimas K. Immune-Related Protein Interaction Network in Severe COVID-19 Patients toward the Identification of Key Proteins and Drug Repurposing. Biomolecules 2022; 12:biom12050690. [PMID: 35625619 PMCID: PMC9138873 DOI: 10.3390/biom12050690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is still an active global public health issue. Although vaccines and therapeutic options are available, some patients experience severe conditions and need critical care support. Hence, identifying key genes or proteins involved in immune-related severe COVID-19 is necessary to find or develop the targeted therapies. This study proposed a novel construction of an immune-related protein interaction network (IPIN) in severe cases with the use of a network diffusion technique on a human interactome network and transcriptomic data. Enrichment analysis revealed that the IPIN was mainly associated with antiviral, innate immune, apoptosis, cell division, and cell cycle regulation signaling pathways. Twenty-three proteins were identified as key proteins to find associated drugs. Finally, poly (I:C), mitomycin C, decitabine, gemcitabine, hydroxyurea, tamoxifen, and curcumin were the potential drugs interacting with the key proteins to heal severe COVID-19. In conclusion, IPIN can be a good representative network for the immune system that integrates the protein interaction network and transcriptomic data. Thus, the key proteins and target drugs in IPIN help to find a new treatment with the use of existing drugs to treat the disease apart from vaccination and conventional antiviral therapy.
Collapse
Affiliation(s)
- Pakorn Sagulkoo
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Biomedical Informatics, Department of Family Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Apichat Suratanee
- Department of Mathematics, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand;
- Intelligent and Nonlinear Dynamics Innovations Research Center, Science and Technology Research Institute, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Kitiporn Plaimas
- Advance Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Omics Science and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence:
| |
Collapse
|
42
|
Firzli TR, Sathappan S, Siddiqui F. A Case of the Use of Extracorporeal Carbon Dioxide Removal in a Patient With COVID-19 Acute Respiratory Distress Syndrome. Cureus 2022; 14:e24645. [PMID: 35663663 PMCID: PMC9153857 DOI: 10.7759/cureus.24645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2022] [Indexed: 11/20/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe complication of coronavirus disease 2019 (COVID-19) infection marked by increased fluid diffusely in alveolar spaces. The management of ARDS can be complicated by mechanical hyperinflation, and thus a mainstay of treatment has included low tidal volume mechanical ventilation. This, however, can lead to ventilation-associated hypercapnia, which may result in respiratory acidosis. COVID-19-associated ARDS (CARDs) has been described in the literature, and guidelines tend to mimic ARDS management. However, the heterogeneous nature of COVID-19 pulmonary disease with respect to dead space, compliance, and shunting could alter guidelines. As low tidal volume remains a cornerstone in CARDS management, hypercapnic acidosis remains a risk. An emerging technology, extracorporeal CO2 removal devices (ECCO2R), has been granted emergency use authorization by the FDA for the management of CARDS. We present a 44-year-old male patient presenting positive for COVID-19. Following admission, the patient's oxygen status continually deteriorated and the patient went into acute respiratory distress, eventually requiring invasive mechanical ventilation. The patient became hypercapnic and acidotic due to low tidal volume ventilation. ECCO2R was used to manage the patient's hypercapnia, resulting in significant amelioration of his partial pressure of carbon dioxide (PCO2) and pH. The patient was eventually transferred to extracorporeal membrane oxygenation (ECMO) certified facility and survived after a prolonged hospital and rehabilitation course. In the management of CARDS patients who require mechanical respiration, there are many unanswered questions as to the appropriate ventilation strategy. Current practice recommends low tidal volume ventilation, carrying, and increased risk of hypercapnic respiratory acidosis as occurred in our patient. We believe that ECCO2R may be an appropriate bridge between low tidal volume ventilation and ECMO to stabilize acid-base disturbances in ventilated patients.
Collapse
|
43
|
Chernov AS, Minakov AA, Kazakov VA, Rodionov MV, Rybalkin IN, Vlasik TN, Yashin DV, Saschenko LP, Kudriaeva AA, Belogurov AA, Smirnov IV, Loginova SY, Schukina VN, Savenko SV, Borisevich SV, Zykov KA, Gabibov AG, Telegin GB. A new mouse unilateral model of diffuse alveolar damage of the lung. Inflamm Res 2022; 71:627-639. [PMID: 35434745 PMCID: PMC9013507 DOI: 10.1007/s00011-022-01568-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Objective and design The existing biological models of diffuse alveolar damage (DAD) in mice have many shortcomings. To offset these shortcomings, we have proposed a simple, nonsurgical, and reproducible method of unilateral total damage of the left lung in ICR mice. This model is based on the intrabronchial administration of a mixture of bacterial lipopolysaccharide (LPS) from the cell wall of S. enterica and α-galactosylceramide (inducing substances) to the left lung. Methods Using computer tomography of the lungs with endobronchial administration of contrast material, we have been able to perform an operative intravital verification of the targeted delivery of the inducer. The model presented is characterized by more serious and homogeneous damage of the affected lung compared to the existing models of focal pneumonia; at the same time, our model is characterized by longer animal survival since the right lung remains intact. Results The model is also characterized by diffuse alveolar damage of the left lung, animal survival of 100%, abrupt increases in plasma levels of TNFa, INFg, and IL-6, and significant myocardial overload in the right heart. It can be used to assess the efficacy of innovative drugs for the treatment of DAD and ARDS as the clinical manifestations that are developed in patients infected with SARS-CoV-2. Morphological patterns of lungs in the noninfectious (“sterile”) model of DAD induced by LPS simultaneously with α-galactosylceramide (presented here) and in the infectious model of DAD induced by SARS-CoV-2 have been compared. Conclusion The DAD model we have proposed can be widely used for studying the efficacy of candidate molecules for the treatment of infectious respiratory diseases, such as viral pneumonias of different etiology, including SARS-CoV-2.
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Invasive pulmonary aspergillosis (IPA) can affect patients with severe coronavirus disease 2019 (COVID-19), but many questions remain open about its very variable incidence across the world, the actual link between the viral infection and the fungal superinfection, the significance of Aspergillus recovery in a respiratory sample, and the management of such cases. This review addresses these questions and aims at providing some clues for the practical diagnostic and therapeutic approaches of COVID-19-associated pulmonary aspergillosis (CAPA) in a clinical perspective. RECENT FINDINGS Definitions have been proposed for possible/probable/proven CAPA, but distinction between colonization and invasive fungal infection is difficult and not possible in most cases in the absence of histopathological proof or positive galactomannan in serum. Most importantly, the recovery of an Aspergillus by a direct (culture, PCR) or indirect (galactomannan) test in a respiratory sample is an indicator of worse outcome, which justifies a screening for early detection and initiation of preemptive antifungal therapy in such cases. SUMMARY The COVID-19 pandemic has increased our awareness of IPA among ICU patients. Although current recommendations are mainly based on experts' opinions, prospective studies are needed to get more evidence-based support for the diagnostic approach and management of CAPA.
Collapse
|
45
|
Yuniarti L, Haerudin H, Triyani Y, Garna H, Dirgavarisya GB, Fernanda DR, Ramandhita AP, Karima H, Resa N, Tejasari M. SARS-CoV-2 Gene Expression as a Prognosis Predictor for COVID-19. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.7667] [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
Introduction: Real time quantitative PCR is the gold standard for detection of SARS-CoV-2 which is specific, sensitive, and simple quantitative. The target of RT-qPCR is to assess the expression level of the SARS-CoV-2 gene through cycle threshold values (CT-value). The purpose of this study was to analyze the association of the level of SARS-CoV-2 gene expression and the severity of COVID-19 in patients hospitalized.
Method: This research is an analytic observational study with cross sectional method. While the research sample was taken using a consecutive sampling technique from the Medical Records of Sumedang Hospital and Cideres Hospital, West Java, Indonesia from December 2020 to March 2021. Patient parameters include analysis of age, sex, comorbidity, and disease severity. The severity of the patient is classified based on complaints and oxygen saturation. The expression level of the SARS-CoV-2 N gene and E gene were assessed by calculating the relative quantification by comparing the expression of the E and N gene with the expression of the internal control gene by Livak formula (2-ΔΔCT Formula).
Result: The Spearman correlation test showed that there was a relationship between the expression of SARS-CoV-2 genes E and N genes with the severity of COVID-19 patients (with r=0.374 and p<0.0001) and (with r=0.452 and p<0.0001).
Conclusions: There is an correlation between the level of expression of genes E and gene N with the severity of patients.
Collapse
|
46
|
Hussain M, Khurram Syed S, Fatima M, Shaukat S, Saadullah M, Alqahtani AM, Alqahtani T, Bin Emran T, Alamri AH, Barkat MQ, Wu X. Acute Respiratory Distress Syndrome and COVID-19: A Literature Review. J Inflamm Res 2022; 14:7225-7242. [PMID: 34992415 PMCID: PMC8710428 DOI: 10.2147/jir.s334043] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an overwhelming inflammatory disorder of the lung due to direct and indirect insults to the lungs. ARDS is characterized by increased vascular permeability, protein-rich edema, diffuse alveolar infiltrate, and loss of aerated lung tissue, leading to decreased lung compliance, tachypnea, and severe hypoxemia. COVID-19 is generally associated with ARDS, and it has gained prime importance since it started. The mortality rate is alarmingly high in COVID-19-related ARDS patients regardless of advances in mechanical ventilation. Several pharmacological agents, including corticosteroids, nitric oxide, neuromuscular blocker, anti-TNF, statins, and exogenous surfactant, have been studied and some are under investigation, like ketoconazole, lisofylline, N-acetylcysteine, prostaglandins, prostacyclin, and fish oil. The purpose of this review is to appraise the understanding of the pathophysiology of ARDS, biomarkers, and clinical trials of pharmacological therapies of ARDS and COVID-19-related ARDS.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Shahzada Khurram Syed
- Department of Basic Medical Sciences, School of Health Sciences, University of Management and Technology Lahore, Lahore, 54000, Pakistan
| | - Mobeen Fatima
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Saira Shaukat
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Malik Saadullah
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, 38000, Pakistan
| | - Ali M Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Ali H Alamri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Muhammad Qasim Barkat
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City, 310000, People's Republic of China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City, 310000, People's Republic of China
| |
Collapse
|
47
|
Zöllkau J, Hagenbeck C, Hecher K, Pecks U, Schlembach D, Simon A, Schlösser R, Schleußner E. [Recommendations for SARS-CoV-2/COVID-19 during Pregnancy, Birth and Childbed - Update November 2021 (Long Version)]. Z Geburtshilfe Neonatol 2021; 226:e1-e35. [PMID: 34918334 DOI: 10.1055/a-1688-9398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the onset of the SARS-CoV-2 pandemic, the German Society of Gynecology and Obstetrics and the Society for Peri-/Neonatal Medicine have published and repeatedly updated recommendations for the management of SARS-CoV-2 positive pregnancies and neonates. As a continuation of existing recommendations, the current update addresses key issues related to the prenatal, perinatal, and postnatal care of pregnant women, women who have given birth, women who have recently given birth, women who are breastfeeding with SARS-CoV-2 and COVID-19, and their unborn or newborn infants, based on publications through September 2021. Recommendations and opinions were carefully derived from currently available scientific data and subsequently adopted by expert consensus. This guideline - here available in the long version - is intended to be an aid to clinical decision making. Interpretation and therapeutic responsibility remain with the supervising local medical team, whose decisions should be supported by these recommendations. Adjustments may be necessary due to the rapid dynamics of new evidence. The recommendations are supported by the endorsement of the professional societies: German Society for Perinatal Medicine (DGPM), German Society of Gynecology and Obstetrics (DGGG), German Society for Prenatal and Obstetric Medicine (DGPGM), German Society for Pediatric Infectiology (DGPI), Society for Neonatology and Pediatric Intensive Care Medicine (GNPI).
Collapse
Affiliation(s)
- Janine Zöllkau
- Klinik für Geburtsmedizin, Universitätsklinikum Jena, Deutschland
| | - Carsten Hagenbeck
- Klinik für Frauenheilkunde und Geburtshilfe, Universität Düsseldorf, Deutschland
| | - Kurt Hecher
- Klinik für Geburtshilfe und Pränatalmedizin, Universitätsklinikum Hamburg-Eppendorf, Deutschland
| | - Ulrich Pecks
- Klinik für Gynäkologie und Geburtshilfe, Campus Kiel, Universitätsklinikum Schleswig-Holstein, Deutschland
| | - Dietmar Schlembach
- Klinik für Geburtsmedizin, Vivantes Klinikum Neukölln, Berlin, Deutschland
| | - Arne Simon
- Klinik für Pädiatrische Onkologie und Hämatologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Deutschland
| | - Rolf Schlösser
- Schwerpunkt Neonatologie, Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Frankfurt, Deutschland
| | | |
Collapse
|
48
|
Thapa K, Verma N, Singh TG, Kaur Grewal A, Kanojia N, Rani L. COVID-19-Associated acute respiratory distress syndrome (CARDS): Mechanistic insights on therapeutic intervention and emerging trends. Int Immunopharmacol 2021; 101:108328. [PMID: 34768236 PMCID: PMC8563344 DOI: 10.1016/j.intimp.2021.108328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023]
Abstract
AIMS The novel Coronavirus disease 2019 (COVID-19) has caused great distress worldwide. Acute respiratory distress syndrome (ARDS) is well familiar but when it happens as part of COVID-19 it has discrete features which are unmanageable. Numerous pharmacological treatments have been evaluated in clinical trials to control the clinical effects of CARDS, but there is no assurance of their effectiveness. MATERIALS AND METHODS A systematic review of the literature of the Medline, Scopus, Bentham, PubMed, and EMBASE (Elsevier) databases was examined to understand the novel therapeutic approaches used in COVID-19-Associated Acute Respiratory Distress Syndrome and their outcomes. KEY FINDINGS Current therapeutic options may not be enough to manage COVID-19-associated ARDS complications in group of patients and therefore, the current review has discussed the pathophysiological mechanism of COVID-19-associated ARDS, potential pharmacological treatment and the emerging molecular drug targets. SIGNIFICANCE The rationale of this review is to talk about the pathophysiology of CARDS, potential pharmacological treatment and the emerging molecular drug targets. Currently accessible treatment focuses on modulating immune responses, rendering antiviral effects, anti-thrombosis or anti-coagulant effects. It is expected that considerable number of studies conducting globally may help to discover effective therapies to decrease mortality and morbidity occurring due to CARDS. Attention should be also given on molecular drug targets that possibly will help to develop efficient cure for COVID-19-associated ARDS.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India; Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Nitin Verma
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | | | | | - Neha Kanojia
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Lata Rani
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| |
Collapse
|
49
|
Goudman L, De Smedt A, Noppen M, Moens M. Is Central Sensitisation the Missing Link of Persisting Symptoms after COVID-19 Infection? J Clin Med 2021; 10:jcm10235594. [PMID: 34884296 PMCID: PMC8658135 DOI: 10.3390/jcm10235594] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 12/18/2022] Open
Abstract
Patients recovered from a COVID-19 infection often report vague symptoms of fatigue or dyspnoea, comparable to the manifestations in patients with central sensitisation. The hypothesis was that central sensitisation could be the underlying common aetiology in both patient populations. This study explored the presence of symptoms of central sensitisation, and the association with functional status and health-related quality of life, in patients post COVID-19 infection. Patients who were previously infected with COVID-19 filled out the Central Sensitisation Inventory (CSI), the Post-COVID-19 Functional Status (PCFS) Scale and the EuroQol with five dimensions, through an online survey. Eventually, 567 persons completed the survey. In total, 29.73% of the persons had a score of <40/100 on the CSI and 70.26% had a score of ≥40/100. Regarding functional status, 7.34% had no functional limitations, 9.13% had negligible functional limitations, 37.30% reported slight functional limitations, 42.86% indicated moderate functional limitations and 3.37% reported severe functional limitations. Based on a one-way ANOVA test, there was a significant effect of PCFS Scale group level on the total CSI score (F(4,486) = 46.17, p < 0.001). This survey indicated the presence of symptoms of central sensitisation in more than 70% of patients post COVID-19 infection, suggesting towards the need for patient education and multimodal rehabilitation, to target nociplastic pain.
Collapse
Affiliation(s)
- Lisa Goudman
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium;
- STIMULUS Research Group (reSearch and TeachIng neuroModULation Uz bruSsel), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium;
- Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Pain in Motion (PAIN) Research Group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Research Foundation—Flanders (FWO), 1090 Brussels, Belgium
- Correspondence: ; Tel.: +32-2477-5514
| | - Ann De Smedt
- STIMULUS Research Group (reSearch and TeachIng neuroModULation Uz bruSsel), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium;
- Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Department of Physical Medicine and Rehabilitation, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Marc Noppen
- Chief Executive Officer, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium;
| | - Maarten Moens
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium;
- STIMULUS Research Group (reSearch and TeachIng neuroModULation Uz bruSsel), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium;
- Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Pain in Motion (PAIN) Research Group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Department of Radiology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
50
|
Palaiodimou L, Stefanou M, Katsanos AH, Fragkou PC, Papadopoulou M, Moschovos C, Michopoulos I, Kokotis P, Bakirtzis C, Naska A, Vassilakopoulos TI, Chroni E, Tsiodras S, Tsivgoulis G. Prevalence, clinical characteristics and outcomes of Guillain-Barré syndrome spectrum associated with COVID-19: A systematic review and meta-analysis. Eur J Neurol 2021; 28:3517-3529. [PMID: 33837630 PMCID: PMC8250909 DOI: 10.1111/ene.14860] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/25/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Mounting evidence supports an association between Guillain-Barré syndrome spectrum (GBSs) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, GBSs in the setting of coronavirus disease 2019 (COVID-19) remains poorly characterized, whilst GBSs prevalence amongst COVID-19 patients has not been previously systematically evaluated using a meta-analytical approach. METHODS A systematic review and meta-analysis of observational cohort and case series studies reporting on the occurrence, clinical characteristics and outcomes of patients with COVID-19-associated GBSs was performed. A random-effects model was used to calculate pooled estimates and odds ratios (ORs) with corresponding 95% confidence intervals (CIs), compared to non-COVID-19, contemporary or historical GBSs patients. RESULTS Eighteen eligible studies (11 cohorts, seven case series) were identified including a total of 136,746 COVID-19 patients. Amongst COVID-19 patients, including hospitalized and non-hospitalized cases, the pooled GBSs prevalence was 0.15‰ (95% CI 0%-0.49‰; I2 = 96%). Compared with non-infected contemporary or historical controls, patients with SARS-CoV-2 infection had increased odds for demyelinating GBSs subtypes (OR 3.27, 95% CI 1.32%-8.09%; I2 = 0%). In SARS-CoV-2-infected patients, olfactory or concomitant cranial nerve involvement was noted in 41.4% (95% CI 3.5%-60.4%; I2 = 46%) and 42.8% (95% CI 32.8%-53%; I2 = 0%) of the patients, respectively. Clinical outcomes including in-hospital mortality were comparable between COVID-19 GBSs patients and non-infected contemporary or historical GBSs controls. CONCLUSION GBSs prevalence was estimated at 15 cases per 100,000 SARS-CoV-2 infections. COVID-19 appears to be associated with an increased likelihood of GBSs and with demyelinating GBSs variants in particular.
Collapse
Affiliation(s)
- Lina Palaiodimou
- Second Department of Neurology, ‘Attikon’ University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Maria‐Ioanna Stefanou
- Second Department of Neurology, ‘Attikon’ University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Aristeidis H. Katsanos
- Second Department of Neurology, ‘Attikon’ University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
- Division of NeurologyMcMaster University/Population Health Research InstituteHamiltonCanada
| | - Paraskevi C. Fragkou
- Fourth Department of Internal Medicine, 'Attikon' University HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Marianna Papadopoulou
- Second Department of Neurology, ‘Attikon’ University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
- Department of PhysiotherapyUniversity of West AtticaAthensGreece
| | - Christos Moschovos
- Second Department of Neurology, ‘Attikon’ University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Ioannis Michopoulos
- Second Department of Psychiatry, ‘Attikon’ Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Panagiotis Kokotis
- First Department of Neurology, ‘Eginition’ University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Christos Bakirtzis
- Second Department of NeurologyAristotle University of ThessalonikiThessalonikiGreece
| | - Androniki Naska
- Department of Hygiene, Epidemiology and Medical Statistics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Theodoros I. Vassilakopoulos
- Third Department of Critical Care Medicine, Evgenideio Hospital, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Elisabeth Chroni
- Department of Neurology, School of MedicineUniversity of PatrasRio‐PatrasGreece
| | - Sotirios Tsiodras
- Fourth Department of Internal Medicine, 'Attikon' University HospitalNational and Kapodistrian University of AthensAthensGreece
- Hellenic Centre for Disease Control and PreventionAthensGreece
| | - Georgios Tsivgoulis
- Second Department of Neurology, ‘Attikon’ University Hospital, School of MedicineNational and Kapodistrian University of AthensAthensGreece
- Department of NeurologyUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| |
Collapse
|