1
|
Mishra S, Chinthala A, Bhattacharya M. Drug-target prediction through self supervised learning with dual task ensemble approach. Comput Biol Chem 2024; 113:108244. [PMID: 39454455 DOI: 10.1016/j.compbiolchem.2024.108244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/15/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
Drug-Target interaction (DTI) prediction, a transformative approach in pharmaceutical research, seeks novel therapeutic applications for computational method based virtual screening, existing drugs to address untreated diseases and discovery of existing drugs side effects. The proposed model predict DTI through Heterogeneous biological network by combining drug, genes and disease related knowledge. For the purpose of embedding extraction Self-supervised learning (SSL) has been used which, trains models through pretext tasks, eliminating the need for manual annotations. The pretext tasks are related to either structural based information or similarity based information. To mitigate GNN vulnerability to non-robustness, ensemble learning can be incorporated into GNNs, harnessing multiple models to enhance robustness. This paper introduces a Graph neural network based architecture consisting of task based module and ensemble module for link prediction of DTI. The ensemble module of dual task combinations, both in cold start and warm start scenarios achieve very good performance as it provide 0.960 in cold start and 0.970 in warm start mean AUCROC score with less deviation.
Collapse
Affiliation(s)
- Surabhi Mishra
- ABV- Indian Institute of Information Technology and Management., Morena Road, Gwalior, 474015, India.
| | - Ashish Chinthala
- ABV- Indian Institute of Information Technology and Management., Morena Road, Gwalior, 474015, India.
| | - Mahua Bhattacharya
- ABV- Indian Institute of Information Technology and Management., Morena Road, Gwalior, 474015, India.
| |
Collapse
|
2
|
Farias PCS, Cabral LP, Neves PAF, Januário CAB, Cordeiro BM, Silva Júnior WJDA, Baseggio C, Paiva Júnior SSL, Araújo PSRDE, Lorena VMBDE, Balbino VQ, Lima Neto RG. Genetic variant in the AGT gene (rs699-GG) is associated with severe COVID-19 in Brazilian patients. AN ACAD BRAS CIENC 2024; 96:e20240274. [PMID: 39630802 DOI: 10.1590/0001-3765202420240274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/30/2024] [Indexed: 12/07/2024] Open
Abstract
The COVID-19 pandemic has been the largest pandemic of the past century, and various genetic factors have played a significant role in this context. This study aimed to analyze the frequency and association between specific SNPs rs3806268 (NLRP3), rs4925543 (NLRP3), rs12150220 (NLRP1), rs455060 (NLRC4), rs699 (AGT), rs1137101 (LEPR), and rs1801133 (MTHFR) and severe/critical outcomes in Brazilian patients with COVID-19. A total of 100 patients were included in the study, comprising 66 cases and 34 controls. DNA was extracted, sequenced, and genotyped via next-generation sequencing (NGS). For non-parametric data, the Mann-Whitney and Kruskal-Wallis tests were used. Fisher's test and multivariate logistic regression, considering AIC and BIC criteria, were employed for risk analysis. Odds Ratios (OR) were calculated, with significance set at p<0.05. Among the seven evaluated SNPs, only rs699-GG (AGT) (OR=8.07; p=0.04) was significantly associated with an increased risk of developing severe/critical COVID-19. Moreover, a borderline protective association was noted between rs1801133-GA (MTHFR) and the disease, although lacking statistical significance. In conclusion, the SNP rs699-GG (AGT) was associated with an increased risk of severe/critical COVID-19.
Collapse
Affiliation(s)
- Pablo C S Farias
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Leandro P Cabral
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Patrícia A F Neves
- Instituto Aggeu Magalhães (FIOCRUZ-PE), Departamento de Imunologia, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
- Universidade Federal de Pernambuco, Departamento de Medicina Tropical, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Caio A B Januário
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Beatriz M Cordeiro
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Wilson J DA Silva Júnior
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Carolina Baseggio
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Sérgio S L Paiva Júnior
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Paulo S R DE Araújo
- Universidade Federal de Pernambuco, Departamento de Medicina Tropical, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Virgínia Maria B DE Lorena
- Instituto Aggeu Magalhães (FIOCRUZ-PE), Departamento de Imunologia, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Valdir Q Balbino
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Reginaldo G Lima Neto
- Universidade Federal de Pernambuco, Departamento de Medicina Tropical, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| |
Collapse
|
3
|
Injinari N, Asadollahi S, Sefid F, Arshadi M, Hosseini SS, Ghoshouni H, Soltani F, Namiranian N, Sheikhha MH, Aghaeimeybodi F. Impact of FCGR2A rs1801274 and IL-6R rs2228145 polymorphisms on tocilizumab response in the Iranian population with severe COVID-19. BMC Infect Dis 2024; 24:1168. [PMID: 39415081 PMCID: PMC11481263 DOI: 10.1186/s12879-024-10073-0] [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: 02/26/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Although several genetic biomarkers have been reported in the tocilizumab (TCZ) response in rheumatoid arthritis, no studies have addressed the pharmacogenomics effect of TCZ in COVID-19. METHODS In this prospective longitudinal study, 95 individuals with severe COVID-19 were selected between 2020-2022. The recovery process was measured at 24 h, 48 h, and 10 days before and after taking TCZ. All participants were genotyped using RFLP-PCR. Different genotypes of FCGR2A rs1801274 and IL-6R rs2228145 were compared in terms of the recovery process. RESULTS 43.2% of patients were male and 56.8% were female with an average age of 58.20(± 16.214) years. The GA genotype for FCGR2A rs1801274 increased the risk of death (OR = 2.83, P = 0.038) and ventilation (OR = 2.71, P = 0.047) in TCZ-treated individuals. However, there was no risk of death and ventilation with IL-6R rs2228145 (P > 0.05). Additionally, docking analysis showed that not only IL6R but also FCGR2A can be a ligand for TCZ. CONCLUSION This study provides valuable insights into the impact of genetic variations on the response rate of TCZ in COVID-19 patients. The GA genotype for FCGR2A rs1801274 was associated with poor treatment outcomes.
Collapse
Affiliation(s)
- Nastaran Injinari
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samira Asadollahi
- Research Center for Food Hygiene and Safety, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Sefid
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maedeh Arshadi
- Department of Epidemiology and Biostatistics, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Saeedeh Sadat Hosseini
- Research Center for Food Hygiene and Safety, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamed Ghoshouni
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Soltani
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nasim Namiranian
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Hasan Sheikhha
- Abortion Research Centre, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Fatemeh Aghaeimeybodi
- Department of Internal Medicine, Shahid Sadoughi University of Medical Sciences, Daneshjoo Blvd, Yazd, Iran.
| |
Collapse
|
4
|
Elemam NM, Bouzid A, Alsafar H, Ahmed SBM, Hafezi S, Venkatachalam T, Eldohaji L, Al Hamidi T, Gerges PH, Halabi N, Hadj-Kacem H, Talaat IM, Taneera J, Sulaiman N, Maghazachi AA, Hamid Q, Hamoudi R, Saber-Ayad M. Association of specific ACE2 and TMPRSS2 variants with circulatory cytokines of COVID-19 Emirati patients. Front Immunol 2024; 15:1348229. [PMID: 38855114 PMCID: PMC11157456 DOI: 10.3389/fimmu.2024.1348229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
Introduction The COVID-19 pandemic represented one of the most significant challenges to researchers and healthcare providers. Several factors determine the disease severity, whereas none alone can explain the tremendous variability. The Single nucleotide variants (SNVs) in angiotensin-converting enzyme-2 (ACE2) and transmembrane serine protease type-2 (TMPRSS2) genes affect the virus entry and are considered possible risk factors for COVID-19. Methods We compiled a panel of gene variants from both genes and used in-silico analysis to predict their significance. We performed biological validation to assess their capacity to alter the ACE2 interaction with the virus spike protein. Subsequently, we conducted a retrospective comparative genome analysis on those variants in the Emirati patients with different disease severity (total of 96) along with 69 healthy control subjects. Results Our results showed that the Emirati population lacks the variants that were previously reported as associated with disease severity, whereas a new variant in ACE2 "Chr X:g.15584534" was associated with disease severity specifically among female patients. In-silico analysis revealed that the new variant can determine the ACE2 gene transcription. Several cytokines (GM-CSF and IL-6) and chemokines (MCP-1/CCL2, IL-8/CXCL8, and IP-10/CXCL10) were markedly increased in COVID-19 patients with a significant correlation with disease severity. The newly reported genetic variant of ACE2 showed a positive correlation with CD40L, IL-1β, IL-2, IL-15, and IL-17A in COVID-19 patients. Conclusion Whereas COVID-19 represents now a past pandemic, our study underscores the importance of genetic factors specific to a population, which can influence both the susceptibility to viral infections and the level of severity; subsequently expected required preparedness in different areas of the world.
Collapse
Affiliation(s)
- Noha M. Elemam
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Amal Bouzid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Habiba Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Emirates Bio-Research Centre, Ministry of Interior, Abu Dhabi, United Arab Emirates
| | - Samrein BM Ahmed
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Health, Wellbeing and Life Sciences, Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield, United Kingdom
| | - Shirin Hafezi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Thenmozhi Venkatachalam
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Physiology and Immunology College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Leen Eldohaji
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Tasneem Al Hamidi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Peter Habib Gerges
- School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
| | - Nour Halabi
- Al Jalila Genomics Center of Excellence, Al Jalila Children’s Specialty Hospital, Dubai, United Arab Emirates
| | - Hassen Hadj-Kacem
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Iman M. Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Jalal Taneera
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Nabil Sulaiman
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Family Medicine, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Azzam A. Maghazachi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Maha Saber-Ayad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
5
|
Zhang C, Zhang Y, Zhuang R, Yang K, Chen L, Jin B, Ma Y, Zhang Y, Tang K. Alterations in CX3CL1 Levels and Its Role in Viral Pathogenesis. Int J Mol Sci 2024; 25:4451. [PMID: 38674036 PMCID: PMC11050295 DOI: 10.3390/ijms25084451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
CX3CL1, also named fractalkine or neurotactin, is the only known member of the CX3C chemokine family that can chemoattract several immune cells. CX3CL1 exists in both membrane-anchored and soluble forms, with each mediating distinct biological activities. CX3CL1 signals are transmitted through its unique receptor, CX3CR1, primarily expressed in the microglia of the central nervous system (CNS). In the CNS, CX3CL1 acts as a regulator of microglia activation in response to brain disorders or inflammation. Recently, there has been a growing interest in the role of CX3CL1 in regulating cell adhesion, chemotaxis, and host immune response in viral infection. Here, we provide a comprehensive review of the changes and function of CX3CL1 in various viral infections, such as human immunodeficiency virus (HIV), SARS-CoV-2, influenza virus, and cytomegalovirus (CMV) infection, to highlight the emerging roles of CX3CL1 in viral infection and associated diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yun Zhang
- Department of Immunology, The Fourth Military Medical University, Xi’an 710032, China; (C.Z.); (Y.Z.); (R.Z.); (K.Y.); (L.C.); (B.J.); (Y.M.)
| | - Kang Tang
- Department of Immunology, The Fourth Military Medical University, Xi’an 710032, China; (C.Z.); (Y.Z.); (R.Z.); (K.Y.); (L.C.); (B.J.); (Y.M.)
| |
Collapse
|
6
|
Pulito-Cueto V, Sebastián Mora-Gil M, Ferrer-Pargada D, Remuzgo-Martínez S, Genre F, Lera-Gómez L, Alonso-Lecue P, Batista-Liz JC, Tello-Mena S, Abascal-Bolado B, Izquierdo S, Ruiz-Cubillán JJ, Armiñanzas-Castillo C, Blanco R, González-Gay MA, López-Mejías R, Cifrián JM. Inflammasome-Related Genetic Polymorphisms as Severity Biomarkers of COVID-19. Int J Mol Sci 2024; 25:3731. [PMID: 38612539 PMCID: PMC11011752 DOI: 10.3390/ijms25073731] [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: 02/15/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The most critical forms of coronavirus disease 2019 (COVID-19) are associated with excessive activation of the inflammasome. Despite the COVID-19 impact on public health, we still do not fully understand the mechanisms by which the inflammatory response influences disease prognosis. Accordingly, we aimed to elucidate the role of polymorphisms in the key genes of the formation and signaling of the inflammasome as biomarkers of COVID-19 severity. For this purpose, a large and well-defined cohort of 377 COVID-19 patients with mild (n = 72), moderate (n = 84), severe (n = 100), and critical (n = 121) infections were included. A total of 24 polymorphisms located in inflammasome-related genes (NLRP3, NLRC4, NLRP1, CARD8, CASP1, IL1B, IL18, NFKB1, ATG16L1, and MIF) were genotyped in all of the patients and in the 192 healthy controls (HCs) (who were without COVID-19 at the time of and before the study) by RT-qPCR. Our results showed that patients with mild, moderate, severe, and critical COVID-19 presented similar allelic and genotypic distribution in all the variants studied. No statistically significant differences in the haplotypic distribution of NLRP3, NLRC4, NLRP1, CARD8, CASP1, IL1B, and ATG16L1 were observed between COVID-19 patients, who were stratified by disease severity. Each stratified group of patients presented a similar genetic distribution to the HCs. In conclusion, our results suggest that the inflammasome polymorphisms studied are not associated with the worsening of COVID-19.
Collapse
Affiliation(s)
- Verónica Pulito-Cueto
- Immunopathology Group, Marqués de Valdecilla University Hospital-Valdecilla Research Institute (IDIVAL), 39008 Santander, Spain; (M.S.M.-G.); (P.A.-L.); (J.C.B.-L.); (R.B.); (R.L.-M.); (J.M.C.)
- Department of Rheumatology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain
| | - María Sebastián Mora-Gil
- Immunopathology Group, Marqués de Valdecilla University Hospital-Valdecilla Research Institute (IDIVAL), 39008 Santander, Spain; (M.S.M.-G.); (P.A.-L.); (J.C.B.-L.); (R.B.); (R.L.-M.); (J.M.C.)
- Department of Rheumatology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain
| | - Diego Ferrer-Pargada
- Department of Pneumology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain; (D.F.-P.); (S.T.-M.); (B.A.-B.); (S.I.); (J.J.R.-C.)
| | | | - Fernanda Genre
- Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (S.R.-M.); (F.G.)
| | - Leticia Lera-Gómez
- Department of Microbiology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain;
| | - Pilar Alonso-Lecue
- Immunopathology Group, Marqués de Valdecilla University Hospital-Valdecilla Research Institute (IDIVAL), 39008 Santander, Spain; (M.S.M.-G.); (P.A.-L.); (J.C.B.-L.); (R.B.); (R.L.-M.); (J.M.C.)
- Department of Pneumology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain; (D.F.-P.); (S.T.-M.); (B.A.-B.); (S.I.); (J.J.R.-C.)
| | - Joao Carlos Batista-Liz
- Immunopathology Group, Marqués de Valdecilla University Hospital-Valdecilla Research Institute (IDIVAL), 39008 Santander, Spain; (M.S.M.-G.); (P.A.-L.); (J.C.B.-L.); (R.B.); (R.L.-M.); (J.M.C.)
- Department of Rheumatology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain
| | - Sandra Tello-Mena
- Department of Pneumology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain; (D.F.-P.); (S.T.-M.); (B.A.-B.); (S.I.); (J.J.R.-C.)
| | - Beatriz Abascal-Bolado
- Department of Pneumology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain; (D.F.-P.); (S.T.-M.); (B.A.-B.); (S.I.); (J.J.R.-C.)
| | - Sheila Izquierdo
- Department of Pneumology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain; (D.F.-P.); (S.T.-M.); (B.A.-B.); (S.I.); (J.J.R.-C.)
| | - Juan José Ruiz-Cubillán
- Department of Pneumology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain; (D.F.-P.); (S.T.-M.); (B.A.-B.); (S.I.); (J.J.R.-C.)
| | | | - Ricardo Blanco
- Immunopathology Group, Marqués de Valdecilla University Hospital-Valdecilla Research Institute (IDIVAL), 39008 Santander, Spain; (M.S.M.-G.); (P.A.-L.); (J.C.B.-L.); (R.B.); (R.L.-M.); (J.M.C.)
- Department of Rheumatology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain
| | - Miguel A. González-Gay
- School of Medicine, University of Cantabria, 39011 Santander, Spain;
- Department of Rheumatology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Raquel López-Mejías
- Immunopathology Group, Marqués de Valdecilla University Hospital-Valdecilla Research Institute (IDIVAL), 39008 Santander, Spain; (M.S.M.-G.); (P.A.-L.); (J.C.B.-L.); (R.B.); (R.L.-M.); (J.M.C.)
- Department of Rheumatology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain
| | - José M. Cifrián
- Immunopathology Group, Marqués de Valdecilla University Hospital-Valdecilla Research Institute (IDIVAL), 39008 Santander, Spain; (M.S.M.-G.); (P.A.-L.); (J.C.B.-L.); (R.B.); (R.L.-M.); (J.M.C.)
- Department of Pneumology, Marqués de Valdecilla University Hospital, 39008 Santander, Spain; (D.F.-P.); (S.T.-M.); (B.A.-B.); (S.I.); (J.J.R.-C.)
- School of Medicine, University of Cantabria, 39011 Santander, Spain;
| |
Collapse
|
7
|
Azzeri A, Mohamed NA, Wan Rosli SH, Abdul Samat MN, Rashid ZZ, Mohamad Jamali MA, Md Zoqratt MZH, Mohammad Nasir MA, Ranjit Singh HK, Azmi L. Unravelling the link between SARS-CoV-2 mutation frequencies, patient comorbidities, and structural dynamics. PLoS One 2024; 19:e0291892. [PMID: 38483913 PMCID: PMC10939192 DOI: 10.1371/journal.pone.0291892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Genomic surveillance is crucial for tracking emergence and spread of novel variants of pathogens, such as SARS-CoV-2, to inform public health interventions and to enforce control measures. However, in some settings especially in low- and middle- income counties, where sequencing platforms are limited, only certain patients get to be selected for sequencing surveillance. Here, we show that patients with multiple comorbidities potentially harbour SARS-CoV-2 with higher mutation rates and thus deserve more attention for genomic surveillance. The relationship between the patient comorbidities, and type of amino acid mutations was assessed. Correlation analysis showed that there was a significant tendency for mutations to occur within the ORF1a region for patients with higher number of comorbidities. Frequency analysis of the amino acid substitution within ORF1a showed that nsp3 P822L of the PLpro protease was one of the highest occurring mutations. Using molecular dynamics, we simulated that the P822L mutation in PLpro represents a system with lower Root Mean Square Deviation (RMSD) fluctuations, and consistent Radius of gyration (Rg), Solvent Accessible Surface Area (SASA) values-indicate a much stabler protein than the wildtype. The outcome of this study will help determine the relationship between the clinical status of a patient and the mutations of the infecting SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Amirah Azzeri
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai, Negeri Sembilan, Malaysia
| | - Nurul Azmawati Mohamed
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai, Negeri Sembilan, Malaysia
| | - Saarah Huurieyah Wan Rosli
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai, Negeri Sembilan, Malaysia
| | - Muttaqillah Najihan Abdul Samat
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Zetti Zainol Rashid
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | | | - Muhammad Zarul Hanifah Md Zoqratt
- Fast Genomics Solutions, Subang Jaya, Selangor Darul Ehsan, Malaysia
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Muhammad Azamuddeen Mohammad Nasir
- Fast Genomics Solutions, Subang Jaya, Selangor Darul Ehsan, Malaysia
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Harpreet Kaur Ranjit Singh
- Fast Genomics Solutions, Subang Jaya, Selangor Darul Ehsan, Malaysia
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Liyana Azmi
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Nilai, Negeri Sembilan, Malaysia
| |
Collapse
|
8
|
Wierzbicka A, Semik-Gurgul E, Świątkiewicz M, Szmatoła T, Steg A, Oczkowicz M. Changes in DNA Methylation and mRNA Expression in Lung Tissue after Long-Term Supplementation with an Increased Dose of Cholecalciferol. Int J Mol Sci 2023; 25:464. [PMID: 38203636 PMCID: PMC10778667 DOI: 10.3390/ijms25010464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Maintaining an appropriate concentration of vitamin D is essential for the proper functioning of the body, regardless of age. Nowadays, there are more and more indications that vitamin D supplementation at higher than standard doses may show protective and therapeutic effects. Our study identified differences in the body's response to long-term supplementation with cholecalciferol at an increased dose. Two groups of pigs were used in the experiment. The first group received a standard dose of cholecalciferol (grower, 2000 IU/kg feed, and finisher, 1500 IU/kg feed), and the second group received an increased dose (grower, 3000 IU/kg feed, and finisher, 2500 IU/kg feed). After slaughter, lung samples were collected and used for RRBS and mRNA sequencing. Analysis of the methylation results showed that 2349 CpG sites had significantly altered methylation patterns and 1116 (47.51%) identified DMSs (Differentially Methylated Sites) were related to genes and their regulatory sites. The mRNA sequencing results showed a significant change in the expression of 195 genes. The integrated analysis identified eleven genes with DNA methylation and mRNA expression differences between the analyzed groups. The results of this study suggested that an increased vitamin D intake may be helpful for the prevention of lung cancer and pulmonary fibrosis. These actions may stem from the influence of vitamin D on the expression of genes associated with collagen production, such as SHMT1, UGT1A6, and ITIH2.The anti-cancer properties of vitamin D are also supported by changes in KLHL3 and TTPA gene expression.
Collapse
Affiliation(s)
- Alicja Wierzbicka
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| | - Ewelina Semik-Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| | - Małgorzata Świątkiewicz
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland;
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
- Center for Experimental and Innovative Medicine, The University of Agriculture in Kraków, Rędzina 1c, 30-248 Kraków, Poland
| | - Anna Steg
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| | - Maria Oczkowicz
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| |
Collapse
|
9
|
Naidoo L, Arumugam T, Ramsuran V. Host Genetic Impact on Infectious Diseases among Different Ethnic Groups. ADVANCED GENETICS (HOBOKEN, N.J.) 2023; 4:2300181. [PMID: 38099246 PMCID: PMC10716055 DOI: 10.1002/ggn2.202300181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/11/2023] [Indexed: 12/17/2023]
Abstract
Infectious diseases such as malaria, tuberculosis (TB), human immunodeficiency virus (HIV), and the coronavirus disease of 2019 (COVID-19) are problematic globally, with high prevalence particularly in Africa, attributing to most of the death rates. There have been immense efforts toward developing effective preventative and therapeutic strategies for these pathogens globally, however, some remain uncured. Disease susceptibility and progression for malaria, TB, HIV, and COVID-19 vary among individuals and are attributed to precautionary measures, environment, host, and pathogen genetics. While studying individuals with similar attributes, it is suggested that host genetics contributes to most of an individual's susceptibility to disease. Several host genes are identified to associate with these pathogens. Interestingly, many of these genes and polymorphisms are common across diseases. This paper analyzes genes and genetic variations within host genes associated with HIV, TB, malaria, and COVID-19 among different ethnic groups. The differences in host-pathogen interaction among these groups, particularly of Caucasian and African descent, and which gene polymorphisms are prevalent in an African population that possesses protection or risk to disease are reviewed. The information in this review could potentially help develop personalized treatment that could effectively combat the high disease burden in Africa.
Collapse
Affiliation(s)
- Lisa Naidoo
- School of Laboratory Medicine and Medical SciencesCollege of Health SciencesUniversity of KwaZulu‐NatalDurban4041South Africa
| | - Thilona Arumugam
- School of Laboratory Medicine and Medical SciencesCollege of Health SciencesUniversity of KwaZulu‐NatalDurban4041South Africa
| | - Veron Ramsuran
- School of Laboratory Medicine and Medical SciencesCollege of Health SciencesUniversity of KwaZulu‐NatalDurban4041South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurban4041South Africa
| |
Collapse
|
10
|
Josuttis D, Schwedler C, Heymann G, Gümbel D, Schmittner MD, Kruse M, Hoppe B. Vascular Endothelial Growth Factor as Potential Biomarker for COVID-19 Severity. J Intensive Care Med 2023; 38:1165-1173. [PMID: 37448220 PMCID: PMC10345830 DOI: 10.1177/08850666231186787] [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: 04/04/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
INTRODUCTION COVID-19 is characterized by immunological responses to viral replication and coherent with endothelitis, microvascular disturbance of lung vasculature and coagulopathy. Vascular Endothelial Growth Factor (VEGF) is a proangiogenic mediator regulating endothelial changes. It is induced by proinflammatory signaling and hypoxia. We sought to determine whether VEGF levels differ between SARS-CoV-2-positive patients of different disease severity and whether VEGF might be useful in risk stratification. METHODS After retrospective screening of all SARS-CoV-2-positive patients treated in Unfallkrankenhaus Berlin in 2020, we included those with documented VEGF measurement. We extracted laboratory values and clinical parameters. An exploratory data analysis was performed to detect possible relations between VEGF level and clinical disease features. RESULTS We included 167 SARS-CoV-2-positive patients of which 139 suffered from COVID-19. Seventy-one of the COVID-19 patients had to be treated in the intensive care unit (ICU), those patients exhibited higher VEGF levels than those being admitted to normal wards (535 vs 279 pg/L, P < .001). APACHE-2 (Acute Physiology And Chronic Health Evaluation Score) correlated with mortality and patients with high values showed higher VEGF concentrations on admission (456 vs 875 pg/L, p = 0.006). Receiver operating characteristic analytic revealed that the occurrence of organ dysfunctions like acute respiratory distress syndrome (ARDS), shock, or acute kidney injury could be predicted by VEGF. It was significantly higher in patients who later died compared to survivors (637 vs 389 pg/mL, P = 0.041) and predicted mortality with same accuracy as established markers. In our cohort, association of VEGF above 277 pg/L on admission with risk of ARDS could be confirmed in logistic regression adjusting for possible confounding factors (odds ratio 3.1, 95% confidence interval: 1.34-7.7). DISCUSSION Even though there are several limitations to this retrospective study it revealed that in COVID-19 patients VEGF can contribute to the prediction of necessity of ICU, mortality and the prediction of ARDS, kidney injury or shock. Its use in risk stratification and potential pathogenetic involvement should be further investigated.
Collapse
Affiliation(s)
- David Josuttis
- Department of Anesthesiology, Intensive Care and Pain Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | | | - Guido Heymann
- Department of Laboratory Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Denis Gümbel
- Department of Trauma, Reconstructive Surgery and Rehabilitation Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Trauma and Orthopaedic Surgery, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Marc Dominik Schmittner
- Department of Anesthesiology, Intensive Care and Pain Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Marianne Kruse
- Department of Anesthesiology, Intensive Care and Pain Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Berthold Hoppe
- Health and Medical University Potsdam, Potsdam, Germany
- Department of Laboratory Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| |
Collapse
|
11
|
Salihefendić L, Čeko I, Bešić L, Mulahuseinović N, Durgut S, Pećar D, Prnjavorac L, Kandić E, Meseldžić N, Bego T, Prnjavorac B, Marjanović D, Konjhodžić R, Ašić A. Identification of human genetic variants modulating the course of COVID-19 infection with importance in other viral infections. Front Genet 2023; 14:1240245. [PMID: 37795240 PMCID: PMC10545899 DOI: 10.3389/fgene.2023.1240245] [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: 06/14/2023] [Accepted: 08/11/2023] [Indexed: 10/06/2023] Open
Abstract
Introduction: COVID-19 has been a major focus of scientific research since early 2020. Due to its societal, economic, and clinical impact worldwide, research efforts aimed, among other questions, to address the effect of host genetics in susceptibility and severity of COVID-19. Methods: We, therefore, performed next-generation sequencing of coding and regulatory regions of 16 human genes, involved in maintenance of the immune system or encoding receptors for viral entry into the host cells, in a subset of 60 COVID-19 patients from the General Hospital Tešanj, Bosnia and Herzegovina, classified into three groups of clinical conditions of different severity ("mild," "moderate," and "severe"). Results: We confirmed that the male sex and older age are risk factors for severe clinical picture and identified 13 variants on seven genes (CD55, IL1B, IL4, IRF7, DDX58, TMPRSS2, and ACE2) with potential functional significance, either as genetic markers of modulated susceptibility to SARS-CoV-2 infection or modifiers of the infection severity. Our results include variants reported for the first time as potentially associated with COVID-19, but further research and larger patient cohorts are required to confirm their effect. Discussion: Such studies, focused on candidate genes and/or variants, have a potential to answer the questions regarding the effect of human genetic makeup on the expected infection outcome. In addition, loci we identified here were previously reported to have clinical significance in other diseases and viral infections, thus confirming a general, broader significance of COVID-19-related research results following the end of the pandemic period.
Collapse
Affiliation(s)
- Lana Salihefendić
- ALEA Genetic Center, Sarajevo, Bosnia and Herzegovina
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | - Ivana Čeko
- ALEA Genetic Center, Sarajevo, Bosnia and Herzegovina
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | - Larisa Bešić
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | | | - Selma Durgut
- ALEA Genetic Center, Sarajevo, Bosnia and Herzegovina
| | - Dino Pećar
- ALEA Genetic Center, Sarajevo, Bosnia and Herzegovina
| | | | - Enis Kandić
- ALEA Genetic Center, Sarajevo, Bosnia and Herzegovina
| | - Neven Meseldžić
- Department of Pharmaceutical Biochemistry and Laboratory Diagnostics, Faculty of Pharmacy, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Tamer Bego
- Department of Pharmaceutical Biochemistry and Laboratory Diagnostics, Faculty of Pharmacy, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Damir Marjanović
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
- Institute for Anthropological Research, University of Zagreb, Zagreb, Croatia
| | - Rijad Konjhodžić
- ALEA Genetic Center, Sarajevo, Bosnia and Herzegovina
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | - Adna Ašić
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
12
|
Montezano AC, Camargo LL, Mary S, Neves KB, Rios FJ, Stein R, Lopes RA, Beattie W, Thomson J, Herder V, Szemiel AM, McFarlane S, Palmarini M, Touyz RM. SARS-CoV-2 spike protein induces endothelial inflammation via ACE2 independently of viral replication. Sci Rep 2023; 13:14086. [PMID: 37640791 PMCID: PMC10462711 DOI: 10.1038/s41598-023-41115-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
COVID-19, caused by SARS-CoV-2, is a respiratory disease associated with inflammation and endotheliitis. Mechanisms underling inflammatory processes are unclear, but angiotensin converting enzyme 2 (ACE2), the receptor which binds the spike protein of SARS-CoV-2 may be important. Here we investigated whether spike protein binding to ACE2 induces inflammation in endothelial cells and determined the role of ACE2 in this process. Human endothelial cells were exposed to SARS-CoV-2 spike protein, S1 subunit (rS1p) and pro-inflammatory signaling and inflammatory mediators assessed. ACE2 was modulated pharmacologically and by siRNA. Endothelial cells were also exposed to SARS-CoV-2. rSP1 increased production of IL-6, MCP-1, ICAM-1 and PAI-1, and induced NFkB activation via ACE2 in endothelial cells. rS1p increased microparticle formation, a functional marker of endothelial injury. ACE2 interacting proteins involved in inflammation and RNA biology were identified in rS1p-treated cells. Neither ACE2 expression nor ACE2 enzymatic function were affected by rSP1. Endothelial cells exposed to SARS-CoV-2 virus did not exhibit viral replication. We demonstrate that rSP1 induces endothelial inflammation via ACE2 through processes that are independent of ACE2 enzymatic activity and viral replication. We define a novel role for ACE2 in COVID-19- associated endotheliitis.
Collapse
Affiliation(s)
- Augusto C Montezano
- Research Institute of the McGill University Health Centre (RI-MUHC), Site Glen-Block E-Office: E01.3362, 1001, Boul. Decarie, Montreal, QC, H4A3J1, Canada.
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK.
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre (RI-MUHC), Site Glen-Block E-Office: E01.3362, 1001, Boul. Decarie, Montreal, QC, H4A3J1, Canada
| | - Sheon Mary
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Karla B Neves
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Francisco J Rios
- Research Institute of the McGill University Health Centre (RI-MUHC), Site Glen-Block E-Office: E01.3362, 1001, Boul. Decarie, Montreal, QC, H4A3J1, Canada
| | - Ross Stein
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Rheure A Lopes
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Wendy Beattie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Jacqueline Thomson
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Vanessa Herder
- MRC Centre for Virus Research, University of Glasgow, Glasgow, UK
| | | | - Steven McFarlane
- MRC Centre for Virus Research, University of Glasgow, Glasgow, UK
| | | | - Rhian M Touyz
- Research Institute of the McGill University Health Centre (RI-MUHC), Site Glen-Block E-Office: E01.3362, 1001, Boul. Decarie, Montreal, QC, H4A3J1, Canada.
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK.
- McGill University, Montreal, Canada.
| |
Collapse
|
13
|
Wilson GN. A Clinical Qualification Protocol Highlights Overlapping Genomic Influences and Neuro-Autonomic Mechanisms in Ehlers-Danlos and Long COVID-19 Syndromes. Curr Issues Mol Biol 2023; 45:6003-6023. [PMID: 37504295 PMCID: PMC10378515 DOI: 10.3390/cimb45070379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
A substantial fraction of the 15% with double-jointedness or hypermobility have the traditionally ascertained joint-skeletal, cutaneous, and cardiovascular symptoms of connective tissue dysplasia and its particular manifestation as Ehlers-Danlos syndrome (EDS). The holistic ascertainment of 120 findings in 1261 EDS patients added neuro-autonomic symptoms like headaches, muscle weakness, brain fog, chronic fatigue, dyspnea, and bowel irregularity to those of arthralgia and skin laxity, 15 of these symptoms shared with those of post-infectious SARS-CoV-2 (long COVID-19). Underlying articulo-autonomic mechanisms guided a clinical qualification protocol that qualified DNA variants in 317 genes as having diagnostic utility for EDS, six of them identical (F2-LIFR-NLRP3-STAT1-T1CAM1-TNFRSF13B) and eighteen similar to those modifying COVID-19 severity/EDS, including ADAMTS13/ADAMTS2-C3/C1R-IKBKG/IKBKAP-PIK3C3/PIK3R1-POLD4/POLG-TMPRSS2/TMPRSS6-WNT3/WNT10A. Also, contributing to EDS and COVID-19 severity were forty and three genes, respectively, impacting mitochondrial functions as well as parts of an overlapping gene network, or entome, that are hypothesized to mediate the cognitive-behavioral, neuro-autonomic, and immune-inflammatory alterations of connective tissue in these conditions. The further characterization of long COVID-19 natural history and genetic predisposition will be necessary before these parallels to EDS can be carefully delineated and translated into therapies.
Collapse
Affiliation(s)
- Golder N Wilson
- Department of Pediatrics, Texas Tech University Health Sciences Center, Lubbock, and KinderGenome Genetics Private Practice, 5347 W Mockingbird, Dallas, TX 75209, USA
| |
Collapse
|
14
|
Su Y, Wu J, Li X, Li J, Zhao X, Pan B, Huang J, Kong Q, Han J. DTSEA: A network-based drug target set enrichment analysis method for drug repurposing against COVID-19. Comput Biol Med 2023; 159:106969. [PMID: 37105108 PMCID: PMC10121077 DOI: 10.1016/j.compbiomed.2023.106969] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023]
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic is still wreaking havoc worldwide. Therefore, the urgent need for efficient treatments pushes researchers and clinicians into screening effective drugs. Drug repurposing may be a promising and time-saving strategy to identify potential drugs against this disease. Here, we developed a novel computational approach, named Drug Target Set Enrichment Analysis (DTSEA), to identify potent drugs against COVID-19. DTSEA first mapped the disease-related genes into a gene functional interaction network, and then it used a network propagation algorithm to rank all genes in the network by calculating the network proximity of genes to disease-related genes. Finally, an enrichment analysis was performed on drug target sets to prioritize disease-candidate drugs. It was shown that the top three drugs predicted by DTSEA, including Ataluren, Carfilzomib, and Aripiprazole, were significantly enriched in the immune response pathways indicating the potential for use as promising COVID-19 inhibitors. In addition to these drugs, DTSEA also identified several drugs (such as Remdesivir and Olumiant), which have obtained emergency use authorization (EUA) for COVID-19. These results indicated that DTSEA could effectively identify the candidate drugs for COVID-19, which will help to accelerate the development of drugs for COVID-19. We then performed several validations to ensure the reliability and validity of DTSEA, including topological analysis, robustness analysis, and prediction consistency. Collectively, DTSEA successfully predicted candidate drugs against COVID-19 with high accuracy and reliability, thus making it a formidable tool to identify potential drugs for a specific disease and facilitate further investigation.
Collapse
Affiliation(s)
- Yinchun Su
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, PR China
| | - Jiashuo Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Xiangmei Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Ji Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Xilong Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Bingyue Pan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Junling Huang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, PR China.
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China.
| |
Collapse
|
15
|
Sarkar S, Lucchetta M, Maier A, Abdrabbou MM, Baumbach J, List M, Schaefer MH, Blumenthal DB. Online bias-aware disease module mining with ROBUST-Web. Bioinformatics 2023; 39:btad345. [PMID: 37233198 PMCID: PMC10246579 DOI: 10.1093/bioinformatics/btad345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/24/2023] [Accepted: 05/25/2023] [Indexed: 05/27/2023] Open
Abstract
SUMMARY We present ROBUST-Web which implements our recently presented ROBUST disease module mining algorithm in a user-friendly web application. ROBUST-Web features seamless downstream disease module exploration via integrated gene set enrichment analysis, tissue expression annotation, and visualization of drug-protein and disease-gene links. Moreover, ROBUST-Web includes bias-aware edge costs for the underlying Steiner tree model as a new algorithmic feature, which allow to correct for study bias in protein-protein interaction networks and further improves the robustness of the computed modules. AVAILABILITY AND IMPLEMENTATION Web application: https://robust-web.net. Source code of web application and Python package with new bias-aware edge costs: https://github.com/bionetslab/robust-web, https://github.com/bionetslab/robust_bias_aware.
Collapse
Affiliation(s)
- Suryadipto Sarkar
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91301, Germany
| | - Marta Lucchetta
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan 20139, Italy
| | - Andreas Maier
- Institute for Computational Systems Biology, University of Hamburg, Hamburg 22607, Germany
| | - Mohamed M Abdrabbou
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91301, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Hamburg 22607, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Martin H Schaefer
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan 20139, Italy
| | - David B Blumenthal
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91301, Germany
| |
Collapse
|
16
|
Purbey PK, Roy K, Gupta S, Paul MK. Mechanistic insight into the protective and pathogenic immune-responses against SARS-CoV-2. Mol Immunol 2023; 156:111-126. [PMID: 36921486 PMCID: PMC10009586 DOI: 10.1016/j.molimm.2023.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
COVID-19 is a severe respiratory illness that has emerged as a devasting health problem worldwide. The disease outcome is heterogeneous, which is most likely dependent on the immunity of an individual. Asymptomatic and mildly/moderate symptomatic (non-severe) patients likely develop an effective early immune response and clear the virus. However, severe symptoms dominate due to a failure in the generation of an effective and specific early immune response against SARS-CoV-2. Moreover, a late surge in pathogenic inflammation involves dysregulated innate and adaptive immune responses leading to local and systemic tissue damage and the emergence of severe disease symptoms. In this review, we describe the potential mechanisms of protective and pathogenic immune responses in "mild/moderate" and "severe" symptomatic SARS-CoV-2 infected people, respectively, and discuss the immune components that are currently targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Prabhat K Purbey
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| | - Koushik Roy
- Microbiology and Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Sandeep Gupta
- Department of Neurobiology, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Manash K Paul
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
17
|
Low RN, Low RJ, Akrami A. A review of cytokine-based pathophysiology of Long COVID symptoms. Front Med (Lausanne) 2023; 10:1011936. [PMID: 37064029 PMCID: PMC10103649 DOI: 10.3389/fmed.2023.1011936] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/27/2023] [Indexed: 04/03/2023] Open
Abstract
The Long COVID/Post Acute Sequelae of COVID-19 (PASC) group includes patients with initial mild-to-moderate symptoms during the acute phase of the illness, in whom recovery is prolonged, or new symptoms are developed over months. Here, we propose a description of the pathophysiology of the Long COVID presentation based on inflammatory cytokine cascades and the p38 MAP kinase signaling pathways that regulate cytokine production. In this model, the SARS-CoV-2 viral infection is hypothesized to trigger a dysregulated peripheral immune system activation with subsequent cytokine release. Chronic low-grade inflammation leads to dysregulated brain microglia with an exaggerated release of central cytokines, producing neuroinflammation. Immunothrombosis linked to chronic inflammation with microclot formation leads to decreased tissue perfusion and ischemia. Intermittent fatigue, Post Exertional Malaise (PEM), CNS symptoms with "brain fog," arthralgias, paresthesias, dysautonomia, and GI and ophthalmic problems can consequently arise as result of the elevated peripheral and central cytokines. There are abundant similarities between symptoms in Long COVID and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). DNA polymorphisms and viral-induced epigenetic changes to cytokine gene expression may lead to chronic inflammation in Long COVID patients, predisposing some to develop autoimmunity, which may be the gateway to ME/CFS.
Collapse
Affiliation(s)
| | - Ryan J. Low
- Gatsby Computational Neuroscience Unit, University College London, London, United Kingdom
- Sainsbury Wellcome Centre, University College London, London, United Kingdom
| | - Athena Akrami
- Sainsbury Wellcome Centre, University College London, London, United Kingdom
| |
Collapse
|
18
|
Enichen E, Harvey C, Demmig-Adams B. COVID-19 Spotlights Connections between Disease and Multiple Lifestyle Factors. Am J Lifestyle Med 2023; 17:231-257. [PMID: 36883129 PMCID: PMC9445631 DOI: 10.1177/15598276221123005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The SARS-CoV-2 virus (severe acute respiratory syndrome coronavirus 2), and the disease it causes (COVID-19), have had a profound impact on global human society and threaten to continue to have such an impact with newly emerging variants. Because of the widespread effects of SARS-CoV-2, understanding how lifestyle choices impact the severity of disease is imperative. This review summarizes evidence for an involvement of chronic, non-resolving inflammation, gut microbiome disruption (dysbiosis with loss of beneficial microorganisms), and impaired viral defenses, all of which are associated with an imbalanced lifestyle, in severe disease manifestations and post-acute sequelae of SARS-CoV-2 (PASC). Humans' physiological propensity for uncontrolled inflammation and severe COVID-19 are briefly contrasted with bats' low propensity for inflammation and their resistance to viral disease. This insight is used to identify positive lifestyle factors with the potential to act in synergy for restoring balance to the immune response and gut microbiome, and thereby protect individuals against severe COVID-19 and PASC. It is proposed that clinicians should consider recommending lifestyle factors, such as stress management, balanced nutrition and physical activity, as preventative measures against severe viral disease and PASC.
Collapse
Affiliation(s)
- Elizabeth Enichen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| | - Caitlyn Harvey
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| | - Barbara Demmig-Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| |
Collapse
|
19
|
Papadopoulos KI, Papadopoulou A, Aw TC. Beauty and the beast: host microRNA-155 versus SARS-CoV-2. Hum Cell 2023; 36:908-922. [PMID: 36847920 PMCID: PMC9969954 DOI: 10.1007/s13577-023-00867-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/29/2023] [Indexed: 02/28/2023]
Abstract
Severe acute respiratory coronavirus 2 (SARS-CoV-2) infection in the young and healthy usually results in an asymptomatic or mild viral syndrome, possibly through an erythropoietin (EPO)-dependent, protective evolutionary landscape. In the old and in the presence of co-morbidities, however, a potentially lethal coronavirus disease 2019 (COVID-19) cytokine storm, through unrestrained renin-angiotensin aldosterone system (RAAS) hyperactivity, has been described. Multifunctional microRNA-155 (miR-155) elevation in malaria, dengue virus (DENV), the thalassemias, and SARS-CoV-1/2, plays critical antiviral and cardiovascular roles through its targeted translational repression of over 140 genes. In the present review, we propose a plausible miR-155-dependent mechanism whereby the translational repression of AGRT1, Arginase-2 and Ets-1, reshapes RAAS towards Angiotensin II (Ang II) type 2 (AT2R)-mediated balanced, tolerable, and SARS-CoV-2-protective cardiovascular phenotypes. In addition, it enhances EPO secretion and endothelial nitric oxide synthase activation and substrate availability, and negates proinflammatory Ang II effects. Disrupted miR-155 repression of AT1R + 1166C-allele, significantly associated with adverse cardiovascular and COVID-19 outcomes, manifests its decisive role in RAAS modulation. BACH1 and SOCS1 repression creates an anti-inflammatory and cytoprotective milieu, robustly inducing antiviral interferons. MiR-155 dysregulation in the elderly, and in comorbidities, allows unimpeded RAAS hyperactivity to progress towards a particularly aggressive COVID-19 course. Elevated miR-155 in thalassemia plausibly engenders a favorable cardiovascular profile and protection against malaria, DENV, and SARS-CoV-2. MiR-155 modulating pharmaceutical approaches could offer novel therapeutic options in COVID-19.
Collapse
Affiliation(s)
- K. I. Papadopoulos
- THAI StemLife, 566/3 Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd., Wangthonglang, Bangkok, 10310 Thailand
| | - A. Papadopoulou
- Occupational and Environmental Health Services, Feelgood Lund, Ideon Science Park, Scheelevägen 17, 223 63 Lund, Sweden
| | - T. C. Aw
- grid.413815.a0000 0004 0469 9373Department of Laboratory Medicine, Changi General Hospital, 2 Simei Street 3, Singapore, 529889 Singapore
- grid.4280.e0000 0001 2180 6431Department of Medicine, National University of Singapore, Singapore, 119228 Singapore
| |
Collapse
|
20
|
Xiang F, Long B, He J, Cheng F, Zhang S, Liu Q, Chen Z, Li H, Chen M, Peng M, Yin W, Liu D, Ren H. Impaired antibody responses were observed in patients with type 2 diabetes mellitus after receiving the inactivated COVID-19 vaccines. Virol J 2023; 20:22. [PMID: 36750902 PMCID: PMC9902824 DOI: 10.1186/s12985-023-01983-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Patients with type 2 diabetes mellitus (T2DM) have been reported to be more susceptible to 2019 novel coronavirus (2019-nCoV) and more likely to develop severe pneumonia. However, the safety and immunological responses of T2DM patients after receiving the inactivated vaccines are not quite definite. Therefore, we aimed to explore the safety, antibody responses, and B-cell immunity of T2DM patients who were vaccinated with inactivated coronavirus disease 2019 (COVID-19) vaccines. METHODS Eighty-nine patients with T2DM and 100 healthy controls (HCs) were enrolled, all of whom had received two doses of full-course inactivated vaccines. At 21-105 days after full-course vaccines: first, the safety of the vaccines was assessed by questionnaires; second, the titers of anti-receptor binding domain IgG (anti-RBD-IgG) and neutralizing antibodies (NAbs) were measured; third, we detected the frequency of RBD-specific memory B cells (RBD-specific MBCs) to explore the cellular immunity of T2DM patients. RESULTS The overall incidence of adverse events was similar between T2DM patients and HCs, and no serious adverse events were recorded in either group. Compared with HCs, significantly lower titers of anti-RBD-IgG (p = 0.004) and NAbs (p = 0.013) were observed in T2DM patients. Moreover, the frequency of RBD-specific MBCs was lower in T2DM patients than in HCs (p = 0.027). Among the 89 T2DM patients, individuals with lower body mass index (BMI) had higher antibody titers (anti-RBD-IgG: p = 0.009; NAbs: p = 0.084). Furthermore, we found that sex, BMI, and days after vaccination were correlated with antibody titers. CONCLUSIONS Inactivated COVID-19 vaccines were safe in patients with T2DM, but the antibody responses and memory B-cell responses were significantly decreased compared to HCs. TRIAL REGISTRATION NUMBER AND DATE NCT05043246. September 14, 2021. (Clinical Trials.gov).
Collapse
Affiliation(s)
- Feng Xiang
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Boyu Long
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jiaoxia He
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Feifei Cheng
- grid.203458.80000 0000 8653 0555Department of Endocrine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Sijing Zhang
- grid.203458.80000 0000 8653 0555Department of Endocrine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qing Liu
- grid.203458.80000 0000 8653 0555Department of Endocrine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhiwei Chen
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hu Li
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Min Chen
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mingli Peng
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Wenwei Yin
- grid.203458.80000 0000 8653 0555Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Dongfang Liu
- Department of Endocrine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Hong Ren
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
21
|
Papadopoulos KI, Papadopoulou A, Aw TC. A protective erythropoietin evolutionary landscape, NLRP3 inflammasome regulation, and multisystem inflammatory syndrome in children. Hum Cell 2023; 36:26-40. [PMID: 36310304 PMCID: PMC9618415 DOI: 10.1007/s13577-022-00819-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2022]
Abstract
The low incidence of pediatric severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection and the associated multisystem inflammatory syndrome (MIS-C) lack a unifying pathophysiological explanation, impeding effective prevention and therapy. Activation of the NACHT, LRR, and PYD domains-containing protein (NLRP) 3 inflammasome in SARS-CoV-2 with perturbed regulation in MIS-C, has been reported. We posit that, early age physiological states and genetic determinants, such as certain polymorphisms of renin-angiotensin aldosterone system (RAAS) molecules, promote a controlled RAAS hyperactive state, and form an evolutionary landscape involving an age-dependent erythropoietin (EPO) elevation, mediating ancestral innate immune defenses that, through appropriate NLRP3 regulation, mitigate tissue injury and pathogen invasion. SARS-CoV-2-induced downregulation of angiotensin-converting enzyme (ACE)2 expression in endothelial cells (EC), impairment of endothelial nitric oxide (NO) synthase (eNOS) activity and downstream NO bioavailability, may promote a hyperactive RAAS with elevated angiotensin II and aldosterone that, can trigger, and accelerate NLRP3 inflammasome activation, while EPO-eNOS/NO abrogate it. Young age and a protective EPO evolutionary landscape may successfully inhibit SARS-CoV-2 and contain NLRP3 inflammasome activation. By contrast, increasing age and falling EPO levels, in genetically susceptible children with adverse genetic variants and co-morbidities, may lead to unopposed RAAS hyperactivity, NLRP3 inflammasome dysregulation, severe endotheliitis with pyroptotic cytokine storm, and development of autoantibodies, as already described in MIS-C. Our haplotype estimates, predicted from allele frequencies in population databases, are in concordance with MIS-C incidence reports in Europeans but indicate lower risks for Asians and African Americans. Targeted Mendelian approaches dissecting the influence of relevant genetic variants are needed.
Collapse
Affiliation(s)
- Konstantinos I Papadopoulos
- Department of Research and Development, THAI StemLife Co., Ltd., 566/3 THAI StemLife Bldg., Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd., Wangthonglang, 10310, Bangkok, Thailand.
| | - Alexandra Papadopoulou
- Occupational and Environmental Health Services, Feelgood Lund, Ideon Science Park, Scheelevägen 17, 223 63, Lund, Sweden
| | - Tar-Choon Aw
- Department of Laboratory Medicine, Changi General Hospital, Singapore, 529889, Singapore
- Department of Medicine, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|
22
|
Srivastava A, Hollenbach JA. The immunogenetics of COVID-19. Immunogenetics 2022; 75:309-320. [PMID: 36534127 PMCID: PMC9762652 DOI: 10.1007/s00251-022-01284-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/19/2022] [Indexed: 12/23/2022]
Abstract
The worldwide coronavirus disease 2019 pandemic was sparked by the severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2) that first surfaced in December 2019 (COVID-19). The effects of COVID-19 differ substantially not just between patients individually but also between populations with different ancestries. In humans, the human leukocyte antigen (HLA) system coordinates immune regulation. Since HLA molecules are a major component of antigen-presenting pathway, they play an important role in determining susceptibility to infectious disease. It is likely that differential susceptibility to SARS-CoV-2 infection and/or disease course in COVID-19 in different individuals could be influenced by the variations in the HLA genes which are associated with various immune responses to SARS-CoV-2. A growing number of studies have identified a connection between HLA variation and diverse COVID-19 outcomes. Here, we review research investigating the impact of HLA on individual responses to SARS-CoV-2 infection and/or progression, also discussing the significance of MHC-related immunological patterns and its use in vaccine design.
Collapse
Affiliation(s)
- Anshika Srivastava
- grid.266102.10000 0001 2297 6811University of California San Francisco, San Francisco, CA USA
| | - Jill A. Hollenbach
- grid.266102.10000 0001 2297 6811University of California San Francisco, San Francisco, CA USA
| |
Collapse
|
23
|
Gushchin VA, Pochtovyi AA, Kustova DD, Ogarkova DA, Tarnovetskii IY, Belyaeva ED, Divisenko EV, Vasilchenko LA, Shidlovskaya EV, Kuznetsova NA, Tkachuk AP, Slutskiy EA, Speshilov GI, Komarov AG, Tsibin AN, Zlobin VI, Logunov DY, Gintsburg AL. Dynamics of SARS-CoV-2 Major Genetic Lineages in Moscow in the Context of Vaccine Prophylaxis. Int J Mol Sci 2022; 23:14670. [PMID: 36498998 PMCID: PMC9736394 DOI: 10.3390/ijms232314670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
Findings collected over two and a half years of the COVID-19 pandemic demonstrated that the level immunity resulting from vaccination and infection is insufficient to stop the circulation of new genetic variants. The short-term decline in morbidity was followed by a steady increase. The early identification of new genetic lineages that will require vaccine adaptation in the future is an important research target. In this study, we summarised data on the variability of genetic line composition throughout the COVID-19 pandemic in Moscow, Russia, and evaluated the virological and epidemiological features of dominant variants in the context of selected vaccine prophylaxes. The prevalence of the Omicron variant highlighted the low effectiveness of the existing immune layer in preventing infection, which points to the necessity of optimising the antigens used in vaccines in Moscow. Logistic growth curves showing the rate at which the new variant displaces the previously dominant variants may serve as early indicators for selecting candidates for updated vaccines, along with estimates of efficacy, reduced viral neutralising activity against the new strains, and viral load in previously vaccinated patients.
Collapse
Affiliation(s)
- Vladimir A. Gushchin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
- Department of Virology, Biological Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Andrei A. Pochtovyi
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
- Department of Virology, Biological Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Daria D. Kustova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
- Department of Virology, Biological Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Darya A. Ogarkova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | | | - Elizaveta D. Belyaeva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Elizaveta V. Divisenko
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Lyudmila A. Vasilchenko
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Elena V. Shidlovskaya
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Nadezhda A. Kuznetsova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Artem P. Tkachuk
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | | | | | | | | | - Vladimir I. Zlobin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Denis Y. Logunov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Alexander L. Gintsburg
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
- Department of Infectiology and Virology, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov, First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| |
Collapse
|
24
|
Alcalá-Santiago Á, Rodríguez-Barranco M, Rava M, Jiménez-Sousa MÁ, Gil Á, Sánchez MJ, Molina-Montes E. Vitamin D Deficiency and COVID-19: A Biological Database Study on Pathways and Gene-Disease Associations. Int J Mol Sci 2022; 23:ijms232214256. [PMID: 36430729 PMCID: PMC9699081 DOI: 10.3390/ijms232214256] [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: 11/04/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Vitamin D (VD) is a fat-soluble vitamin, and pivotal for maintaining health. Several genetic markers have been related to a deficient VD status; these markers could confer an increased risk to develop osteoporosis and other chronic diseases. A VD deficiency could also be a determinant of a severe COVID-19 disease. This study aimed to interrogate genetic/biological databases on the biological implications of a VD deficiency and its association with diseases, to further explore its link with COVID-19. The genetic variants of both a VD deficiency and COVID-19 were identified in the genome-wide association studies (GWAS) catalog and other sources. We conducted enrichment analyses (considering corrected p-values < 0.05 as statistically significant) of the pathways, and gene-disease associations using tools, such as FUMA, REVIGO, DAVID and DisGeNET, and databases, such as the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO). There were 26 and 46 genes associated with a VD deficiency and COVID-19, respectively. However, there were no genes shared between the two. Genes related to a VD deficiency were involved in the metabolism of carbohydrates, retinol, drugs and xenobiotics, and were associated with the metabolic syndrome and related factors (obesity, hypertension and diabetes mellitus), as well as with neoplasms. There were few enriched pathways and disease connections for the COVID-19-related genes, among which some of the aforementioned comorbidities were also present. In conclusion, genetic factors that influence the VD levels in the body are most prominently associated with nutritional and metabolic diseases. A VD deficiency in high-risk populations could be therefore relevant in a severe COVID-19, underlining the need to examine whether a VD supplementation could reduce the severity of this disease.
Collapse
Affiliation(s)
- Ángela Alcalá-Santiago
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Institute of Nutrition and Food Technology (INYTA) ‘José Mataix’, Biomedical Research Centre, University of Granada, Avenida del Conocimiento s/n, 18071 Granada, Spain
| | - Miguel Rodríguez-Barranco
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Andalusian School of Public Health, Cuesta del Observatorio 4, 18012 Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Correspondence: (M.R.-B.); (M.J.S.)
| | - Marta Rava
- National Center of Epidemiology (CNE), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - María Ángeles Jiménez-Sousa
- CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
- Unit of Viral Infection and Immunity, National Center for Microbiology (CNM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Ángel Gil
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Institute of Nutrition and Food Technology (INYTA) ‘José Mataix’, Biomedical Research Centre, University of Granada, Avenida del Conocimiento s/n, 18071 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- CIBER de Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - María José Sánchez
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Andalusian School of Public Health, Cuesta del Observatorio 4, 18012 Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, Faculty of Medicine, University of Granada, 18011 Granada, Spain
- Correspondence: (M.R.-B.); (M.J.S.)
| | - Esther Molina-Montes
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Institute of Nutrition and Food Technology (INYTA) ‘José Mataix’, Biomedical Research Centre, University of Granada, Avenida del Conocimiento s/n, 18071 Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
25
|
Severity of COVID-19 patients with coexistence of asthma and vitamin D deficiency. INFORMATICS IN MEDICINE UNLOCKED 2022; 34:101116. [PMID: 36338941 PMCID: PMC9616486 DOI: 10.1016/j.imu.2022.101116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19)-driven global pandemic triggered innumerable health complications, imposing great challenges in managing other respiratory diseases like asthma. Furthermore, increases in the underlying inflammation involved in the fatality of COVID-19 have been linked with lack of vitamin D. In this research work, we intend to investigate the possible genetic linkage of asthma and vitamin D deficiency with the severity and fatality of COVID-19 using a network-based approach. We identified and analysed 41 and 14 differentially expressed genes (DEGs) of COVID-19 being common with asthma and vitamin D deficiency, respectively, through the comparative differential gene expression analysis and their footprints on signalling pathways. Gene set enrichment analysis for GO terms and signalling pathways reveals key biological activities, including inflammatory response-related pathways (e.g., cytokine- and chemokine-mediated signalling pathways, IL-17, and TNF signalling pathways). Besides, the Protein–Protein Interaction network analysis of those DEGs reveals hub proteins, some of which are reported as inflammatory antiviral interferon-stimulated biomarkers that potentially drive the cytokine storm leading to COVID-19 severity and fatality, and contributes in the early stage of viral replication, respectively. Moreover, the regulatory network analysis found these DEGs associated with antiviral and tumour inhibitory transcription factors and micro-RNAs. Finally, drug–target enrichment analysis yields tetradioxin, estradiol, arsenenous acid, and zinc, which have been reported to be effective in suppressing the pro-inflammatory cytokines production, and other respiratory tract infections. Our results yield shared biomarker-driven key hypotheses followed by network-based analytics, demystifying the mechanistic details of COVID-19 comorbidity of asthma and vitamin D deficiency with their potential therapeutic implications.
Collapse
|
26
|
Presti EL, Nuzzo D, Al Mahmeed W, Al-Rasadi K, Al-Alawi K, Banach M, Banerjee Y, Ceriello A, Cesur M, Cosentino F, Firenze A, Galia M, Goh SY, Janez A, Kalra S, Kapoor N, Kempler P, Lessan N, Lotufo P, Papanas N, Rizvi AA, Sahebkar A, Santos RD, Stoian AP, Toth PP, Viswanathan V, Rizzo M. Molecular and pro-inflammatory aspects of COVID-19: The impact on cardiometabolic health. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166559. [PMID: 36174875 PMCID: PMC9510069 DOI: 10.1016/j.bbadis.2022.166559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/03/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022]
Abstract
Obesity, type 2 diabetes (T2DM), hypertension (HTN), and Cardiovascular Disease (CVD) often cluster together as “Cardiometabolic Disease” (CMD). Just under 50% of patients with CMD increased the risk of morbidity and mortality right from the beginning of the COVID-19 pandemic as it has been reported in most countries affected by the SARS-CoV2 virus. One of the pathophysiological hallmarks of COVID-19 is the overactivation of the immune system with a prominent IL-6 response, resulting in severe and systemic damage involving also cytokines such as IL2, IL4, IL8, IL10, and interferon-gamma were considered strong predictors of COVID-19 severity. Thus, in this mini-review, we try to describe the inflammatory state, the alteration of the adipokine profile, and cytokine production in the obese state of infected and not infected patients by SARS-CoV2 with the final aim to find possible influences of COVID-19 on CMD and CVD. The immunological-based discussion of the molecular processes could inspire the study of promising targets for managing CMD patients and its complications during COVID-19.
Collapse
Affiliation(s)
- Elena Lo Presti
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Domenico Nuzzo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Wael Al Mahmeed
- Heart and Vascular Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | | | - Kamila Al-Alawi
- Department of Training and Studies, Royal Hospital, Ministry of Health, Muscat, Oman
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Yajnavalka Banerjee
- Department of Biochemistry, Mohamed Bin Rashid University, Dubai, United Arab Emirates
| | | | - Mustafa Cesur
- Clinic of Endocrinology, Ankara Güven Hospital, Ankara, Turkey
| | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, University of Stockholm, Sweden
| | - Alberto Firenze
- Unit of Research and International Cooperation, University Hospital of Palermo, Italy
| | - Massimo Galia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bind), University of Palermo, Italy
| | - Su-Yen Goh
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Slovenia
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital & BRIDE, Karnal, India
| | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter Kempler
- Department of Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Nader Lessan
- The Research Institute, Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates
| | - Paulo Lotufo
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, Brazil
| | - Nikolaos Papanas
- Diabetes Center, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| | - Ali A Rizvi
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raul D Santos
- Heart Institute (InCor) University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil; Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Anca P Stoian
- Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, Bucharest, Romania
| | - Peter P Toth
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, Italy.
| | | |
Collapse
|
27
|
Vitello GA, Federico C, Bruno F, Vinci M, Musumeci A, Ragalmuto A, Sturiale V, Brancato D, Calì F, Saccone S. Allelic Variations in the Human Genes TMPRSS2 and CCR5, and the Resistance to Viral Infection by SARS-CoV-2. Int J Mol Sci 2022; 23:ijms23169171. [PMID: 36012436 PMCID: PMC9409186 DOI: 10.3390/ijms23169171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/18/2022] [Accepted: 08/13/2022] [Indexed: 12/28/2022] Open
Abstract
During the first wave of COVID-19 infection in Italy, the number of cases and the mortality rates were among the highest compared to the rest of Europe and the world. Several studies demonstrated a severe clinical course of COVID-19 associated with old age, comorbidities, and male gender. However, there are cases of virus infection resistance in subjects living in close contact with infected subjects. Thus, to explain the predisposition to virus infection and to COVID-19 disease progression, we must consider, in addition to the genetic variability of the virus and other environmental or comorbidity conditions, the allelic variants of specific human genes, directly or indirectly related to the life cycle of the virus. Here, we analyzed three human genetic polymorphisms belonging to the TMPRSS2 and CCR5 genes in a sample population from Sicily (Italy) to investigate possible correlations with the resistance to viral infection and/or to COVID-19 disease progression as recently described in other human populations. Our results did not show any correlations of the rs35074065, rs12329760, and rs333 polymorphisms with SARS-CoV-2 infection or with COVID-19 disease severity. Further studies on other human genetic polymorphisms should be performed to identify the major human determinants of SARS-CoV-2 viral resistance.
Collapse
Affiliation(s)
| | - Concetta Federico
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Francesca Bruno
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Mirella Vinci
- Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| | - Antonino Musumeci
- Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| | - Alda Ragalmuto
- Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| | - Valentina Sturiale
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Desiree Brancato
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Francesco Calì
- Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| | - Salvatore Saccone
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| |
Collapse
|
28
|
The Prevalence and Impact of Coinfection and Superinfection on the Severity and Outcome of COVID-19 Infection: An Updated Literature Review. Pathogens 2022; 11:pathogens11040445. [PMID: 35456120 PMCID: PMC9027948 DOI: 10.3390/pathogens11040445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 02/07/2023] Open
Abstract
Patients with viral illness are at higher risk of secondary infections—whether bacterial, viral, or parasitic—that usually lead to a worse prognosis. In the setting of Corona Virus Disease 2019 (COVID-19), the Severe Acute Respiratory Syndrome Coronavirus-type 2 (SARS-CoV-2) infection may be preceded by a prior microbial infection or has a concurrent or superinfection. Previous reports documented a significantly higher risk of microbial coinfection in SARS-CoV-2-positive patients. Initial results from the United States (U.S.) and Europe found a significantly higher risk of mortality and severe illness among hospitalized patients with SARS-CoV-2 and bacterial coinfection. However, later studies found contradictory results concerning the impact of coinfection on the outcomes of COVID-19. Thus, we conducted the present literature review to provide updated evidence regarding the prevalence of coinfection and superinfection amongst patients with SARS-CoV-2, possible mechanisms underlying the higher risk of coinfection and superinfection in SARS-CoV-2 patients, and the impact of coinfection and superinfection on the outcomes of patients with COVID-19.
Collapse
|
29
|
Keskin A, Ustun GU, Aci R, Duran U. Homocysteine as a marker for predicting disease severity in patients with COVID-19. Biomark Med 2022; 16:559-568. [PMID: 35343243 DOI: 10.2217/bmm-2021-0688] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: Our study was designed on the hypothesis that homocysteine levels are a prognostic parameter that can predict the severity of COVID-19 disease. Materials & methods: 117 COVID-19 patients and 34 non COVID-19 individuals were included in the study. Receiver operating characteristic (ROC) analysis was performed for homocysteine, D-dimer and monocyte/lymphocyte ratio (MLR) levels. Results: According to the ROC analysis, in COVID-19 patients group, Area under curve (AUC) values were 0.835 for homocysteine, 0.859 for D-dimer and 0.882 for MLR. According to the ROC analysis, in which homocysteine, MLR and D-dimer parameters were evaluated together, AUC values were 0.951 in the mild disease group, 1,000 in severe disease group and 0.967 in COVID-19 patients group. Conclusion: It was concluded that homocysteine level is an important parameter in the follow-up of COVID-19 disease.
Collapse
Affiliation(s)
- Adem Keskin
- Department of Medicine Biochemistry, Aydin Adnan Menderes University Institute of Health Sciences, Aydin, 09100, Turkey
| | - Goksenin U Ustun
- Department of Biochemistry, Samsun Training & Research Hospital, Samsun, 55090, Turkey
| | - Recai Aci
- Department of Biochemistry, Samsun Training & Research Hospital, Samsun, 55090, Turkey
| | - Utku Duran
- Department of Veterinary Medicine, Zonguldak Bulent Ecevit University, Çaycuma Food & Agriculture Vocational School, Zonguldak, 67900, Turkey
| |
Collapse
|