1
|
Chatow L, Nudel A, Eyal N, Lupo T, Ramirez S, Zelinger E, Nesher I, Boxer R. Terpenes and cannabidiol against human corona and influenza viruses-Anti-inflammatory and antiviral in vitro evaluation. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2024; 41:e00829. [PMID: 38318445 PMCID: PMC10840330 DOI: 10.1016/j.btre.2024.e00829] [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: 05/07/2023] [Revised: 09/19/2023] [Accepted: 01/16/2024] [Indexed: 02/07/2024]
Abstract
The activity of the terpenes and Cannabidiol (CBD) against human coronavirus (HCoV) strain OC43 and influenza A (H1N1) was evaluated in human lung fibroblasts (MRC-5 cells). Also, we examined whether these ingredients inhibit pro-inflammatory cytokines in peripheral blood mononuclear cells (PBMC). The tested preparations exhibited both anti-inflammatory and antiviral effects. The combination of terpenes was effective against both HCoV-OC43 and influenza A (H1N1) virus. The addition of CBD improved the antiviral activity in some, but not all cases. This variation in activity may suggest an antiviral mechanism. In addition, there was a strong correlation between the quantitative results from a cell-viability assay and the cytopathic effect after 72 h, as observed under a microscope. The anti-inflammatory properties of terpenes were demonstrated using a pro-inflammatory cytokine-inhibition assay, which revealed significant cytokine inhibition and enhanced by the addition of CBD.
Collapse
Affiliation(s)
| | - Adi Nudel
- Eybna Technologies Ltd., Kfar Saba, Israel
| | - Nadav Eyal
- Eybna Technologies Ltd., Kfar Saba, Israel
| | - Tal Lupo
- Eybna Technologies Ltd., Kfar Saba, Israel
| | | | - Einat Zelinger
- CSI Center for Scientific Imaging Faculty of Agriculture, Hebrew University, Rehovot, Israel
| | | | - Richard Boxer
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
2
|
Khafaei M, Asghari R, Zafari F, Sadeghi M. Impact of IL-6 rs1800795 and IL-17A rs2275913 gene polymorphisms on the COVID-19 prognosis and susceptibility in a sample of Iranian patients. Cytokine 2024; 174:156445. [PMID: 38056249 DOI: 10.1016/j.cyto.2023.156445] [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: 08/08/2023] [Revised: 09/23/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND From asymptomatic to acute and life-threatening pulmonary infection, the clinical manifestations of COVID-19 are highly variable. Interleukin (IL)-6 and IL-17A are key drivers of hyper inflammation status in COVID-19, and their elevated levels are hallmarks of the infection progression. To explore whether prognosis and susceptibility to COVID-19 are linked to IL-6 rs1800795 and IL-17A rs2275913, these single-nucleotide polymorphisms (SNPs) were assessed in a sample of Iranian COVID-19 patients. METHODS This study enrolled two hundred and eighty COVID-19 patients (140 non-severe and 140 severe). Genotyping for IL-6 rs1800795 and IL-17A rs2275913 was performed using tetra primer-amplification refractory mutation system-polymerase chain reaction (tetra-ARMS-PCR). IL-6 and IL-17A circulating levels were measured using enzyme-linked immunosorbent assay (ELISA). Also, mortality predictors of COVID-19 were investigated. RESULTS The rs1800795 GG genotype (78/140 (55.7 %)) and G allele (205/280 (73.2 %)) were significantly associated with a positive risk of COVID-19 severe infection (OR = 2.19, 95 %CI: 1.35-3.54, P =.006 and OR = 1.79, 95 %CI: 1.25-2.56, P <.001, respectively). Also, rs1800795 GG genotype was significantly linked to disease mortality (OR = 1.95, 95 %CI: 1.06-3.61, P =.04). The rs2275913 GA genotype was protective against severe COVID-19 (OR = 0.5, 95 %CI: 0.31--0.80, P =.012). However, the present study did not reveal any significant link between rs2275913 genotypes with disease mortality. INR ≥ 1.2 (OR = 2.19, 95 %CI: 1.61-3.78, P =.007), D-dimer ≥ 565.5 ng/mL (OR = 3.12, 95 %CI: 1.27-5.68, P =.019), respiratory rate ≥ 29 (OR = 1.19, 95 %CI: 1.12-1.28, P =.001), IL-6 serum concentration ≥ 28.5 pg/mL (OR = 1.97, 95 %CI: 1.942-2.06, P =.013), and IL-6 rs1800795 GG genotype (OR = 1.95, 95 %CI: 1.06-3.61, P =.04) were predictive of COVID-19 mortality. CONCLUSION The rs1800795 GG genotype and G allele were associated with disease severity, and INR, D-dimer, respiratory rate, IL-6 serum concentration, and IL-6 rs1800795 GG genotype were predictive of COVID-19 mortality.
Collapse
Affiliation(s)
- Mostafa Khafaei
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Reza Asghari
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Fariba Zafari
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Morteza Sadeghi
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Zhang Q, Bignotti A, Yada N, Ye Z, Liu S, Han Z, Zheng XL. Dynamic Assessment of Plasma von Willebrand Factor and ADAMTS13 Predicts Mortality in Hospitalized Patients with SARS-CoV-2 Infection. J Clin Med 2023; 12:7174. [PMID: 38002786 PMCID: PMC10672082 DOI: 10.3390/jcm12227174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Plasma levels of von Willebrand factor (VWF) are significantly elevated in patients with coronavirus disease 2019 (COVID-19). However, dynamic changes and prognostic value of this biomarker in hospitalized patients with COVID-19 have not been determined. METHODS A total of 124 patients infected with SARS-CoV-2 were prospectively recruited for the study. Serial blood samples were obtained at the time of admission (D1), 3-4 days following standard-care treatments (D2), and 1-2 days prior to discharge or any time collected prior to death (D3). Plasma VWF antigen, ADAMTS13 antigen, and ADAMTS13 proteolytic activity, as well as the ratio of VWF/ADAMTS13 were determined, followed by various statistical analyses. RESULTS On admission, plasma levels of VWF in COVID-19 patients were significantly elevated compared with those in the healthy controls, but no statistical significance was detected among patients with different disease severity. Plasma ADAMTS13 activity but not its antigen levels were significantly lower in patients with severe or critical COVID-19 compared with that in other patient groups. Interestingly, the ratios of plasma VWF antigen to ADAMTS13 antigen were significantly higher in patients with severe or critical COVID-19 than in those with mild to moderate disease. More importantly, plasma levels of VWF and the ratios of VWF/ADAMTS13 were persistently elevated in patients with COVID-19 throughout hospitalization. Kaplan-Meier and Cox proportional hazard regression analyses demonstrated that an increased plasma level of VWF or ratio of VWF/ADAMTS13 at D2 and D3 was associated with an increased mortality rate. CONCLUSIONS Persistent endotheliopathy, marked by the elevated levels of plasma VWF or VWF/ADAMTS13 ratio, is present in all hospitalized patients following SARS-CoV-2 infection, which is strongly associated with mortality.
Collapse
Affiliation(s)
- Quan Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Antonia Bignotti
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Noritaka Yada
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Zhan Ye
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Szumam Liu
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, 670 West Baltimore Street, Baltimore, MD 21201, USA
| | - X. Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
4
|
Bagheri-Hosseinabadi Z, Moadab F, Amiri A, Abbasifard M. The prevalence and contributing risk factors of coronavirus disease 2019 infection in patients with metabolic syndrome. BMC Endocr Disord 2023; 23:100. [PMID: 37142990 PMCID: PMC10157563 DOI: 10.1186/s12902-023-01351-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Components of metabolic syndrome (MetS) was reported to contribute to severe and worse outcomes of coronavirus disease 2019 (COVID-19). Hereby, we evaluated the association of MetS and its components with susceptibility to COVID-19. METHODS Here, 1000 subjects with MetS were recruited that were diagnosed via the International Diabetes Federation (IDF) criterion. Real-time PCR was exerted to detect SARS-CoV-2 in the nasopharyngeal swabs. RESULTS Among the MetS patients, 206 (20.6%) cases were detected to have COVID-19. Smoking (OR = 5.04, 95%CI = 3.53-7.21, P < 0.0001) and CVD (OR = 1.62, 95%CI = 1.09-2.40, P = 0.015) were associated with increased chance of COVID-19 infection in the MetS patients. BMI was significantly higher (P = 0.0001) in MetS cases with COVID-19 than those without COVID-19. Obesity was associated with increased susceptibility to COVID-19 in MetS patients (OR = 2.00, 95%CI = 1.47-2.74, P < 0.0001). Total cholesterol, TG, LDL were significantly higher in the MetS cases with COVID-19 than those without COVID-19. Dyslipidemia was associated with increased chance of COVID-19 (OR = 1.50, 95%CI = 1.10-2.05, P = 0.0104). FBS level was significantly higher in the MetS cases with COVID-19. T2DM was associated with increased risk of COVID-19 in MetS patients (OR = 1.43, 95%CI = 1.01-2.00, P = 0.0384). Hypertension was associated with increased chance of COVID-19 in the MetS patients (OR = 1.44, 95%CI = 1.05-1.98, P = 0.0234). CONCLUSIONS MetS and its components, like obesity, diabetes, dyslipidemia, cardiovascular complications were associated with increased chance of COVID-19 infection development and probably with aggravated symptoms in such patients.
Collapse
Affiliation(s)
- Zahra Bagheri-Hosseinabadi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Moadab
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Amiri
- Department of Orthodontics, College of Stomatology, The First Affiliated Stomatological Hospital, Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
5
|
Phytocannabinoids Act Synergistically with Non-Steroidal Anti-Inflammatory Drugs Reducing Inflammation in 2D and 3D In Vitro Models. Pharmaceuticals (Basel) 2022; 15:ph15121559. [PMID: 36559009 PMCID: PMC9787964 DOI: 10.3390/ph15121559] [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: 11/16/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Lung inflammation is associated with elevated pro-inflammatory cytokines and chemokines. Treatment with FCBD:std (standard mix of cannabidiol [CBD], cannabigerol [CBG] and tetrahydrocannabivarin [THCV]) leads to a marked reduction in the inflammation of alveolar epithelial cells, but not in macrophages. In the present study, the combined anti-inflammatory effect of FCBD:std with two corticosteroids (dexamethasone and budesonide) and two non-steroidal anti-inflammatory drugs (NSAID; ibuprofen and diclofenac), was examined. Enzyme-linked immunosorbent assay (ELISA) was used to determine protein levels. Gene expression was determined by quantitative real-time PCR. Inhibition of cyclo-oxygenase (COX) activity was determined in vitro. FCBD:std and diclofenac act synergistically, reducing IL-8 levels in macrophages and lung epithelial cells. FCBD:std plus diclofenac also reduced IL-6, IL-8 and CCL2 expression levels in co-cultures of macrophages and lung epithelial cells, in 2D and 3D models. Treatment by FCBD:std and/or NSAID reduced COX-1 and COX-2 gene expression but not their enzymatic activity. FCBD:std and diclofenac exhibit synergistic anti-inflammatory effects on macrophages and lung epithelial cells, yet this combined activity needs to be examined in pre-clinical studies and clinical trials.
Collapse
|
6
|
Upadhyay M, Gupta S. Endoplasmic reticulum secretory pathway: Potential target against SARS-CoV-2. Virus Res 2022; 320:198897. [PMID: 35988898 PMCID: PMC9387115 DOI: 10.1016/j.virusres.2022.198897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has recently emerged throughout the world, resulting in more than 400 million cases and over 6 million deaths worldwide as of January 2022. Coronaviruses subvert or use certain aspects of the unfolded protein response in the endoplasmic reticulum to overcome protein translation shutdown to benefit their replication. New virions use the ER-Golgi intermediate compartment to assemble and gain transportation to the cell membrane. Extensive remodeling of the ER has been demonstrated during SARS-CoV-2 infection. In this review article, we discuss the role of the endoplasmic reticulum secretory pathway in the replication cycle of SARS-CoV-2. Currently, there is a dearth of therapeutic options for intervening with SARS-CoV-2 infection. To accelerate drug development, efforts around the globe have been focusing on repurposing drugs that have already been approved for clinical use by regulatory agencies. Targeting the ERS pathway is reasonable, as prior work has shown that SARS-CoV-2 egress is dependent on this pathway. Here we discuss the feasibility of off-patent, FDA-approved, pharmacological inhibitors of the ERS pathway to suppress the SARS-CoV-2 replication cycle, a promising approach that warrants investigation.
Collapse
Affiliation(s)
- Maarisha Upadhyay
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, National University of Ireland-Galway, Galway, Ireland
| | - Sanjeev Gupta
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, National University of Ireland-Galway, Galway, Ireland.
| |
Collapse
|
7
|
Singh P, Salman KA, Shameem M, Warsi MS. Withania somnifera (L.) Dunal as Add-On Therapy for COPD Patients: A Randomized, Placebo-Controlled, Double-Blind Study. Front Pharmacol 2022; 13:901710. [PMID: 35784687 PMCID: PMC9243480 DOI: 10.3389/fphar.2022.901710] [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: 03/22/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background: The current gold-standard therapies for chronic obstructive pulmonary disease (COPD) lack disease-modifying potential and exert adverse side effects. Moreover, COPD patients are at a higher risk of severe outcomes if they get infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, the cause of the current epidemic. This is the first study to document clinical research on an adaptogenic and steroidal activity–containing herb as a complementary medicine for COPD treatment. Objective: We aimed to evaluate the efficacy of Withania somnifera (L.) Dunal [Solanaceae] (WS) as an add-on therapy for COPD patients. Methods: A randomized, placebo-controlled, and double-blind clinical study was conducted. A total of 150 patients were randomly assigned to three groups: control, placebo, and WS group. In addition to conventional medicines, WS root capsules or starch capsules were given twice a day to the WS group and the placebo group, respectively. Their lung functioning, quality of life, exercise tolerance, systemic oxidative stress (OS), and systemic inflammation were assessed before and after 12 weeks of intervention. WS root phytochemicals were identified by LC-ESI-MS. The inhibitory activity of these phytochemicals against angiotensin-converting enzyme 2 (ACE-2); the SARS-CoV-2 receptor; myeloperoxidase (MPO); and interleukin-6 (IL-6) was evaluated by in silico docking to investigate the mechanism of action of WS. Results: The pulmonary functioning, quality of life, and exercise tolerance improved, and inflammation reduced notably the most in the WS group. Systemic oxidative stress subsided significantly only in the WS group. Although a minor placebo effect was observed in the SGRQ test, but it was not present in other tests. Withanolides found in the WS roots demonstrated substantial inhibitory activity against the proteins ACE-2, MPO, and IL-6, compared to that of a standard drug or known inhibitor. Moreover, FEV1% predicted had significant correlation with systemic antioxidative status (positive correlation) and malondialdehyde (MDA, negative correlation), suggesting that the antioxidative potential of WS has significant contribution to improving lung functioning. Conclusion: Our study clinically demonstrated that WS root when given along with conventional drugs ameliorated COPD significantly more in comparison to the conventional drugs alone, in GOLD 2 and 3 categories of COPD patients. In silico, it has potent inhibitory activity against SARS-CoV-2 receptor, ACE-2, MPO, and IL-6.
Collapse
Affiliation(s)
- Priyam Singh
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
- *Correspondence: Priyam Singh,
| | - Khushtar Anwar Salman
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| | - Mohammad Shameem
- Department of Tuberculosis and Respiratory Diseases, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| | - Mohd Sharib Warsi
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
8
|
Ruscitti P, Di Cola I, Di Muzio C, Italiano N, Ursini F, Giacomelli R, Cipriani P. Expanding the spectrum of the hyperferritinemic syndrome, from pathogenic mechanisms to clinical observations, and therapeutic implications. Autoimmun Rev 2022; 21:103114. [PMID: 35595050 DOI: 10.1016/j.autrev.2022.103114] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/15/2022] [Indexed: 01/19/2023]
Abstract
From the introduction of hyperferritinemic syndrome concept, a growing body of evidence has suggested the role of ferritin as a pathogenic mediator and a relevant clinical feature in the management of patients with inflammatory diseases. From a pathogenic point of view, ferritin may directly stimulate the aberrant immune response by triggering the production of pro-inflammatory mediators in inducing a vicious pathogenic loop and contributing to the occurrence of cytokine storm syndrome. The latter has been recently defined as a clinical picture characterised by elevated circulating cytokine levels, acute systemic inflammatory symptoms, and secondary organ dysfunction beyond that which could be attributed to a normal response to a pathogen It is noteworthy that the occurrence of hyperferritinemia may be correlated with the development of the cytokine storm syndrome in the context of an inflammatory disease. In addition to adult onset Still's disease, macrophage activation syndrome, catastrophic anti-phospholipids syndrome, and septic shock, recent evidence has suggested this association between ferritin and life-threatening evolution in patients with systemic lupus erythematosus, with anti-MDA5 antibodies in the context of poly-dermatomyositis, with severe COVID-19, and with multisystem inflammatory syndrome. The possible underlying common inflammatory mechanisms, associated with hyperferritinemia, may led to the similar clinical picture observed in these patients. Furthermore, similar therapeutic strategies could be suggested inhibiting pro-inflammatory cytokines and improving long-term outcomes in these disorders. Thus, it could be possible to expand the spectrum of the hyperferritinemic syndrome to those diseases burdened by a dreadful clinical picture correlated with hyperferritinemia and the occurrence of the cytokine storm syndrome. In addition, the assessment of ferritin may provide useful information to the physicians in clinical practice to manage these patients. Therefore, ferritin may be considered a relevant clinical feature to be used as biomarker in dissecting the unmet needs in the management of these disorders. Novel evidence may thus support an expansion of the spectrum of the hyperferritinemic syndrome to these diseases burdened by a life-threatening clinical picture correlated with hyperferritinemia and the occurrence of the cytokine storm syndrome.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudia Di Muzio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Noemi Italiano
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesco Ursini
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
9
|
Antiviral phytocompounds “ellagic acid” and “(+)-sesamin” of Bridelia retusa identified as potential inhibitors of SARS-CoV-2 3CL pro using extensive molecular docking, molecular dynamics simulation studies, binding free energy calculations, and bioactivity prediction. Struct Chem 2022; 33:1445-1465. [PMID: 35571865 PMCID: PMC9086128 DOI: 10.1007/s11224-022-01959-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/28/2022] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected billions and has killed millions to date. Studies are being carried out to find therapeutic molecules that can potentially inhibit the replication of SARS-CoV-2. 3-chymotrypsin-like protease (3CL pro) involved in the polyprotein cleavage process is believed to be the key target for viral replication, and hence is an attractive target for the discovery of antiviral molecules. In the present study, we aimed to identify natural phytocompounds from Bridelia retusa as potential inhibitors of SARS-CoV-2 3CL pro (PDB ID: 6M2N) using in silico techniques. Molecular docking studies conducted with three different tools in triplicates revealed that ellagic acid (BR6) and (+)-sesamin (BR13) has better binding affinity than the co-crystal inhibitor “3WL” of 6M2N. BR6 and BR13 were found to have a high LD50 value with good bioavailability. 3WL, BR6, and BR13 bind to the same active binding site and interacted with the HIS41-CYS145 catalytic dyad including other crucial amino acids. Molecular dynamics simulation studies revealed stability of protein–ligand complexes as evidenced from root-mean-square deviations, root-mean-square fluctuations (RMSF), protein secondary structure elements, ligand-RMSF, protein–ligand contacts, ligand torsions, and ligand properties. BR6 (−22.3064 kcal/mol) and BR13 (−19.1274 kcal/mol) showed a low binding free energy value. The Bayesian statistical model revealed BR6 and BR13 as better protease inhibitors than 3WL. Moreover, BR6 and BR13 had already been reported to elicit antiviral activities. Therefore, we conclude that ellagic acid and (+)-sesamin as natural antiviral phytocompounds with inhibitory potential against SARS-CoV-2 3CL pro.
Collapse
|
10
|
Levosimendan Ameliorates Cardiopulmonary Function but Not Inflammatory Response in a Dual Model of Experimental ARDS. Biomedicines 2022; 10:biomedicines10051031. [PMID: 35625767 PMCID: PMC9138326 DOI: 10.3390/biomedicines10051031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022] Open
Abstract
The calcium sensitiser levosimendan, which is used as an inodilator to treat decompensated heart failure, may also exhibit anti-inflammatory properties. We examined whether treatment with levosimendan improves cardiopulmonary function and is substantially beneficial to the inflammatory response in acute respiratory response syndrome (ARDS). Levosimendan was administered intravenously in a new experimental porcine model of ARDS. For comparison, we used milrinone, another well-known inotropic agent. Our results demonstrated that levosimendan intravenously improved hemodynamics and lung function in a porcine ARDS model. Significant beneficial alterations in the inflammatory response and lung injury were not detected.
Collapse
|
11
|
Hromić-Jahjefendić A, Barh D, Ramalho Pinto CH, Gabriel Rodrigues Gomes L, Picanço Machado JL, Afolabi OO, Tiwari S, Aljabali AAA, Tambuwala MM, Serrano-Aroca Á, Redwan EM, Uversky VN, Lundstrom K. Associations and Disease-Disease Interactions of COVID-19 with Congenital and Genetic Disorders: A Comprehensive Review. Viruses 2022; 14:910. [PMID: 35632654 PMCID: PMC9146233 DOI: 10.3390/v14050910] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
Since December 2019, the COVID-19 pandemic, which originated in Wuhan, China, has resulted in over six million deaths worldwide. Millions of people who survived this SARS-CoV-2 infection show a number of post-COVID complications. Although, the comorbid conditions and post-COVID complexities are to some extent well reviewed and known, the impact of COVID-19 on pre-existing congenital anomalies and genetic diseases are only documented in isolated case reports and case series, so far. In the present review, we analyzed the PubMed indexed literature published between December 2019 and January 2022 to understand this relationship from various points of view, such as susceptibility, severity and heritability. Based on our knowledge, this is the first comprehensive review on COVID-19 and its associations with various congenital anomalies and genetic diseases. According to reported studies, some congenital disorders present high-risk for developing severe COVID-19 since these disorders already include some comorbidities related to the structure and function of the respiratory and cardiovascular systems, leading to severe pneumonia. Other congenital disorders rather cause psychological burdens to patients and are not considered high-risk for the development of severe COVID-19 infection.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina
| | - Debmalya Barh
- Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, India
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.R.G.); (S.T.)
| | - Cecília Horta Ramalho Pinto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Lucas Gabriel Rodrigues Gomes
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.R.G.); (S.T.)
| | - Jéssica Lígia Picanço Machado
- Department of Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Oladapo Olawale Afolabi
- Department of Physiology and Biophysics, Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Sandeep Tiwari
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (L.G.R.G.); (S.T.)
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK;
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Elrashdy M. Redwan
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab 21934, Alexandria, Egypt
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | | |
Collapse
|
12
|
Bagwe PV, Bagwe PV, Ponugoti SS, Joshi SV. Peptide-Based Vaccines and Therapeutics for COVID-19. Int J Pept Res Ther 2022; 28:94. [PMID: 35463185 PMCID: PMC9017722 DOI: 10.1007/s10989-022-10397-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 12/20/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been prevalent in the humans since 2019 and has given rise to a pandemic situation. With the discovery and ongoing use of drugs and vaccines against SARS-CoV-2, there is still no surety of its complete suppression of this disease or if there is a need for additional booster doses. There is an urgent need for alternative treatment strategies against COVID-19. Peptides and peptidomimetics have several advantages as therapeutic agents because of their target selectivity, better interactions, and lower toxicity. Minor structural alterations to peptides can help prevent their fast metabolism and provide long-action. This comprehensive review provides an overview of different peptide-based vaccines and therapeutics against SARS-CoV-2. It discusses the design and mechanism of action of the peptide-based vaccines, peptide immunomodulators, anti-inflammatory agents, and peptides as entry inhibitors of SARS-CoV-2. Moreover, the mechanism of action, sequences and current clinical trial studies are also summarized. The review also discusses the future aspects of peptide-based vaccines and therapeutics for COVID-19. Graphical Abstract
Collapse
Affiliation(s)
- Pritam V. Bagwe
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, Maharashtra 400019 India
| | - Priyal V. Bagwe
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341 USA
| | - Sai Srinivas Ponugoti
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, Maharashtra 400019 India
| | - Shreerang V. Joshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, Maharashtra 400019 India
| |
Collapse
|
13
|
ÇOLAK S, TEKGÖZ E, ÇINAR M, YILMAZ G, TECER D, BIÇAKÇI F, CUCE F, FİDAN G, DOĞAN D, SAVAŞÇI Ü, ARSLAN Y, TAŞÇI C, UYAR E, KARACAER Z, ŞENKAL S, YILMAZ S. Efficacy of tocilizumab in severe COVID-19: a retrospective study. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2022. [DOI: 10.32322/jhsm.1064728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
14
|
Lu R, Zheng XL. Plasma Levels of Big Endothelin-1 Are Associated with Renal Insufficiency and In-Hospital Mortality of Immune Thrombotic Thrombocytopenic Purpura. Thromb Haemost 2022; 122:344-352. [PMID: 33984867 PMCID: PMC9514555 DOI: 10.1055/a-1508-8347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Immune thrombotic thrombocytopenic purpura (iTTP) is caused by severe deficiency of plasma ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) activity. Despite advances in early diagnosis and management, the mortality rate of acute iTTP remains high in a large part of world where access to some of the most novel therapies is limited. To determine the role of plasma big endothelin-1 (bigET-1) or its bioactive product ET-1 as a biomarker and/or a pathogenic factor in acute iTTP, plasma levels of bigET-1 were determined using an immunoassay in patients with iTTP on admission and during remission, as well as in healthy controls; moreover, the biological effect of ET-1 in thrombus formation was determined by a microfluidic assay. We show that plasma levels of bigET-1 were dramatically increased in patients with acute iTTP on admission, which was significantly decreased during clinical response/remission; elevated admission levels of plasma bigET-1 were associated with low estimated glomerular filtration rate, the need for intensive care unit admission or intubation, and in-hospital mortality. Moreover, an addition of a bioactive product ET-1 to cultured endothelial cells in a microfluidic channel significantly accelerated the rate of thrombus formation under arterial flow. Our results demonstrate for the first time a potential role of measuring plasma bigET-1 in patients with acute iTTP in assessing the disease severity and risk of in-hospital mortality, which may help stratify patients for a more aggressive monitoring and therapeutic strategy; also, the bioactive ET-1, derived from bigET-1, may result in acute renal injury in TTP patient, likely through its vasoconstriction and prothrombotic properties.
Collapse
Affiliation(s)
- Ruinan Lu
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A.,Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - X. Long Zheng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A.,Correspondence should be sent to: X. Long Zheng, M.D., Ph.D., Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, 4000 Cambridge Street, MS 3045, Kansas City, KS 66160, Tel. +1 913-588-7124, or
| |
Collapse
|
15
|
Ning Q, Wu D, Wang X, Xi D, Chen T, Chen G, Wang H, Lu H, Wang M, Zhu L, Hu J, Liu T, Ma K, Han M, Luo X. The mechanism underlying extrapulmonary complications of the coronavirus disease 2019 and its therapeutic implication. Signal Transduct Target Ther 2022; 7:57. [PMID: 35197452 PMCID: PMC8863906 DOI: 10.1038/s41392-022-00907-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a highly transmissible disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that poses a major threat to global public health. Although COVID-19 primarily affects the respiratory system, causing severe pneumonia and acute respiratory distress syndrome in severe cases, it can also result in multiple extrapulmonary complications. The pathogenesis of extrapulmonary damage in patients with COVID-19 is probably multifactorial, involving both the direct effects of SARS-CoV-2 and the indirect mechanisms associated with the host inflammatory response. Recognition of features and pathogenesis of extrapulmonary complications has clinical implications for identifying disease progression and designing therapeutic strategies. This review provides an overview of the extrapulmonary complications of COVID-19 from immunological and pathophysiologic perspectives and focuses on the pathogenesis and potential therapeutic targets for the management of COVID-19.
Collapse
Affiliation(s)
- Qin Ning
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Di Wu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Xi
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guang Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongwu Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Lu
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Zhu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Liu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Ma
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meifang Han
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Luo
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Castellar-López J, Villamizar-Villamizar W, Amaranto-Pallares A, Rosales-Rada W, De Los Angeles Vélez Verbel M, Chang A, Jiménez FT, Mendoza-Torres E. Recent Insights into COVID-19 in Children and Clinical Recommendations. Curr Pediatr Rev 2022; 18:121-137. [PMID: 34872479 DOI: 10.2174/1573396317666211206124347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/30/2021] [Accepted: 09/01/2021] [Indexed: 01/08/2023]
Abstract
Pediatric coronavirus disease 2019 (COVID-19) and multisystem inflammatory syndrome in children (MIS-C) have been recognized in multiple countries globally. In this review, we provide recent insights into SARS-CoV-2 infection in children from epidemiological, clinical, and laboratory perspectives, including reports on the disease course and therapy. We highlight key features of SARS-CoV-2 infection in children, the relationship between MIS-C and Kawasaki disease, and summarize treatment guidelines for COVID-19 in children from institutional protocols from Colombia, case reports, recommendations based on expert consensus, and official statements from organizations such as the World Health Organization (WHO), United States Center for Disease Control (CDC), Colombian Association of Infectious Diseases, and the Colombian Society of Pediatrics. Finally, we discuss gaps in research with suggestions for future research on the pathogenesis underlying pediatric COVID-19.
Collapse
Affiliation(s)
- Jairo Castellar-López
- Faculty of Exact and Natural Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Barranquilla, Barranquilla, Colombia
| | | | - Aldo Amaranto-Pallares
- Faculty of Health Sciences, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Wendy Rosales-Rada
- Faculty of Exact and Natural Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Barranquilla, Barranquilla, Colombia.,Department of Medicine, Grupo de Investigación en Biotecnología. Division of Health Sciences. Universidad del Norte, Colombia
| | | | - Aileen Chang
- Department of Medicine, George Washington University, Washington D.C., USA
| | - Franklin Torres Jiménez
- Faculty of Health Sciences, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Evelyn Mendoza-Torres
- Faculty of Health Sciences, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| |
Collapse
|
17
|
Mohamed N, Hamad MA, Ghaleb AH, Esmat G, Elsabahy M. Applications of nanoengineered therapeutics and vaccines: special emphasis on COVID-19. IMMUNOMODULATORY EFFECTS OF NANOMATERIALS 2022. [PMCID: PMC9212255 DOI: 10.1016/b978-0-323-90604-3.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nanomedicine provides innovative strategies that had significantly improved drug and gene delivery and allowed control over the engineering of therapeutics, diagnostics, vaccines, and other medical devices, for a diversity of medical applications. This review focuses on the current attempts to develop potent nanoengineered vaccines and therapeutics against coronaviruses, and the recent fabrication strategies and design principles to control acute infections from the escalating SARS-CoV-2 pandemic. Nanomedical approaches provide versatile platforms that can be utilized to enhance the overall potency, safety, and stability of vaccines, thus augmenting the desired immune response. Their modulable conformational features of size, shape, surface charge, antigen display, and composition allow for precise tuning and optimization of the nanoconstructs for the management of a variety of diseases and pathological conditions. The ability to control the release of their encapsulated cargoes and the possibility of surface decoration with various moieties support the construction of multifunctional nanomaterials that ultimately boost and prolong the immune response elicited and/or therapeutic effects, selectively at the diseased tissues and target sites.
Collapse
|
18
|
Para O, Caruso L, Pestelli G, Tangianu F, Carrara D, Maddaluni L, Tamburello A, Castelnovo L, Fedi G, Guidi S, Pestelli C, Pennella B, Ciarambino T, Nozzoli C, Dentali F. Ferritin as prognostic marker in COVID-19: the FerVid study. Postgrad Med 2022; 134:58-63. [PMID: 34613875 PMCID: PMC8544665 DOI: 10.1080/00325481.2021.1990091] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/09/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND In COVID-19 patients the progressive clinical deterioration seems secondary to the activation of a cytokine storm. Ferritin is considered a direct mediator of the immune system and some evidences suggested a shared physio-pathogenic basis between COVID-19 and 'Hyperferritinemic Syndromes.' The aim of our study was to evaluate the prognostic role of ferritin in COVID-19 patients. METHODS We retrospectively studied consecutive COVID-19 patients admitted to four Italian Internal Medicine Units. Role of potential prognostic markers was evaluated with binary logistic regression analysis and results were expressed as odds ratios (ORs) with the corresponding 95% confidence intervals (CIs). Poor outcome was defined as death or need to transfer in the intensive care unit. RESULTS Two hundred patients were included (mean age 68.75 ± 13.22 years). Ferritin value was highly elevated (>3000 ng/mL) in 8% of our population; 13% of patients were transferred to intensive care units and 12% of patients died. At multivariate analysis, highly elevated ferritin levels (OR 16.67 C.I. 4.89-57.57 p < 0.001) and hemoglobin < 10 g/dL (OR 8.88 C.I. 2.02-39.09 p = 0.004) were independently associated with a bad outcome.Patients with ferritin values > 3000 ng/ml appeared to have an inflammatory activation with elevated values of CRP and D-dimer and low values of lymphocyte count. CONCLUSION Our results confirm the prognostic role of ferritin in hospitalized COVID-19 patients. Patients with high ferritin levels should be considered critically ill and treated in an adequate setting. Furthermore, COVID-19 seems to share some characteristics with hyperferritinemic syndromes with potential therapeutic implications.
Collapse
Affiliation(s)
- Ombretta Para
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | - Lorenzo Caruso
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | - Giulia Pestelli
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | - Flavio Tangianu
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Carrara
- U.O.C. General Medicine, Versilia Hospital, Viareggio, Italy
| | - Lucia Maddaluni
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | | | - Laura Castelnovo
- Department of Internal Medicine, Hospital of Legnano, Legnano, Italy
| | - Giacomo Fedi
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | - Stefano Guidi
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | - Caterina Pestelli
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | - Benedetta Pennella
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | - Carlo Nozzoli
- Internal Medicine Unit 1, University Hospital Careggi, Florence, Italy
| | - Francesco Dentali
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
19
|
Nguyen N, Nguyen H, Ukoha C, Hoang L, Patel C, Ikram FG, Acharya P, Dhillon A, Sidhu M. Relation of interleukin-6 levels in COVID-19 patients with major adverse cardiac events. Proc AMIA Symp 2021; 35:6-9. [PMID: 34966215 PMCID: PMC8425472 DOI: 10.1080/08998280.2021.1961571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Elevated interleukin-6 (IL-6) levels may correlate with disease severity in COVID-19. We analyzed whether there was an association between elevated IL-6 levels and major adverse cardiac events (MACE) and/or mortality in COVID-19 patients. A retrospective chart review was performed on COVID-19 patients among four hospitals in one health system from March to May 2020, extracting information on baseline characteristics, MACE (i.e., myocardial infarction, stroke, deep venous thrombosis/pulmonary embolism, or shock requiring vasopressor support), mortality, and IL-6 levels. Of the 496 patients hospitalized with COVID-19, 191 patients had an IL-6 level drawn and 68% had elevated IL-6 levels. The elevated IL-6 population had higher odds of developing a MACE compared to the normal IL-6 population (P < 0.0001, odds ratio [OR] = 5.91, 95% confidence interval [CI] = 2.65–14.11). The elevated IL-6 population also had higher mortality rates (28.2% vs 5%, P = 0.0001, OR = 7.47, 95% CI = 2.19–39.32) and an increased incidence of a MACE and/or mortality (58.78% vs 20.00%, P < 0.0001, OR = 5.7, 95% CI 2.65–12.83) compared to the normal IL-6 population. Elevated IL-6 levels in COVID-19 patients may be associated with MACE and/or mortality. Monitoring IL-6 levels in COVID-19 patients may help risk-stratify patients.
Collapse
Affiliation(s)
- Nicholas Nguyen
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, Texas
| | - Hao Nguyen
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, Texas
| | - Chijioke Ukoha
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, Texas
| | - Lawrence Hoang
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, Texas
| | - Chirag Patel
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, Texas
| | - Farukh G Ikram
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, Texas
| | - Priyanka Acharya
- Clinical Research Institute, Methodist Health System, Dallas, Texas
| | - Anmol Dhillon
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, Texas
| | - Manavjot Sidhu
- Department of Cardiology, Methodist Dallas Medical Center, Dallas, Texas
| |
Collapse
|
20
|
Tewari D, Boger L, Brady S, Livengood J, Killian ML, Nair MS, Thirumalapura N, Kuchipudi SV, Zellers C, Schroder B, Torchetti M, Carpenter A, Kunkel A, Brightbill K. Transmission of SARS-CoV-2 from humans to a 16-year-old domestic cat with comorbidities in Pennsylvania, USA. Vet Med Sci 2021; 8:899-906. [PMID: 34910368 PMCID: PMC8959284 DOI: 10.1002/vms3.695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background and Objectives Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), besides causing human infection, has been shown to naturally infect several susceptible animal species including large cats (tigers, lions, pumas, spotted leopards), dogs, cats, ferrets, gorillas and minks. Cats and minks are continuing to be the most reported species with SARS‐CoV‐2 infections among animals but it needs to be investigated further. Methods and Results We report the detection of SARS‐CoV‐2 from a domestic cat that exhibited respiratory disease after being exposed to SARS‐CoV‐2 virus from humans in the same household. SARS‐CoV‐2 RNA was detected in two oropharyngeal swabs collected at two time points, 11 days apart; the first, when the cat was reported to be sick and the second, before euthanasia due to poor prognosis. The viral nucleic acid detected at two time points showed no genomic variation and resembled the clade GH circulating in humans in the United States. Clinical and pathological findings noted in this 16‐year‐old cat were consistent with respiratory and cardiac insufficiency. Conclusions SARS‐CoV‐2 viral infection was likely an incidental clinical finding, as the virus was not detected in fixed lungs, heart, or kidney tissues. Only fresh lung tissue collected at necropsy showed the presence of viral nucleic acid, albeit at a very low level. Further research is needed to clarify the clinical course of SARS‐CoV‐2 in companion animals of advanced age and underlying cardiac disease.
Collapse
Affiliation(s)
- Deepanker Tewari
- Pennsylvania Department of Agriculture, Pennsylvania Veterinary Laboratory, Harrisburg, Pennsylvania, USA
| | - Lore Boger
- Pennsylvania Department of Agriculture, Pennsylvania Veterinary Laboratory, Harrisburg, Pennsylvania, USA
| | - Steven Brady
- Silver Spring Clinic, Palmyra, Pennsylvania, USA
| | - Julia Livengood
- Pennsylvania Department of Agriculture, Pennsylvania Veterinary Laboratory, Harrisburg, Pennsylvania, USA
| | - Mary Lea Killian
- Diagnostic Virology Laboratory, National Veterinary Services Laboratories, Iowa, USA
| | - Meera Surendran Nair
- Animal Diagnostic Laboratory, Pennsylvania State University, State College, Pennsylvania, USA
| | - Nagaraja Thirumalapura
- Pennsylvania Department of Agriculture, Pennsylvania Veterinary Laboratory, Harrisburg, Pennsylvania, USA
| | - Suresh V Kuchipudi
- Animal Diagnostic Laboratory, Pennsylvania State University, State College, Pennsylvania, USA
| | - Corey Zellers
- Pennsylvania Department of Agriculture, Pennsylvania Veterinary Laboratory, Harrisburg, Pennsylvania, USA
| | - Betsy Schroder
- Pennsylvania Department of Health, Bureau of Epidemiology, Harrisburg, Pennsylvania, USA
| | - Mia Torchetti
- Diagnostic Virology Laboratory, National Veterinary Services Laboratories, Iowa, USA
| | | | - Amber Kunkel
- Centers for Disease Control, Atlanta, Georgia, USA
| | - Kevin Brightbill
- Pennsylvania Department of Agriculture, Pennsylvania Veterinary Laboratory, Harrisburg, Pennsylvania, USA
| |
Collapse
|
21
|
Jamil Z, Almajhdi FN, Khalid S, Asghar M, Ahmed J, Waheed Y. Comparison of Low-Versus High-Dose Steroids in the Clinical Outcome of Hospitalized COVID-19 Patients. Antibiotics (Basel) 2021; 10:antibiotics10121510. [PMID: 34943722 PMCID: PMC8698954 DOI: 10.3390/antibiotics10121510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
(1) Objectives: Patients with COVID-19 infection have been given various formulations and dosages of steroids over the last year and a half. This study aims to compare the effects of different formulations and doses of steroids on the 30 day in-hospital clinical outcome of patients with severe COVID-19 infection. (2) Material and Methods: An analysis of a retrospective cohort was carried out on patients with severe COVID-19 infection in a high-dependency unit (HDU) between February and July 2021. In total, 557 patients were included in this study. Patients who did not receive steroids (124) were excluded. Patients were divided into three groups based on dosages of steroids (Dexamethasone = 6 mg/day, Dexamethasone > 6 mg/day, and Methylprednisolone = 500 mg/day), given for 10 days. First, clinical outcome was evaluated on the 10th day of steroid administration in relation to mode of oxygen delivery. Then, Kaplan-Meier analysis was employed to determine 30 day in-hospital survival in relation to the use of steroid. (3) Results: Three groups were statistically equal according to biochemical characteristics. After 10 days of Methylprednisolone = 500 mg/day vs. Dexamethasone = 6 mg/day, 10.9% vs. 6.2% of patients required invasive ventilation (p = 0.01). The 30 day in-hospital mortality was lowest, 3%, in individuals receiving Dexamethasone = 6 mg/day, compared to 3.9% in individuals receiving Dexamethasone > 6 mg/day and 9.9% in individuals receiving Methylprednisolone = 500 mg/day, respectively. The median elapsed time was longer than 28 days between admission and outcome for Dexamethasone = 6 mg/day, compared to 18 days for Dexamethasone > 6 mg/day and 17 days for Methylprednisolone = 500 mg/day (p = < 0.0001). Dexamethasone = 6 mg/day was found to be a positive predictor of clinical outcome in COVID-19 patients on regression analysis. (4) Conclusions: Low-dose Dexamethasone (6 mg/day) is more effective than high-dose Dexamethasone and Methylprednisolone in improving the survival outcome of severe COVID-19 cases.
Collapse
Affiliation(s)
- Zubia Jamil
- Department of Medicine, Foundation University Medical College, Foundation University Islamabad, Islamabad 44000, Pakistan; (Z.J.); (S.K.)
| | - Fahad N. Almajhdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Samreen Khalid
- Department of Medicine, Foundation University Medical College, Foundation University Islamabad, Islamabad 44000, Pakistan; (Z.J.); (S.K.)
| | - Muhammad Asghar
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Instututet, 17177 Stockholm, Sweden;
- Department of Infectious Diseases, Karolinska University Hospital, 17164 Stockholm, Sweden
| | - Jamal Ahmed
- Department of Pulmonology, Fauji Foundation Hospital, Rawalpindi 45000, Pakistan;
| | - Yasir Waheed
- Multidisciplinary Lab, Foundation University Medical College, Foundation University Islamabad, Islamabad 44000, Pakistan
- Correspondence:
| |
Collapse
|
22
|
Zolnikova OY, Maslennikov RV, Ivashkin VT, Dzhakhaya NL, Kiseleva OY, Potskhverashvili ND, Shorokhova SA. Dexamethasone in Treatment of Comorbid SARS-CoV-2 Patients Aged over 50 Years with Lung Injury over 50 %. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2021. [DOI: 10.22416/1382-4376-2021-31-3-43-50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aim. An efficacy assessment of steroid therapy in SARS-CoV-2 patients aged over 50 years with lung damage over 50 % (grade 3–4).Materials and methods. A retrospective study of 158 SARS-CoV-2 patients hospitalised in April—June 2020 was conducted under the inclusion criteria: age over 50 years, chest computed tomography (CT) for COVID-19-asso-ciated pneumonia, C-reactive protein (CRP) >50 mg/L, anticoagulant therapy, no contraindications to steroids, no biologic therapy. Cohort 1 patients (n = 96) received dexamethasone 4–12 mg/day, cohort 2 (n = 62) — a standard non-dexamethasone therapy.Results. Dexamethasone treatment associated with a significant alleviation of COVID-19-associated pneumonia in CT score (p = 0.001), reduced fibrinogen (p = 0.001), a trend to CRP reduction by day 8–10 and lower demand for oxygen therapy, including ventilatory support (p = 0.001). Mortality rate was 19.8 and 75.8 % in cohorts 1 and 2, respectively (p = 0.001).Conclusion. Dexamethasone therapy 4–12 mg/day in SARS-CoV-2 patients aged 50+ years with grade 3–4 CT changes receiving LMWH from start of hospitalisation significantly improved CT scores and reduced mortality.
Collapse
Affiliation(s)
- O. Yu. Zolnikova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - R. V. Maslennikov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - V. T. Ivashkin
- Sechenov First Moscow State Medical University (Sechenov University)
| | - N. L. Dzhakhaya
- Sechenov First Moscow State Medical University (Sechenov University)
| | - O. Yu. Kiseleva
- Sechenov First Moscow State Medical University (Sechenov University)
| | | | - S. A. Shorokhova
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
23
|
Cho DH, Choi J, Gwon JG. Metabolic syndrome and the risk of COVID-19 infection: A nationwide population-based case-control study. Nutr Metab Cardiovasc Dis 2021; 31:2596-2604. [PMID: 34348879 PMCID: PMC8158348 DOI: 10.1016/j.numecd.2021.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Metabolic syndrome (MetS) is a chronic, low-grade inflammatory disease. This study aimed to investigate the impact of MetS on the risk and severity of COVID-19. METHODS AND RESULTS We investigated a nationwide cohort with COVID-19 including all patients who underwent the test for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Korea. The COVID-19 group included 4070 patients with positive SARS-CoV-2 test results, and the age- and sex-matched control group included 27,618 subjects with negative SARS-CoV-2 test results. The endpoints were SARS-CoV-2 positivity and the severity of COVID-19. The prevalence of MetS was 24.7% and 24.5% in the COVID-19 and control groups, respectively. The presence of MetS was not associated with the risk of developing COVID-19. Among the components of MetS, central obesity was associated with a higher risk of COVID-19 infection (adjusted odds ratio [aOR], 1.17; 95% confidence interval [CI], 1.06-1.28, P = 0.001). The presence of MetS was significantly associated with severe COVID-19 (aOR, 1.25; 95% CI, 0.78-2.00, P = 0.352). Among the individual components of MetS, prediabetes/diabetes mellitus was associated with a higher risk of severe COVID-19 (aOR, 1.61; 95% CI, 1.21-2.13, P = 0.001). The risk of severe COVID-19 linearly increased according to the number of metabolic components (P for trend = 0.005). CONCLUSION In this nationwide cohort study, the individuals with MetS had a significant increase in the risk of severe COVID-19 infection. These patients, particularly those with central obesity and insulin resistance, deserve special attention amid the COVID-19 pandemic.
Collapse
Affiliation(s)
- Dong-Hyuk Cho
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Gangwon, Republic of Korea
| | - Jimi Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jun Gyo Gwon
- Division of Transplantation and Vascular Surgery, Department of Surgery, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Rotondo JC, Martini F, Maritati M, Mazziotta C, Di Mauro G, Lanzillotti C, Barp N, Gallerani A, Tognon M, Contini C. SARS-CoV-2 Infection: New Molecular, Phylogenetic, and Pathogenetic Insights. Efficacy of Current Vaccines and the Potential Risk of Variants. Viruses 2021; 13:1687. [PMID: 34578269 PMCID: PMC8473168 DOI: 10.3390/v13091687] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/16/2021] [Accepted: 08/21/2021] [Indexed: 12/11/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a newly discovered coronavirus responsible for the coronavirus disease 2019 (COVID-19) pandemic. COVID-19 has rapidly become a public health emergency of international concern. Although remarkable scientific achievements have been reached since the beginning of the pandemic, the knowledge behind this novel coronavirus, in terms of molecular and pathogenic characteristics and zoonotic potential, is still relatively limited. Today, there is a vaccine, or rather several vaccines, which, for the first time in the history of highly contagious infectious diseases that have plagued mankind, has been manufactured in just one year. Currently, four vaccines are licensed by regulatory agencies, and they use RNA or viral vector technologies. The positive effects of the vaccination campaign are being felt in many parts of the world, but the disappearance of this new infection is still far from being a reality, as it is also threatened by the presence of novel SARS-CoV-2 variants that could undermine the effectiveness of the vaccine, hampering the immunization control efforts. Indeed, the current findings indicate that SARS-CoV-2 is adapting to transmission in humans more efficiently, while further divergence from the initial archetype should be considered. In this review, we aimed to provide a collection of the current knowledge regarding the molecular, phylogenetic, and pathogenetic insights into SARS-CoV-2. The most recent findings obtained with respect to the impact of novel emerging SARS-CoV-2 variants as well as the development and implementation of vaccines are highlighted.
Collapse
Affiliation(s)
- John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Martina Maritati
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Giulia Di Mauro
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Carmen Lanzillotti
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Nicole Barp
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Altea Gallerani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Carlo Contini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
25
|
Liu D, Zeng X, Ding Z, Lv F, Mehta JL, Wang X. Adverse Cardiovascular Effects of Anti-COVID-19 Drugs. Front Pharmacol 2021; 12:699949. [PMID: 34512335 PMCID: PMC8424204 DOI: 10.3389/fphar.2021.699949] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or COVID-19 infection is the cause of the ongoing global pandemic. Mortality from COVID-19 infection is particularly high in patients with cardiovascular diseases. In addition, COVID-19 patients with preexisting cardiovascular comorbidities have a higher risk of death. Main cardiovascular complications of COVID-19 are myocardial infarction, myocarditis, acute myocardial injury, arrhythmias, heart failure, stroke, and venous thromboembolism. Therapeutic interventions in terms of drugs for COVID-19 have many cardiac adverse effects. Here, we review the relative therapeutic efficacy and adverse effects of anti-COVID-19 drugs.
Collapse
Affiliation(s)
- Dongling Liu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xiang Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, China
| | - Zufeng Ding
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States
| | - Fenghua Lv
- Department of Cardiology, Xinxiang Medical University First Affiliated Hospital, Weihui, China
| | - Jawahar L. Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
- Department of Cardiology, Xinxiang Medical University First Affiliated Hospital, Weihui, China
| |
Collapse
|
26
|
Grygiel-Górniak B, Shaikh O, Kumar NN, Hsu SH, Samborski W. Use of the rheumatic drug tocilizumab for treatment of SARS-CoV-2 patients. Reumatologia 2021; 59:252-259. [PMID: 34538956 PMCID: PMC8436792 DOI: 10.5114/reum.2021.108554] [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: 05/18/2021] [Accepted: 07/19/2021] [Indexed: 11/27/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly infectious respiratory disease caused by a new coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has been observed to vary in its degree of symptoms. One of the most important clinical manifestations is pneumonia and the subsequent worsening of the hyperinflammatory state and cytokine storm. Tocilizumab (TCB) is a recombinant humanized, anti-human monoclonal antibody of the immunoglobulin G1k (IgG1k) subclass that acts against soluble and membrane-bound interleukin six receptors (IL-6R). There is wide use of TCB in rheumatic diseases. However, recently this medication has been used in the treatment of SARS-CoV-2 infection. Tocilizumab application in COVID-19 patients with a high risk of a cytokine storm shows a positive response in reducing the mortality rate. Moreover, TCB minimizes the time needed to recover, improves oxygenation, shortens the duration of vasopressor support, and reduces the likelihood of invasive mechanical ventilation. Therefore we provide an overview of recent studies to understand the efficacy of this drug under various circumstances, including COVID-19 and rheumatic pathologies. This article also explores and integrates the different treatment possibilities in prominent anti-inflammatory and immune-modulatory-related symptoms. The preliminary data demonstrate promising results regarding the efficacy of TCB use in severe COVID-19 patients. Nevertheless, randomized controlled trials, with adequate sample sizes and sufficient follow-up periods, are needed to form conclusions and establish treatment recommendations.
Collapse
Affiliation(s)
- Bogna Grygiel-Górniak
- Department of Rheumatology, Rehabilitation and Internal Medicine, Poznan University of Medical Sciences, Poland
| | - Osama Shaikh
- Department of Rheumatology, Rehabilitation and Internal Medicine, Poznan University of Medical Sciences, Poland
| | - Nikita Niranjan Kumar
- Department of Rheumatology, Rehabilitation and Internal Medicine, Poznan University of Medical Sciences, Poland
| | - Shao Heng Hsu
- Department of Rheumatology, Rehabilitation and Internal Medicine, Poznan University of Medical Sciences, Poland
| | - Włodzimierz Samborski
- Department of Rheumatology, Rehabilitation and Internal Medicine, Poznan University of Medical Sciences, Poland
| |
Collapse
|
27
|
Maione F, Casillo GM, Raucci F, Salvatore C, Ambrosini G, Costa L, Scarpa R, Caso F, Bucci M. Interleukin-17A (IL-17A): A silent amplifier of COVID-19. Biomed Pharmacother 2021; 142:111980. [PMID: 34364043 PMCID: PMC8318692 DOI: 10.1016/j.biopha.2021.111980] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/27/2022] Open
Abstract
One of the hallmarks of COVID-19 is the cytokine storm that provokes primarily pneumonia followed by systemic inflammation. Emerging evidence has identified a potential link between elevated interleukin-17A (IL-17A) levels and disease severity and progression. Considering that per se, IL-17A can activate several inflammatory pathways, it is plausible to hypothesize an involvement of this cytokine in COVID-19 clinical outcomes. Thus, IL-17A could represent a marker of disease progression and/or a target to develop therapeutic strategies. This hypothesis paper aims to propose this "unique" cytokine as a silent amplifier of the COVID-19 immune response and (potentially) related therapy.
Collapse
Affiliation(s)
- Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Cristian Salvatore
- New.Fa.DEm SRL, Viale Ferrovie Dello Stato, 1, 80014 Giugliano in Campania, Naples, Italy
| | - Giovanna Ambrosini
- New.Fa.DEm SRL, Viale Ferrovie Dello Stato, 1, 80014 Giugliano in Campania, Naples, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Francesco Caso
- Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini, 5, 80131 Naples, Italy.
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
28
|
Aksakal A, Kerget B, Kerget F, Aşkın S. Evaluation of the relationship between macrophage migration inhibitory factor level and clinical course in patients with COVID-19 pneumonia. J Med Virol 2021; 93:6519-6524. [PMID: 34241898 PMCID: PMC8426684 DOI: 10.1002/jmv.27189] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022]
Abstract
The COVID‐19 pandemic, which has ravaged our world for more than a year, still shapes our agenda with a scale of intensity that fluctuates over time. In our study, we aimed to determine the correlation between serum migration inhibitory factor (MIF) level and disease severity in COVID‐19 with different prognoses. Between 15 October 2020 and 20 January 2021, 110 patients over the age of 18 who were diagnosed with COVID‐19 and 40 volunteer healthcare personnel were included in our study. MIF levels were measured by enzyme‐linked immunosorbent assay. In the comparison of serum MIF values in the patient and control group, it was observed that the MIF level was significantly higher in patients with both moderate and severe COVID‐19 levels compared to the control group (p = 0.001, 0.001). In the comparison of serum MIF values of moderate to severe COVID‐19 patients, it was observed that MIF level was higher in severe patients (p = 0.001). In the receiver operating characteristic curve analysis performed to differentiate between severe and moderate COVID‐19 patients with MIF levels, the area under the curve was observed as 0.78. When the cutoff value of the MIF level was taken as 4.455 ng/ml, the sensitivity was 83% and the specificity was 62%. Failure to adequately balance the pro‐inflammatory cytokines synthesized in COVID‐19 with anti‐inflammatory effect is the most important reason for the aggravation of the disease course. Playing a role in pro‐inflammatory cytokine synthesis, MIF can provide important information about the disease prognosis in the early period. Failure to adequately balance the proinflammatory cytokines synthesized in COVID‐19 with anti‐inflammatory effect is the most important reason for the aggravation of the disease course. Playing a role in proinflammatory cytokine synthesis, MIF can provide important information about the disease prognosis in the early period.
Collapse
Affiliation(s)
- Alperen Aksakal
- Department of Pulmonary Diseases, Health Sciences University Erzurum Regional Education and Research Hospital, Yakutiye, Erzurum, Turkey
| | - Buğra Kerget
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Ferhan Kerget
- Department of Infection Diseases and Clinical Microbiology, Health Sciences University Erzurum Regional Education and Research Hospital, Yakutiye, Erzurum, Turkey
| | - Seda Aşkın
- Department of Biochemistry, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| |
Collapse
|
29
|
Ruscitti P, Conforti A, Cipriani P, Giacomelli R, Tasso M, Costa L, Caso F. Pathogenic implications, incidence, and outcomes of COVID-19 in autoimmune inflammatory joint diseases and autoinflammatory disorders. Adv Rheumatol 2021; 61:45. [PMID: 34238376 PMCID: PMC8264991 DOI: 10.1186/s42358-021-00204-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
As the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread rapidly, there are still many unresolved questions of how this virus would impact on autoimmune inflammatory joint diseases and autoinflammatory disorders. The main aim of this paper is to describe the main studies focusing their attention on COVID-19 incidence and outcomes of rheumatoid arthritis (RA), spondylarthritis (SpA), and autoinflammatory disease cohorts. We also revised possible pathogenic mechanisms associated with. Available data suggest that, in patients with RA and SpA, the immunosuppressive therapy, older age, male sex, and the presence of comorbidities (hypertension, lung disease, diabetes, CVD, and chronic renal insufficiency/end-stage renal disease) could be associated with an increased risk of infections and high rate of hospitalization. Other studies have shown that lower odds of hospitalization were associated with bDMARD or tsDMARDs monotherapy, driven largely by anti-TNF therapies. For autoinflammatory diseases, considering the possibility that COVID-19 could be associated with a cytokine storm syndrome, the question of the susceptibility and severity of SARS-CoV-2 infection in patients displaying innate immunity disorders has been raised. In this context, data are very scarce and studies available did not clarify if having an autoinflammatory disorder could be or not a risk factor to develop a more severe COVID-19. Taking together these observations, further studies are likely to be needed to fully characterize these specific patient groups and associated SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Alessandro Conforti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome 'Campus Biomedico', Rome, Italy
| | - Marco Tasso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
30
|
Laveneziana P, Straus C, Meiners S. How and to What Extent Immunological Responses to SARS-CoV-2 Shape Pulmonary Function in COVID-19 Patients. Front Physiol 2021; 12:628288. [PMID: 34267671 PMCID: PMC8276038 DOI: 10.3389/fphys.2021.628288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/31/2021] [Indexed: 12/25/2022] Open
Abstract
COVID-19 is a disease caused by a new coronavirus SARS-CoV-2, primarily impacting the respiratory system. COVID-19 can result in mild illness or serious disease leading to critical illness and requires admission to ICU due to respiratory failure. There is intense discussion around potential factors predisposing to and protecting from COVID-19. The immune response and the abnormal respiratory function with a focus on respiratory function testing in COVID-19 patients will be at the center of this Perspective article of the Frontiers in Physiology Series on “The Tribute of Physiology for the Understanding of COVID-19 Disease.” We will discuss current advances and provide future directions and present also our perspective in this field.
Collapse
Affiliation(s)
- Pierantonio Laveneziana
- Assistance Publique - Hôpitaux de Paris (AP-HP), Groupe Hospitalier Universitaire APHP-Sorbonne Université, Sites Pitié-Salpêtrière, Saint-Antoine et Tenon, Service des Explorations Fonctionnelles de la Respiration, de l'Exercice et de la Dyspnée (Département R3S), Paris, France.,Sorbonne Université, INSERM, UMRS 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Christian Straus
- Assistance Publique - Hôpitaux de Paris (AP-HP), Groupe Hospitalier Universitaire APHP-Sorbonne Université, Sites Pitié-Salpêtrière, Saint-Antoine et Tenon, Service des Explorations Fonctionnelles de la Respiration, de l'Exercice et de la Dyspnée (Département R3S), Paris, France.,Sorbonne Université, INSERM, UMRS 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
31
|
Sharanya CS, Sabu A, Haridas M. Potent phytochemicals against COVID-19 infection from phyto-materials used as antivirals in complementary medicines: a review. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021; 7:113. [PMID: 34095323 PMCID: PMC8170460 DOI: 10.1186/s43094-021-00259-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/10/2021] [Indexed: 12/23/2022] Open
Abstract
Background Following the outbreak of the COVID-19 pandemic, there was a surge of research activity to find methods/drugs to treat it. There has been drug-repurposing research focusing on traditional medicines. Concomitantly, many researchers tried to find in silico evidence for traditional medicines. There is a great increase in article publication to commensurate the new-found research interests. This situation inspired the authors to have a comprehensive understanding of the multitude of publications related to the COVID-19 pandemic with a wish to get promising drug leads. Main body This review article has been conceived and made as a hybrid of the review of the selected papers advertised recently and produced in the interest of the COVID-19 situation, and in silico work done by the authors. The outcome of the present review underscores a recommendation for thorough MDS analyses of the promising drug leads. The inclusion of in silico work as an addition to the review was motivated by a recently published article of Toelzer and colleagues. The in silico investigation of free fatty acids is novel to the field and it buttresses the further MDS analysis of drug leads for managing the COVID-19 pandemic. Conclusion The review performed threw light on the need for MDS analyses to be considered together with the application of other in silico methods of prediction of pharmacologic properties directing towards the sites of drug-receptor regulation. Also, the present analysis would help formulate new recipes for complementary medicines.
Collapse
Affiliation(s)
- C S Sharanya
- Inter University Centre for Bioscience and Department of Biotechnology & Microbiology, Dr. Janaki Ammal Campus, Kannur University, Thalassery, 670661 India
| | - A Sabu
- Inter University Centre for Bioscience and Department of Biotechnology & Microbiology, Dr. Janaki Ammal Campus, Kannur University, Thalassery, 670661 India
| | - M Haridas
- Inter University Centre for Bioscience and Department of Biotechnology & Microbiology, Dr. Janaki Ammal Campus, Kannur University, Thalassery, 670661 India
| |
Collapse
|
32
|
Rubilar T, Barbieri ES, Gazquez A, Avaro M. Sea Urchin Pigments: Echinochrome A and Its Potential Implication in the Cytokine Storm Syndrome. Mar Drugs 2021; 19:267. [PMID: 34064550 PMCID: PMC8151293 DOI: 10.3390/md19050267] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Echinochrome A (EchA) is a pigment from sea urchins. EchA is a polyhydroxylated 1,4-naphthoquinone that contains several hydroxyl groups appropriate for free-radical scavenging and preventing redox imbalance. EchA is the most studied molecule of this family and is an active principle approved to be used in humans, usually for cardiopathies and glaucoma. EchA is used as a pharmaceutical drug. Methods: A comprehensive literature and patent search review was undertaken using PubMed, as well as Google Scholar and Espacenet search engines to review these areas. Conclusions: In the bloodstream, EchA can mediate cellular responses, act as a radical scavenger, and activate the glutathione pathway. It decreases ROS imbalance, prevents and limits lipid peroxidation, and enhances mitochondrial functions. Most importantly, EchA contributes to the modulation of the immune system. EchA can regulate the generation of regulatory T cells, inhibit pro-inflammatory IL-1β and IL-6 cytokine production, while slightly reducing IL-8, TNF-α, INF-α, and NKT, thus correcting immune imbalance. These characteristics suggest that EchA is a candidate drug to alleviate the cytokine storm syndrome (CSS).
Collapse
Affiliation(s)
- Tamara Rubilar
- Laboratorio de Química de Organismos Marinos, Instituto Patagónico del Mar, Universidad Nacional de la Patagonia San Juan Bosco (UNPSJB), Puerto Madryn 9120, Chubut, Argentina;
- Laboratorio de Oceanografía Biológica, Centro Para el Estudio de Sistemas Marinos (CESIMAR), CONICET, Puerto Madryn 9120, Chubut, Argentina;
| | - Elena S. Barbieri
- Laboratorio de Oceanografía Biológica, Centro Para el Estudio de Sistemas Marinos (CESIMAR), CONICET, Puerto Madryn 9120, Chubut, Argentina;
- Laboratorio de Virología, Instituto Patagónico del Mar, Universidad Nacional de la Patagonia San Juan Bosco (UNPSJB), Puerto Madryn 9120, Chubut, Argentina
| | - Ayelén Gazquez
- Instituto Tecnológico de Chascomús, The Chascomús Technological Institute (INTECH), CONICET-UNSAM, Chascomús 7130, Buenos Aires, Argentina;
| | - Marisa Avaro
- Laboratorio de Química de Organismos Marinos, Instituto Patagónico del Mar, Universidad Nacional de la Patagonia San Juan Bosco (UNPSJB), Puerto Madryn 9120, Chubut, Argentina;
| |
Collapse
|
33
|
Wang X, He Z, Zhao X. Immunoregulatory therapy strategies that target cytokine storms in patients with COVID-19 (Review). Exp Ther Med 2021; 21:319. [PMID: 33732292 PMCID: PMC7903484 DOI: 10.3892/etm.2021.9750] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
A cytokine storm is an uncontrolled, excessive immune response that contributes to the pathogenesis of coronavirus disease 2019 (COVID-19). Viral infections lead to the loss of negative feedback in immune regulation and an abnormal elevation of the levels of multiple cytokines. In COVID-19, this causes diffuse damage to alveolar functions and may culminate in multiple organ dysfunction. Immunoregulatory therapies target the cytokine storms induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, and include monoclonal antibodies, recombinant granulocyte-macrophage colony stimulating factor, interferon, mesenchymal stem cell-based therapy, thymosin, immunoglobulins and blood purification therapies. These approaches may be effective in the alleviation of COVID-19 symptoms. In this review, cytokine storms caused by SARS-CoV-2 infections are evaluated and discussed, and advances in immunoregulatory therapy strategies for patients with COVID-19 are reviewed.
Collapse
Affiliation(s)
- Xianyao Wang
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
- National Joint Local Engineering Laboratory for Cell Engineering and Biomedicine Technique, Guizhou Province Key Laboratory of Regenerative Medicine, Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, Guizhou 550004, P.R. China
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Zhixu He
- National Joint Local Engineering Laboratory for Cell Engineering and Biomedicine Technique, Guizhou Province Key Laboratory of Regenerative Medicine, Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, Guizhou 550004, P.R. China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xing Zhao
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
- National Joint Local Engineering Laboratory for Cell Engineering and Biomedicine Technique, Guizhou Province Key Laboratory of Regenerative Medicine, Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, Guizhou 550004, P.R. China
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
34
|
Hemophagocytic Lymphohistiocytosis in the Emergency Department: Recognizing and Evaluating a Hidden Threat. J Emerg Med 2021; 60:743-751. [PMID: 33745765 PMCID: PMC7972988 DOI: 10.1016/j.jemermed.2021.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/04/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022]
Abstract
Background Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hematologic disorder resulting from an ineffective and pathologic activation of the immune response system that may mimic common emergency department presentations, including sepsis, acute liver failure, disseminated intravascular coagulation, and flu-like illnesses such as coronavirus disease 2019 (COVID-19). Objective This narrative review provides a summary of the disease and recommendations for the recognition and diagnostic evaluation of HLH with a focus on the emergency clinician. Discussion Though the condition is rare, mortality rates are high, ranging from 20% to 80% and increasing with delays in treatment. Importantly, HLH has been recognized as a severe variation of the cytokine storm associated with COVID-19. Common features include a history of infection or malignancy, fever, splenomegaly or hepatomegaly, hyperferritinemia, cytopenias, coagulopathies, abnormal liver enzymes, and hypertriglyceridemia. Using specific features of the history, physical examination, laboratory studies, and tools such as the HScore, HLH-2004/2009, and hyperferritinemia thresholds, the emergency clinician can risk-stratify patients and admit for definitive testing. Once diagnosed, disease specific treatment can be initiated. Conclusion This review describes the relevant pathophysiology, common presentation findings, and a framework for risk stratification in the emergency department.
Collapse
|
35
|
Hasbal NB, Turgut D, Oguz EG, Ulu S, Gungor O. Effect of Calcineurin Inhibitors and Mammalian Target of Rapamycin Inhibitors on the Course of COVID-19 in Kidney Transplant Recipients. Ann Transplant 2021; 26:e929279. [PMID: 33707409 PMCID: PMC7962418 DOI: 10.12659/aot.929279] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
Coronavirus disease 19 (COVID-19) has been an ongoing pandemic since December 2019. Unfortunately, kidney transplant recipients are a high-risk group during the disease course, and scientific data are still limited in this patient group. Beyond the dosage of immunosuppressive drugs, pharmacological immunosuppression may also alter the infection response in the COVID-19 course. The effects of immunosuppressive agents on the development and process of infection should not be decided only by determining how potent they are and how much they suppress the immune system; it is also thought that the direct effect of the virus, increased oxidative stress, and cytokine storm play a role in the pathogenesis of COVID-19 disease. There are data about immunosuppressive drugs like calcineurin inhibitors (CNI) or mammalian target of rapamycin inhibitors (mTORi) therapy related to their beneficial effects during any infection course. Limited data suggest that the use of CNI or mTORi may have beneficial effects on the process. In this hypothetical review, the probable impacts of CNI and mTORi on the pathogenesis of the COVID-19 were investigated.
Collapse
Affiliation(s)
- Nuri Baris Hasbal
- Clinic of Nephrology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Didem Turgut
- Department of Nephrology, Baskent University School of Medicine, Ankara, Turkey
| | - Ebru Gok Oguz
- Department of Nephrology, University of Health Sciences, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Sena Ulu
- Department of Nephrology, Afyonkarahisar Health Sciences University School of Medicine, Afyonkarahisar, Turkey
| | - Ozkan Gungor
- Department of Nephrology, Kahramanmaras Sutcu Imam University School of Medicine, Kahramanmaras, Turkey
| |
Collapse
|
36
|
Han M, Ma K, Wang X, Yan W, Wang H, You J, Wang Q, Chen H, Guo W, Chen T, Ning Q, Luo X. Immunological Characteristics in Type 2 Diabetes Mellitus Among COVID-19 Patients. Front Endocrinol (Lausanne) 2021; 12:596518. [PMID: 33776910 PMCID: PMC7992040 DOI: 10.3389/fendo.2021.596518] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/04/2021] [Indexed: 01/08/2023] Open
Abstract
Clinical Trial Registration www.ClinicalTrials.gov, identifier: NCT04365634. Context Diabetes mellitus was associated with increased severity and mortality of disease in COVID-19 pneumonia. So far the effect of type 2 diabetes (T2DM) or hyperglycemia on the immune system among COVID-19 disease has remained unclear. Objective We aim to explore the clinical and immunological features of type 2 diabetes mellitus (T2DM) among COVID-19 patients. Design and Methods In this retrospective study, the clinical and immunological characteristics of 306 hospitalized confirmed COVID-19 patients (including 129 diabetic and 177 non-diabetic patients) were analyzed. The serum concentrations of laboratory parameters including cytokines and numbers of immune cells were measured and compared between diabetic and non-diabetic groups. Results Compared with non-diabetic group, diabetic cases more frequently had lymphopenia and hyperglycemia, with higher levels of urea nitrogen, myoglobin, D-dimer and ferritin. Diabetic cases indicated the obviously elevated mortality and the higher levels of cytokines IL-2R, IL-6, IL-8, IL-10, and TNF-α, as well as the distinctly reduced Th1/Th2 cytokines ratios compared with non-diabetic cases. The longitudinal assays showed that compared to that at week 1, the levels of IL-6 and IL-8 were significantly elevated at week 2 after admission in non-survivors of diabetic cases, whereas there were greatly reductions from week 1 to week 2 in survivors of diabetic cases. Compared with survival diabetic patients, non-survival diabetic cases displayed distinct higher serum concentrations of IL-2R, IL-6, IL-8, IL-10, TNF-α, and lower Th1/Th2 cytokines ratios at week 2. Samples from a subset of participants were evaluated by flow cytometry for the immune cells. The counts of peripheral total T lymphocytes, CD4+ T cells, CD8+ T cells and NK cells were markedly lower in diabetic cases than in non-diabetic cases. The non-survivors showed the markedly declined counts of CD8+ T cells and NK cells than survivors. Conclusion The elevated cytokines, imbalance of Th1/Th2 cytokines ratios and reduced of peripheral numbers of CD8+ T cells and NK cells might contribute to the pathogenic mechanisms of high mortality of COVID-19 patients with T2DM.
Collapse
Affiliation(s)
- Meifang Han
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Ma
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongwu Wang
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie You
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuxia Wang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huilong Chen
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Guo
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Center for the Diagnosis of Genetic Metabolic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Atila C, Sailer CO, Bassetti S, Tschudin-Sutter S, Bingisser R, Siegemund M, Osswald S, Rentsch K, Rueegg M, Schaerli S, Kuster GM, Twerenbold R, Christ-Crain M. Prevalence and outcome of dysnatremia in patients with COVID-19 compared to controls. Eur J Endocrinol 2021; 184:409-418. [PMID: 33449918 PMCID: PMC9494345 DOI: 10.1530/eje-20-1374] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/12/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The pandemic of coronavirus disease (COVID-19) has rapidly spread globally and infected millions of people. The prevalence and prognostic impact of dysnatremia in COVID-19 is inconclusive. Therefore, we investigated the prevalence and outcome of dysnatremia in COVID-19. DESIGN The prospective, observational, cohort study included consecutive patients with clinical suspicion of COVID-19 triaged to a Swiss Emergency Department between March and July 2020. METHODS Collected data included clinical, laboratory and disease severity scoring parameters on admission. COVID-19 cases were identified based on a positive nasopharyngeal swab test for SARS-CoV-2, patients with a negative swab test served as controls. The primary analysis was to assess the prognostic impact of dysnatremia on 30-day mortality using a cox proportional hazard model. RESULTS 172 (17%) cases with COVID-19 and 849 (83%) controls were included. Patients with COVID-19 showed a higher prevalence of hyponatremia compared to controls (28.1% vs 17.5%, P < 0.001); while comparable for hypernatremia (2.9% vs 2.1%, P = 0.34). In COVID-19 but not in controls, hyponatremia was associated with a higher 30-day mortality (HR: 1.4, 95% CI: 1.10-16.62, P = 0.05). In both groups, hypernatremia on admission was associated with higher 30-day mortality (COVID-19 - HR: 11.5, 95% CI: 5.00-26.43, P < 0.001; controls - HR: 5.3, 95% CI: 1.60-17.64, P = 0.006). In both groups, hyponatremia and hypernatremia were significantly associated with adverse outcome, for example, intensive care unit admission, longer hospitalization and mechanical ventilation. CONCLUSION Our results underline the importance of dysnatremia as predictive marker in COVID-19. Treating physicians should be aware of appropriate treatment measures to be taken for patients with COVID-19 and dysnatremia.
Collapse
Affiliation(s)
- Cihan Atila
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- University of Basel, Department of Clinical Research, Basel, Switzerland
| | - Clara O Sailer
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- University of Basel, Department of Clinical Research, Basel, Switzerland
| | - Stefano Bassetti
- Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Sarah Tschudin-Sutter
- University of Basel, Department of Clinical Research, Basel, Switzerland
- Division of Infectious Disease & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Roland Bingisser
- Emergency Department, University Hospital Basel, Basel, Switzerland
| | - Martin Siegemund
- Department of Intensive Care, University Hospital Basel, Basel, Switzerland
| | - Stefan Osswald
- Department of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Katharina Rentsch
- Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Marco Rueegg
- Emergency Department, University Hospital Basel, Basel, Switzerland
| | - Sabrina Schaerli
- Emergency Department, University Hospital Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Department of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Raphael Twerenbold
- Department of Cardiology, University Hospital Basel, Basel, Switzerland
- Correspondence should be addressed to R Twerenbold;
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- University of Basel, Department of Clinical Research, Basel, Switzerland
- Correspondence should be addressed to M Christ-Crain;
| |
Collapse
|
38
|
Lam G, Zhou Y, Wang JX, Tsui YP. Targeting mesenchymal stem cell therapy for severe pneumonia patients. World J Stem Cells 2021; 13:139-154. [PMID: 33708343 PMCID: PMC7933990 DOI: 10.4252/wjsc.v13.i2.139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/03/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
Pneumonia is the inflammation of the lungs and it is the world's leading cause of death for children under 5 years of age. The latest coronavirus disease 2019 (COVID-19) virus is a prominent culprit to severe pneumonia. With the pandemic running rampant for the past year, more than 1590000 deaths has occurred worldwide up to December 2020 and are substantially attributable to severe pneumonia and induced cytokine storm. Effective therapeutic approaches in addition to the vaccines and drugs under development are hence greatly sought after. Therapies harnessing stem cells and their derivatives have been established by basic research for their versatile capacity to specifically inhibit inflammation due to pneumonia and prevent alveolar/pulmonary fibrosis while enhancing antibacterial/antiviral immunity, thus significantly alleviating the severe clinical conditions of pneumonia. In recent clinical trials, mesenchymal stem cells have shown effectiveness in reducing COVID-19-associated pneumonia morbidity and mortality; positioning these cells as worthy candidates for combating one of the greatest challenges of our time and shedding light on their prospects as a next-generation therapy to counter future challenges.
Collapse
Affiliation(s)
- Guy Lam
- School of Biomedical Sciences, University of Hong Kong, Hong Kong 999077, China
| | - Yuan Zhou
- Research and Development, Help Therapeutics Co. Ltd., Nanjing 211100, Jiangsu Province, China
| | - Jia-Xian Wang
- Research and Development, Help Therapeutics Co. Ltd., Nanjing 211100, Jiangsu Province, China
| | - Yat-Ping Tsui
- Research and Development, Help Therapeutics Co. Ltd., Nanjing 211100, Jiangsu Province, China.
| |
Collapse
|
39
|
Abstract
SARS-CoV-2 has claimed 2,137,908 lives in more than a year. Some COVID-19 patients experience sudden and rapid deterioration with the onset of fatal cytokine storm syndrome (CSS), which have increased interest in CSS’s mechanisms, diagnosis and therapy. Although the prototypic concept of CSS was first proposed 116 years ago, we have only begun to study and understand CSS for less than 30 years. Actually, diseases under CSS umbrella include familial/primary and secondary hemophagocytic lymphohistiocytosis (HLH), macrophage activation syndrome (MAS), infection-associated hemophagocytic syndrome, cytokine release syndrome (CRS), and cytokine storm (CS). Hematologic malignancies and autoimmune diseases that cause CSS are named malignancy-associated hemophagocytic syndrome (MAHS) and MAS, respectively. In-depth research on the pathogenesis of HLH/CSS has greatly increased the number of patients that were able to be definitively diagnosed with HLH/CSS. However, it should be emphasized that HLH/CSS diagnosis is difficult at the early stages due to the non-specific clinical signs and symptoms, which tends to result in missed and incorrect diagnoses. Therefore, clinicians should not only possess extensive clinical experience to ensure high sensitivity to the characteristics of HLH/CSS but must also be familiar with HLH-2004/2009 diagnostic criteria, and HScore methods. The paper concisely comment evolution of CSS classifications, cytokines associated with CSS, evolution of CSS diagnostic criteria and importance of the correct identification of hemophagocytes in diagnosing CSS, which is timely and may benefit clinicians familiar HLH-2004/2009 diagnostic criteria, and HScore methods. In addition, clinicians must also understand that there are some limitations to these diagnostic criteria. Abbreviations: aBMT: autologous bone marrow transplantation; CAR-T: chimeric antigen receptor-engineered T-cell; COVID-19: coronavirus disease 2019; CSS: cytokine storm syndrome; HLH: hemophagocytic lymphohistiocytosis; MAS: macrophage activation syndrome; CRS: cytokine release syndrome; CS: cytokine storm; MAHS: malignancy-associated hemophagocytic syndrome; IAHS: infection-associated hemophagocytic syndrome; fHLH/pHLH: familial/primary hemophagocytic lymphohistiocytosis; sHLH: secondary hemophagocytic lymphohistiocytosis; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; TCR-T, T-cell receptor-engineered T-cell
Collapse
Affiliation(s)
- Xi Yongzhi
- Department of Immunology and National Center for Biomedicine Analysis; Fifth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
40
|
Ryabkova VA, Churilov LP, Shoenfeld Y. Influenza infection, SARS, MERS and COVID-19: Cytokine storm - The common denominator and the lessons to be learned. Clin Immunol 2021; 223:108652. [PMID: 33333256 PMCID: PMC7832378 DOI: 10.1016/j.clim.2020.108652] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/29/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023]
Abstract
The outbreak of COVID-19 reminds us that the emerging and reemerging respiratory virus infections pose a continuing threat to human life. Cytokine storm syndromes of viral origin seem to have a common pathogenesis of the imbalanced immune response with the exaggerated inflammatory reaction combined with the reduction and functional exhaustion of T cells. Immunomodulatory therapy is gaining interest in COVID-19, but this strategy has received less attention in other respiratory viral infections than it deserved. In this review we suggest that based on the similarities of the immune dysfunction in the severe cases of different respiratory viral infections, some lessons from the immunomodulatory therapy of COVID-19 (particularly regarding the choice of an immunomodulatory drug, the selection of patients and optimal time window for this kind of therapy) could be applied for some cases of severe influenza infection and probably for some future outbreaks of novel severe respiratory viral infections.
Collapse
Affiliation(s)
- Varvara A Ryabkova
- Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint-Petersburg, Russian Federation
| | - Leonid P Churilov
- Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint-Petersburg, Russian Federation
| | - Yehuda Shoenfeld
- Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint-Petersburg, Russian Federation; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Affiliated to Tel-Aviv University School of Medicine, Tel-Hashomer, Israel.
| |
Collapse
|
41
|
CENTRAL HEMODYNAMICS AND OXYGEN TRANSPORT IN PATIENTS WITH ACUTE RESPIRATORY DISTRESS SYNDROME CAUSED BY COVID-19 AND THEIR IMPACT ON THE COURSE AND OUTCOMES OF THE DISEASE. EUREKA: HEALTH SCIENCES 2021. [DOI: 10.21303/2504-5679.2021.001622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The aim. Determine hemodynamic status and its impact on oxygen transport, frequency of adverse events and outcomes in patients with severe SARS-CoV-2 associated with acute respiratory distress syndrome (ARDS).
Materials and methods. A single-center prospective comparative study was conducted with 29 patients enrolled over the period of July—October 2020 who suffered a severe course of coronavirus disease and bilateral pneumonia associated with ARDS. Based on the estimated cardiac index (CI), patients were allocated to two groups: Group 1 included 14 patients with severe ARDS and CI 1.9 [1.5–2.5] L/min/m2, whereas Group 2 included 15 patients with CI 4.2 [3.2–8.1] L/min/m2 (p=0.001). Patient`s intensive care was regulated by the relevant orders of the Ministry of Health of Ukraine. Statistical analysis of the results was carried out using Statistica 10 software. Statistical significance of parameters was assessed using the non-parametric Wilcoxon criterion. Results were considered significant at p values <0.05. Data are presented as M [25–75]. Relative risk (RR) and odds ratio (OR) of adverse events were calculated.
Results. The severe course of coronavirus disease is associated with significant oxygen transport disorders that increase with hypovolemia. Despite the increase in oxygen delivery in the group with normal CI its high tissue extraction remained, which may be a sign of development mitochondrial distress.
Conclusions. Patients admitted to the ICU with severe COVID-19 may be in a state of hypovolemia and require individual assessment of hemodynamic status and the appointment of infusion therapy. Increased oxygen delivery in patients with normal cardiac index was associated with decreased adverse events rate and statistically significant decrease of mortality rate
Collapse
|
42
|
SARS-CoV-2 Mediated Hyperferritinemia and Cardiac Arrest: Preliminary Insights. Drug Discov Today 2021; 26:1265-1274. [PMID: 33493677 PMCID: PMC7826001 DOI: 10.1016/j.drudis.2021.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023]
|
43
|
Cannabis compounds exhibit anti-inflammatory activity in vitro in COVID-19-related inflammation in lung epithelial cells and pro-inflammatory activity in macrophages. Sci Rep 2021; 11:1462. [PMID: 33446817 PMCID: PMC7809280 DOI: 10.1038/s41598-021-81049-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/03/2021] [Indexed: 01/29/2023] Open
Abstract
Cannabis sativa is widely used for medical purposes and has anti-inflammatory activity. This study intended to examine the anti-inflammatory activity of cannabis on immune response markers associated with coronavirus disease 2019 (COVID-19) inflammation. An extract fraction from C. sativa Arbel strain (FCBD) substantially reduced (dose dependently) interleukin (IL)-6 and -8 levels in an alveolar epithelial (A549) cell line. FCBD contained cannabidiol (CBD), cannabigerol (CBG) and tetrahydrocannabivarin (THCV), and multiple terpenes. Treatments with FCBD and a FCBD formulation using phytocannabinoid standards (FCBD:std) reduced IL-6, IL-8, C-C Motif Chemokine Ligands (CCLs) 2 and 7, and angiotensin I converting enzyme 2 (ACE2) expression in the A549 cell line. Treatment with FCBD induced macrophage (differentiated KG1 cell line) polarization and phagocytosis in vitro, and increased CD36 and type II receptor for the Fc region of IgG (FcγRII) expression. FCBD treatment also substantially increased IL-6 and IL-8 expression in macrophages. FCBD:std, while maintaining anti-inflammatory activity in alveolar epithelial cells, led to reduced phagocytosis and pro-inflammatory IL secretion in macrophages in comparison to FCBD. The phytocannabinoid formulation may show superior activity versus the cannabis-derived fraction for reduction of lung inflammation, yet there is a need of caution proposing cannabis as treatment for COVID-19.
Collapse
|
44
|
Rios CI, Cassatt DR, Hollingsworth BA, Satyamitra MM, Tadesse YS, Taliaferro LP, Winters TA, DiCarlo AL. Commonalities Between COVID-19 and Radiation Injury. Radiat Res 2021; 195:1-24. [PMID: 33064832 PMCID: PMC7861125 DOI: 10.1667/rade-20-00188.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023]
Abstract
As the multi-systemic components of COVID-19 emerge, parallel etiologies can be drawn between SARS-CoV-2 infection and radiation injuries. While some SARS-CoV-2-infected individuals present as asymptomatic, others exhibit mild symptoms that may include fever, cough, chills, and unusual symptoms like loss of taste and smell and reddening in the extremities (e.g., "COVID toes," suggestive of microvessel damage). Still others alarm healthcare providers with extreme and rapid onset of high-risk indicators of mortality that include acute respiratory distress syndrome (ARDS), multi-organ hypercoagulation, hypoxia and cardiovascular damage. Researchers are quickly refocusing their science to address this enigmatic virus that seems to unveil itself in new ways without discrimination. As investigators begin to identify early markers of disease, identification of common threads with other pathologies may provide some clues. Interestingly, years of research in the field of radiation biology documents the complex multiorgan nature of another disease state that occurs after exposure to high doses of radiation: the acute radiation syndrome (ARS). Inflammation is a key common player in COVID-19 and ARS, and drives the multi-system damage that dramatically alters biological homeostasis. Both conditions initiate a cytokine storm, with similar pro-inflammatory molecules increased and other anti-inflammatory molecules decreased. These changes manifest in a variety of ways, with a demonstrably higher health impact in patients having underlying medical conditions. The potentially dramatic human impact of ARS has guided the science that has identified many biomarkers of radiation exposure, established medical management strategies for ARS, and led to the development of medical countermeasures for use in the event of a radiation public health emergency. These efforts can now be leveraged to help elucidate mechanisms of action of COVID-19 injuries. Furthermore, this intersection between COVID-19 and ARS may point to approaches that could accelerate the discovery of treatments for both.
Collapse
Affiliation(s)
- Carmen I. Rios
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Brynn A. Hollingsworth
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Yeabsera S. Tadesse
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Lanyn P. Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| |
Collapse
|
45
|
Fedele D, De Francesco A, Riso S, Collo A. Obesity, malnutrition, and trace element deficiency in the coronavirus disease (COVID-19) pandemic: An overview. Nutrition 2021; 81:111016. [PMID: 33059127 PMCID: PMC7832575 DOI: 10.1016/j.nut.2020.111016] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/24/2020] [Accepted: 08/29/2020] [Indexed: 02/06/2023]
Abstract
The world is currently facing the coronavirus disease (COVID-19) pandemic which places great pressure on health care systems and workers, often presents with severe clinical features, and sometimes requires admission into intensive care units. Derangements in nutritional status, both for obesity and malnutrition, are relevant for the clinical outcome in acute illness. Systemic inflammation, immune system impairment, sarcopenia, and preexisting associated conditions, such as respiratory, cardiovascular, and metabolic diseases related to obesity, could act as crucial factors linking nutritional status and the course and outcome of COVID-19. Nevertheless, vitamins and trace elements play an essential role in modulating immune response and inflammatory status. Overall, evaluation of the patient's nutritional status is not negligible for its implications on susceptibility, course, severity, and responsiveness to therapies, in order to perform a tailored nutritional intervention as an integral part of the treatment of patients with COVID-19. The aim of this study was to review the current data on the relevance of nutritional status, including trace elements and vitamin status, in influencing the course and outcome of the disease 3 mo after the World Health Organization's declaration of COVID-19 as a pandemic.
Collapse
Affiliation(s)
- Debora Fedele
- Dietetic and Clinical Nutrition Unit, San Giovanni Battista Hospital, Città della Salute e della Scienza, Turin, Italy.
| | - Antonella De Francesco
- Dietetic and Clinical Nutrition Unit, San Giovanni Battista Hospital, Città della Salute e della Scienza, Turin, Italy
| | - Sergio Riso
- Dietetic and Clinical Nutrition Unit, Maggiore della Carità Hospital, Novara, Italy
| | - Alessandro Collo
- Dietetic and Clinical Nutrition Unit, Maggiore della Carità Hospital, Novara, Italy
| |
Collapse
|
46
|
Caricchio R, Gallucci M, Dass C, Zhang X, Gallucci S, Fleece D, Bromberg M, Criner GJ. Preliminary predictive criteria for COVID-19 cytokine storm. Ann Rheum Dis 2021; 80:88-95. [PMID: 32978237 DOI: 10.1136/annrheumdis-2020-218323] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To develop predictive criteria for COVID-19-associated cytokine storm (CS), a severe hyperimmune response that results in organ damage in some patients infected with COVID-19. We hypothesised that criteria for inflammation and cell death would predict this type of CS. METHODS We analysed 513 hospitalised patients who were positive for COVID-19 reverse transcriptase PCR and for ground-glass opacity by chest high-resolution CT. To achieve an early diagnosis, we analysed the laboratory results of the first 7 days of hospitalisation. We implemented logistic regression and principal component analysis to determine the predictive criteria. We used a 'genetic algorithm' to derive the cut-offs for each laboratory result. We validated the criteria with a second cohort of 258 patients. RESULTS We found that the criteria for macrophage activation syndrome, haemophagocytic lymphohistiocytosis and the HScore did not identify the COVID-19 cytokine storm (COVID-CS). We developed new predictive criteria, with sensitivity and specificity of 0.85 and 0.80, respectively, comprising three clusters of laboratory results that involve (1) inflammation, (2) cell death and tissue damage, and (3) prerenal electrolyte imbalance. The criteria identified patients with longer hospitalisation and increased mortality. These results highlight the relevance of hyperinflammation and tissue damage in the COVID-CS. CONCLUSIONS We propose new early predictive criteria to identify the CS occurring in patients with COVID-19. The criteria can be readily used in clinical practice to determine the need for an early therapeutic regimen, block the hyperimmune response and possibly decrease mortality.
Collapse
Affiliation(s)
- Roberto Caricchio
- Medicine/Rheumatology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Marcello Gallucci
- Department of Psychology, University of Milano-Bicocca, Milan, Italy
| | - Chandra Dass
- Radiology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Xinyan Zhang
- Medicine/Rheumatology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Stefania Gallucci
- Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - David Fleece
- Pediatrics, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Michael Bromberg
- Medicine/Hematology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Gerard J Criner
- Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
47
|
Omarjee L, Perrot F, Meilhac O, Mahe G, Bousquet G, Janin A. Immunometabolism at the cornerstone of inflammaging, immunosenescence, and autoimmunity in COVID-19. Aging (Albany NY) 2020; 12:26263-26278. [PMID: 33361522 PMCID: PMC7803547 DOI: 10.18632/aging.202422] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/10/2020] [Indexed: 01/10/2023]
Abstract
Inflammaging constitutes the common factor for comorbidities predisposing to severe COVID-19. Inflammaging leads to T-cell senescence, and immunosenescence is linked to autoimmune manifestations in COVID-19. As in SLE, metabolic dysregulation occurs in T-cells. Targeting this T-cell dysfunction opens the field for new therapeutic strategies to prevent severe COVID-19. Immunometabolism-mediated approaches such as rapamycin, metformin and dimethyl fumarate, may optimize COVID-19 treatment of the elderly and patients at risk for severe disease.
Collapse
Affiliation(s)
- Loukman Omarjee
- Vascular Medicine Department, CHU Rennes, French National Health and Medical Research (Inserm), Clinical Investigation Center (CIC) 1414, University of Rennes 1, Rennes F-35033, France
- NuMeCan Institute, Exogenous and Endogenous Stress and Pathological Responses in Hepato-Gastrointestinal Diseases (EXPRES) Team, French National Health and Medical Research (Inserm) U1241, University of Rennes 1, Rennes F-35033, France
| | | | - Olivier Meilhac
- University of Reunion Island, INSERM, UMR 1188 Reunion, Indian Ocean Diabetic Atherothrombosis Therapies (DéTROI), CHU de La Réunion, Saint-Denis de La Réunion F-97400, France
| | - Guillaume Mahe
- Vascular Medicine Department, CHU Rennes, French National Health and Medical Research (Inserm), Clinical Investigation Center (CIC) 1414, University of Rennes 1, Rennes F-35033, France
| | - Guilhem Bousquet
- AP-HP Hôpital Avicenne, Oncologie Médicale, Bobigny F-93000, France
- Sorbonne University Paris Nord, INSERM, U942, Cardiovascular Markers in Stressed Conditions, MASCOT, Bobigny F-93000, France
| | - Anne Janin
- Sorbonne University Paris Nord, INSERM, U942, Cardiovascular Markers in Stressed Conditions, MASCOT, Bobigny F-93000, France
- Department of Pathology, Paris Diderot University, Sorbonne Paris Cité, Paris F-75010, France
| |
Collapse
|
48
|
Callaghan PJ, Rybakovsky E, Ferrick B, Thomas S, Mullin JM. Retinoic acid improves baseline barrier function and attenuates TNF-α-induced barrier leak in human bronchial epithelial cell culture model, 16HBE 14o. PLoS One 2020; 15:e0242536. [PMID: 33301441 PMCID: PMC7728186 DOI: 10.1371/journal.pone.0242536] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Retinoic acid (RA) has been shown to improve epithelial and endothelial barrier function and development and even suppress damage inflicted by inflammation on these barriers through regulating immune cell activity. This paper thus sought to determine whether RA could improve baseline barrier function and attenuate TNF-α-induced barrier leak in the human bronchial epithelial cell culture model, 16HBE14o- (16HBE). We show for the first time that RA increases baseline barrier function of these cell layers indicated by an 89% increase in transepithelial electrical resistance (TER) and 22% decrease in 14C-mannitol flux. A simultaneous, RA-induced 70% increase in claudin-4 attests to RA affecting the tight junctional (TJ) complex itself. RA was also effective in alleviating TNF-α-induced 16HBE barrier leak, attenuating 60% of the TNF-α-induced leak to 14C-mannitol and 80% of the leak to 14C-inulin. Interleukin-6-induced barrier leak was also reduced by RA. Treatment of 16HBE cell layers with TNF-α resulted in dramatic decrease in immunostaining for occludin and claudin-4, as well as a downward “band-shift” in occludin Western immunoblots. The presence of RA partially reversed TNF-α’s effects on these select TJ proteins. Lastly, RA completely abrogated the TNF-α-induced increase in ERK-1,2 phosphorylation without significantly decreasing the TNF-driven increase in total ERK-1,2. This study suggests RA could be effective as a prophylactic agent in minimizing airway barrier leak and as a therapeutic in preventing leak triggered by inflammatory cascades. Given the growing literature suggesting a “cytokine storm” may be related to COVID-19 morbidity, RA may be a useful adjuvant for use with anti-viral therapies.
Collapse
Affiliation(s)
- Patrick J. Callaghan
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
| | - Elizabeth Rybakovsky
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
| | - Bryan Ferrick
- Department of Biomedical Engineering, Drexel University, Philadelphia, PA, United States of America
| | - Sunil Thomas
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
| | - James M. Mullin
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
- * E-mail:
| |
Collapse
|
49
|
Bonavita AG. Ac2-26 mimetic peptide of annexin A1 to treat severe COVID-19: A hypothesis. Med Hypotheses 2020; 145:110352. [PMID: 33129009 PMCID: PMC7577270 DOI: 10.1016/j.mehy.2020.110352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/17/2020] [Indexed: 02/07/2023]
Abstract
The Coronavirus Diseases-2019 (COVID-19) pandemic leads many researchers around the world to study the SARS-CoV-s2 infection and pathology to find a treatment for it. This generates a massive production of papers including pre-clinical, clinical and revisions but till now no specific treatment were identified. Meanwhile, like other coronavirus infections, COVID-19 leads to the cytokine storm syndrome resulting in hyperinflammation, exacerbated immune response and multiple organ dysfunctions indicating that drugs that modulate this response, as glucocorticoids could be a treatment option. However glucocorticoids have several side effects or usage limitations. In this sense a drug with anti-inflammatory effects and capable to reduce inflammation but with less after-effects could be a powerful tool to combat COVID-19. Thus the Ac2-26 Mimetic Peptide of Annexin A1 emerges as a possible therapy. The peptide has many anti-inflammatory effects described including the reduction of interleukin (IL)-6, one of the main mediators of cytokine storm syndrome. Therefore the hypothesis to use the Ac2-26 peptide to treat severe COVID-19 will be highlighted in this paper.
Collapse
Affiliation(s)
- Andre Gustavo Bonavita
- Grupo de Pesquisa em Farmacologia de Produtos Bioativos, Campus UFRJ-Macaé Professor Aloizio Teixeira Macaé, Universidade Federal do Rio de Janeiro, Rua Aloísio da Silva Gomes, 50, Macaé, RJ, Brazil.
| |
Collapse
|
50
|
Hosseini A, Hashemi V, Shomali N, Asghari F, Gharibi T, Akbari M, Gholizadeh S, Jafari A. Innate and adaptive immune responses against coronavirus. Biomed Pharmacother 2020; 132:110859. [PMID: 33120236 PMCID: PMC7580677 DOI: 10.1016/j.biopha.2020.110859] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 01/08/2023] Open
Abstract
Coronaviruses (CoVs) are a member of the Coronaviridae family with positive-sense single- stranded RNA. In recent years, the CoVs have become a global problem to public health. The immune responses (innate and adaptive immunity) are essential for elimination and clearance of CoVs infections, however, uncontrolled immune responses can result in aggravating acute lung injury and significant immunopathology. Gaining profound understanding about the interaction between CoVs and the innate and adaptive immune systems could be a critical step in the field of treatment. In this review, we present an update on the host innate and adaptive immune responses against SARS-CoV, MERS-CoV and newly appeared SARS-CoV-2.
Collapse
Affiliation(s)
- Arezoo Hosseini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Asghari
- Department of Immunology, School of Medicine, Tarbiat Modares University of Medical Sciences, Tehran, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saber Gholizadeh
- Department of Medical Entomology and Vector Control, School of Public Health, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Jafari
- Department of Toxicology and Cellular and Molecular Research Center, School of Public Health, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|