1
|
Mahmood MK, Fatih MT, Kurda HA, Mahmood NK, Shareef FU, Faraidun H, Tassery H, Tardivo D, Lan R, Noori ZF, Qadir BH, Hassan AD. Role of viruses in periodontitis: An extensive review of herpesviruses, human immunodeficiency virus, coronavirus-19, papillomavirus and hepatitis viruses. World J Virol 2024; 13:99070. [PMID: 39722755 PMCID: PMC11551682 DOI: 10.5501/wjv.v13.i4.99070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/02/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024] Open
Abstract
Periodontitis is the inflammation of the supporting structures around the dentition. Several microbial agents, mostly bacteria, have been identified as causative factors for periodontal disease. On the other hand, oral cavity is a rich reservoir for viruses since it contains a wide variety of cell types that can be targeted by viruses. Traditionally, the focus of research about the oral flora has been on bacteria because the most widespread oral diseases, like periodontitis and dental caries, are outcomes of bacterial infection. However, recently and especially after the emergence of coronavirus disease 2019, there is a growing tendency toward including viruses also into the scope of oral microbiome investigations. The global high prevalence of periodontitis and viral infections may point out to a concomitant or synergistic effect between the two. Although the exact nature of the mechanism still is not clearly understood, this could be speculated through the manipulation of the immune system by viruses; hence facilitating the furthermore colonization of the oral tissues by bacteria. This review provides an extensive and detailed update on the role of the most common viruses including herpes family (herpes simplex, varicella-zoster, Epstein-Barr, cytomegalovirus), Human papillomaviruses, Human immunodeficiency virus and severe acute respiratory syndrome coronavirus 2 in the initiation, progression and prognosis of periodontitis.
Collapse
Affiliation(s)
| | - Mohammed Taib Fatih
- College of Dentistry, Komar University of Science and technology, Sulaimani 46001, Iraq
| | | | - Nwsiba Khalid Mahmood
- Department of Biology, College of Science, Sulaimani University, Sulaimani 46001, Iraq
| | - Farman Uthman Shareef
- Department of Medical Laboratory Science, College of Science, Charmo University, Chamchamal/Sulaimani 46001, Iraq
| | - Hemin Faraidun
- Department of Biology, University of Freiburg, Mina Biotech, Freiburg 79098, Germany
| | - Herve Tassery
- Department of Odontology, Timone Hospital, Aix Marseille University, APHM, Marseille 13000, France, LBN Laboratory, Montpellier 34000, France
| | - Delphine Tardivo
- Department of Odontology, Timone Hospital, Aix Marseille University, APHM, CNRS, EFS, ADES, Marseille 13000, France
| | - Romain Lan
- Department of Odontology, Timone Hospital, Aix Marseille University, APHM, CNRS, EFS, ADES, Marseille 13000, France
| | - Zana Fuad Noori
- Department of Dentistry, American University of Sulaimani Iraq AUIS, Sulaimani 46001, Iraq
| | - Balen Hamid Qadir
- College of Dentistry, Komar University of Science and technology, Sulaimani 46001, Iraq
| | - Arman Dlshad Hassan
- Department of Biomedical Science, University of Denver, Denver, CO 80014, United States
| |
Collapse
|
2
|
Botto L, Bulbarelli A, Lonati E, Cazzaniga E, Palestini P. Correlation between Exposure to UFP and ACE/ACE2 Pathway: Looking for Possible Involvement in COVID-19 Pandemic. TOXICS 2024; 12:560. [PMID: 39195662 PMCID: PMC11359209 DOI: 10.3390/toxics12080560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024]
Abstract
The overlap between the geographic distribution of COVID-19 outbreaks and pollution levels confirmed a correlation between exposure to atmospheric particulate matter (PM) and the SARS-CoV-2 pandemic. The RAS system is essential in the pathogenesis of inflammatory diseases caused by pollution: the ACE/AngII/AT1 axis activates a pro-inflammatory pathway, which is counteracted by the ACE2/Ang(1-7)/MAS axis, which activates an anti-inflammatory and protective pathway. However, ACE2 is also known to act as a receptor through which SARS-CoV-2 enters host cells to replicate. Furthermore, in vivo systems have demonstrated that exposure to PM increases ACE2 expression. In this study, the effects of acute and sub-acute exposure to ultrafine particles (UFP), originating from different anthropogenic sources (DEP and BB), on the levels of ACE2, ACE, COX-2, HO-1, and iNOS in the lungs and other organs implicated in the pathogenesis of COVID-19 were analyzed in the in vivo BALB/c male mice model. Exposure to UFP alters the levels of ACE2 and/or ACE in all examined organs, and exposure to sub-acute DEP also results in the release of s-ACE2. Furthermore, as evidenced in this and our previous works, COX-2, HO-1, and iNOS levels also demonstrated organ-specific alterations. These proteins play a pivotal role in the UFP-induced inflammatory and oxidative stress responses, and their dysregulation is linked to the development of severe symptoms in individuals infected with SARS-CoV-2, suggesting a heightened vulnerability or a more severe clinical course of the disease. UFP and SARS-CoV-2 share common pathways; therefore, in a "risk stratification" concept, daily exposure to air pollution may significantly increase the likelihood of developing a severe form of COVID-19, explaining, at least in part, the greater lethality of the virus observed in highly polluted areas.
Collapse
Affiliation(s)
- Laura Botto
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
| | - Alessandra Bulbarelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
- POLARIS Research Centre, University of Milano-Bicocca, 20900 Monza, Italy
| | - Elena Lonati
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
| | - Emanuela Cazzaniga
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
| | - Paola Palestini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
- POLARIS Research Centre, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
3
|
Ayeldeen G, Shaker OG, Amer E, Zaafan MA, Herzalla MR, Keshk MA, Abdelhamid AM. The Impact of lncRNA-GAS5/miRNA-200/ACE2 Molecular Pathway on the Severity of COVID-19. Curr Med Chem 2024; 31:1142-1151. [PMID: 37190816 DOI: 10.2174/0929867330666230515144133] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 03/03/2023] [Accepted: 03/13/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARSCoV- 2), which is responsible for coronavirus disease (COVID-19), potentially has severe adverse effects, leading to public health crises worldwide. In COVID-19, deficiency of ACE-2 is linked to increased inflammation and cytokine storms via increased angiotensin II levels and decreased ACE-2/Mas receptor axis activity. MiRNAs are small sequences of noncoding RNAs that regulate gene expression by binding to the targeted mRNAs. MiR-200 dysfunction has been linked to the development of ARDS following acute lung injury and has been proposed as a key regulator of ACE2 expression. LncRNA growth arrest-specific transcript 5 (GAS5) has been recently studied for its modulatory effect on the miRNA-200/ACE2 axis. OBJECTIVE The current study aims to investigate the role of lncRNA GAS5, miRNA-200, and ACE2 as new COVID-19 diagnostic markers capable of predicting the severity of SARS-CoV-2 complications. METHODS A total of 280 subjects were classified into three groups: COVID-19-negative controls (n = 80), and COVID-19 patients (n=200) who required hospitalization were classified into two groups: group (2) moderate cases (n = 112) and group (3) severe cases (n = 88). RESULTS The results showed that the serum GAS5 expression was significantly down-expressed in COVID-19 patients; as a consequence, the expression of miR-200 was reported to be overexpressed and its targeted ACE2 was down-regulated. The ROC curve was drawn to examine the diagnostic abilities of GAS5, miR-200, and ACE2, yielding high diagnostic accuracy with high sensitivity and specificity. CONCLUSION lncRNA-GAS5, miRNA-200, and ACE2 panels presented great diagnostic potential as they demonstrated the highest diagnostic accuracy for discriminating moderate COVID-19 and severe COVID-19 cases.
Collapse
Affiliation(s)
- Ghada Ayeldeen
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Olfat G Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eman Amer
- Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Egypt
| | - Mai A Zaafan
- Pharmacology & Toxicology Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Mohamed R Herzalla
- Internal Medicine Department, Endocrinology & Diabetes Unit, Zagazig University, 6th of October City, Egypt
| | - Mofida A Keshk
- Department of Molecular Diagnostics and Therapeutics, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City (USC), El-Sadat City, Egypt
| | - Amr M Abdelhamid
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| |
Collapse
|
4
|
Cheraghi Z, Ziai SA, Fazeli Z, Gheisoori A. Unveiling sex-based impact of TYK2 rs2304256 polymorphism on interferon beta-1alpha responsiveness in COVID-19 patients. GENE REPORTS 2023; 33:101846. [DOI: 10.1016/j.genrep.2023.101846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Chang ZY, Alhamami FAMS, Chin KL. Aptamer-Based Strategies to Address Challenges in COVID-19 Diagnosis and Treatments. Interdiscip Perspect Infect Dis 2023; 2023:9224815. [PMID: 37554129 PMCID: PMC10406522 DOI: 10.1155/2023/9224815] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 08/10/2023] Open
Abstract
Coronavirus disease (COVID-19), a highly contagious and rapidly spreading disease with significant fatality in the elderly population, has swept across the world since 2019. Since its first appearance, the causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has undergone multiple mutations, with Omicron as the predominant circulating variant of concern at the moment. The gold standard for diagnosis of COVID-19 by real-time polymerase chain reaction (RT-PCR) to detect the virus is laborious and requires well-trained personnel to perform sophisticated procedures. Also, the genetic variants of SARS-CoV-2 that arise regularly could result in false-negative detection. Meanwhile, the current COVID-19 treatments such as conventional medicine, complementary and alternative medicine, passive antibody therapy, and respiratory therapy are associated with adverse effects. Thus, there is an urgent need to discover novel diagnostic and therapeutic approaches against SARS-CoV-2 and its variants. Over the past 30 years, nucleic acid-based aptamers have gained increasing attention and serve as a promising alternative to the antibodies in the diagnostic and therapeutic fields with their uniqueness of being small, nonimmunogenicity, and thermally stable. Aptamer targeting the SARS-CoV-2 structural proteins or the host receptor proteins represent a powerful tool to control COVID-19 infection. In this review, challenges faced by currently available diagnostic and therapeutic tools for COVID-19 are underscored, along with how aptamers can shed a light on the current COVID-19 pandemic, focusing on the critical factors affecting the discovery of high-affinity aptamers and their potential applications to control COVID-19 infection.
Collapse
Affiliation(s)
- Zi Yuan Chang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | | | - Kai Ling Chin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
6
|
Rezaei M, Mohammadpour H, Eftekhari M, Pourabdollah M, Nasr Azadani F, Tabarsi P, Marjani M, Ziai SA. The role of angiotensin I converting enzyme insertion/deletion polymorphism in the severity and outcomes of COVID-19 patients. Front Genet 2022; 13:1035796. [DOI: 10.3389/fgene.2022.1035796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/07/2022] [Indexed: 12/04/2022] Open
Abstract
The pandemic of coronavirus disease in 2019 has led to a global crisis. COVID-19 shows distinct clinical manifestations of the severity of symptoms. Numerous patients with no associated risk factors demonstrate acute respiratory distress syndrome (ARDS). The role of genetic factors in determining the severity and outcome of the disease remains unresolved. The purpose of this study was to see if a correlation exists between Angiotensin I Converting Enzyme (ACE) insertion/deletion (I/D) polymorphism and the severity of COVID-19 patients’ symptoms. 120 COVID-19 patients admitted to Masih Daneshvari Hospital in Tehran with their consent to participate entered the study. Based on the World Health Organization classification, patients were divided into moderate and severe groups, which were primarily affected by O2 saturation levels. The effects of the patients’ ACE insertion/deletion polymorphism, background disease, Angiotensin receptor blocker (ARB) drug consumption, and demographic parameters on the severity risk were calculated statistically. The ACE D allele was associated with an increased risk of disease severity (OR = 6.766, p = 0.012), but had no effect on mortality.
Collapse
|
7
|
Zareei S, Pourmand S, Amanlou M. Design of novel disturbing peptides against ACE2 SARS-CoV-2 spike-binding region by computational approaches. Front Pharmacol 2022; 13:996005. [PMID: 36438825 PMCID: PMC9692113 DOI: 10.3389/fphar.2022.996005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/24/2022] [Indexed: 10/12/2023] Open
Abstract
The SARS-CoV-2, the virus which is responsible for COVID-19 disease, employs its spike protein to recognize its receptor, angiotensin-converting enzyme 2 (ACE2), and subsequently enters the host cell. In this process, the receptor-binding domain (RBD) of the spike has an interface with the α1-helix of the peptidase domain (PD) of ACE2. This study focuses on the disruption of the protein-protein interaction (PPI) of RBD-ACE2. Among the residues in the template (which was extracted from the ACE2), those with unfavorable energies were selected for substitution by mutagenesis. As a result, a library of 140 peptide candidates was constructed and the binding affinity of each candidate was evaluated by molecular docking and molecular dynamics simulations against the α1-helix of ACE2. Finally, the most potent peptides P23 (GFNNYFPHQSYGFMPTNGVGY), P28 (GFNQYFPHQSYGFPPTNGVGY), and P31 (GFNRYFPHQSYGFCPTNGVGY) were selected and their dynamic behaviors were studied. The results showed peptide inhibitors increased the radius, surface accessible area, and overall mobility of residues of the protein. However, no significant alteration was seen in the key residues in the active site. Meanwhile, they can be proposed as promising agents against COVID-19 by suppressing the viral attachment and curbing the infection at its early stage. The designed peptides showed potency against beta, gamma, delta, and omicron variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Sara Zareei
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Saeed Pourmand
- Department of Chemical Engineering, Faculty of Chemical and Petroleum Engineering, University of Tabriz, Tabriz, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Deng Z, Li Q, Li Y, Deng Z, Chen X, Zhao Z, Wang G, Wang D, Liu J, Li W. Single-cell RNA-seq data analysis characterizing bronchoalveolar epithelial cells in patients with SARS-CoV-2 infection. J Inflamm (Lond) 2022; 19:13. [PMID: 36064702 PMCID: PMC9441843 DOI: 10.1186/s12950-022-00310-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/10/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Angiotensin-converting enzyme 2 (ACE2) has been reported to be the main receptor for SARS-CoV-2 infection of host cells. Understanding the changes in bronchoalveolar epithelial cells after SARS-CoV-2 infection of host cells and the intercellular communication relationship between these epithelial cell changes and immune cells is of great significance for the development of therapeutic methods.
Methods
We explored the single-cell RNA sequence (scRNA-seq) of cells infected with bronchoalveolar lavage fluid (BaLF) of patients with different severities of SARS-CoV-2 and healthy people.
Results
We found 11 clusters of epithelial cells in the BaLF, and they were derived from the S group. In the S group, the proportion of cells with positive ACE2 expression was relatively high. ACE2 was relatively more expressed in epithelial cell clusters 1, 3, and 7. Clusters 4 and 5 represented the original state, and there were two differentiation directions: one was cluster 2, and the others were clusters 1, 3, and 6. Cluster 7 was the intermediate state. Clusters 1, 3, 6, and 7 had high similarities (> 0.9), and their main signaling pathways focused on inflammatory activation and immune response. Cluster 2 was relatively specific and was up-regulated in differential genes that were mainly related to apoptosis. The ligand-receptor expression pattern of TNFRSF10D-TNFSF10 showed a special inter-cell regulatory relationship between epithelial cell cluster 2 and macrophages.
Conclusion
This study revealed the changes in epithelial cells derived from alveolar lavage fluid after SARS-CoV-2 infection and the communication relationship with other immune cells.
Collapse
|
9
|
Romani L, Del Chierico F, Macari G, Pane S, Ristori MV, Guarrasi V, Gardini S, Pascucci GR, Cotugno N, Perno CF, Rossi P, Villani A, Bernardi S, Campana A, Palma P, Putignani L. The Relationship Between Pediatric Gut Microbiota and SARS-CoV-2 Infection. Front Cell Infect Microbiol 2022; 12:908492. [PMID: 35873161 PMCID: PMC9304937 DOI: 10.3389/fcimb.2022.908492] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
This is the first study on gut microbiota (GM) in children affected by coronavirus disease 2019 (COVID-19). Stool samples from 88 patients with suspected severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and 95 healthy subjects were collected (admission: 3–7 days, discharge) to study GM profile by 16S rRNA gene sequencing and relationship to disease severity. The study group was divided in COVID-19 (68), Non–COVID-19 (16), and MIS-C (multisystem inflammatory syndrome in children) (4). Correlations among GM ecology, predicted functions, multiple machine learning (ML) models, and inflammatory response were provided for COVID-19 and Non–COVID-19 cohorts. The GM of COVID-19 cohort resulted as dysbiotic, with the lowest α-diversity compared with Non–COVID-19 and CTRLs and by a specific β-diversity. Its profile appeared enriched in Faecalibacterium, Fusobacterium, and Neisseria and reduced in Bifidobacterium, Blautia, Ruminococcus, Collinsella, Coprococcus, Eggerthella, and Akkermansia, compared with CTRLs (p < 0.05). All GM paired-comparisons disclosed comparable results through all time points. The comparison between COVID-19 and Non–COVID-19 cohorts highlighted a reduction of Abiotrophia in the COVID-19 cohort (p < 0.05). The GM of MIS-C cohort was characterized by an increase of Veillonella, Clostridium, Dialister, Ruminococcus, and Streptococcus and a decrease of Bifidobacterium, Blautia, Granulicatella, and Prevotella, compared with CTRLs. Stratifying for disease severity, the GM associated to “moderate” COVID-19 was characterized by lower α-diversity compared with “mild” and “asymptomatic” and by a GM profile deprived in Neisseria, Lachnospira, Streptococcus, and Prevotella and enriched in Dialister, Acidaminococcus, Oscillospora, Ruminococcus, Clostridium, Alistipes, and Bacteroides. The ML models identified Staphylococcus, Anaerostipes, Faecalibacterium, Dorea, Dialister, Streptococcus, Roseburia, Haemophilus, Granulicatella, Gemmiger, Lachnospira, Corynebacterium, Prevotella, Bilophila, Phascolarctobacterium, Oscillospira, and Veillonella as microbial markers of COVID-19. The KEGG ortholog (KO)–based prediction of GM functional profile highlighted 28 and 39 KO-associated pathways to COVID-19 and CTRLs, respectively. Finally, Bacteroides and Sutterella correlated with proinflammatory cytokines regardless disease severity. Unlike adult GM profiles, Faecalibacterium was a specific marker of pediatric COVID-19 GM. The durable modification of patients’ GM profile suggested a prompt GM quenching response to SARS-CoV-2 infection since the first symptoms. Faecalibacterium and reduced fatty acid and amino acid degradation were proposed as specific COVID-19 disease traits, possibly associated to restrained severity of SARS-CoV-2–infected children. Altogether, this evidence provides a characterization of the pediatric COVID-19–related GM.
Collapse
Affiliation(s)
- Lorenza Romani
- Infectious Disease Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Federica Del Chierico
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, IRCCS, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | - Stefania Pane
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Maria Vittoria Ristori
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, IRCCS, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | | | - Giuseppe Rubens Pascucci
- Research Unit of Congenital and Perinatal Infections, Bambino Gesu` Children’s Hospital, IRCCS, Rome, Italy
| | - Nicola Cotugno
- Research Unit of Congenital and Perinatal Infections, Bambino Gesu` Children’s Hospital, IRCCS, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome ‘‘Tor Vergata’’, Rome, Italy
| | - Carlo Federico Perno
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Multimodal Laboratory Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paolo Rossi
- Chair of Pediatrics, Department of Systems Medicine, University of Rome ‘‘Tor Vergata’’, Rome, Italy
- Academic Department of Pediatrics, Bambino Gesu` Children’s Hospital, IRCCS, Rome, Italy
| | - Alberto Villani
- Pediatric Emergency Department and General Pediatrics, Children Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Stefania Bernardi
- Infectious Disease Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Andrea Campana
- Department of Pediatrics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paolo Palma
- Research Unit of Congenital and Perinatal Infections, Bambino Gesu` Children’s Hospital, IRCCS, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome ‘‘Tor Vergata’’, Rome, Italy
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Lorenza Putignani,
| | | |
Collapse
|
10
|
In silico discovery of multi-targeting inhibitors for the COVID-19 treatment by molecular docking, molecular dynamics simulation studies, and ADMET predictions. Struct Chem 2022. [DOI: 10.1007/s11224-022-01996-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
11
|
Hosseini P, Fallahi MS, Erabi G, Pakdin M, Zarezadeh SM, Faridzadeh A, Entezari S, Ansari A, Poudineh M, Deravi N. Multisystem Inflammatory Syndrome and Autoimmune Diseases Following COVID-19: Molecular Mechanisms and Therapeutic Opportunities. Front Mol Biosci 2022; 9:804109. [PMID: 35495619 PMCID: PMC9046575 DOI: 10.3389/fmolb.2022.804109] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/14/2022] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), has led to huge concern worldwide. Some SARS-CoV-2 infected patients may experience post–COVID-19 complications such as multisystem inflammatory syndrome, defined by symptoms including fever and elevated inflammatory markers (such as elevation of C reactive protein (CRP), erythrocyte sedimentation rate, fibrinogen, procalcitonin test, D-dimer, ferritin, lactate dehydrogenase or IL-6, presence of neutrophilia, lymphopenia, decreased albumin, and multiple organ dysfunction). Post–COVID-19 complications may also manifest as autoimmune diseases such as Guillain-Barré syndrome and systemic lupus erythematosus. Signaling disorders, increased inflammatory cytokines secretion, corticosteroid use to treat COVID-19 patients, or impaired immune responses are suggested causes of autoimmune diseases in these patients. In this review, we discuss the molecular and pathophysiological mechanisms and therapeutic opportunities for multisystem inflammatory syndrome and autoimmune diseases following SARS-CoV-2 infection with the aim to provide a clear view for health care providers and researchers.
Collapse
Affiliation(s)
- Parastoo Hosseini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Gisou Erabi
- Student Research Committee, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Majid Pakdin
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sarina Entezari
- Student Research Committee, School of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Niloofar Deravi,
| |
Collapse
|
12
|
Ayala-Ramírez P, González M, Escudero C, Quintero-Arciniegas L, Giachini FR, Alves de Freitas R, Damiano AE, García-Robles R. Severe Acute Respiratory Syndrome Coronavirus 2 Infection in Pregnancy. A Non-systematic Review of Clinical Presentation, Potential Effects of Physiological Adaptations in Pregnancy, and Placental Vascular Alterations. Front Physiol 2022; 13:785274. [PMID: 35431989 PMCID: PMC9005899 DOI: 10.3389/fphys.2022.785274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
In December 2019, the novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) rapidly spread to become a pandemic. To date, increasing evidence has described the potential negative impact of SARS-CoV-2 infection on pregnant women. Although the pathophysiology of coronavirus disease 2019 (COVID-19) is not entirely understood, there is emerging evidence that it causes a severe systemic inflammatory response associated with vascular alterations that could be of special interest considering some physiological changes in pregnancy. Additionally, these alterations may affect the physiology of the placenta and are associated with pregnancy complications and abnormal histologic findings. On the other hand, data about the vaccine against SARS-CoV-2 are limited, but the risks of administering COVID-19 vaccines during pregnancy appear to be minimal. This review summarizes the current literature on SARSCoV2 virus infection, the development of COVID-19 and its relationship with physiological changes, and angiotensin-converting enzyme 2 (ACE2) function during pregnancy. We have particularly emphasized evidence coming from Latin American countries.
Collapse
Affiliation(s)
- Paola Ayala-Ramírez
- School of Medicine, Human Genetics Institute, Pontificia Universidad Javeriana, Bogotá, Colombia
- *Correspondence: Paola Ayala-Ramírez,
| | - Marcelo González
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
- Marcelo González,
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
- Laboratory of Vascular Physiology, Department of Basic Sciences, Faculty of Sciences, Universidad del Bio-Bio, Chillan, Chile
| | - Laura Quintero-Arciniegas
- Perinatal Medicine Seedbed, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
- Department of Physiological Sciences, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Fernanda R. Giachini
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil
| | | | - Alicia E. Damiano
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)- CONICET- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Reggie García-Robles
- Department of Physiological Sciences, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
13
|
Butnariu AB, Look A, Grillo M, Tabish TA, McGarvey MJ, Pranjol MZI. SARS-CoV-2-host cell surface interactions and potential antiviral therapies. Interface Focus 2022; 12:20200081. [PMID: 34956606 PMCID: PMC8662392 DOI: 10.1098/rsfs.2020.0081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
In this review, we reveal the latest developments at the interface between SARS-CoV-2 and the host cell surface. In particular, we evaluate the current and potential mechanisms of binding, fusion and the conformational changes of the spike (S) protein to host cell surface receptors, especially the human angiotensin-converting enzyme 2 (ACE2) receptor. For instance, upon the initial attachment, the receptor binding domain of the S protein forms primarily hydrogen bonds with the protease domain of ACE2 resulting in conformational changes within the secondary structure. These surface interactions are of paramount importance and have been therapeutically exploited for antiviral design, such as monoclonal antibodies. Additionally, we provide an insight into novel therapies that target viral non-structural proteins, such as viral RNA polymerase. An example of which is remdesivir which has now been approved for use in COVID-19 patients by the US Food and Drug Administration. Establishing further understanding of the molecular details at the cell surface will undoubtably aid the development of more efficacious and selectively targeted therapies to reduce the burden of COVID-19.
Collapse
Affiliation(s)
| | - Alex Look
- School of Life Sciences, University of Sussex, Falmer, UK
| | - Marta Grillo
- School of Life Sciences, University of Sussex, Falmer, UK
| | - Tanveer A. Tabish
- Faculty of Engineering, Department of Materials, Royal School of Mines, Imperial College London, London, UK
| | - Michael J. McGarvey
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | | |
Collapse
|
14
|
Monroe I, Dale M, Schwabe M, Schenkel R, Schenarts PJ. The COVID-19 Patient in the Surgical Intensive Care Unit. Surg Clin North Am 2022; 102:1-21. [PMID: 34800379 PMCID: PMC8479422 DOI: 10.1016/j.suc.2021.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
COVID-19 continues to rampage around the world. Noncritical care-trained physicians may be deployed into the intensive care unit to manage these complex patients. Although COVID-19 is primarily a respiratory disease, it is also associated with significant pathology in the brain, heart, vasculature, lungs, gastrointestinal tract, and kidneys. This article provides an overview of COVID-19 using an organ-based, systematic approach.
Collapse
|
15
|
Huang Y, Chen S, Xiao L, Qin W, Li L, Wang Y, Ma L, Yuan X. A Novel Prognostic Signature for Survival Prediction and Immune Implication Based on SARS-CoV-2–Related Genes in Kidney Renal Clear Cell Carcinoma. Front Bioeng Biotechnol 2022; 9:744659. [PMID: 35141213 PMCID: PMC8819071 DOI: 10.3389/fbioe.2021.744659] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/31/2021] [Indexed: 12/28/2022] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) is a common aggressive malignancy of the urinary system. COVID-19, a highly infectious and severe disease caused by SARS-CoV-2, has become a significant challenge for global public health. Cancer patients have been reported to be more vulnerable to SARS-CoV-2 infection and have a higher risk for serious complications than the general population. However, the correlation between KIRC and COVID-19 remains incompletely elucidated. In this study, we comprehensively investigated the expression and prognostic significance of 333 SARS-CoV-2 infection–related genes in KIRC using the TCGA dataset and identified 31 SARS-CoV-2–related differently expressed genes between KIRC and normal renal tissues. Based on these genes, we constructed and validated a 5-gene prognostic signature (including ACADM, CENPF, KDELC1, PLOD2, and TRMT1) to distinguish low- and high-risk KIRC patients of poor survival in TCGA and E-MTAB-1980 cohorts. Gene set enrichment analysis (GSEA) showed that some inflammatory/immune-related pathways were significantly enriched in the high-risk group. The ESTIMATE analysis indicated that patients in the high-risk group had higher stromal and immune cell scores, therefore lower tumor purity. Moreover, they presented higher proportions of macrophages M0, regulatory T cells (Tregs), and T follicular helper cells and higher expression of immune checkpoints CTLA-4, LAG-3, TIGIT, and PDCD1 than low-risk patients. Besides, we also developed a nomogram to expand clinical applicability, which exhibits excellent predictive accuracy for survival. In conclusion, we identified a novel prognostic signature and nomogram based on SARS-CoV-2–related genes as reliable prognostic predictors for KIRC patients and provided potential therapeutic targets for KIRC and COVID-19.
Collapse
Affiliation(s)
- Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yali Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xianglin Yuan,
| |
Collapse
|
16
|
Brock M, Bahammam S, Sima C. The Relationships Among Periodontitis, Pneumonia and COVID-19. FRONTIERS IN ORAL HEALTH 2022; 2:801815. [PMID: 35128525 PMCID: PMC8813972 DOI: 10.3389/froh.2021.801815] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022] Open
Abstract
Periodontitis is a chronic inflammatory disease of the supporting structures of the teeth that affects approximately half of adults 30 years and older. There is increasing interest in the direct and indirect relationships between periodontitis and systemic diseases, including respiratory diseases. The aim of this study was to assess the evidence on links among periodontitis, pneumonia, and COVID-19. Oral and periodontal bacteria may be linked to respiratory disease directly by aspiration of pathogens into the lungs causing pneumonia. As SARS-CoV-2 began to spread worldwide in 2020, questions have arisen of how periodontal disease may also be connected to SARS-CoV-2 infection and severity, including potential replication and dissemination of the virus from periodontal pockets. Some proposed mechanisms include the oral cavity acting as a reservoir or point of entry for SARS-CoV-2, overgrowth of periodontal pathogens, and increased production of proinflammatory cytokines. Due to potential links between periodontal disease and respiratory infections like pneumonia and SARS-CoV-2, oral hygiene and management of periodontitis remain essential to help reduce infection and transmission of SARS-CoV-2.
Collapse
Affiliation(s)
- Mikaela Brock
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, United States
| | - Shaima Bahammam
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, United States
- Department of Dentistry, King Fasial Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Corneliu Sima
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, United States
- *Correspondence: Corneliu Sima
| |
Collapse
|
17
|
Zhang R, Chen X, Zuo W, Ji Z, Qu Y, Su Y, Yang M, Zuo P, Ma G, Li Y. Inflammatory activation and immune cell infiltration are main biological characteristics of SARS-CoV-2 infected myocardium. Bioengineered 2022; 13:2486-2497. [PMID: 35037831 PMCID: PMC8974226 DOI: 10.1080/21655979.2021.2014621] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) can target cardiomyocytes (CMs) to directly invade the heart resulting in high mortality. This study aims to explore the biological characteristics of SARS-CoV-2 infected myocardium based on omics by collecting transcriptome data and analyzing them with a series of bioinformatics tools. Totally, 86 differentially expressed genes (DEGs) were discovered in SARS-CoV-2 infected CMs, and 15 miRNAs were discovered to target 60 genes. Functional enrichment analysis indicated that these DEGs were mainly enriched in the inflammatory signaling pathway. After the protein-protein interaction (PPI) network was constructed, several genes including CCL2 and CXCL8 were regarded as the hub genes. SRC inhibitor saracatinib was predicted to potentially act against the cardiac dysfunction induced by SARS-CoV-2. Among the 86 DEGs, 28 were validated to be dysregulated in SARS-CoV-2 infected hearts. Gene Set Enrichment Analysis (GSEA) analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) showed that malaria, IL-17 signaling pathway, and complement and coagulation cascades were significantly enriched. Immune infiltration analysis indicated that ‘naive B cells’ was significantly increased in the SARS-CoV-2 infected heart. The above results may help to improve the prognosis of patients with COVID-19.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Xi Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Wenjie Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Yangyang Qu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Yamin Su
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Mingming Yang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Pengfei Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Yongjun Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| |
Collapse
|
18
|
Shchetinin E, Baturin V, Arushanyan E, Bolatchiev A, Bobryshev D. Potential and Possible Therapeutic Effects of Melatonin on SARS-CoV-2 Infection. Antioxidants (Basel) 2022; 11:140. [PMID: 35052644 PMCID: PMC8772978 DOI: 10.3390/antiox11010140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
The absence of effective drugs for COVID-19 prevention and treatment requires the search for new candidates among approved medicines. Fundamental studies and clinical observations allow us to approach an understanding of the mechanisms of damage and protection from exposure to SARS-CoV-2, to identify possible points of application for pharmacological interventions. In this review we presented studies on the anti-inflammatory, antioxidant, and immunotropic properties of melatonin. We have attempted to present scientifically proven mechanisms of action for the potential therapeutic use of melatonin during SARS-CoV-2 infection. A wide range of pharmacological properties allows its inclusion as an effective addition to the methods of prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Evgeny Shchetinin
- Department of Pathophysiology, Stavropol State Medical University, 355000 Stavropol, Russia
| | - Vladimir Baturin
- Department of Clinical Pharmacology, Stavropol State Medical University, 355000 Stavropol, Russia
| | - Eduard Arushanyan
- Department of Pharmacology, Stavropol State Medical University, 355000 Stavropol, Russia
| | - Albert Bolatchiev
- Department of Clinical Pharmacology, Stavropol State Medical University, 355000 Stavropol, Russia
| | - Dmitriy Bobryshev
- Center of Personalized Medicine, Stavropol State Medical University, 355000 Stavropol, Russia
| |
Collapse
|
19
|
Ernzen K, Trask AJ, Peeples ME, Garg V, Zhao MT. Human Stem Cell Models of SARS-CoV-2 Infection in the Cardiovascular System. Stem Cell Rev Rep 2021; 17:2107-2119. [PMID: 34365591 PMCID: PMC8349465 DOI: 10.1007/s12015-021-10229-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 11/28/2022]
Abstract
The virus responsible for coronavirus disease 2019 (COVID-19), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected over 190 million people to date, causing a global pandemic. SARS-CoV-2 relies on binding of its spike glycoprotein to angiotensin-converting enzyme 2 (ACE2) for infection. In addition to fever, cough, and shortness of breath, severe cases of SARS-CoV-2 infection may result in the rapid overproduction of pro-inflammatory cytokines. This overactive immune response is known as a cytokine storm, which leads to several serious clinical manifestations such as acute respiratory distress syndrome and myocardial injury. Cardiovascular disorders such as acute coronary syndrome (ACS) and heart failure not only enhance disease progression at the onset of infection, but also arise in hospitalized patients with COVID-19. Tissue-specific differentiated cells and organoids derived from human pluripotent stem cells (hPSCs) serve as an excellent model to address how SARS-CoV-2 damages the lungs and the heart. In this review, we summarize the molecular basis of SARS-CoV-2 infection and the current clinical perspectives of the bidirectional relationship between the cardiovascular system and viral progression. Furthermore, we also address the utility of hPSCs as a dynamic model for SARS-CoV-2 research and clinical translation.
Collapse
Affiliation(s)
- Kyle Ernzen
- Center for Cardiovascular Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
- MCDB Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Aaron J Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mark E Peeples
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Center for Vaccine and Immunity, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Vidu Garg
- Center for Cardiovascular Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
- MCDB Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ming-Tao Zhao
- Center for Cardiovascular Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA.
- MCDB Graduate Program, The Ohio State University, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
20
|
Mohapatra RK, Dhama K, El-Arabey AA, Sarangi AK, Tiwari R, Emran TB, Azam M, Al-Resayes SI, Raval MK, Seidel V, Abdalla M. Repurposing benzimidazole and benzothiazole derivatives as potential inhibitors of SARS-CoV-2: DFT, QSAR, molecular docking, molecular dynamics simulation, and in-silico pharmacokinetic and toxicity studies. JOURNAL OF KING SAUD UNIVERSITY. SCIENCE 2021; 33:101637. [PMID: 34642560 PMCID: PMC8496942 DOI: 10.1016/j.jksus.2021.101637] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 05/20/2023]
Abstract
Density Functional Theory (DFT) and Quantitative Structure-Activity Relationship (QSAR) studies were performed on four benzimidazoles (compounds 1-4) and two benzothiazoles (compounds 5 and 6), previously synthesized by our group. The compounds were also investigated for their binding affinity and interactions with the SARS-CoV-2 Mpro (PDB ID: 6LU7) and the human angiotensin-converting enzyme 2 (ACE2) receptor (PDB ID: 6 M18) using a molecular docking approach. Compounds 1, 2, and 3 were found to bind with equal affinity to both targets. Compound 1 showed the highest predictive docking scores, and was further subjected to molecular dynamics (MD) simulation to explain protein stability, ligand properties, and protein-ligand interactions. All compounds were assessed for their structural, physico-chemical, pharmacokinetic, and toxicological properties. Our results suggest that the investigated compounds are potential new drug leads to target SARS-CoV-2.
Collapse
Affiliation(s)
- Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, Odisha 758002, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Amr Ahmed El-Arabey
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ashish K Sarangi
- Department of Chemistry, School of Applied Sciences, Centurion University of Technology and Management, Odisha, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura 281001, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Mohammad Azam
- Department of Chemistry, College of Science, King Saud University, PO BOX 2455, Riyadh 11451, Saudi Arabia
| | - Saud I Al-Resayes
- Department of Chemistry, College of Science, King Saud University, PO BOX 2455, Riyadh 11451, Saudi Arabia
| | - Mukesh K Raval
- Department of Chemistry, G. M. University, Sambalpur, Odisha, India
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Mohnad Abdalla
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Shandong Province 250012, PR China
| |
Collapse
|
21
|
Butler AE, Moin ASM, Sathyapalan T, Atkin SL. Vitamin D association with the renin angiotensin system in polycystic ovary syndrome. J Steroid Biochem Mol Biol 2021; 214:105965. [PMID: 34619249 DOI: 10.1016/j.jsbmb.2021.105965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
Vitamin D deficiency is a negative endocrine renin-angiotensin system (RAS) modulator and PCOS women are often vitamin D deficient, leading to RAS overactivation in PCOS. A cross-sectional study was performed in 99 PCOS and 68 control women who presented sequentially. Circulating plasma levels of RAS proteins (Angiotensin-converting enzyme 2 (ACE2), renin and angiotensinogen) were measured by Slow Off-rate Modified Aptamer (SOMA)-scan and 25-hydroxyvitamin D [25(OH)D] was measured by tandem mass spectroscopy. The RAS system was found to be overactivated in the PCOS women compared to non-PCOS control women with increased renin and decreased angiotensinogen (p < 0.05); 25-hydroxyvitamin D was also significantly lower in the PCOS group (p < 0.0001). In PCOS women, plasma renin was increased in vitamin D deficient and insufficient groups compared with the vitamin D sufficient group (p < 0.005), but did not differ across non-PCOS control subgroups. In non-PCOS controls, plasma ACE2 decreased from vitamin D insufficiency to deficiency (p < 0.05). Angiotensinogen was not different across the vitamin D sufficiency, insufficiency and deficiency strata for either PCOS or non-PCOS controls. These data show that RAS activation through increased plasma renin levels was seen in vitamin D insufficient and deficient PCOS subjects compared to non-PCOS control women. In addition, decreased plasma ACE2 levels were seen in vitamin D deficiency in non-PCOS controls, which may predispose these vitamin D deficient subjects to increased cardiovascular risk and susceptibility to infectious agents such as COVID-19 where this is a risk factor.
Collapse
Affiliation(s)
| | | | - Thozhukat Sathyapalan
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, UK.
| | - Stephen L Atkin
- Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain.
| |
Collapse
|
22
|
Awwad I, Greuel S, Tacke F, Fischer C. Haemorrhagic ulcerative duodenitis in a patient with COVID-19 infection: clinical improvement following treatment with budesonide. BMJ Open Gastroenterol 2021; 8:bmjgast-2021-000757. [PMID: 34663581 PMCID: PMC8523959 DOI: 10.1136/bmjgast-2021-000757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
We present a case of a male patient in his mid-30s with COVID-19-induced lung failure requiring extracorporeal membrane oxygenation, who needed an emergency oesophagogastroduodenoscopy due to major upper gastrointestinal bleeding. Endoscopy exposed severe ulcerative duodenitis with diffuse mucosal bleeding. While CT angiography did not show any signs of ischaemia, histopathology revealed duodenitis with substantial inflammatory cell infiltrates consisting of neutrophils and CD3+ T lymphocytes with equal CD4+/CD8+ distribution. Since the composition of cell infiltrates coincides with changes in inflammatory patterns of the respiratory mucosa from patients with COVID-19 and in COVID-19-associated enterocolitis, and systemic dexamethasone treatment became standard of care in ventilated intensive care unit patients with COVID-19 infection, we initiated an individualised therapeutic attempt to treat the duodenitis with topical enteral budesonide. Follow-up oesophagogastroduodenoscopies within 4 weeks of enteral budesonide administration revealed a full clinical and histological healing of the duodenal mucosa with marked reduction of neutrophilic and lymphocytic infiltrates. To our knowledge, the current report is the first description of enteral budesonide treatment of duodenitis in a patient with COVID-19 infection and warrants further investigation, whether budesonide might constitute a novel therapeutic strategy for the management of COVID-19-related intestinal mucosal damage.
Collapse
Affiliation(s)
- Ibrahim Awwad
- Department of Gastroenterology and Hepatology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Selina Greuel
- Department of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Gastroenterology and Hepatology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Fischer
- Department of Gastroenterology and Hepatology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
23
|
Motavalli R, Abdelbasset WK, Rahman HS, Achmad MH, Sergeevna NK, Zekiy AO, Adili A, Khiavi FM, Marofi F, Yousefi M, Ghoreishizadeh S, Shomali N, Etemadi J, Jarahian M. The lethal internal face of the coronaviruses: Kidney tropism of the SARS, MERS, and COVID19 viruses. IUBMB Life 2021; 73:1005-1015. [PMID: 34118117 PMCID: PMC8426673 DOI: 10.1002/iub.2516] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 01/08/2023]
Abstract
The kidney is one of the main targets attacked by viruses in patients with a coronavirus infection. Until now, SARS-CoV-2 has been identified as the seventh member of the coronavirus family capable of infecting humans. In the past two decades, humankind has experienced outbreaks triggered by two other extremely infective members of the coronavirus family; the MERS-CoV and the SARS-CoV. According to several investigations, SARS-CoV causes proteinuria and renal impairment or failure. The SARS-CoV was identified in the distal convoluted tubules of the kidney of infected patients. Also, renal dysfunction was observed in numerous cases of MERS-CoV infection. And recently, during the 2019-nCoV pandemic, it was found that the novel coronavirus not only induces acute respiratory distress syndrome (ARDS) but also can induce damages in various organs including the liver, heart, and kidney. The kidney tissue and its cells are targeted massively by the coronaviruses due to the abundant presence of ACE2 and Dpp4 receptors on kidney cells. These receptors are characterized as the main route of coronavirus entry to the victim cells. Renal failure due to massive viral invasion can lead to undesirable complications and enhanced mortality rate, thus more attention should be paid to the pathology of coronaviruses in the kidney. Here, we have provided the most recent knowledge on the coronaviruses (SARS, MERS, and COVID19) pathology and the mechanisms of their impact on the kidney tissue and functions.
Collapse
Affiliation(s)
- Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical SciencesTabrizIran
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation SciencesCollege of Applied Medical Sciences, Prince Sattam bin Abdulaziz UniversityAl KharjSaudi Arabia
- Department of Physical TherapyKasr Al‐Aini Hospital, Cairo UniversityGizaEgypt
| | | | - Muhammad Harun Achmad
- Department of Pediatric DentistryFaculty of Dentistry, Hasanuddin UniversityMakassarIndonesia
| | | | | | - Ali Adili
- Department of oncologyTabriz University of Medical SciencesTabrizIran
| | | | - Faroogh Marofi
- Department of Immunology, Division of Hematology, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mehdi Yousefi
- Department of Immunology, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| | | | - Navid Shomali
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Immunology, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Jalal Etemadi
- Kidney Research Center, Tabriz University of Medical SciencesTabrizIran
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
24
|
Yalcin HC, Sukumaran V, Al-Ruweidi MKAA, Shurbaji S. Do Changes in ACE-2 Expression Affect SARS-CoV-2 Virulence and Related Complications: A Closer Look into Membrane-Bound and Soluble Forms. Int J Mol Sci 2021; 22:6703. [PMID: 34201415 PMCID: PMC8269184 DOI: 10.3390/ijms22136703] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The SARS-CoV-2 virus utilizes angiotensin converting enzyme (ACE-2) for cell entry and infection. This enzyme has important functions in the renin-angiotensin aldosterone system to preserve cardiovascular function. In addition to the heart, it is expressed in many tissues including the lung, intestines, brain, and kidney, however, its functions in these organs are mostly unknown. ACE-2 has membrane-bound and soluble forms. Its expression levels are altered in disease states and by a variety of medications. Currently, it is not clear how altered ACE-2 levels influence ACE-2 virulence and relevant complications. In addition, membrane-bound and soluble forms are thought to have different effects. Most work on this topic in the literature is on the SARS-CoV virus that has a high genetic resemblance to SARS-Co-V-2 and also uses ACE-2 enzyme to enter the cell, but with much lower affinity. More recent studies on SARS-CoV-2 are mainly clinical studies aiming at relating the effect of medications that are thought to influence ACE-2 levels, with COVID-19 outcomes for patients under these medications. This review paper aims to summarize what is known about the relationship between ACE-2 levels and SARS-CoV/SARS-CoV-2 virulence under altered ACE-2 expression states.
Collapse
Affiliation(s)
- Huseyin C. Yalcin
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (M.K.A.A.A.-R.); (S.S.)
| | - Vijayakumar Sukumaran
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (M.K.A.A.A.-R.); (S.S.)
| | - Mahmoud Khatib A. A. Al-Ruweidi
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (M.K.A.A.A.-R.); (S.S.)
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| | - Samar Shurbaji
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (M.K.A.A.A.-R.); (S.S.)
| |
Collapse
|
25
|
Hristov D, Rijal H, Gomez-Marquez J, Hamad-Schifferli K. Developing a Paper-Based Antigen Assay to Differentiate between Coronaviruses and SARS-CoV-2 Spike Variants. Anal Chem 2021; 93:7825-7832. [PMID: 34037382 PMCID: PMC8171108 DOI: 10.1021/acs.analchem.0c05438] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/14/2021] [Indexed: 02/07/2023]
Abstract
COVID-19 first appeared in December of 2019 in Wuhan, China. Since then, it has become a global pandemic. A robust and scalable diagnostics strategy is crucial for containing and monitoring the pandemic. RT-PCR is a known, reliable method for COVID-19 diagnostics, which can differentiate between SARS-CoV-2 and other viruses. However, PCR is location-dependent, time-consuming, and relatively expensive. Thus, there is a need for a more flexible method, which may be produced in an off-the-shelf format and distributed more widely. Paper-based immunoassays can fulfill this function. Here, we present the first steps toward a paper-based test, which can differentiate between different spike proteins of various coronaviruses, SARS-CoV-1, SARS-CoV-2, and CoV-HKU1, with negligible cross-reactivity for HCoV-OC43 and HCoV-229E in a single assay, which takes less than 30 min. Furthermore, our test can distinguish between fractions of the same spike protein. This is done by an altered assay design with four test line locations where each antigen builds a unique, identifiable binding pattern. The effect of several factors, such as running media, immunoprobe concentration, and antigen interference, is considered. We find that running media has a significant effect on the final binding pattern where human saliva provides results while human serum leads to the lowest signal quality.
Collapse
Affiliation(s)
- Delyan Hristov
- Department
of Engineering, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| | - Hom Rijal
- Department
of Chemistry, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| | - Jose Gomez-Marquez
- Little
Devices Lab, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Kimberly Hamad-Schifferli
- Department
of Engineering, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
- School
for the Environment, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| |
Collapse
|
26
|
Fakhri S, Nouri Z, Moradi SZ, Akkol EK, Piri S, Sobarzo-Sánchez E, Farzaei MH, Echeverría J. Targeting Multiple Signal Transduction Pathways of SARS-CoV-2: Approaches to COVID-19 Therapeutic Candidates. Molecules 2021; 26:2917. [PMID: 34068970 PMCID: PMC8156180 DOI: 10.3390/molecules26102917] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Due to the complicated pathogenic pathways of coronavirus disease 2019 (COVID-19), related medicinal therapies have remained a clinical challenge. COVID-19 highlights the urgent need to develop mechanistic pathogenic pathways and effective agents for preventing/treating future epidemics. As a result, the destructive pathways of COVID-19 are in the line with clinical symptoms induced by severe acute coronary syndrome (SARS), including lung failure and pneumonia. Accordingly, revealing the exact signaling pathways, including inflammation, oxidative stress, apoptosis, and autophagy, as well as relative representative mediators such as tumor necrosis factor-α (TNF-α), nuclear factor erythroid 2-related factor 2 (Nrf2), Bax/caspases, and Beclin/LC3, respectively, will pave the road for combating COVID-19. Prevailing host factors and multiple steps of SARS-CoV-2 attachment/entry, replication, and assembly/release would be hopeful strategies against COVID-19. This is a comprehensive review of the destructive signaling pathways and host-pathogen interaction of SARS-CoV-2, as well as related therapeutic targets and treatment strategies, including potential natural products-based candidates.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; (S.F.); (S.Z.M.); (S.P.)
| | - Zeinab Nouri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; (S.F.); (S.Z.M.); (S.P.)
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler, Ankara 06330, Turkey;
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; (S.F.); (S.Z.M.); (S.P.)
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9170022, Chile
| |
Collapse
|
27
|
Yarmohammadi A, Yarmohammadi M, Fakhri S, Khan H. Targeting pivotal inflammatory pathways in COVID-19: A mechanistic review. Eur J Pharmacol 2021; 890:173620. [PMID: 33038418 PMCID: PMC7539138 DOI: 10.1016/j.ejphar.2020.173620] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/15/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022]
Abstract
As an emerging global health crisis, coronavirus disease 2019 (COVID-19) has been labeled a worldwide pandemic. Growing evidence is revealing further pathophysiological mechanisms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Amongst these dysregulated pathways inflammation seems to play a more critical role toward COVID-19 complications. In the present study, precise inflammatory pathways triggered by SARS-CoV-2, along with potential therapeutic candidates have been discussed. Prevailing evidence has indicated a close correlation of inflammatory cascades with severity, pathological progression, and organ damages in COVID-19 patients. From the mechanistic point of view, interleukin-6, interleukin-1β receptor, interferon-gamma, tumor necrosis factor-alpha receptor, toll-like receptor, receptor tyrosine kinases, growth factor receptor, Janus kinase/signal transducers and transcription pathway, mammalian target of rapamycin, cytokine storm and macrophage activation have shown to play critical roles in COVID-19 complications. So, there is an urgent need to provide novel mechanistic-based anti-inflammatory agents. This review highlights inflammatory signaling pathways of SARS-CoV-2. Several therapeutic targets and treatment strategies have also been provided in an attempt to tackle COVID-19 complications.
Collapse
Affiliation(s)
- Akram Yarmohammadi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Mostafa Yarmohammadi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
28
|
Majnooni MB, Fakhri S, Shokoohinia Y, Kiyani N, Stage K, Mohammadi P, Gravandi MM, Farzaei MH, Echeverría J. Phytochemicals: Potential Therapeutic Interventions Against Coronavirus-Associated Lung Injury. Front Pharmacol 2020; 11:588467. [PMID: 33658931 PMCID: PMC7919380 DOI: 10.3389/fphar.2020.588467] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19) in December 2019, millions of people have been infected and died worldwide. However, no drug has been approved for the treatment of this disease and its complications, which urges the need for finding novel therapeutic agents to combat. Among the complications due to COVID-19, lung injury has attained special attention. Besides, phytochemicals have shown prominent anti-inflammatory effects and thus possess significant effects in reducing lung injury caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Also, the prevailing evidence reveales the antiviral effects of those phytochemicals, including anti-SARS-CoV activity, which could pave the road in providing suitable lead compounds in the treatment of COVID-19. In the present study, candidate phytochemicals and related mechanisms of action have been shown in the treatment/protection of lung injuries induced by various methods. In terms of pharmacological mechanism, phytochemicals have shown potential inhibitory effects on inflammatory and oxidative pathways/mediators, involved in the pathogenesis of lung injury during COVID-19 infection. Also, a brief overview of phytochemicals with anti-SARS-CoV-2 compounds has been presented.
Collapse
Affiliation(s)
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yalda Shokoohinia
- Pharmaceutical Sciences Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Ric Scalzo Botanical Research Institute, Southwest College of Naturopathic Medicine, Tempe, AZ, United States
| | - Narges Kiyani
- Ric Scalzo Botanical Research Institute, Southwest College of Naturopathic Medicine, Tempe, AZ, United States
| | - Katrina Stage
- Ric Scalzo Botanical Research Institute, Southwest College of Naturopathic Medicine, Tempe, AZ, United States
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javier Echeverría
- Departamento De Ciencias Del Ambiente, Facultad De Química y Biología, Universidad De Santiago De Chile, Santiago, Chile
| |
Collapse
|