1
|
Hromić-Jahjefendić A, Mahmutović L, Sezer A, Bećirević T, Rubio-Casillas A, Redwan EM, Uversky VN. The intersection of microbiome and autoimmunity in long COVID-19: Current insights and future directions. Cytokine Growth Factor Rev 2025; 82:43-54. [PMID: 39179487 DOI: 10.1016/j.cytogfr.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
Long COVID-19 affects a significant percentage of patients and is characterized by a wide range of symptoms, including weariness and mental fog as well as emotional symptoms like worry and sadness. COVID-19 is closely linked to the autoimmune disorders that are becoming more prevalent worldwide and are linked to immune system hyperactivation, neutrophil extracellular trap (NET) development, and molecular mimicry pathways. Long-term COVID-related autoimmune responses include a watchful immune system referring to the ability of immune system to constantly monitor the body for signs of infection, disease, or abnormal cells; altered innate and adaptive immune cells, autoantigens secreted by living or dead neutrophils, and high concentrations of autoantibodies directed against different proteins. The microbiome, which consists of billions of bacteria living in the human body, is essential for controlling immune responses and supporting overall health. The microbiome can affect the course of long COVID-associated autoimmunity, including the degree of illness, the rate of recovery, and the onset of autoimmune reactions. Although the precise role of the microbiome in long COVID autoimmunity is still being investigated, new studies indicate that probiotics, prebiotics, and dietary changes-interventions that target the microbiome-may be able to reduce autoimmune reactions and enhance long-term outcomes for COVID-19 survivors. More research is required to precisely understand how the microbiome affects COVID-19-related autoimmunity and to create tailored treatment plans.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Abas Sezer
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Tea Bećirević
- Atrijum Polyclinic, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Jalisco 48900, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco 48900, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, 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 of Scientific Research and Technological Applications, New Borg EL-Arab 21934, Alexandria, Egypt.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC07, Tampa, FL, USA.
| |
Collapse
|
2
|
Juanes-Velasco P, Pérez-Arévalo JC, Arias-Hidalgo C, Nuño-Soriano A, Landeira-Viñuela A, Corrales F, Bernardo D, Cuesta-Sancho S, Rojo-Rello S, Lécrevisse Q, Góngora R, Sánchez-Santos JM, De Las Rivas J, Hernández ÁP, Fuentes M. Assessment of Humoral Response at SARS-CoV-2 Infection by Multipronged Functional Proteomics Approaches. J Proteome Res 2025; 24:515-525. [PMID: 39772566 DOI: 10.1021/acs.jproteome.4c00635] [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] [Indexed: 01/11/2025]
Abstract
In the past decade, a major goal in biomedical research has been to understand why individuals differ in disease susceptibility, disease dynamics, and progression. In many pathologies, this variability stems from evolved immune mechanisms that resist inflammatory stress from various diseases that have been encountered throughout life. These may provide advantages against other diseases, reduce comorbidities, and enhance longevity. This study evaluates prior immunity as a prognostic factor in COVID-19 patients, crucial for understanding plasmatic signaling cascades in different disease stages and their impact on disease progression. COVID-19, caused by SARS-CoV-2, primarily affects the respiratory system and presents a wide range of symptoms, posing significant challenges to medicine. This study systematically analyzed prior immunity and inflammation in two independent cohorts of infected patients. A serological profile is determined by protein microarrays, which identify IgM and IgG responses against 37 prevalent microbial pathogens and provide a comprehensive plasma analysis of 21 acute-phase proteins. Our results reveal distinct serological profiles correlating with disease severity, indicating that immune system dysregulation in COVID-19 patients is linked to existing immunity. These findings highlight the relevance of prior immunity for monitoring disease progression, particularly in infections and vaccine failure, and underscore the importance of functional proteomics in determining prognostic biomarkers.
Collapse
Affiliation(s)
- Pablo Juanes-Velasco
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Proteomics Unit-IBSAL, Instituto de Investigación Biomédica de Salamanca, Universidad de Salamanca (IBSAL/USAL), 37007 Salamanca, Spain
| | - Juan Carlos Pérez-Arévalo
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
| | - Carlota Arias-Hidalgo
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Proteomics Unit-IBSAL, Instituto de Investigación Biomédica de Salamanca, Universidad de Salamanca (IBSAL/USAL), 37007 Salamanca, Spain
| | - Ana Nuño-Soriano
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Proteomics Unit-IBSAL, Instituto de Investigación Biomédica de Salamanca, Universidad de Salamanca (IBSAL/USAL), 37007 Salamanca, Spain
| | - Alicia Landeira-Viñuela
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
| | - Fernando Corrales
- Functional Proteomics Laboratory, National Centre for Biotechnology, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain
| | - David Bernardo
- Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, 47003 Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - Sara Cuesta-Sancho
- Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, 47003 Valladolid, Spain
| | - Silvia Rojo-Rello
- Microbiology Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Quentin Lécrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rafael Góngora
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | | | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Centre (IBMCC, CSIC/USAL), Consejo Superior de Investigaciones Científicas (CSIC), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), 37007 Salamanca, Spain
| | - Ángela-Patricia Hernández
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
- Department of Pharmaceutical Sciences: Organic Chemistry, Faculty of Pharmacy, CIETUS, IBSAL, University of Salamanca, 37007 Salamanca, Spain
| | - Manuel Fuentes
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, 37008 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Proteomics Unit-IBSAL, Instituto de Investigación Biomédica de Salamanca, Universidad de Salamanca (IBSAL/USAL), 37007 Salamanca, Spain
| |
Collapse
|
3
|
Huang X, Zhu W, Zhang H, Qiu S, Shao H. SARS-CoV-2 N protein induces alveolar epithelial apoptosis via NLRP3 pathway in ARDS. Int Immunopharmacol 2025; 144:113503. [PMID: 39591821 DOI: 10.1016/j.intimp.2024.113503] [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: 05/26/2024] [Revised: 09/10/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024]
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a severe inflammatory condition often resulting from sepsis and viral infections, including (Severe Acute Respiratory Syndrome Coronavirus 2) SARS-CoV-2. This study investigates the molecular mechanisms by which the SARS-CoV-2 nucleocapsid (N) protein influences alveolar macrophage activation, leading to alveolar epithelial cell apoptosis and exacerbating ARDS. Single-cell RNA sequencing data from ARDS patients were analyzed to identify cell subpopulations and their interactions, revealing significant macrophage-epithelial cell communication through the (NOD-like receptor family pyrin domain containing 3) NLRP3 pathway. Differential gene expression in SARS-CoV-2-infected macrophages highlighted key genes, with WGCNA pinpointing core modules. In vitro experiments demonstrated that N protein overexpression in MH-S macrophages activates the NLRP3 pathway, promoting M1 macrophage polarization and inducing apoptosis in co-cultured MLE-12 epithelial cells. Immunoprecipitation, pull-down assays, Enzyme-Linked Immunosorbent Assay (ELISA), RT-qPCR, Western blotting, and flow cytometry confirmed these findings. In vivo, ARDS mouse models induced by CLP surgery or N protein administration showed increased M1 macrophage infiltration, heightened inflammatory responses, and significant epithelial cell damage, as evidenced by H&E staining, immunofluorescence, RNA-ISH, and ELISA. These results suggest that the SARS-CoV-2 N protein activates the NLRP3 signaling pathway, driving M1 macrophage polarization and the release of pro-inflammatory factors, thereby inducing alveolar epithelial cell apoptosis and worsening ARDS. Targeting this pathway may provide new therapeutic avenues for treating ARDS associated with SARS-CoV-2.
Collapse
Affiliation(s)
- Xiaopei Huang
- Department of Critical Care Medicine, Henan Key Laboratory for Critical Care Medicine,Zhengzhou Key Laboratory for Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Wenliang Zhu
- Department of Critical Care Medicine, Henan Key Laboratory for Critical Care Medicine,Zhengzhou Key Laboratory for Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Huifeng Zhang
- Department of Critical Care Medicine, Henan Key Laboratory for Critical Care Medicine,Zhengzhou Key Laboratory for Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Shi Qiu
- Department of Critical Care Medicine, Henan Key Laboratory for Critical Care Medicine,Zhengzhou Key Laboratory for Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Huanzhang Shao
- Department of Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou 450003, China.
| |
Collapse
|
4
|
Mirza AA, Almalki AH, Noori FA, Neazy SA, Dahm V, Bajin MD, Lin VY. Facial Nerve Palsy Amid the SARS-CoV-2 Pandemic: A Pooled Analysis. J Otolaryngol Head Neck Surg 2025; 54:19160216251315057. [PMID: 39916377 PMCID: PMC11803641 DOI: 10.1177/19160216251315057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 11/23/2024] [Indexed: 02/11/2025] Open
Abstract
IMPORTANCE Idiopathic facial nerve palsy (FNP) has devastating sequelae and is potentially linked to coronavirus disease-19 (COVID-19). OBJECTIVE The rate of FNP was compared in the pandemic versus pre-pandemic periods. Furthermore, the risk of FNP was estimated among the COVID-19 vaccinated group. DESIGN Systematic review and meta-analysis. SETTING An electronic search was conducted in 7 databases: Scopus, Web of Science core collection, PubMed, Cochrane Central Register of Controlled Trials, MEDLINE, Embase, and CINAHL. PARTICIPANTS English observational studies investigating an association between idiopathic FNP and COVID-19 or its vaccination were included, irrespective of patients' demographics. EXPOSURES COVID-19 or COVID-19 vaccine. MAIN OUTCOME MEASURES Change in FNP incidence between the pre-pandemic and pandemic periods; risk of developing FNP in individuals vaccinated against COVID-19 compared to those who were unvaccinated against COVID-19. RESULTS After excluding duplicates, the search yielded 906 related articles, of which 118 articles were included. The risk of FNP was statistically significantly higher during the COVID-19 pandemic than the pre-pandemic period (RR: 1.68, [95% CI: 1.16-2.43], P = .01). A nonsignificant increase in FNP risk was identified among COVID-19 vaccinated individuals compared to unvaccinated individuals (overall OR: 1.07, [95% CI: 0.85-1.35], P = .55). CONCLUSIONS AND RELEVANCE A remarkable increase in FNP rates was identified during the pandemic compared to pre-pandemic, which seemed unlikely to be attributed to COVID-19 vaccination.
Collapse
Affiliation(s)
- Ahmad A. Mirza
- Department of Otolaryngology–Head and Neck Surgery, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Department of Otolaryngology–Head and Neck Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Abdulaziz H. Almalki
- Division of Otolaryngology–Head and Neck Surgery, Department of Surgery, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Faisal A. Noori
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Kingdom of Saudi Arabia
| | - Sultan A. Neazy
- Pediatric Surgery Department, King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia
| | - Valerie Dahm
- Department of Otolaryngology–Head and Neck Surgery, Sunnybrook Health Sciences Center, University of Toronto, Toronto, ON, Canada
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Münir Demir Bajin
- Department of Otolaryngology–Head and Neck Surgery, Sunnybrook Health Sciences Center, University of Toronto, Toronto, ON, Canada
| | - Vincent Y. Lin
- Department of Otolaryngology–Head and Neck Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Otolaryngology–Head and Neck Surgery, Sunnybrook Health Sciences Center, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Song C, Li H, Han Y, Wang K, Yan W, Yang X, Zhang A, Wang H. Host restriction factor Rab11a limits Porcine deltacoronavirus invasion of cells via fusion peptide-mediated membrane fusion. Vet Microbiol 2024; 298:110246. [PMID: 39244909 DOI: 10.1016/j.vetmic.2024.110246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
Porcine deltacoronavirus (PDCoV) poses a serious threat to pork industry and has the potential for cross-species transmission. Yet, the invasion mechanisms and host factors involved are still unknown. In the present work, using siRNA interference and co-immunoprecipitation, we identified Annexin A2 (ANXA2), Prohibitin-2 (PHB2), or Caveolin-2 (CAV2) as host factors positively regulating the internalization of PDCoV. We further found that Rab11a co-localized with PDCoV S and inhibited PDCoV internalization. Subsequently, a pseudoviral infection model (LV-PDCoV S-GFP) was constructed, and ANXA2 or CAV2 promoted PDCoV invasion by downregulating Rab11a. Our results also indicated that ANXA2, CAV2, and Rab11a interact with the S protein via S-FP, thereby regulating virus-host membrane fusion. Through LV-PDCoV S-GFP infection, we found that Rab11a may act as a host restriction factor, and it could regulate the invasion efficiency of PDCoV by adding of exogenous GTP. These findings revealed that Rab11a was an exciting target to restrict fusion of PDCoV with host cell membranes. AVAILABILITY OF DATA AND MATERIAL: Not applicable.
Collapse
Affiliation(s)
- Cailiang Song
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hao Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yun Han
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Kailu Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Wenjun Yan
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Xin Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| | - Anyun Zhang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| | - Hongning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| |
Collapse
|
6
|
Zamanian MH, Norooznezhad AH, Hosseinkhani Z, Hassaninia D, Mansouri F, Vaziri S, Payandeh M, Heydarpour F, Kiani S, Shirvani M, Rajati M, Bakhtiari M, Esmaili F, Yarani R, Mansouri K. Human placental mesenchymal stromal cell-derived small extracellular vesicles as a treatment for severe COVID-19: A double-blind randomized controlled clinical trial. J Extracell Vesicles 2024; 13:e12492. [PMID: 39051747 PMCID: PMC11270582 DOI: 10.1002/jev2.12492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
The current study aimed to investigate the effects of human placental mesenchymal stromal cell-derived small extracellular vesicles (hPMSC-sEVs) as a treatment for COVID-19. This double-blind, randomized, controlled clinical trial was conducted on two groups of patients with COVID-19-associated acute respiratory distress syndrome. After randomization, the control group received standard treatment and placebo, and the intervention arm received standard treatment plus hPMSC-sEVs. The number of hospital deaths was considered the primary outcome. After meeting the exclusion and inclusion criteria, 21 and 24 patients were allocated to intervention and control arms, respectively. Besides admission SpO2 levels, which were significantly lower in the intervention arm (p = 0.008), all the baseline demo-biographic and laboratory variables were similar between the groups. It was shown that hPMSC-sEVs could significantly (p = 0.015) decrease the mortality ratio in the intervention group (4/21 [19.04%]) compared to the controls (13/24 [54.16%]). The mean time to death in the intervention and control groups was 28.06 and 11.10 days, respectively (p < 0.001). This study showed that hPMSC-sEVs are a possible treatment for critically ill patients with COVID-19.
Collapse
Affiliation(s)
| | - Amir Hossein Norooznezhad
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Zohreh Hosseinkhani
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Daryoush Hassaninia
- Infectious Diseases Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Feizollah Mansouri
- Infectious Diseases Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Siavash Vaziri
- Infectious Diseases Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Mehrdad Payandeh
- Bone Marrow Transplantation Department, School of Medicine, KermanshahUniversity of Medical SciencesKermanshahIran
| | - Fatemeh Heydarpour
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Sara Kiani
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Maria Shirvani
- Department of Infectious Disease, School of MedicineKermanshah University of Medical SciencesKermanshahIran
| | - Mojgan Rajati
- Motazedi Hospital, Kermanshah University of Medical SciencesKermanshahIran
| | - Mitra Bakhtiari
- Motazedi Hospital, Kermanshah University of Medical SciencesKermanshahIran
| | - Farzaneh Esmaili
- Motazedi Hospital, Kermanshah University of Medical SciencesKermanshahIran
| | - Reza Yarani
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
- Translational Type 1 Diabetes, Department of Clinical ResearchSteno Diabetes Center CopenhagenHerlevDenmark
| | - Kamran Mansouri
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| |
Collapse
|
7
|
Gozukucuk R, Kılıc HH, Uyanik BS, Cakiroglu B. The İmportance of hematological parameters in the prognosis of patients with severe COVID-19, A single-center retrospective study. Niger J Clin Pract 2023; 26:1297-1302. [PMID: 37794542 DOI: 10.4103/njcp.njcp_22_23] [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: 01/11/2023] [Accepted: 02/24/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Inflammation biomarkers known as acute phase reactants (APRs) show significant variations in serum concentrations during inflammation brought on by both viral and noninfectious diseases. The erythrocyte sedimentation rate (ESR), the C-reactive protein (CRP), the lactate dehydrogenase (LDH), the ferritin, the fibrinogen, the procalcitonin, the D-dimer, and the troponin I are all significant APRs. During inflammation, the serum levels of each of these positive APRs rise. The sensitivity and specificity of hematologic parameters and indices are as high as the inflammatory biomarkers mentioned above for monitoring disease severity and treatment response. AIM We aimed to evaluate the differences in hematological parameters and indices, and to reveal their treatment and prognostic values, especially in deceased patients with COVID-19. MATERIALS AND METHODS The hemogram parameters of 169 critical patients with COVID-19 (125 males and 44 females) who received inpatient treatment at …. UNLABELLED between 1 March 2020 and 31 December 2021 were analyzed retrospectively. The patients were divided into two groups-deceased (77) and surviving (92)-noting demographic data such as age and gender. All analyses were performed using SPSS 25.0. RESULTS Analyses of the hematological parameters used during the treatment processes revealed statistically significant differences between the two patient groups. White blood cell (WBC), neutrophil, and neutrophil-to-lymphocyte ratio (NLR) values were significantly higher (P = 0.019, P = 0.000 and P = 0.000, respectively) for deceased subjects, while lymphocyte, platelet and plateletcrit (PCT) values were significantly lower (for all values, P = 0.000). Platelet volume (MPV) and platelet distribution width (PDW), as well as MPV/PLT, PDW/PLT, MPV/PCT, and PDW/PCT, levels were significantly higher in deceased subjects (P = 0.000). Particularly in our deceased cases, receiver operating characteristic analyses were performed to reveal the importance of such analyses in prognostic status evaluation in COVID-19 since the hematological parameters are quite different. Cut-off values were determined for each parameter, and sensitivity and specificity ratios were calculated. While the sensitivities of MPV/PLT, PDW/PLT, MPV/PCT, and PDW/PCT indices are over 80%, neutrophil and white blood cell sensitivities were found to be lower (74%, 68.8%, respectively). CONCLUSION In addition to NLR, which is an important biomarker, the hematological indices MPV/PLT, PDW/PLT, MPV/PCT, and PDW/PCT can be used to determine the risk of death in patients with severe COVID-19.
Collapse
Affiliation(s)
- R Gozukucuk
- Department of Infectious Disease and Clinical Microbiology, Hisar Hospital Intercontinental, Istanbul Galata Universty, Istanbul, Turkey
| | - H H Kılıc
- Department of Anesthesiology and Reanimation, Hisar Hospital Intercontinental, Istanbul Dogus Universty, Istanbul, Turkey
| | - B S Uyanik
- Department of Clinical Biochemistry, Hisar Hospital Intercontinental, Istanbul Galata Universty, Turkey
| | - B Cakiroglu
- Department of Urology, Hisar Hospital Intercontinental, Istanbul Galata Universty, Turkey
| |
Collapse
|
8
|
Fonseca DLM, Filgueiras IS, Marques AHC, Vojdani E, Halpert G, Ostrinski Y, Baiocchi GC, Plaça DR, Freire PP, Pour SZ, Moll G, Catar R, Lavi YB, Silverberg JI, Zimmerman J, Cabral-Miranda G, Carvalho RF, Khan TA, Heidecke H, Dalmolin RJS, Luchessi AD, Ochs HD, Schimke LF, Amital H, Riemekasten G, Zyskind I, Rosenberg AZ, Vojdani A, Shoenfeld Y, Cabral-Marques O. Severe COVID-19 patients exhibit elevated levels of autoantibodies targeting cardiolipin and platelet glycoprotein with age: a systems biology approach. NPJ AGING 2023; 9:21. [PMID: 37620330 PMCID: PMC10449916 DOI: 10.1038/s41514-023-00118-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 07/12/2023] [Indexed: 08/26/2023]
Abstract
Age is a significant risk factor for the coronavirus disease 2019 (COVID-19) severity due to immunosenescence and certain age-dependent medical conditions (e.g., obesity, cardiovascular disorder, and chronic respiratory disease). However, despite the well-known influence of age on autoantibody biology in health and disease, its impact on the risk of developing severe COVID-19 remains poorly explored. Here, we performed a cross-sectional study of autoantibodies directed against 58 targets associated with autoimmune diseases in 159 individuals with different COVID-19 severity (71 mild, 61 moderate, and 27 with severe symptoms) and 73 healthy controls. We found that the natural production of autoantibodies increases with age and is exacerbated by SARS-CoV-2 infection, mostly in severe COVID-19 patients. Multiple linear regression analysis showed that severe COVID-19 patients have a significant age-associated increase of autoantibody levels against 16 targets (e.g., amyloid β peptide, β catenin, cardiolipin, claudin, enteric nerve, fibulin, insulin receptor a, and platelet glycoprotein). Principal component analysis with spectrum decomposition and hierarchical clustering analysis based on these autoantibodies indicated an age-dependent stratification of severe COVID-19 patients. Random forest analysis ranked autoantibodies targeting cardiolipin, claudin, and platelet glycoprotein as the three most crucial autoantibodies for the stratification of severe COVID-19 patients ≥50 years of age. Follow-up analysis using binomial logistic regression found that anti-cardiolipin and anti-platelet glycoprotein autoantibodies significantly increased the likelihood of developing a severe COVID-19 phenotype with aging. These findings provide key insights to explain why aging increases the chance of developing more severe COVID-19 phenotypes.
Collapse
Affiliation(s)
- Dennyson Leandro M Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil.
| | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Alexandre H C Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Elroy Vojdani
- Regenera Medical 11860 Wilshire Blvd., Ste. 301, Los Angeles, CA, 90025, USA
| | - Gilad Halpert
- Ariel University, Ari'el, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Saint Petersburg State University Russia, Saint Petersburg, Russia
| | - Yuri Ostrinski
- Ariel University, Ari'el, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Saint Petersburg State University Russia, Saint Petersburg, Russia
| | - Gabriela Crispim Baiocchi
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Desirée Rodrigues Plaça
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paula P Freire
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Shahab Zaki Pour
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Guido Moll
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Rusan Catar
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Yael Bublil Lavi
- Scakler faculty of medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan I Silverberg
- Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Robson F Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Taj Ali Khan
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
| | - Harald Heidecke
- CellTrend Gesellschaft mit beschränkter Haftung (GmbH), Luckenwalde, Germany
| | - Rodrigo J S Dalmolin
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Andre Ducati Luchessi
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, R.N., Natal, Brazil
| | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, USA
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Howard Amital
- Ariel University, Ari'el, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Department of Medicine B, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriela Riemekasten
- Department of Rheumatology, University Medical Center Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Israel Zyskind
- Maimonides Medical Center, Brooklyn, NY, USA
- Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Aristo Vojdani
- Department of Immunology, Immunosciences Laboratory, Inc., Los Angeles, CA, USA
- Cyrex Laboratories, LLC 2602 S. 24th St., Phoenix, AZ, 85034, USA
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Otavio Cabral-Marques
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil.
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
- Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal, Brazil.
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil.
- Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil.
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, SP, Brazil.
| |
Collapse
|
9
|
van Heerden PV, Abutbul A, Naama A, Maayan S, Makram N, Nachshon A, abu Jabal K, Hershkovitz O, Binder L, Shabat Y, Reicher B, Mevorach D. Apoptotic cells for treatment of acute respiratory distress syndrome associated with COVID-19. Front Immunol 2023; 14:1242551. [PMID: 37600829 PMCID: PMC10433372 DOI: 10.3389/fimmu.2023.1242551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background Hyper-inflammatory immune response, a hallmark of severe COVID-19, is associated with increased mortality. Acute respiratory distress syndrome (ARDS) is a common manifestation. We undertook two phase I/II studies in five and then 16 subjects with severe/critical COVID-19 to assess the safety and preliminary efficacy of apoptotic cells (Allocetra™-OTS, Enlivex Therapeutics), a cellular immunomodulatory therapy that reprograms macrophages to reduce hyper-inflammatory response severity. Methods Eligible patients presenting to the Emergency Room with severe COVID-19 and respiratory dysfunction received one intravenous administration of Allocetra™-OTS and were monitored for adverse events (AEs) for 28 days. The primary aim was to determine the safety profile of treatment; secondary aims were recovery from ARDS, intensive care unit (ICU) and hospital length-of-stay, and mortality. Immune modulator markers were measured to elucidate the mechanism of action of Allocetra™-OTS. Results 21 patients with severe-critical COVID-19 of Gamma, Alpha and Delta variants, were treated with a single dose of apoptotic cells. 19/21 patients had mild-to-severe ARDS at presentation. Median age was 53 years, 16/21 were males, 16/21 were overweight/obese. No serious related adverse events (SAEs) were reported. All 21 study subjects survived to day 28 (end of study); 19/21 recovered completely. Comparable mortality rates at the hospital were 3.8%-8.9% for age- and gender-matched patients, and 39%-55% for critical patients. Recovering patients exhibited rapid ARDS resolution and parallel resolution of inflammation markers and elevated cytokines/chemokines. Conclusion In patients with severe/critical COVID-19 associated with ARDS, Allocetra™-OTS was safe, well-tolerated, and showed promising results for resolution of respiratory failure and inflammation. Trial registration https://clinicaltrials.gov/ct2/show/study/NCT04513470, https://clinicaltrials.gov/ct2/show/study/NCT04590053, Identifiers NCT04513470, NCT04590053.
Collapse
Affiliation(s)
| | - Avraham Abutbul
- Medical Intensive Care Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ahmad Naama
- Department of Emergency Medicine, Hadassah-Hebrew University Medical Center and Hebrew University-Hadassah Faculty of Medicine, Jerusalem, Israel
| | - Shlomo Maayan
- Infectious Diseases Division, Barzilai Medical Center, Ashkelon, Israel
| | - Nassar Makram
- Infectious Diseases Division, Barzilai Medical Center, Ashkelon, Israel
| | - Akiva Nachshon
- General Intensive Care Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Kamal abu Jabal
- Ziv Medical Center and Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | | | - Lior Binder
- Enlivex Therapeutics Ltd., Ness Ziona, Israel
| | | | | | - Dror Mevorach
- Enlivex Therapeutics Ltd., Ness Ziona, Israel
- Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- The Institute of Rheumatology-Immunology-Rheumatology, The Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center and Hebrew University-Hadassah Faculty of Medicine, Jerusalem, Israel
| |
Collapse
|
10
|
Akkemik O, Acikgoz A. Effect of the Ongoing Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Pandemic on Dental Service Utilization in Patients With Comorbidities. Cureus 2023; 15:e39926. [PMID: 37292106 PMCID: PMC10245075 DOI: 10.7759/cureus.39926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND The keystone of safe and effective patient management is to approach a patient with up-to-date medical information. Assessment of patients for their medical conditions has changed during the coronavirus disease 2019 (COVID-19) pandemic and the need for appropriate research infrastructure has increased. Considering an updated list of high-risk underlying conditions in the post-COVID-19 era, this study aimed to evaluate the utilization of dental services by patients with comorbidities during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. METHODS Data of patients with comorbidities seeking dental care at a dental school during the COVID-19 pandemic were retrospectively evaluated. Demographic variables (age, gender) and medical history of the participants were recorded. The patients were classified according to their diagnosis. Data were analyzed using descriptive statistics and Chi-square analysis. The significance level was determined at α=0.05. RESULTS The study included data from 1067 patient visits between September 1, 2020 and November 1, 2021. Among these patients, 406 (38.1%) were males and 661 (61.9%) were females, with a mean age of 38.28 ± 14.36 years. Comorbidities were identified in 38.3% of the patients with predominance in females (74.1% n=303). Single comorbidity was observed in 28.1% while multi-morbidity was detected in 10.2% of the cohort. The most prevalent comorbidity was hypertension (9.7%), followed by diabetes (6.5%), thyroid disorders (5%), various psychological diseases (4.5%), COVID-19 infection (4.5%), and different allergies (4%). The presence of one or more co-morbidities was observed mostly in the 50-59 years age group. CONCLUSIONS The seeking of dental care among the adult population with comorbidities was high during the SARS-CoV-2 pandemic. It would be beneficial to develop a template for obtaining a medical history from patients by taking full account of the consequences of the pandemic. The dental profession needs to respond accordingly.
Collapse
Affiliation(s)
- Ozlem Akkemik
- Dentomaxillofacial Radiology, Baris Medical Imaging Center, Izmir, TUR
| | - Aydan Acikgoz
- Dentomaxillofacial Radiology, Faculty of Dentistry, Yeni Yuzyil University, Istanbul, TUR
| |
Collapse
|
11
|
Juanes-Velasco P, Arias-Hidalgo C, Landeira-Viñuela A, Nuño-Soriano A, Fuentes-Vacas M, Góngora R, Hernández ÁP, Fuentes M. Functional proteomics based on protein microarray technology for biomedical research. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:49-65. [PMID: 38220432 DOI: 10.1016/bs.apcsb.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
This chapter traces a route through Proteomics from its origins to the present day. The different proteomics applications are discussed with a focus on microarray technology. Analytical microarrays, functional microarrays and reverse phase microarrays and their different applications are discussed. Several studies are mentioned where the great versatility of this approach is shown. Finally, the advantages and future challenges of microarray technology are outlined.
Collapse
Affiliation(s)
- Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Carlota Arias-Hidalgo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Ana Nuño-Soriano
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Marina Fuentes-Vacas
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Rafa Góngora
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Ángela-Patricia Hernández
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain; Department of Pharmaceutical Sciences: Organic Chemistry, Faculty of Pharmacy, University of Salamanca, CIETUS, IBSAL, Salamanca, Spain
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain; Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain.
| |
Collapse
|
12
|
Taghadosi M, Safarzadeh E, Asgarzadeh A, Roghani SA, Shamsi A, Jalili C, Assar S, Soufivand P, Pournazari M, Feizollahi P, Nicknam MH, Asghariazar V, Vaziri S, Shahriari H, Mohammadi A. Partners in crime: Autoantibodies complicit in COVID-19 pathogenesis. Rev Med Virol 2023; 33:e2412. [PMID: 36471421 PMCID: PMC9877745 DOI: 10.1002/rmv.2412] [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: 06/20/2022] [Revised: 11/06/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
Autoantibodies (AABs) play a critical role in the pathogenesis of autoimmune diseases (AIDs) and serve as a diagnostic and prognostic tool in assessing these complex disorders. Viral infections have long been recognized as a principal environmental factor affecting the production of AABs and the development of autoimmunity. COVID-19 has primarily been considered a hyperinflammatory syndrome triggered by a cytokine storm. In the following, the role of maladaptive B cell response and AABs became more apparent in COVID-19 pathogenesis. The current review will primarily focus on the role of extrafollicular B cell response, Toll-like receptor-7 (TLR-7) activation, and neutrophil extracellular traps (NETs) formation in the development of AABs following SARS-CoV-2 infection. In the following, this review will clarify how these AABs dysregulate immune response to SARS-CoV-2 by disrupting cytokine function and triggering neutrophil hyper-reactivity. Finally, the pathologic effects of these AABs will be further described in COVID-19 associate clinical manifestations, including venous and arterial thrombosis, a multisystem inflammatory syndrome in children (MIS-C), acute respiratory distress syndrome (ARDS), and recently described post-acute sequelae of COVID-19 (PASC) or long-COVID.
Collapse
Affiliation(s)
- Mahdi Taghadosi
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Safarzadeh
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Asgarzadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Seyed Askar Roghani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Afsaneh Shamsi
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Cyrus Jalili
- Department of Anatomy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shirin Assar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parviz Soufivand
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehran Pournazari
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Feizollahi
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hossein Nicknam
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Molecular Immunology Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Asghariazar
- Deputy of Research and Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Siavash Vaziri
- Infectious Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Shahriari
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
13
|
Kit Y, Starykovych M, Manko N, Orfin A, Alexanyan T, Bozhko L, Turchyna T, Kit O, Krishnankutty R, Anand A, Sibirny A, Souchelnytskyi S, Stoika R. Elevation of truncated (48 kDa) form of unconventional myosin 1C in blood serum correlates with severe Covid-19. J Immunol Methods 2023; 514:113437. [PMID: 36736950 PMCID: PMC9889275 DOI: 10.1016/j.jim.2023.113437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/25/2022] [Accepted: 01/28/2023] [Indexed: 02/04/2023]
Abstract
In Covid-19 and autoimmune patients, there are several similarities revealed in the immune responses (Liu et al., 2021; Woodruff et al., 2020). Earlier, we firstly detected a truncated (48 kDa) form of the unconventional Myosin 1C (48/Myo1C) in a fraction of proteins soluble in 10% 2,2,2-trichloroacetic acid (TCA). These proteins were obtained from blood serum of patients with autoimmune diseases, such as multiple sclerosis, systemic lupus erythematosus, and rheumatoid arthritis (Kit et al., 2018). Here, we demonstrated that content of 48/Myo1C was also elevated in blood serum of the severe Covid-19 patients. Whereas in blood of 28 clinically healthy human individuals regularly tested for Covid-19 infection, the amount of this protein was undetectable or very low, in blood of 16 of 28 patients hospitalized with severe course of this disease, its amount was significantly increased. Dexamethasone, steroid hormone which is widely used for treatment of severe Covid-19 patients, induced time-dependent elevation of the 48/Myo1C in blood of such patients. The 48/Myo1C dose-dependently suppressed the viability of anti-CD3-activated lymphocytes of human peripheral blood. Recently, we used affinity chromatography on the magnetic poly(glycidyl-methacrylate) (mag-PGMA-NH2) microparticles functionalized with Myo1C and MALDI-TOF mass spectrometry with molecular modeling in silico in order to identify potential molecular partners of the 48/Myo1C. It was found that 48/Myo1C might bind to component 3 of the complement system and the anti-thrombin-III (Starykovych et al., 2021). Thus, the mechanisms of the pathogenic action of truncated form of Myo1C in severe COVID-19 patients may involve a suppression of the immune cells, as well as modulation of complement and coagulation cascades.
Collapse
Affiliation(s)
- Yuriy Kit
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov st., 14/16, Lviv 79005, Ukraine
| | - Marina Starykovych
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov st., 14/16, Lviv 79005, Ukraine
| | - Nazar Manko
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov st., 14/16, Lviv 79005, Ukraine
| | - Andrii Orfin
- Municipal Non-commercial Enterprise of Lviv Regional Council "Lviv Regional Infection Clinical Hospital", Pekarska St., 54, 79010, Lviv, Ukraine
| | - Tamila Alexanyan
- Municipal Non-commercial Enterprise of Lviv Regional Council "Lviv Regional Infection Clinical Hospital", Pekarska St., 54, 79010, Lviv, Ukraine
| | - Lydmyla Bozhko
- Lviv Regional Phthysio-pulmonology Clinical Medical and Diagnostic Center, Zelena st., 477, 79035, Lviv, Ukraine
| | - Tetiana Turchyna
- Lviv Regional Phthysio-pulmonology Clinical Medical and Diagnostic Center, Zelena st., 477, 79035, Lviv, Ukraine
| | - Oleg Kit
- Interregional Academy of Personnel Management, Frometivska st., 2, Kyiv 01001, Ukraine
| | - Roopesh Krishnankutty
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 2713, Qatar; University of Edinburgh, Edinburgh EH4 2XU, the, UK
| | - Anjana Anand
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 2713, Qatar
| | - Andrey Sibirny
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov st., 14/16, Lviv 79005, Ukraine
| | | | - Rostyslav Stoika
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov st., 14/16, Lviv 79005, Ukraine.
| |
Collapse
|
14
|
Elizalde-Díaz JP, Miranda-Narváez CL, Martínez-Lazcano JC, Martínez-Martínez E. The relationship between chronic immune response and neurodegenerative damage in long COVID-19. Front Immunol 2022; 13:1039427. [PMID: 36591299 PMCID: PMC9800881 DOI: 10.3389/fimmu.2022.1039427] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
In the past two years, the world has faced the pandemic caused by the severe acute respiratory syndrome 2 coronavirus (SARS-CoV-2), which by August of 2022 has infected around 619 million people and caused the death of 6.55 million individuals globally. Although SARS-CoV-2 mainly affects the respiratory tract level, there are several reports, indicating that other organs such as the heart, kidney, pancreas, and brain can also be damaged. A characteristic observed in blood serum samples of patients suffering COVID-19 disease in moderate and severe stages, is a significant increase in proinflammatory cytokines such as interferon-α (IFN-α), interleukin-1β (IL-1β), interleukin-2 (IL-2), interleukin-6 (IL-6) and interleukin-18 (IL-18), as well as the presence of autoantibodies against interferon-α (IFN-α), interferon-λ (IFN-λ), C-C motif chemokine ligand 26 (CCL26), CXC motif chemokine ligand 12 (CXCL12), family with sequence similarity 19 (chemokine (C-C motif)-like) member A4 (FAM19A4), and C-C motif chemokine ligand 1 (CCL1). Interestingly, it has been described that the chronic cytokinemia is related to alterations of blood-brain barrier (BBB) permeability and induction of neurotoxicity. Furthermore, the generation of autoantibodies affects processes such as neurogenesis, neuronal repair, chemotaxis and the optimal microglia function. These observations support the notion that COVID-19 patients who survived the disease present neurological sequelae and neuropsychiatric disorders. The goal of this review is to explore the relationship between inflammatory and humoral immune markers and the major neurological damage manifested in post-COVID-19 patients.
Collapse
Affiliation(s)
- José Pedro Elizalde-Díaz
- Laboratory of Cell Communication & Extracellular Vesicles, Division of Basic Science, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| | - Clara Leticia Miranda-Narváez
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Juan Carlos Martínez-Lazcano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Eduardo Martínez-Martínez
- Laboratory of Cell Communication & Extracellular Vesicles, Division of Basic Science, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| |
Collapse
|
15
|
Juanes‐Velasco P, García‐Vaquero ML, Landeira‐Viñuela A, Lopez‐Campos JL, Marín C, Lecrevisse Q, Arias‐Hidalgo C, Montalvillo E, Góngora R, Hernández Á, Fuentes M. Systematic evaluation of plasma signaling cascades by functional proteomics approaches: SARS-CoV-2 infection as model. Proteomics Clin Appl 2022; 16:e2100100. [PMID: 36168869 PMCID: PMC9537801 DOI: 10.1002/prca.202100100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE Acute phase reactants (APRs) play a critical role in inflammation. The difference in their physiological functions or the different dynamic ranges of these proteins in plasma makes it difficult to detect them simultaneously and to use several of these proteins as a tool in clinical practice. EXPERIMENTAL DESIGN A novel multiplex assay has been designed and optimized to carry out a high-throughput and simultaneous screening of APRs, allowing the detection of each of them at the same time and in their corresponding dynamic range. RESULTS Using Sars-CoV-2 infection as a model, it has been possible to profile different patterns of acute phase proteins that vary significantly between healthy and infected patients. In addition, severity profiles (acute respiratory distress syndrome and sepsis) have been established. CONCLUSIONS AND CLINICAL RELEVANCE Differential profiles in acute phase proteins can serve as a diagnostic and prognostic tool, among patient stratification. The design of this new platform for their simultaneous detection paves the way for them to be more extensive use in clinical practice.
Collapse
Affiliation(s)
- Pablo Juanes‐Velasco
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Marina L. García‐Vaquero
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Alicia Landeira‐Viñuela
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - José Luis Lopez‐Campos
- Unidad Médico‐Quirúrgica de Enfermedades Respiratorias. Instituto de Biomedicina de Sevilla (IBiS)Hospital Universitario Virgen del Rocío/Universidad de SevillaSpain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)Instituto de Salud Carlos IIIMadridSpain
| | - Carmen Marín
- Instituto de Biomedicina de Sevilla (IBiS)Hospital Universitario Virgen del RocíoSevillaSpain
| | - Quentin Lecrevisse
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Carlota Arias‐Hidalgo
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Enrique Montalvillo
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Rafael Góngora
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Ángela‐Patricia Hernández
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain,Department of Pharmaceutical Sciences: Organic Chemistry; Faculty of PharmacyUniversity of Salamanca, CIETUS, IBSALSalamanca37007Spain
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain,Proteomics UnitCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)Salamanca37007Spain
| |
Collapse
|
16
|
Correlates with Vaccine Protective Capacity and COVID-19 Disease Symptoms Identified by Serum Proteomics in Vaccinated Individuals. Molecules 2022; 27:molecules27185933. [PMID: 36144669 PMCID: PMC9500703 DOI: 10.3390/molecules27185933] [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: 08/01/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
In the last two years, the coronavirus disease 19 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been a scientific and social challenge worldwide. Vaccines have been the most effective intervention for reducing virus transmission and disease severity. However, genetic virus variants are still circulating among vaccinated individuals with different disease symptomatology. Understanding the protective- or disease-associated mechanisms in vaccinated individuals is relevant to advances in vaccine development and implementation. To address this objective, serum-protein profiles were characterized by quantitative proteomics and data-analysis algorithms in four cohorts of uninfected and SARS-CoV-2-infected vaccinated individuals with asymptomatic, non-severe, and severe disease symptomatology. The results show that immunoglobulins were the most overrepresented proteins in infected cohorts when compared to PCR-negative individuals. The immunoglobulin profile varied between different infected cohorts and correlated with protective- or disease-associated capacity. Overrepresented immunoglobulins in PCR-positive individuals correlated with protective response against SARS-CoV-2, other viruses, and thrombosis in asymptomatic cases. In non-severe cases, correlates of protection against SARS-CoV-2 and HBV together with risk of myasthenia gravis and allergy and autoantibodies were observed. Patients with severe symptoms presented risk for allergy, chronic idiopathic thrombocytopenic purpura, and autoantibodies. The analysis of underrepresented immunoglobulins in PCR-positive compared to PCR-negative individuals identified vaccine-induced protective epitopes in various coronavirus proteins, including the spike receptor-binding domain RBD. Non-immunoglobulin proteins were associated with COVID-19 symptoms and biological processes. These results evidence host-associated differences in response to vaccination and the possibility of improving vaccine efficacy against SARS-CoV-2.
Collapse
|
17
|
Darmarajan T, Paudel KR, Candasamy M, Chellian J, Madheswaran T, Sakthivel LP, Goh BH, Gupta PK, Jha NK, Devkota HP, Gupta G, Gulati M, Singh SK, Hansbro PM, Oliver BGG, Dua K, Chellappan DK. Autoantibodies and autoimmune disorders in SARS-CoV-2 infection: pathogenicity and immune regulation. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:54072-54087. [PMID: 35657545 PMCID: PMC9163295 DOI: 10.1007/s11356-022-20984-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/17/2022] [Indexed: 04/16/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease associated with the respiratory system caused by the SARS-CoV-2 virus. The aim of this review article is to establish an understanding about the relationship between autoimmune conditions and COVID-19 infections. Although majority of the population have been protected with vaccines against this virus, there is yet a successful curative medication for this disease. The use of autoimmune medications has been widely considered to control the infection, thus postulating possible relationships between COVID-19 and autoimmune diseases. Several studies have suggested the correlation between autoantibodies detected in patients and the severity of the COVID-19 disease. Studies have indicated that the SARS-CoV-2 virus can disrupt the self-tolerance mechanism of the immune system, thus triggering autoimmune conditions. This review discusses the current scenario and future prospects of promising therapeutic strategies that may be employed to regulate such autoimmune conditions.
Collapse
Affiliation(s)
- Thiviya Darmarajan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Subang Jaya, Bandar Sunway, Selangor, Malaysia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2007, Australia
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Lakshmana Prabu Sakthivel
- Department of Pharmaceutical Technology, University College of Engineering (BIT Campus), Anna University, Tiruchirappalli, 620024, India
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, 201310, Uttar Pradesh, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Knowledge Park III, Greater Noida, 201310, Uttar Pradesh, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto City, Kumamoto, 862-0973, Japan
- Program for Leading Graduate Schools, Health Life Science: Interdisciplinary and Glocal Oriented (HIGO) Program, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Sydney, NSW, 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Sydney, NSW, 2007, Australia
| | - Philip Michael Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2007, Australia
| | - Brian Gregory George Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Sydney, NSW, 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
18
|
McGill JR, Lagassé HAD, Hernandez N, Hopkins L, Jankowski W, McCormick Q, Simhadri V, Golding B, Sauna ZE. A structural homology approach to identify potential cross-reactive antibody responses following SARS-CoV-2 infection. Sci Rep 2022; 12:11388. [PMID: 35794133 PMCID: PMC9259575 DOI: 10.1038/s41598-022-15225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/21/2022] [Indexed: 11/09/2022] Open
Abstract
The emergence of the novel SARS-CoV-2 virus is the most important public-health issue of our time. Understanding the diverse clinical presentations of the ensuing disease, COVID-19, remains a critical unmet need. Here we present a comprehensive listing of the diverse clinical indications associated with COVID-19. We explore the theory that anti-SARS-CoV-2 antibodies could cross-react with endogenous human proteins driving some of the pathologies associated with COVID-19. We describe a novel computational approach to estimate structural homology between SARS-CoV-2 proteins and human proteins. Antibodies are more likely to interrogate 3D-structural epitopes than continuous linear epitopes. This computational workflow identified 346 human proteins containing a domain with high structural homology to a SARS-CoV-2 Wuhan strain protein. Of these, 102 proteins exhibit functions that could contribute to COVID-19 clinical pathologies. We present a testable hypothesis to delineate unexplained clinical observations vis-à-vis COVID-19 and a tool to evaluate the safety-risk profile of potential COVID-19 therapies.
Collapse
Affiliation(s)
- Joseph R McGill
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - H A Daniel Lagassé
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Nancy Hernandez
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Louis Hopkins
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Wojciech Jankowski
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Quinn McCormick
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Vijaya Simhadri
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Basil Golding
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Zuben E Sauna
- Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
19
|
Naveed Z, Sarwar M, Ali Z, Saeed D, Choudhry K, Sarfraz A, Sarfraz Z, Felix M, Cherrez-Ojeda I. Anakinra treatment efficacy in reduction of inflammatory biomarkers in COVID-19 patients: A meta-analysis. J Clin Lab Anal 2022; 36:e24434. [PMID: 35435272 PMCID: PMC9110982 DOI: 10.1002/jcla.24434] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 01/23/2023] Open
Abstract
Introduction Anakinra is being empirically considered for the treatment of COVID‐19 patients. The aim is to assess the efficacy of anakinra treatment on inflammatory marker reduction, including c‐reactive protein (CRP) concentrations, serum ferritin, and serum d‐dimer levels. Methods Adhering to PRISMA 2020 statement guidelines, a systematic search was conducted across the following databases from December 2019 until January 10, 2022: PubMed/MEDLINE, Cochrane Central, Web of Science, Scopus, and EMBASE. The following keywords were employed: Anakinra, COVID*, SARS‐CoV‐2, inflammatory, CRP, D‐dimer, Ferritin, hematological, laboratory, clinical, trials. The findings were collated and presented in a tabulated manner, and statistically analyzed using Review Manger 5.4 (Cochrane). Results In total, 2032 patients were included (881 in the anakinra and 1151 in the control/standard care group); 69.1% of them were males. Overall, the mean difference from admission until last follow‐up in CRP values was −9.66, where notable reductions were seen in the anakinra group (SMD = −0.46, p < 0.00001, N = 655). Serum ferritin mean values were reduced by 1467.16 in the anakinra group (SMD = −0.31, p = 0.004, N = 537). D‐dimer mean values were largely reduced by 4.04 in the anakinra group (SMD = −0.38, p = 0.0004, N = 375). Conclusion This study finds that anakinra is potentially a strong candidate as an anti‐inflammatory agent to reduce mortality in COVID‐19 patients, specifically in patients with elevated inflammatory biomarkers.
Collapse
Affiliation(s)
| | | | - Zahid Ali
- Kyrgyz State Medical Academy, Bishkek, Kyrgyzstan
| | - Danish Saeed
- Shaikh Khalifa Bin Zayed Al-Nahyan Medical and Dental College, Lahore, Pakistan
| | | | | | | | - Miguel Felix
- Universidad Espíritu Santo, Samborondón, Ecuador.,Respiralab Research Group, Guayaquil, Ecuador
| | - Ivan Cherrez-Ojeda
- Universidad Espíritu Santo, Samborondón, Ecuador.,Respiralab Research Group, Guayaquil, Ecuador
| |
Collapse
|