51
|
Sha A, Liu Y, Zhao X. SARS-CoV-2 and gastrointestinal diseases. Front Microbiol 2023; 14:1177741. [PMID: 37323898 PMCID: PMC10267706 DOI: 10.3389/fmicb.2023.1177741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/21/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative agent of the novel coronavirus disease (COVID-19) pandemic, which has caused serious challenges for public health systems worldwide. LITERATURE REVIEW SARS-CoV-2 invades not only the respiratory system, but also the digestive system, causing a variety of gastrointestinal diseases. SIGNIFICANCE Understanding the gastrointestinal diseases caused by SARS-CoV-2, and the damage mechanisms of SARS-CoV-2 to the gastrointestinal tracts and gastrointestinal glands are crucial to treating the gastrointestinal diseases caused by SARS-CoV-2. CONCLUSION This review summarizes the gastrointestinal diseases caused by SARS-CoV-2, including gastrointestinal inflammatory disorders, gastrointestinal ulcer diseases, gastrointestinal bleeding, and gastrointestinal thrombotic diseases, etc. Furthermore, the mechanisms of gastrointestinal injury induced by SARS-COV-2 were analyzed and summarized, and the suggestions for drug prevention and treatment were put forward for the reference of clinical workers.
Collapse
Affiliation(s)
- Ailong Sha
- School of Teacher Education, Chongqing Three Gorges University, Chongqing, China
- School of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Yi Liu
- School of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Xuewen Zhao
- School of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| |
Collapse
|
52
|
Chen Y, Zhang C, Wang N, Feng Y. Deciphering suppressive effects of Lianhua Qingwen Capsule on COVID-19 and synergistic effects of its major botanical drug pairs. Chin J Nat Med 2023; 21:383-400. [PMID: 37245876 PMCID: PMC10214843 DOI: 10.1016/s1875-5364(23)60455-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Indexed: 05/30/2023]
Abstract
The COVID-19 pandemic has resulted in excess deaths worldwide. Conventional antiviral medicines have been used to relieve the symptoms, with limited therapeutic effect. In contrast, Lianhua Qingwen Capsule is reported to exert remarkable anti-COVID-19 effect. The current review aims to: 1) uncover the main pharmacological actions of Lianhua Qingwen Capsule for managing COVID-19; 2) verify the bioactive ingredients and pharmacological actions of Lianhua Qingwen Capsule by network analysis; 3) investigate the compatibility effect of major botanical drug pairs in Lianhua Qingwen Capsule; and 4) clarify the clinical evidence and safety of the combined therapy of Lianhua Qingwen Capsule and conventional drugs. Numerous bioactive ingredients in Lianhu Qingwen, such as quercetin, naringenin, β-sitosterol, luteolin, and stigmasterol, were identified to target host cytokines, and to regulate the immune defence in response to COVID-19. Genes including androgen receptor (AR), myeloperoxidase (MPO), epidermal growth factor receptor (EGFR), insulin (INS), and aryl hydrocarbon receptor (AHR) were found to be significantly involved in the pharmacological actions of Lianhua Qingwen Capsule against COVID-19. Four botanical drug pairs in Lianhua Qingwen Capsule were shown to have synergistic effect for the treatment of COVID-19. Clinical studies demonstrated the medicinal effect of the combined use of Lianhua Qingwen Capsule and conventional drugs against COVID-19. In conclusion, the four main pharmacological mechanisms of Lianhua Qingwen Capsule for managing COVID-19 are revealed. Therapeutic effect has been noted against COVID-19 in Lianhua Qingwen Capsule.
Collapse
Affiliation(s)
- Yuanyuan Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China.
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
53
|
Lala M, Bhattacharjee S, Ghosh C, Sen A, Sarkar I. In-silico studies on wild orange ( Citrus macroptera Mont.) compounds against COVID-19 pro-inflammation targets. J Biomol Struct Dyn 2023; 41:3511-3523. [PMID: 35297321 DOI: 10.1080/07391102.2022.2051744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 03/05/2022] [Indexed: 12/14/2022]
Abstract
One-fifth of COVID-19 patients suffer a severe course of COVID-19 (SARS-CoV-2) infection; however, the specific causes remain unclear. Despite numerous papers that have been flooded in different scientific journals clear clinical picture of COVID-19 aftermath persists to remain fuzzy. The survivors of severe COVID-19infection having defeated the virus are just the starting of an uncharted recovery path. Currently, there is no drug available that is safe to consume to combat this pandemic. However, researchers still struggling to find specific therapeutic solutions. The present study employed an in silico approach to assessing the inhibitory potential of the phytochemicals obtained from GC-MS analysis of Citrus macroptera against inflammatory proteins like COX-2, NMDAR and VCAM-1 which remains in a hyperactive state even after a patient is fully cured of this deadly mRNA virus. An extensive molecular docking investigation of the phyto-compounds at the active binding pockets of the inflammatory proteins revealed the promising inhibitory potential of the phytochemicals. Reasonable physicochemical attributes of the compounds following Lipinski's rule of five, VEBER and PAINS analysis further established them as potential therapeutic candidates against aforesaid inflammatory proteins. MM-GBSA binding free energy estimation revealed that Limonene was the most promising candidate displaying the highest binding efficacy with the concerned VCAM-1 protein included in the present analysis. An interesting finding is the phytochemicals exhibited better binding energy scores with the concerned COX-2, VCAM-1 and NMDA receptor proteins than the conventional drugs that are specifically targeted against them. Our in silico results suggest that all the natural phyto-compounds derived from C. macroptera could be employed in Post covid inflammation complexities after appropriate pre-clinical and clinical trials for further scientific validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mousikha Lala
- Department of Botany, University of North Bengal, Siliguri, India
| | - Soumita Bhattacharjee
- Department of Botany, University of North Bengal, Siliguri, India
- Department of Tea Science, University of North Bengal, Siliguri, India
| | - Chandra Ghosh
- Department of Tea Science, University of North Bengal, Siliguri, India
| | - Arnab Sen
- Department of Botany, University of North Bengal, Siliguri, India
- Biswa Bangla GenomeCentre, University of North Bengal, Siliguri, India
| | - Indrani Sarkar
- Department of Botany, University of North Bengal, Siliguri, India
| |
Collapse
|
54
|
Elbadawy HM, Khattab A, El-Agamy DS, Eltahir HM, Alhaddad A, Aljohani FD, Almuzaini TM, Abouzied MM, Aldhafiri A. IL-6 at the center of cytokine storm: Circulating inflammation mediators as biomarkers in hospitalized COVID-19 patients. J Clin Lab Anal 2023; 37:e24881. [PMID: 37096731 DOI: 10.1002/jcla.24881] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/11/2023] [Accepted: 03/26/2023] [Indexed: 04/26/2023] Open
Abstract
INTRODUCTION The management of hospitalized COVID-19 patients depends largely on controlling the intensified inflammatory response known as the cytokine storm. Candidate inflammatory cytokines can serve as new biomarkers for the management of hospitalized COVID-19 patients. METHODS Patients (80) were recruited into three groups: room air (RA), oxygen (OX) and mechanical ventilator (MV). Blood analysis was performed for RBC, WBC, Hb, Platelets, serum albumin and creatinine, INR, PTT, and hematocrit. ELISA was used to quantify a panel of inflammatory mediators including GM-SCF, IFN-α, IFNγ, IL-1β, IL-1R, IL-2, IL-2Ra, IL-6, IL-8, IL-10, IL-12p70, IL-13, MCP-1, MIP-1a, and TNF-α. Correlations between laboratory results and the levels of circulating inflammation mediators were investigated. RESULTS Patients on MV had low RBC, Hb, albumin, and HCT and high WBC count, PTT, and INR when compared to RA and OX groups. A statistical positive correlation was found between WBC and the levels of IL-6 and MCP-1. RBCs correlated negatively with IL-6 and IL-10 and positively with IL-8. Higher TNF-α correlated with lower platelet counts while higher levels of IL-1Rα and IL-10 were associated with lower Hb levels. Increases in IFN-γ and TNF-α were indicative of compromised kidney functions as creatinine levels increased significantly. Most significant correlations were found between IL-6 and lab results, showing positive correlation with WBC and INR, and negative correlation with RBC, albumin, and HCT. CONCLUSIONS Having the most significant correlations, IL-6 high levels in mechanically ventilated patients were shown to affect laboratory results, and, therefore, is suggested as a severity biomarker of COVID-19.
Collapse
Affiliation(s)
- Hossein M Elbadawy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | | | - Dina S El-Agamy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Heba M Eltahir
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Aisha Alhaddad
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | | | | | - Mekky M Abouzied
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Ahmed Aldhafiri
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
55
|
Niidome S, Usui Y, Tsubota K, Sugawara R, Goto H. COVID-19 Developed During Systemic Steroid Therapy for Vogt-Koyanagi-Harada Disease: A Case Report. Ocul Immunol Inflamm 2023:1-5. [PMID: 37084286 DOI: 10.1080/09273948.2023.2200488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
PURPOSE The use of immunomodulatory therapy in the setting of coexistence of uveitis and coronavirus disease (COVID-19) remains controversial. We report a case of COVID-19 during systemic steroid therapy for Vogt-Koyanagi-Harada disease (VKH). CASE REPORT A 43-year-old female was diagnosed with VKH and started on steroid pulse therapy (1,000 mg/day) followed by high-dose oral corticosteroids. Two weeks after discharge from the hospital, she was readmitted to the intensive care unit with severe acute respiratory syndrome due to SARS-CoV-2 infection confirmed by PCR test, and fortunately both VKH and COVID-19-induced respiratory disease improved. CONCLUSION Given the absence of international agreement on how to manage COVID-19 patients with steroid-dependent VKH, existing clinical guidelines should be reviewed thoroughly to formulate useful strategies for managing VKH patients on steroid treatment who contract COVID-19. Furthermore, the outcomes of patients with steroid-dependent autoimmune uveitis including VKH who develop COVID-19 should be analyzed.
Collapse
Affiliation(s)
| | - Yoshihiko Usui
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| | - Kinya Tsubota
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| | - Risa Sugawara
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Goto
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
56
|
Mohammed MA. Fighting cytokine storm and immunomodulatory deficiency: By using natural products therapy up to now. Front Pharmacol 2023; 14:1111329. [PMID: 37124230 PMCID: PMC10134036 DOI: 10.3389/fphar.2023.1111329] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/14/2023] [Indexed: 05/02/2023] Open
Abstract
A novel coronavirus strain (COVID-19) caused severe illness and mortality worldwide from 31 December 2019 to 21 March 2023. As of this writing, 761,071,826 million cases have been diagnosed worldwide, with 6,879,677 million deaths accorded by WHO organization and has spread to 228 countries. The number of deaths is closely connected to the growth of innate immune cells in the lungs, mainly macrophages, which generate inflammatory cytokines (especially IL-6 and IL-1β) that induce "cytokine storm syndrome" (CSS), multi-organ failure, and death. We focus on promising natural products and their biologically active chemical constituents as potential phytopharmaceuticals that target virus-induced pro-inflammatory cytokines. Successful therapy for this condition is currently rare, and the introduction of an effective vaccine might take months. Blocking viral entrance and replication and regulating humoral and cellular immunity in the uninfected population are the most often employed treatment approaches for viral infections. Unfortunately, no presently FDA-approved medicine can prevent or reduce SARS-CoV-2 access and reproduction. Until now, the most important element in disease severity has been the host's immune response activation or suppression. Several medicines have been adapted for COVID-19 patients, including arbidol, favipiravir, ribavirin, lopinavir, ritonavir, hydroxychloroquine, chloroquine, dexamethasone, and anti-inflammatory pharmaceutical drugs, such as tocilizumab, glucocorticoids, anakinra (IL-1β cytokine inhibition), and siltuximab (IL-6 cytokine inhibition). However, these synthetic medications and therapies have several side effects, including heart failure, permanent retinal damage in the case of hydroxyl-chloroquine, and liver destruction in the case of remdesivir. This review summarizes four strategies for fighting cytokine storms and immunomodulatory deficiency induced by COVID-19 using natural product therapy as a potential therapeutic measure to control cytokine storms.
Collapse
Affiliation(s)
- Mona A. Mohammed
- Medicinal and Aromatic Plants Research Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza, Egypt
| |
Collapse
|
57
|
Fu Y, Fang Y, Gong S, Xue T, Wang P, She L, Huang J. Deep learning-based network pharmacology for exploring the mechanism of licorice for the treatment of COVID-19. Sci Rep 2023; 13:5844. [PMID: 37037848 PMCID: PMC10086012 DOI: 10.1038/s41598-023-31380-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/10/2023] [Indexed: 04/12/2023] Open
Abstract
Licorice, a traditional Chinese medicine, has been widely used for the treatment of COVID-19, but all active compounds and corresponding targets are still not clear. Therefore, this study proposed a deep learning-based network pharmacology approach to identify more potential active compounds and targets of licorice. 4 compounds (quercetin, naringenin, liquiritigenin, and licoisoflavanone), 2 targets (SYK and JAK2) and the relevant pathways (P53, cAMP, and NF-kB) were predicted, which were confirmed by previous studies to be associated with SARS-CoV-2-infection. In addition, 2 new active compounds (glabrone and vestitol) and 2 new targets (PTEN and MAP3K8) were further validated by molecular docking and molecular dynamics simulations (simultaneous molecular dynamics), as well as the results showed that these active compounds bound well to COVID-19 related targets, including the main protease (Mpro), the spike protein (S-protein) and the angiotensin-converting enzyme 2 (ACE2). Overall, in this study, glabrone and vestitol from licorice were found to inhibit viral replication by inhibiting the activation of Mpro, S-protein and ACE2; related compounds in licorice may reduce the inflammatory response and inhibit apoptosis by acting on PTEN and MAP3K8. Therefore, licorice has been proposed as an effective candidate for the treatment of COVID-19 through PTEN, MAP3K8, Mpro, S-protein and ACE2.
Collapse
Affiliation(s)
- Yu Fu
- Alibaba Business School, Hangzhou Normal University, Hangzhou, 310000, China
| | - Yangyue Fang
- Alibaba Business School, Hangzhou Normal University, Hangzhou, 310000, China
| | - Shuai Gong
- Alibaba Business School, Hangzhou Normal University, Hangzhou, 310000, China
| | - Tao Xue
- Alibaba Business School, Hangzhou Normal University, Hangzhou, 310000, China
| | - Peng Wang
- Alibaba Business School, Hangzhou Normal University, Hangzhou, 310000, China
| | - Li She
- Alibaba Business School, Hangzhou Normal University, Hangzhou, 310000, China
| | - Jianping Huang
- Alibaba Business School, Hangzhou Normal University, Hangzhou, 310000, China.
| |
Collapse
|
58
|
Majumder R, Ghosh S, Singh MK, Das A, Roy Chowdhury S, Saha A, Saha RP. Revisiting the COVID-19 Pandemic: An Insight into Long-Term Post-COVID Complications and Repurposing of Drugs. COVID 2023; 3:494-519. [DOI: 10.3390/covid3040037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
SARS-CoV-2 is a highly contagious and dangerous coronavirus that has been spreading around the world since late December 2019. Severe COVID-19 has been observed to induce severe damage to the alveoli, and the slow loss of lung function led to the deaths of many patients. Scientists from all over the world are now saying that SARS-CoV-2 can spread through the air, which is a very frightening prospect for humans. Many scientists thought that this virus would evolve during the first wave of the pandemic and that the second wave of reinfection with the coronavirus would also be very dangerous. In late 2020 and early 2021, researchers found different genetic versions of the SARS-CoV-2 virus in many places around the world. Patients with different types of viruses had different symptoms. It is now evident from numerous case studies that many COVID-19 patients who are released from nursing homes or hospitals are more prone to developing multi-organ dysfunction than the general population. Understanding the pathophysiology of COVID-19 and its impact on various organ systems is crucial for developing effective treatment strategies and managing long-term health consequences. The case studies highlighted in this review provide valuable insights into the ongoing health concerns of individuals affected by COVID-19.
Collapse
Affiliation(s)
- Rajib Majumder
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Sanmitra Ghosh
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Manoj K. Singh
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Arpita Das
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Swagata Roy Chowdhury
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Abinit Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Rudra P. Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| |
Collapse
|
59
|
Lu Y, Xu S, Sun H, Shan J, Shen C, Ji J, Lin L, Xu J, Peng L, Dai C, Xie T. Analysis of temporal metabolic rewiring for human respiratory syncytial virus infection by integrating metabolomics and proteomics. Metabolomics 2023; 19:30. [PMID: 36991292 PMCID: PMC10057675 DOI: 10.1007/s11306-023-01991-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/05/2023] [Indexed: 03/31/2023]
Abstract
INTRODUCTION Human respiratory syncytial virus (HRSV) infection causes significant morbidity, and no effective treatments are currently available. Viral infections induce substantial metabolic changes in the infected cells to optimize viral production. Metabolites that reflect the interactions between host cells and viruses provided an opportunity to identify the pathways underlying severe infections. OBJECTIVE To better understand the metabolic changes caused by HRSV infection, we analyzed temporal metabolic profiling to provide novel targets for therapeutic strategies for inhaled HRSV infection. METHODS The epithelial cells and BALB/c mice were infected with HRSV. Protein and mRNA levels of inflammation factors were measured by using quantitative reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay. Untargeted metabolomics, lipidomics and proteomics were performed using liquid chromatography coupled with mass spectrometry to profile the metabolic phenotypic alterations in HRSV infection. RESULTS In this study, we evaluated the inflammatory responses in vivo and in vitro and investigated the temporal metabolic rewiring of HRSV infection in epithelial cells. We combined metabolomics and proteomic analyses to demonstrate that the redox imbalance was further provoked by increasing glycolysis and anaplerotic reactions. These responses created an oxidant-rich microenvironment that elevated reactive oxygen species levels and exacerbated glutathione consumption. CONCLUSION These observations indicate that adjusting for metabolic events during a viral infection could represent a valuable approach for reshaping the outcome of infections.
Collapse
Affiliation(s)
- Yao Lu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shan Xu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Huan Sun
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cunsi Shen
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lili Lin
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jianya Xu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Linxiu Peng
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Dai
- Experimental Teaching Center of Life Science, College of Life Sciences, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Tong Xie
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
60
|
Thymosin Alpha 1 Restores the Immune Homeostasis in lymphocytes during Post-Acute Sequelae of SARS-CoV-2 infection. Int Immunopharmacol 2023; 118:110055. [PMID: 36989892 PMCID: PMC10030336 DOI: 10.1016/j.intimp.2023.110055] [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: 01/24/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023]
Abstract
The complex alterations of the immune system and the immune-mediated multiorgan injury plays a key role in host response to SARS-CoV-2 infection and in the pathogenesis of COVID-19, being also associated with adverse outcomes. Thymosin alpha 1 (Tα1) is one of the molecules used in the treatment of COVID-19, as it is known to restore the homeostasis of the immune system during infections and cancer. The use of Tα1 in COVID-19 patients had been widely used in China and in COVID-19 patients, it has been shown to decrease hospitalization rate, especially in those with greater disease severity, and reduce mortality by restoring lymphocytopenia and more specifically, depleted T cells. Persistent dysregulation with depletion of naive B and T cell subpopulations and expansion of memory T cells suggest a chronic stimulation of the immune response in individuals with post-acute sequelae of SARS-CoV-2 infection (PASC). Our data obtained from an ex vivo study, showed that in PASC individuals with a chronically altered immune response, Tα1 improve the restoration of an appropriate response, most evident in those with more severe illness and who need respiratory support during acute phase, and in those with specific systemic and psychiatric symptoms of PASC, confirming Tα1 treatment being more effective in compromised patients. The results obtained, along with promising reports on recent trials on Tα1 administration in patients with COVID-19, offer new insights into intervention also for those patients with long-lasting inflammation with post-infectious symptoms, some of which have a delayed onset.
Collapse
Key Words
- post-acute sars-cov-2 symptoms
- thymosin alpha 1
- immune regulation
- anti-inflammatory response
- a-cov, acute covid-19
- aa, ambient air
- cdc, center for desease control and prevention
- em, effector memory
- tfh, follicular helper lymphocytes
- hd, healthy donors
- pasc, post-acute sequelae of sars-cov-2 infection
- pcc, post-covid conditions
- pd-1, programmed cell death-1
- ards, respiratory stress syndrome
- resp sup, respiratory support
- rpmi, roswell park memorial institute
- sev, severe acute phase of infection
- tem, terminal effector memory
- tα1, thymosin alpha 1
Collapse
|
61
|
Maria NI, Rapicavoli RV, Alaimo S, Bischof E, Stasuzzo A, Broek JA, Pulvirenti A, Mishra B, Duits AJ, Ferro A, RxCOVEA Framework. Application of the PHENotype SIMulator for rapid identification of potential candidates in effective COVID-19 drug repurposing. Heliyon 2023; 9:e14115. [PMID: 36911878 PMCID: PMC9986505 DOI: 10.1016/j.heliyon.2023.e14115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
The current, rapidly diversifying pandemic has accelerated the need for efficient and effective identification of potential drug candidates for COVID-19. Knowledge on host-immune response to SARS-CoV-2 infection, however, remains limited with few drugs approved to date. Viable strategies and tools are rapidly arising to address this, especially with repurposing of existing drugs offering significant promise. Here we introduce a systems biology tool, the PHENotype SIMulator, which -by leveraging available transcriptomic and proteomic databases-allows modeling of SARS-CoV-2 infection in host cells in silico to i) determine with high sensitivity and specificity (both>96%) the viral effects on cellular host-immune response, resulting in specific cellular SARS-CoV-2 signatures and ii) utilize these cell-specific signatures to identify promising repurposable therapeutics. Powered by this tool, coupled with domain expertise, we identify several potential COVID-19 drugs including methylprednisolone and metformin, and further discern key cellular SARS-CoV-2-affected pathways as potential druggable targets in COVID-19 pathogenesis.
Collapse
Key Words
- 2DG, 2-Deoxy-Glucose
- ACE2, Angiotensin-converting enzyme 2
- COVID-19
- COVID-19, Coronavirus disease 2019
- Caco-2, Human colon epithelial carcinoma cell line
- Calu-3, Epithelial cell line
- Cellular SARS-CoV-2 signatures
- Cellular host-immune response
- Cellular simulation models
- DEGs, Differentially Expressed Genes
- DEPs, Differentially expressed proteins
- Drug repurposing
- HCQ-CQ, (Hydroxy)chloroquine
- IFN, Interferon
- ISGs, IFN-stimulated genes
- MITHrIL, Mirna enrIched paTHway Impact anaLysis
- MOI, Multiplicity of infection
- MP, Methylprednisolone
- NHBE, Normal human bronchial epithelial cells
- PHENSIM, PHENotype SIMulator
- SARS-CoV-2, Severe acute respiratory syndrome coronavirus 2
- Systems biology
- TLR, Toll-like Receptor
Collapse
Affiliation(s)
- Naomi I. Maria
- Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health, Manhasset, NY, USA
- Red Cross Blood Bank Foundation Curaçao, Willemstad, Curaçao
- Department of Medical Microbiology and Immunology, St. Antonius Ziekenhuis, Niewegein, the Netherlands
- Corresponding author. Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA.
| | - Rosaria Valentina Rapicavoli
- Department of Physics and Astronomy, University of Catania, Italy
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Salvatore Alaimo
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Evelyne Bischof
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, Naples, Italy
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Pudong, Shanghai, China
- Insilico Medicine, Hong Kong Special Administrative Region, China
| | | | - Jantine A.C. Broek
- Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA
| | - Alfredo Pulvirenti
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Bud Mishra
- Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA
- Simon Center for Quantitative Biology, Cold Spring Harbor Lab, Long Island, USA
- Corresponding author. Courant Institute of Mathematical Sciences, Room 405, 251 Mercer Street, NY, USA.
| | - Ashley J. Duits
- Red Cross Blood Bank Foundation Curaçao, Willemstad, Curaçao
- Curaçao Biomedical Health Research Institute, Willemstad, Curaçao
- Institute for Medical Education, University Medical Center Groningen, Groningen, the Netherlands
| | - Alfredo Ferro
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | | |
Collapse
|
62
|
COVID-19 signalome: Potential therapeutic interventions. Cell Signal 2023; 103:110559. [PMID: 36521656 PMCID: PMC9744501 DOI: 10.1016/j.cellsig.2022.110559] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/21/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The COVID-19 pandemic has triggered intensive research and development of drugs and vaccines against SARS-CoV-2 during the last two years. The major success was especially observed with development of vaccines based on viral vectors, nucleic acids and whole viral particles, which have received emergent authorization leading to global mass vaccinations. Although the vaccine programs have made a big impact on COVID-19 spread and severity, emerging novel variants have raised serious concerns about vaccine efficacy. Due to the urgent demand, drug development had originally to rely on repurposing of antiviral drugs developed against other infectious diseases. For both drug and vaccine development the focus has been mainly on SARS-CoV-2 surface proteins and host cell receptors involved in viral attachment and entry. In this review, we expand the spectrum of SARS-CoV-2 targets by investigating the COVID-19 signalome. In addition to the SARS-CoV-2 Spike protein, the envelope, membrane, and nucleoprotein targets have been subjected to research. Moreover, viral proteases have presented the possibility to develop different strategies for the inhibition of SARS-CoV-2 replication and spread. Several signaling pathways involving the renin-angiotensin system, angiotensin-converting enzymes, immune pathways, hypoxia, and calcium signaling have provided attractive alternative targets for more efficient drug development.
Collapse
|
63
|
Hatta MHM, Matmin J, Malek NANN, Kamisan FH, Badruzzaman A, Batumalaie K, Ling Lee S, Abdul Wahab R. COVID‐19: Prevention, Detection, and Treatment by Using Carbon Nanotubes‐Based Materials. ChemistrySelect 2023. [DOI: 10.1002/slct.202204615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Affiliation(s)
- Mohd Hayrie Mohd Hatta
- Centre for Research and Development Asia Metropolitan University 81750 Johor Bahru Johor Malaysia
| | - Juan Matmin
- Department of Chemistry Faculty of Science Universiti Teknologi Malaysia 81310 UTM Johor Bahru Johor Malaysia
- Centre for Sustainable Nanomaterials Ibnu Sina Institute for Scientific and Industrial Research Universiti Teknologi Malaysia 81310 UTM Johor Bahru Johor Malaysia
| | - Nik Ahmad Nizam Nik Malek
- Centre for Sustainable Nanomaterials Ibnu Sina Institute for Scientific and Industrial Research Universiti Teknologi Malaysia 81310 UTM Johor Bahru Johor Malaysia
- Department of Biosciences, Faculty of Science Universiti Teknologi Malaysia 81310 UTM Johor Bahru Johor Malaysia
| | - Farah Hidayah Kamisan
- Department of Biomedical Sciences Faculty of Health Sciences Asia Metropolitan University 81750 Johor Bahru Johor Malaysia
| | - Aishah Badruzzaman
- Centre for Foundation, Language and General Studies Asia Metropolitan University 81750 Johor Bahru Johor Malaysia
| | - Kalaivani Batumalaie
- Department of Biomedical Sciences Faculty of Health Sciences Asia Metropolitan University 81750 Johor Bahru Johor Malaysia
| | - Siew Ling Lee
- Department of Chemistry Faculty of Science Universiti Teknologi Malaysia 81310 UTM Johor Bahru Johor Malaysia
- Centre for Sustainable Nanomaterials Ibnu Sina Institute for Scientific and Industrial Research Universiti Teknologi Malaysia 81310 UTM Johor Bahru Johor Malaysia
| | - Roswanira Abdul Wahab
- Department of Chemistry Faculty of Science Universiti Teknologi Malaysia 81310 UTM Johor Bahru Johor Malaysia
| |
Collapse
|
64
|
Arlegui H, Mahé E, Richard MA, De Rycke Y, Viguier M, Beylot-Barry M, Dupuy A, Beneton N, Joly P, Jullien D, Bachelez H, Sbidian É, Chosidow O, Paul C, Tubach F. Impact of the first wave of the COVID-19 pandemic on the treatment of psoriasis with systemic therapies in France: Results from the PSOBIOTEQ cohort. Ann Dermatol Venereol 2023:S0151-9638(23)00009-1. [PMID: 36914553 PMCID: PMC9928748 DOI: 10.1016/j.annder.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/18/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND The nature of the COVID-19 pandemic led to concerns among patients and physicians about the potential impact of immunosuppressive treatments for chronic diseases such as psoriasis on the risk of severe COVID-19. OBJECTIVES To describe treatment modifications and determine the incidence of COVID-19 infection among psoriasis patients during the first wave of the pandemic, and identify the factors associated with these events. METHODS Data from PSOBIOTEQ cohort relating to the first COVID-19 wave in France (March to June, 2020), as well as a patient-centred COVID-19 questionnaire, were used to evaluate the impact of lockdown on changes (discontinuations, delays or reductions) in systemic therapies, and to determine the incidence of COVID-19 cases among these patients. Logistic regression models were used to assess associated factors. RESULTS Among the 1751 respondents (89.3%), 282 patients (16.9%) changed their systemic treatment for psoriasis, with 46.0% of these changes being initiated by the patients themselves. Patients were more likely to experience psoriasis flare-ups during the first wave if they changed their treatment during this period (58.7% vs 14.4%; P < 0.0001). Changes to systemic therapies were less frequent among patients with cardiovascular diseases (P < 0.001), and those aged ≥ 65 years (P = 0.02). Overall, 45 patients (2.9%) reported having COVID-19, and eight (17.8%) required hospitalization. Risk factors for COVID-19 infection were close contact with a positive case (P < 0.001) and living in a region with a high incidence of COVID-19 (P < 0.001). Factors associated with a lower risk of COVID-19 were avoiding seeing a physician (P = 0.002), systematically wearing a mask during outings (P = 0.011) and being a current smoker (P = 0.046). CONCLUSIONS Discontinuation of systemic psoriasis treatments during the first COVID-19 wave (16.9%) - mainly decided by patients themselves (46.0%) - was associated with a higher incidence of disease flares (58.7% vs 14.4%). This observation and factors associated with a higher risk of COVID-19 highlight the need to maintain and adapt patient-physician communication during health crises according to patient profiles, with the aim of avoiding unnecessary treatment discontinuations and ensuring that patients are informed about the risk of infection and the importance of complying with hygiene rules.
Collapse
Affiliation(s)
- H Arlegui
- AP-HP, Hôpital Pitié-Salpêtrière, Centre de Pharmaco-épidémiologie (Cephépi), INSERM, CIC-1901, Paris, France
| | - E Mahé
- Service de Dermatologie, Hôpital Victor Dupouy, Argenteuil, France
| | - M-A Richard
- Service de Dermatologie, EA 3279: CEReSS - Health Service Research and Quality of Life Centre, Timone Hospital, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, Marseille, France
| | - Y De Rycke
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique, AP-HP, Hôpital Pitié-Salpêtrière, Département de Santé Publique, Centre de Pharmaco-épidémiologie (Cephépi), CIC-1901, 75013 Paris, France
| | - M Viguier
- Service de Dermatologie-Vénéréologie, Hôpital Robert Debré, Université Reims Champagne Ardenne, 51100 Reims, France
| | - M Beylot-Barry
- Service de Dermatologie, CHU de Bordeaux, Translational Research on Oncodermatology and Rare skin diseases, Bordeaux Institute of Oncology, UMR 1312 INSERM, Université Bordeaux, Bordeaux, France
| | - A Dupuy
- France Univ Rennes, CHU Rennes, 35000 Rennes, France
| | - N Beneton
- REPERES Pharmaco-Epidemiology and Health Services Research, University Rennes and French School of Public Health, Rennes, France
| | - P Joly
- Service de Dermatologie, Hôpital du Mans, Le Mans, France
| | - D Jullien
- Service de Dermatologie, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon F-69003; Lyon-1 University; INSERM U1111 - CIRI, Lyon F-69007, France
| | - H Bachelez
- Service de Dermatologie, Assistance Publique- Hôpitaux de Paris, Hôpital Saint-Louis, Université Paris Cité, Paris, France; INSERM UMR1163, Institut Imagine, Paris, France
| | - É Sbidian
- Service de Dermatologie, Assistance Publique- Hôpitaux de Paris, Hôpital Saint-Louis, Université Paris Cité, Paris, France; Hôpital Henri Mondor, Clinical Investigation Centre, Université Paris-Est Créteil (UPEC), Epidemiology in Dermatology and Evaluation of Therapeutics (EpiDermE) - EA 7379, Créteil, France
| | - O Chosidow
- Service de Dermatologie, Assistance Publique- Hôpitaux de Paris, Hôpital Saint-Louis, Université Paris Cité, Paris, France; Groupe de recherche Dynamyc, EA7380, Faculté de Santé de Créteil, École nationale vétérinaire d'Alfort, USC ANSES, Université Paris-Est Créteil, Créteil, France
| | - C Paul
- Service de Dermatologie, Toulouse University Hospital (CHU), Paul Sabatier University, Toulouse, France
| | - F Tubach
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique, AP-HP, Hôpital Pitié-Salpêtrière, Département de Santé Publique, Centre de Pharmaco-épidémiologie (Cephépi), CIC-1901, 75013 Paris, France.
| | | |
Collapse
|
65
|
Zhao K, Zhang D, Xu X, Wang S, Liu Z, Ren X, Zhang X, Lu Z, Ren S, Qin C. Exploring the potential mechanisms of impairment on genitourinary system associated with coronavirus disease 2019 infection: Bioinformatics and molecular simulation analyses. Asian J Urol 2023; 10:S2214-3882(23)00023-1. [PMID: 36776826 PMCID: PMC9902342 DOI: 10.1016/j.ajur.2022.12.004] [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: 08/27/2022] [Revised: 11/13/2022] [Accepted: 12/02/2022] [Indexed: 02/10/2023] Open
Abstract
Objective The novel coronavirus (severe acute respiratory syndrome coronavirus 2) has been spreading worldwide since December 2019, posing a serious danger to human health and socioeconomic development. A large number of clinical trials have revealed that coronavirus disease 2019 (COVID-19) results in multi-organ damage including the urogenital system. This study aimed to explore the potential mechanisms of genitourinary damage associated with COVID-19 infection through bioinformatics and molecular simulation analysis. Methods We used multiple publicly available databases to explore the expression patterns of ACE2, TMPRSS2, and CD147 (Basigin [BSG]) in major organs in the healthy and disease-specific populations, particularly the genitourinary organs. Single-cell RNA sequencing was used to analyze the cell-specific expression patterns of ACE2, TMPRSS2, CD147, cytokine receptors, and cytokine interacting proteins in genitourinary organs, such as the bladder, kidney, prostate, and testis. Additionally, gene set enrichment analysis was used to investigate the relationship between testosterone levels and COVID-19 vulnerability in patients with prostate cancer. Results The results revealed that ACE2, TMPRSS2, and CD147 were highly expressed in normal urogenital organs. Then, they were also highly expressed in multiple tumors and chronic kidney diseases. Additionally, ACE2, TMPRSS2, and CD147 were significantly expressed in a range of cells in urogenital organs according to single-cell RNA sequencing. Cytokine receptors and cytokine interacting proteins, especially CCL2, JUN, and TIMP1, were commonly highly expressed in urogenital organs. Finally, gene set enrichment analysis results showed that high testosterone levels in prostate cancer patients were significantly related to the JAK/STAT signaling pathway and the Toll-like receptor signaling pathway which were associated with COVID-19. Conclusion Our study provides new insights into the potential mechanisms of severe acute respiratory syndrome coronavirus 2 damage to urogenital organs from multiple perspectives, which may draw the attention of urologists to COVID-19 and contribute to the development of targeted drugs.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Dong Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Xinchi Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Shangqian Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Zhanpeng Liu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Xiaohan Ren
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Xu Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Zhongwen Lu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Shancheng Ren
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| |
Collapse
|
66
|
Kumar V, Yasmeen N, Chaudhary AA, Alawam AS, Al-Zharani M, Suliman Basher N, Harikrishnan S, Goud MD, Pandey A, Lakhawat SS, Sharma PK. Specialized pro-resolving lipid mediators regulate inflammatory macrophages: A paradigm shift from antibiotics to immunotherapy for mitigating COVID-19 pandemic. Front Mol Biosci 2023; 10:1104577. [PMID: 36825200 PMCID: PMC9942001 DOI: 10.3389/fmolb.2023.1104577] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
The most severe clinical manifestations of the horrifying COVID-19 disease, that claimed millions of lives during the pandemic time, were Acute respiratory distress syndrome (ARDS), Coagulopathies, septic shock leading eventually to death. ARDS was a consequence of Cytokine storm. The viral SARS-COV2infection lead to avalanche of cytokines and eicosanoids causing "cytokine storm" and "eicosanoid storm." Cytokine storm is one of the macrophage-derived inflammatory responses triggered by binding of virus particles to ACE2 receptors of alveolar macrophages, arise mainly due to over production of various pro-inflammatory mediators like cytokines, e.g., interleukin (IL)-1, IL-2, and tumor necrosis factor (TNF)- α, causing pulmonary edema, acute respiratory distress, and multi-organ failure. Cytokine storm was regarded as the predictor of severity of the disease and was deemed one of the causes of the high mortality rates due to the COVID-19. The basis of cytokine storm is imbalanced switching between an inflammation increasing - pro-inflammatory (M1) and an inflammation regulating-anti-inflammatory (M2) forms of alveolar macrophages which further deteriorates if opportunistic secondary bacterial infections prevail in the lungs. Lack of sufficient knowledge regarding the virus and its influence on co-morbidities, clinical treatment of the diseases included exorbitant use of antibiotics to mitigate secondary bacterial infections, which led to the unwarranted development of multidrug resistance (MDR) among the population across the globe. Antimicrobial resistance (AMR) needs to be addressed from various perspectives as it may deprive future generations of the basic health immunity. Specialized pro-resolving mediators (SPMs) are generated from the stereoselective enzymatic conversions of essential fatty acids that serve as immune resolvents in controlling acute inflammatory responses. SPMs facilitate the clearance of injured tissue and cell debris, the removal of pathogens, and augment the concentration of anti-inflammatory lipid mediators. The SPMs, e.g., lipoxins, protectins, and resolvins have been implicated in exerting inhibitory influence on with cytokine storm. Experimental evidence suggests that SPMS lower antibiotic requirement. Therefore, in this review potential roles of SPMs in enhancing macrophage polarization, triggering immunological functions, hastening inflammation resolution, subsiding cytokine storm and decreasing antibiotic requirement that can reduce AMR load are discussed.
Collapse
Affiliation(s)
- Vikram Kumar
- Amity institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India,*Correspondence: Vikram Kumar,
| | - Nusrath Yasmeen
- Amity institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Abdullah S. Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohammed Al-Zharani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Nosiba Suliman Basher
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - S. Harikrishnan
- Amity institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | | | - Aishwarya Pandey
- INRS, Eau Terre Environnement Research Centre, Québec, QC, Canada
| | | | | |
Collapse
|
67
|
Liu X, Du Z, Wang J, Wang Q, Zheng Y, Niu L, Hao C, Xue D, Zhang Y. Association between trans fatty acids and COVID-19: A multivariate Mendelian randomization study. J Med Virol 2023; 95:e28455. [PMID: 36597904 DOI: 10.1002/jmv.28455] [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: 11/27/2022] [Revised: 12/16/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
Traditional observational studies have suggested a potential association between trans fatty acids (TFAs), which are considered to be health-damaging fatty acids, and coronavirus disease 2019 (COVID-19). However, whether there is a causal relationship between them is currently unclear. We aimed to investigate the causal link between genetically determined TFAs and COVID-19. We performed univariate and multivariate Mendelian randomization (MR) studies using summary statistics from the European Pedigree TFAs (n = 8013), COVID-19 susceptibility (n = 159 840), COVID-19 hospitalization (n = 44 986), and COVID-19 severity (n = 18 152) genome-wide association studies (GWAS). The inverse variance weighted (IVW) method was used as the primary MR analysis, and several other methods were used as supplements. In univariate MR analysis, higher levels of circulating trans, cis-18:2 TFAs were positively associated with a higher COVID-19 hospitalization rate (p < 0.0033; odds ratio [OR] = 1.637; 95% confidence interval [CI]: 1.116-2.401) and COVID-19 severity (p < 0.0033; OR = 2.575; 95% CI: 1.412-4.698). Furthermore, in multivariate MR analysis, trans, cis-18:2 had an independent and significant causal association with a higher COVID-19 hospitalization rate (p = 0.00044; OR = 1.862; 95% CI = 1.316-2.636) and COVID-19 severity (p = 0.0016; OR = 2.268; 95% CI = 1.361-3.779) after the five TFAs were adjusted for each other. Together, our findings provide evidence that trans, cis-18:2 TFAs have an independent and robust causal effect on COVID-19 hospitalization and severity.
Collapse
Affiliation(s)
- Xuxu Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiwei Du
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China.,Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
| | - Qiang Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Zheng
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Niu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chenjun Hao
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingmei Zhang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
68
|
Andrade MCR, Lemos BRP, Silva LM, Pecotte JK. Eliminating Potential Effects of Other Infections During Selection of Nonhuman Primates for COVID-19 Research. Comp Med 2023; 73:45-57. [PMID: 36744555 PMCID: PMC9948906 DOI: 10.30802/aalas-cm-21-000086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The study of nonhuman primates (NHP) can provide significant insights into our understanding numerous infectious agents. The etiological agent of COVID-19, SARS-CoV-2 virus, first emerged in 2019 and has so far been responsible for the deaths of over 4 million people globally. In the frenzied search to understand its pathogenesis and immunology and to find measures for prevention and control of this pandemic disease, NHP, particularly macaques, are the preferred model because they manifest similar clinical signs and immunologic features as humans. However, possible latent, subclinical, and opportunistic infections not previously detected in animals participating in a study may obscure experimental results and confound data interpretations in testing treatments and vaccine studies for COVID-19. Certain pathophysiologic changes that occur with SARS-CoV-2 virus infection are similar to those of simian pathogens. The current review discusses numerous coinfections of COVID-19 with other diseases and describes possible outcomes and mechanisms in COVID-19 studies of NHP that have coinfections. Due to the urgency triggered by the pandemic, screening that is more rigorous than usual is necessary to limit background noise and maximize the reliability of data from NHP COVID-19 studies. Screening for influenza virus, selected respiratory bacteria, and regional endemic pathogens such as vector-borne agents, together with the animal's individual exposure history, should be the main considerations in selecting a NHP for a COVID-19 study. In addition, because NHP are susceptible to the SARS-CoV-2 virus, management and surveillance measures should be established to prevent transmission to healthy animals from infected colony animals and husbandry staff. This review presents compiled data on the use of NHP in COVID-19 studies, emphasizing the need to create the most reliable NHP model for those studies by extensive screening for other pathogens.
Collapse
Key Words
- absl, animal biosafety level
- ace-2, angiotensin-converting enzyme
- ards, acute respiratory distress syndrome
- cnprc, california national primate research center
- e, envelope
- esr, erythrocyte sedimentation rate
- hav, hepatitis a virus
- hbv, hepatitis b virus
- hgf hepatocyte growth factor
- htlv, human t-cell lymphotropic virus
- ifn, interferon
- il, interleukin
- ip, inducible protein
- m, matrix
- mcp, monocyte chemotactic proteins
- mcsf, macrophage colony-stimulating factor
- mip, macrophage inflammatory protein
- n, nucleocapsid
- nsp, non-structural proteins
- rdrp, rna-dependent rna polymerase
- s, spike
- sars-cov-2, severe acute respiratory syndrome-coronavirus-2
- sfv, simian foamy virus
- sop, standard operating procedures
- srv/d, simian retrovirus type d
- stlv, simian t-lymphotropic virus
- tb, tuberculosis
- tgf, transforming growth factor
- tmprss2 transmembrane serine protease 2
Collapse
Affiliation(s)
- Marcia C R Andrade
- Institute of Science and Technology on Biomodels, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil;,
| | - Bárbara R P Lemos
- Institute of Science and Technology on Biomodels, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa M Silva
- Institute of Science and Technology on Biomodels, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; University of Grande Rio, Duque de Caxias, Brazil
| | | |
Collapse
|
69
|
Elsebai MF, Habib ESE. Blood pH and COVID-19. Arch Pharm (Weinheim) 2023; 356:e2200558. [PMID: 36690587 DOI: 10.1002/ardp.202200558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/25/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a worldwide war. Raising the blood pH might be a crucial strategy to chase COVID-19. The human blood is slightly alkaline, which is essential for cell metabolism, normal physiology, and balanced immunity since all of these biological processes are pH-dependent. Varieties of physiologic derangements occur when the blood pH is disrupted. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) proliferates in acidic blood that magnifies the severity of COVID-19. On the other side, blood acidemia is linked to increased morbidity and mortality because of its complications on immunity, especially in the elderly and in critical diseases such as cancer, musculoskeletal degradation, renal, cardiac, and pulmonary disorders, which result in many pathological disorders such as osteomalacia, and disturbing the hematopoiesis. Additionally, acidemia of the blood facilitates viral infection and progression. Thus, correcting the acid-base balance might be a crucial strategy for the treatment of COVID-19, which might be attributed to the distraction of the viral spike protein to its cognate receptor angiotensin-converting enzyme 2 and supporting the over-taxed immunity.
Collapse
Affiliation(s)
- Mahmoud Fahmi Elsebai
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - El-Sayed E Habib
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
70
|
Zhao T, Ding J, Liang Z, Cui X, He K, Chen L, Li J. When type II diabetes mellitus meets COVID-19-Identification of the shared gene signatures and biological mechanism between the two diseases. Eur J Clin Invest 2023; 53:e13955. [PMID: 36656083 DOI: 10.1111/eci.13955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
BACKGROUND According to current studies, more than 20% of all patients diagnosed with COVID-19 globally have diabetes. Further, the mortality rate of these patients is 7.3%. Compared with non-diabetic COVID-19 patients, diabetic COVID-19 patients have higher rates of mortality and severe infection, suggesting that diabetes is associated with the severity of COVID-19 infection. This study aimed to analyse the relationship and susceptibility factors between COVID-19 and T2DM. METHODS Using bioinformatics methods, potential targets for COVID-19 and T2DM were screened from GeneCards database. Potential targets of COVID-19 and T2DM were mapped to each other to identify overlapping targets, and a PPI network was constructed to extract the core target. The clusterProfiler package in R was used to analyse the function and pathway that core target involved. GO enrichment and KEGG pathway analysis were used to elucidate the correlation between COVID-19 and T2DM. RESULTS A total of 277 potential pathogenic targets of COVID-19 were found, 282 potential targets were found for T2DM. Mapping of the potential COVID-19 and T2DM targets revealed 53 overlapping targets, with TNF as the core target. IL-17 signalling pathway was the most significant KEGG pathway involving TNF. CONCLUSIONS The inflammatory cytokine, TNF, was identified as a core target between COVID-19 and T2DM, which induces inflammatory response mainly through the IL-17 signalling pathway, leading to aggravation of infection and increased difficulty in blood glucose control. This study provides a reference for further exploring the potential correlation and endogenous mechanisms between two seemingly independent and unrelated diseases-T2DM and COVID-19.
Collapse
Affiliation(s)
- Tianyu Zhao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - JiPeng Ding
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zuowen Liang
- Department of Andrology, First Hospital of Jilin University, Changchun, China
| | - Xiaoli Cui
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jing Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
71
|
Tyrkalska SD, Candel S, Pedoto A, García-Moreno D, Alcaraz-Pérez F, Sánchez-Ferrer Á, Cayuela ML, Mulero V. Zebrafish models of COVID-19. FEMS Microbiol Rev 2023; 47:fuac042. [PMID: 36323404 PMCID: PMC9841970 DOI: 10.1093/femsre/fuac042] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/23/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Although COVID-19 has only recently appeared, research studies have already developed and implemented many animal models for deciphering the secrets of the disease and provided insights into the biology of SARS-CoV-2. However, there are several major factors that complicate the study of this virus in model organisms, such as the poor infectivity of clinical isolates of SARS-CoV-2 in some model species, and the absence of persistent infection, immunopathology, severe acute respiratory distress syndrome, and, in general, all the systemic complications which characterize COVID-19 clinically. Another important limitation is that SARS-CoV-2 mainly causes severe COVID-19 in older people with comorbidities, which represents a serious problem when attempting to use young and immunologically naïve laboratory animals in COVID-19 testing. We review here the main animal models developed so far to study COVID-19 and the unique advantages of the zebrafish model that may help to contribute to understand this disease, in particular to the identification and repurposing of drugs to treat COVID-19, to reveal the mechanism of action and side-effects of Spike-based vaccines, and to decipher the high susceptibility of aged people to COVID-19.
Collapse
Affiliation(s)
- Sylwia D Tyrkalska
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sergio Candel
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Annamaria Pedoto
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Diana García-Moreno
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisca Alcaraz-Pérez
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Grupo de Telomerasa, Cáncer y Envejecimiento (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Álvaro Sánchez-Ferrer
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Departmento de Bioloquímica y Biología Molecular A, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
| | - María L Cayuela
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Grupo de Telomerasa, Cáncer y Envejecimiento (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Victoriano Mulero
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
72
|
Wu W, Cheng Y, Zhou H, Sun C, Zhang S. The SARS-CoV-2 nucleocapsid protein: its role in the viral life cycle, structure and functions, and use as a potential target in the development of vaccines and diagnostics. Virol J 2023; 20:6. [PMID: 36627683 PMCID: PMC9831023 DOI: 10.1186/s12985-023-01968-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) continues to take a heavy toll on personal health, healthcare systems, and economies around the globe. Scientists are expending tremendous effort to develop diagnostic technologies for detecting positive infections within the shortest possible time, and vaccines and drugs specifically for the prevention and treatment of COVID-19 disease. At the same time, emerging novel variants have raised serious concerns about vaccine efficacy. The SARS-CoV-2 nucleocapsid (N) protein plays an important role in the coronavirus life cycle, and participates in various vital activities after virus invasion. It has attracted a large amount of attention for vaccine and drug development. Here, we summarize the latest research of the N protein, including its role in the SARS-CoV-2 life cycle, structure and function, and post-translational modifications in addition to its involvement in liquid-liquid phase separation (LLPS) and use as a basis for the development of vaccines and diagnostic techniques.
Collapse
Affiliation(s)
- Wenbing Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Ying Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Hong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Changzhen Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Shujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
73
|
COVID-19 diagnostics: Molecular biology to nanomaterials. Clin Chim Acta 2023; 538:139-156. [PMID: 36403665 PMCID: PMC9673061 DOI: 10.1016/j.cca.2022.11.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
The SARS-CoV-2 pandemic has claimed around 6.4 million lives worldwide. The disease symptoms range from mild flu-like infection to life-threatening complications. The widespread infection demands rapid, simple, and accurate diagnosis. Currently used methods include molecular biology-based approaches that consist of conventional amplification by RT-PCR, isothermal amplification-based techniques such as RT-LAMP, and gene editing tools like CRISPR-Cas. Other methods include immunological detection including ELISA, lateral flow immunoassay, chemiluminescence, etc. Radiological-based approaches are also being used. Despite good analytical performance of these current methods, there is an unmet need for less costly and simpler tests that may be performed at point of care. Accordingly, nanomaterial-based testing has been extensively pursued. In this review, we discuss the currently used diagnostic techniques for SARS-CoV-2, their usefulness, and limitations. In addition, nanoparticle-based approaches have been highlighted as another potential means of detection. The review provides a deep insight into the current diagnostic methods and future trends to combat this deadly menace.
Collapse
|
74
|
Chikomba C, Dlamini S, George JA, Pillay T. COVID Diagnostics: From Molecules to Omics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:141-158. [PMID: 37378765 DOI: 10.1007/978-3-031-28012-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The identification and genetic sequencing of a novel coronavirus was key to the diagnosis and management of the global pandemic. An understanding of the SARS-CoV-2 structure and mechanism of injury is vital to explaining the disease course and the pathophysiology of the signs and symptoms observed. This particularly as the presentation, disease course, and severity are noted to be highly variable. The role of the spike protein and angiotensin-converting enzyme 2 (ACE-2) receptor in immune response and viral entry provides great insight into current and future diagnostics and therapeutics. This article reviews the traditional diagnostic methods, which include molecular testing methods, antigen testing, and antibody testing. The gold standard for diagnosis of COVID-19 is reverse transcriptase polymerase chain reaction (RT-PCR). There have been multiple improvements to these principles to help optimize the sensitivity, specificity, and user friendliness of the method. In addition, advancements in gene sequencing and identification have been integral to identifying variants and managing outbreaks. Serological and immunological testing have made significant contributions to the management of the COVID-19 pandemic, each with its unique benefits and limitations. A growing role of the laboratory is in triaging patients to determine which patients will most benefit from hospitalization and specialized care. This is imperative for rationalizing resources during outbreaks. As we learn to live with the pandemic, novel testing methods include the use of multiomic technologies and the greater utility of point of care.
Collapse
Affiliation(s)
- Chemedzai Chikomba
- Department of Chemical Pathology. National Health Laboratory Services and University of Witwatersrand, Johannesburg, South Africa
| | - Siphelele Dlamini
- Department of Chemical Pathology. National Health Laboratory Services and University of Witwatersrand, Johannesburg, South Africa
| | - Jaya A George
- Wits Diagnostic and Innovation Hub, University of Witwatersrand, Johannesburg, South Africa.
| | - Taryn Pillay
- Department of Chemical Pathology. National Health Laboratory Services and University of Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
75
|
Fang C, Xiao G, Wang T, Song L, Peng B, Xu B, Zhang K. Emerging Nano-/Biotechnology Drives Oncolytic Virus-Activated and Combined Cancer Immunotherapy. RESEARCH 2023; 6:0108. [PMID: 37040283 PMCID: PMC10079287 DOI: 10.34133/research.0108] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
Oncolytic viruses (OVs) as one promising antitumor methods have made important contributions to tumor immunotherapy, which arouse increasing attention. They provide the dual mechanisms including direct killing effect toward tumor cells and immune activation for elevating antitumor responses, which have been proved in many preclinical studies. Especially, natural or genetically modified viruses as clinical immune preparations have emerged as a new promising approach objective to oncology treatment. The approval of talimogene laherparepvec (T-VEC) by the U.S. Food and Drug Administration (FDA) for the therapy of advanced melanoma could be considered as a milestone achievement in the clinical translation of OV. In this review, we first discussed the antitumor mechanisms of OVs with an emphasis on targeting, replication, and propagation. We further outlined the state of the art of current OVs in tumor and underlined the activated biological effects especially including immunity. More significantly, the enhanced immune responses based on OVs were systematically discussed from different perspectives such as combination with immunotherapy, genetic engineering of OVs, integration with nanobiotechnology or nanoparticles, and antiviral response counteraction, where their principles were shed light on. The development of OVs in the clinics was also highlighted to analyze the actuality and concerns of different OV applications in clinical trials. At last, the future perspectives and challenges of OVs as an already widely accepted treatment means were discussed. This review will provide a systematic review and deep insight into OV development and also offer new opportunities and guidance pathways to drive the further clinical translation.
Collapse
Affiliation(s)
- Chao Fang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Gaozhe Xiao
- National Center for International Research of Bio-targeting Theranostics,
Guangxi Medical University, No. 22 Shuangyong Road 22, Nanning, Guangxi 530021, China
| | - Taixia Wang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Li Song
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Bo Peng
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Bin Xu
- Department of Urology, Shanghai Ninth People’s Hospital,
Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Huangpu, Shanghai 200011, China
| | - Kun Zhang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
- National Center for International Research of Bio-targeting Theranostics,
Guangxi Medical University, No. 22 Shuangyong Road 22, Nanning, Guangxi 530021, China
| |
Collapse
|
76
|
Yue Y, Ma W, Accorsi EK, Ding M, Hu F, Willett WC, Chan AT, Sun Q, Rich-Edwards J, Smith-Warner SA, Bhupathiraju SN. Long-term diet and risk of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and Coronavirus Disease 2019 (COVID-19) severity. Am J Clin Nutr 2022; 116:1672-1681. [PMID: 35945354 PMCID: PMC9384672 DOI: 10.1093/ajcn/nqac219] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/04/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The role of diet on Coronavirus Disease 2019 (COVID-19) is emerging. We investigated the association between usual diet before the onset of the pandemic and risk and severity of subsequent SARS-CoV-2 infection. METHODS We included 42,935 participants aged 55-99 y in 2 ongoing cohort studies, the Nurses' Health Study II and Health Professionals Follow-up Study, who completed a series of COVID-19 surveys in 2020 and 2021. Using data from FFQs before COVID-19, we assessed diet quality using the Alternative Healthy Eating Index (AHEI)-2010, the alternative Mediterranean Diet (AMED) score, an Empirical Dietary Index for Hyperinsulinemia (EDIH), and an Empirical Dietary Inflammatory Pattern (EDIP). We calculated multivariable-adjusted ORs and 95% CIs for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and severity of COVID-19 after controlling for demographic, medical, and lifestyle factors. RESULTS Among 19,754 participants tested for SARS-CoV-2, 1941 participants reported a positive result. Of these, 1327 reported symptoms needing assistance and another 109 were hospitalized. Healthier diets, represented by higher AHEI-2010 and AMED scores and lower EDIH and EDIP scores, were associated with lower likelihood of SARS-CoV-2 infection (quartile 4 compared with quartile 1: OR: 0.80; 95% CI: 0.69, 0.92 for AHEI-2010; OR: 0.78; 95% CI: 0.67, 0.92 for AMED; OR: 1.36; 95% CI: 1.16, 1.57 for EDIH; and OR: 1.13; 95% CI: 0.99, 1.30 for EDIP; all P-trend ≤ 0.01). In the analysis of COVID-19 severity, participants with healthier diet had lower likelihood of severe infection and were less likely to be hospitalized owing to COVID-19. However, associations were no longer significant after controlling for BMI and pre-existing medical conditions. CONCLUSIONS Diet may be an important modifiable risk factor for SARS-CoV-2 infection, as well as for severity of COVID-19. This association is partially mediated by BMI and pre-existing medical conditions.
Collapse
Affiliation(s)
- Yiyang Yue
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Emma K Accorsi
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Ming Ding
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Frank Hu
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Sun
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Janet Rich-Edwards
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephanie A Smith-Warner
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Shilpa N Bhupathiraju
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
77
|
Graciliano NG, Tenório MCS, Fragoso MBT, Moura FA, Botelho RM, Tanabe ELL, Borbely KSC, Borbely AU, Oliveira ACM, Goulart MOF. The impact on colostrum oxidative stress, cytokines, and immune cells composition after SARS-CoV-2 infection during pregnancy. Front Immunol 2022; 13:1031248. [PMID: 36591280 PMCID: PMC9798093 DOI: 10.3389/fimmu.2022.1031248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Background Limited data are available regarding the differences between immunological, biochemical, and cellular contents of human colostrum following maternal infection during pregnancy with coronavirus 2 disease (COVID-19). Objective To investigate whether maternal COVID-19 infection may affect immunological, biochemical, and cellular contents of human colostrum. Methods Using a case-control study design, we collected colostrum from 14 lactating women with a previous diagnosis of COVID-19 during pregnancy and 12 without a clear diagnosis during September 2020 to May 2021. Colostrum samples were analysed for some enzymes and non-enzymatic oxidative stress markers (SOD, CAT, GPx, MDA, GSH, GSSG, H2O2, MPO) and for IL-1β, IL-6, tumour necrosis factor (TNF)-α, protein induced by interferon gamma (IP)-10, IL-8, IFN-λ1, IL12p70, IFN-α2, IFN-λ2/3, granulocyte macrophage colony stimulating factor (GM-CSF), IFN-β, IL-10 and IFN-γ, along with IgA and IgG for the SARS-CoV-2 S protein. We perform immunophenotyping to assess the frequency of different cell types in the colostrum. Results Colostrum from the COVID-19 symptomatic group in pregnancy contained reduced levels of H2O2, IFN-α2, and GM-CSF. This group had higher levels of GSH, and both NK cell subtypes CD3-CD56brightCD16-CD27+IFN-γ+ and CD3-CD56dimCD16+CD27- were also increased. Conclusion The present results reinforce the protective role of colostrum even in the case of mild SARS-Cov-2 infection, in addition to demonstrating how adaptive the composition of colostrum is after infections. It also supports the recommendation to encourage lactating women to continue breastfeeding after COVID-19 illness.
Collapse
Affiliation(s)
- Nayara Gomes Graciliano
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | | | | | | | - Rayane Martins Botelho
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | - Eloiza Lopes Lira Tanabe
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | | | - Alexandre Urban Borbely
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | - Alane Cabral Menezes Oliveira
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
- College of Nutrition, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | - Marília Oliveira Fonseca Goulart
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio, Alagoas, Brazil
- National Institute of Science and Technology in Bioanalytics (INCT-Bio), Campinas, Sao Paulo, Brazil
| |
Collapse
|
78
|
Chen Y, Qin Y, Fu Y, Gao Z, Deng Y. Integrated Analysis of Bulk RNA-Seq and Single-Cell RNA-Seq Unravels the Influences of SARS-CoV-2 Infections to Cancer Patients. Int J Mol Sci 2022; 23:15698. [PMID: 36555339 PMCID: PMC9779348 DOI: 10.3390/ijms232415698] [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: 10/22/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly contagious and pathogenic coronavirus that emerged in late 2019 and caused a pandemic of respiratory illness termed as coronavirus disease 2019 (COVID-19). Cancer patients are more susceptible to SARS-CoV-2 infection. The treatment of cancer patients infected with SARS-CoV-2 is more complicated, and the patients are at risk of poor prognosis compared to other populations. Patients infected with SARS-CoV-2 are prone to rapid development of acute respiratory distress syndrome (ARDS) of which pulmonary fibrosis (PF) is considered a sequelae. Both ARDS and PF are factors that contribute to poor prognosis in COVID-19 patients. However, the molecular mechanisms among COVID-19, ARDS and PF in COVID-19 patients with cancer are not well-understood. In this study, the common differentially expressed genes (DEGs) between COVID-19 patients with and without cancer were identified. Based on the common DEGs, a series of analyses were performed, including Gene Ontology (GO) and pathway analysis, protein-protein interaction (PPI) network construction and hub gene extraction, transcription factor (TF)-DEG regulatory network construction, TF-DEG-miRNA coregulatory network construction and drug molecule identification. The candidate drug molecules (e.g., Tamibarotene CTD 00002527) obtained by this study might be helpful for effective therapeutic targets in COVID-19 patients with cancer. In addition, the common DEGs among ARDS, PF and COVID-19 patients with and without cancer are TNFSF10 and IFITM2. These two genes may serve as potential therapeutic targets in the treatment of COVID-19 patients with cancer. Changes in the expression levels of TNFSF10 and IFITM2 in CD14+/CD16+ monocytes may affect the immune response of COVID-19 patients. Specifically, changes in the expression level of TNFSF10 in monocytes can be considered as an immune signature in COVID-19 patients with hematologic cancer. Targeting N6-methyladenosine (m6A) pathways (e.g., METTL3/SERPINA1 axis) to restrict SARS-CoV-2 reproduction has therapeutic potential for COVID-19 patients.
Collapse
Affiliation(s)
- Yu Chen
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
- Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Yujia Qin
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Yuanyuan Fu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Zitong Gao
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
- Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| |
Collapse
|
79
|
Sun Y, An X, Jin D, Duan L, Zhang Y, Yang C, Duan Y, Zhou R, Zhao Y, Zhang Y, Kang X, Jiang L, Lian F. Model exploration for discovering COVID-19 targeted traditional Chinese medicine. Heliyon 2022; 8:e12333. [PMID: 36530927 PMCID: PMC9737519 DOI: 10.1016/j.heliyon.2022.e12333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/15/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
In terms of treatment, a particularly targeted drug is needed to combat the COVID-19 pandemic. Although there are currently no specific drugs for COVID-19, traditional Chinese medicine(TCM) is clearly effective. It is recommended that through data analysis and mining of TCM cases (expert experience) and population evidence (RCT and cohort studies), core prescriptions for various efficacy can be obtained. Starting from a multidimensional model of regulating immunity, improving inflammation, and protecting multiple organs, this paper constructs a multidimensional model of targeted drug discovery, integrating molecular, cellular, and animal efficacy evaluation. Through functional activity testing, biophysical detection of compound binding to target proteins, multidimensional pharmacodynamic evaluation systems of cells (Vero E6, Vero, Vero81, Huh7, and caca2) and animals (mice infected with the new coronavirus, rhesus macaques, and hamsters), the effectiveness of effective preparations was evaluated, and various efficacy effects including lung moisturizing, dehumidification and detoxification were obtained. Using modern technology, it is now possible to understand how the immune system is controlled, how inflammation is reduced, and how various organs are protected. Complete early drug characterization and finally obtain effective targeted TCM. This article provides a demonstration resource for the development of new drugs specifically for TCM.
Collapse
Affiliation(s)
- Yuting Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Xuedong An
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - De Jin
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Liyun Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Yuehong Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Cunqing Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Yingying Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Rongrong Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Yiru Zhao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Yuqing Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Xiaomin Kang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Linlin Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China
| | - Fengmei Lian
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing 100053, China,Corresponding author.
| |
Collapse
|
80
|
Lei S, Chen X, Wu J, Duan X, Men K. Small molecules in the treatment of COVID-19. Signal Transduct Target Ther 2022; 7:387. [PMID: 36464706 PMCID: PMC9719906 DOI: 10.1038/s41392-022-01249-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 12/11/2022] Open
Abstract
The outbreak of COVID-19 has become a global crisis, and brought severe disruptions to societies and economies. Until now, effective therapeutics against COVID-19 are in high demand. Along with our improved understanding of the structure, function, and pathogenic process of SARS-CoV-2, many small molecules with potential anti-COVID-19 effects have been developed. So far, several antiviral strategies were explored. Besides directly inhibition of viral proteins such as RdRp and Mpro, interference of host enzymes including ACE2 and proteases, and blocking relevant immunoregulatory pathways represented by JAK/STAT, BTK, NF-κB, and NLRP3 pathways, are regarded feasible in drug development. The development of small molecules to treat COVID-19 has been achieved by several strategies, including computer-aided lead compound design and screening, natural product discovery, drug repurposing, and combination therapy. Several small molecules representative by remdesivir and paxlovid have been proved or authorized emergency use in many countries. And many candidates have entered clinical-trial stage. Nevertheless, due to the epidemiological features and variability issues of SARS-CoV-2, it is necessary to continue exploring novel strategies against COVID-19. This review discusses the current findings in the development of small molecules for COVID-19 treatment. Moreover, their detailed mechanism of action, chemical structures, and preclinical and clinical efficacies are discussed.
Collapse
Affiliation(s)
- Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xiaohua Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
81
|
Chattopadhyay P, Khare K, Kumar M, Mishra P, Anand A, Maurya R, Gupta R, Sahni S, Gupta A, Wadhwa S, Yadav A, Devi P, Tardalkar K, Joshi M, Sethi T, Pandey R. Single-cell multiomics revealed the dynamics of antigen presentation, immune response and T cell activation in the COVID-19 positive and recovered individuals. Front Immunol 2022; 13:1034159. [PMID: 36532041 PMCID: PMC9755500 DOI: 10.3389/fimmu.2022.1034159] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Despite numerous efforts to describe COVID-19's immunological landscape, there is still a gap in our understanding of the virus's infections after-effects, especially in the recovered patients. This would be important to understand as we now have huge number of global populations infected by the SARS-CoV-2 as well as variables inclusive of VOCs, reinfections, and vaccination breakthroughs. Furthermore, single-cell transcriptome alone is often insufficient to understand the complex human host immune landscape underlying differential disease severity and clinical outcome. Methods By combining single-cell multi-omics (Whole Transcriptome Analysis plus Antibody-seq) and machine learning-based analysis, we aim to better understand the functional aspects of cellular and immunological heterogeneity in the COVID-19 positive, recovered and the healthy individuals. Results Based on single-cell transcriptome and surface marker study of 163,197 cells (124,726 cells after data QC) from the 33 individuals (healthy=4, COVID-19 positive=16, and COVID-19 recovered=13), we observed a reduced MHC Class-I-mediated antigen presentation and dysregulated MHC Class-II-mediated antigen presentation in the COVID-19 patients, with restoration of the process in the recovered individuals. B-cell maturation process was also impaired in the positive and the recovered individuals. Importantly, we discovered that a subset of the naive T-cells from the healthy individuals were absent from the recovered individuals, suggesting a post-infection inflammatory stage. Both COVID-19 positive patients and the recovered individuals exhibited a CD40-CD40LG-mediated inflammatory response in the monocytes and T-cell subsets. T-cells, NK-cells, and monocyte-mediated elevation of immunological, stress and antiviral responses were also seen in the COVID-19 positive and the recovered individuals, along with an abnormal T-cell activation, inflammatory response, and faster cellular transition of T cell subtypes in the COVID-19 patients. Importantly, above immune findings were used for a Bayesian network model, which significantly revealed FOS, CXCL8, IL1β, CST3, PSAP, CD45 and CD74 as COVID-19 severity predictors. Discussion In conclusion, COVID-19 recovered individuals exhibited a hyper-activated inflammatory response with the loss of B cell maturation, suggesting an impeded post-infection stage, necessitating further research to delineate the dynamic immune response associated with the COVID-19. To our knowledge this is first multi-omic study trying to understand the differential and dynamic immune response underlying the sample subtypes.
Collapse
Affiliation(s)
- Partha Chattopadhyay
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kriti Khare
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manish Kumar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Pallavi Mishra
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Alok Anand
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Ranjeet Maurya
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rohit Gupta
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Shweta Sahni
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Ayushi Gupta
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Saruchi Wadhwa
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Aanchal Yadav
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priti Devi
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kishore Tardalkar
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Meghnad Joshi
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Tavpritesh Sethi
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
82
|
Zolfaghari MA, Ghadiri Moghaddam F, Rajput S, Karimi A, Naghi Vishteh M, Mahmoodpoor A, Dolati S, Yousefi M. SARS-CoV-2 vaccines: A double-edged sword throughout rapid evolution of COVID-19. Cell Biol Int 2022; 46:2009-2017. [PMID: 36047303 PMCID: PMC9539123 DOI: 10.1002/cbin.11903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 07/12/2022] [Accepted: 08/24/2022] [Indexed: 11/11/2022]
Abstract
After more than 2 years of the coronavirus disease 2019 pandemic caused by severe acute respiratory syndrome coronavirus 2, several questions have remained unanswered that affected our daily lives. Although substantial vaccine development could resist this challenge, emerging new variants in different countries could be considered as potent concerns regarding the adverse effects of reinfection or postvaccination. Precisely, these concerns address some significant and probable outcomes in vaccinated or reinfected models, followed by some virus challenges, such as antibody-dependent enhancement and cytokine storm. Therefore, the importance of evaluating the effectiveness of neutralizing antibodies (nAbs) elicited by vaccination and the rise of new variants must be addressed.
Collapse
Affiliation(s)
- Mohammad Ali Zolfaghari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Department of Molecular Medicine, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | | | - Shabnam Rajput
- Department of Pediatrics, School of MedicineJahrom University of Medical SciencesJahromIran
| | - Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Biotechnology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohadeseh Naghi Vishteh
- Department of Genetics and Molecular Biology, School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Ata Mahmoodpoor
- Department of AnesthesiologyTabriz University of Medical SciencesTabrizIran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| | - Mehdi Yousefi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Immunology, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
83
|
Khongthaw B, Dulta K, Chauhan PK, Kumar V, Ighalo JO. Lycopene: a therapeutic strategy against coronavirus disease 19 (COVID- 19). Inflammopharmacology 2022; 30:1955-1976. [PMID: 36050507 PMCID: PMC9436159 DOI: 10.1007/s10787-022-01061-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/18/2022] [Indexed: 02/07/2023]
Abstract
Lycopene is a group of phytochemicals found in nature, primarily in fruits and vegetables. Lycopene is thought to protect against a variety of diseases attributed to its antioxidant capabilities. Lycopene has anti-inflammatory, anti-cancer, and immunity-boosting qualities, among other biological and pharmacological benefits. COVID-19 (coronavirus disease 19) is an infectious disease caused by the SARS-CoV-2 virus, which has recently emerged as one of the world's leading causes of death. Patients may be asymptomatic or show signs of respiratory, cytokine release syndrome, gastrointestinal, or even multiple organ failure, all of which can lead to death. In COVID-19, inflammation, and cytokine storm are the key pathogenic mechanisms, according to SARS-CoV-2 infection symptoms. ARDS develops in some vulnerable hosts, which is accompanied by an inflammatory "cytokine syndrome" that causes lung damage. Immunological and inflammatory markers were linked to disease severity in mild and severe COVID-19 cases, implying that inflammatory markers, including IL-6, CRP, ESR, and PCT were significantly linked with COVID-19 severity. Patients with severe illness have reduced levels of several immune subsets, including CD4 + T, NK, and CD8 + cells. As a result, lycopene can be commended for bolstering physiological defenses against COVID-19 infections.
Collapse
Affiliation(s)
- Banlambhabok Khongthaw
- Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh, 173229, India
| | - Kanika Dulta
- Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh, 173229, India
| | - Pankaj Kumar Chauhan
- Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh, 173229, India.
| | - Vinod Kumar
- Department of Life Sciences, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, 248002, India
| | - Joshua O Ighalo
- Department of Chemical Engineering, Nnamdi Azikiwe University, P. M. B. 5025, Awka, Nigeria.
- Department of Chemical Engineering, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
84
|
Noto A, Joo V, Mancarella A, Suffiotti M, Pellaton C, Fenwick C, Perreau M, Pantaleo G. CXCL12 and CXCL13 Cytokine Serum Levels Are Associated with the Magnitude and the Quality of SARS-CoV-2 Humoral Responses. Viruses 2022; 14:2665. [PMID: 36560669 PMCID: PMC9785906 DOI: 10.3390/v14122665] [Citation(s) in RCA: 9] [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: 10/13/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
A better understanding of the immunological markers associated with long-lasting immune responses to SARS-CoV-2 infection is of paramount importance. In the present study, we characterized SARS-CoV-2-specific humoral responses in hospitalized (ICU and non-ICU) and non-hospitalized individuals at six months post-onset of symptoms (POS) (N = 95). We showed that the proportion of individuals with detectable anti-SARS-CoV-2 IgG or neutralizing (NAb) responses and the titers of antibodies were significantly reduced in non-hospitalized individuals, compared to ICU- or non-ICU-hospitalized individuals at 6 months POS. Interestingly, SARS-CoV-2-specific memory B cells persist at 6 months POS in both ICU and non-ICU patients and were enriched in cells harboring an activated and/or exhausted phenotype. The frequency/phenotype of SARS-CoV-2-specific memory B cells and the magnitude of IgG or NAb responses at 6 months POS correlated with the serum immune signature detected at patient admission. In particular, the serum levels of CXCL13, IL-1RA, and G-CSF directly correlated with the frequency of Spike-specific B cells and the magnitude of Spike-specific IgG or NAb, while the serum levels of CXCL12 showed an antagonizing effect. Our results indicate that the balance between CXCL12 and CXCL13 is an early marker associated with the magnitude and the quality of the SARS-CoV-2 humoral memory.
Collapse
Affiliation(s)
- Alessandra Noto
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Victor Joo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Antonio Mancarella
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Celine Pellaton
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
85
|
Tian R, Zhu H, Lu Y, Shi X, Tu P, Li H, Huang H, Chen D. Therapeutic Potential of 2-Methylquinazolin-4(3H)-one as an Antiviral Agent against Influenza A Virus-Induced Acute Lung Injury in Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227857. [PMID: 36431955 PMCID: PMC9697438 DOI: 10.3390/molecules27227857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
Qingdai-Mabo (QM), a traditional Chinese herbal formula composed of medicinal herb and fungus, has been used for treatment of cough and viral pneumonia. However, the underlying mechanism and bioactive components against anti-influenza A virus remain unclear. In the present study, ethyl acetate (EA) extract of QM decoctions was tested for its biological activity against acute lung injury (ALI) and its main components were identified using UPLC-MS/MS. In total, 18 bioactive components were identified, including 2-Methylquinaozlin-4(3H)-one (C1), which showed significant antiviral activity in vitro with an IC50 of 23.8 μg/mL. Furthermore, we validated the efficacy of C1 in ameliorating ALI lesions and inflammation in influenza A virus-infected mice. The results showed that C1 significantly reduced the lung index, downregulated neuraminidase (NA) and nucleoprotein (NP), and decreased the expression of pro-inflammatory molecules IFN-α, TNF-α, MCP-1, IL-6, and IL-8; however, they enhanced levels of IL-10 and IFN-γ in lung homogenate from mice infected by influenza A virus. In addition, C1 inhibited the recruitment of macrophages. These in vitro and in vivo studies suggested that the significant anti-influenza A virus activity contributed to its curative effect on lesions and inflammation of viral pneumonia in mice. Given its potential antiviral activity against influenza A virus, C1 is determined to be a main active component in the EA extract of QM. Taken together, the antiviral activity of C1 suggests its potential as an effective treatment against viral pneumonia via the inhibition of virus replication, but the mechanism C1 on antiviral research needs to be explored further.
Collapse
Affiliation(s)
- Rong Tian
- Department of Natural Medicine, School of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
| | - Haiyan Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
- Correspondence: (H.Z.); (D.C.)
| | - Yan Lu
- Department of Natural Medicine, School of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
| | - Xunlong Shi
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
| | - Peng Tu
- Department of Natural Medicine, School of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
| | - Hong Li
- Department of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
| | - Hai Huang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
| | - Daofeng Chen
- Department of Natural Medicine, School of Pharmacy, Fudan University, No. 3728, Jin Ke Road, Shanghai 201203, China
- Correspondence: (H.Z.); (D.C.)
| |
Collapse
|
86
|
Dantas ACS, Oliveira HBM, Gomes CP, Alves DL, Infante PDB, Caitité RDJA, Fritsch HM, Cucco MS, Silva LSC, Oliveira CNT, Bittencourt RDS, Amorim AT, Nascimento ALP, Marinho FAGC, de Medeiros DS, de Oliveira MGG, Mistro S, de Melo FF, Pereira TTS, Guimarães AMS, Timenetsky J, Moreira PMB, de Oliveira SHP, Alcantara LCJ, Giovanetti M, Santos LA, Fonseca V, Barreto FK, Campos GB, Marques LM. Retrospective Analysis of the SARS-CoV-2 Infection Profile in COVID-19 Positive Patients in Vitoria da Conquista, Northeast Brazil. Viruses 2022; 14:2424. [PMID: 36366521 PMCID: PMC9699198 DOI: 10.3390/v14112424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is responsible for causing Coronavirus Disease-2019 (COVID-19), a heterogeneous clinical condition that manifests varying symptom severity according to the demographic profile of the studied population. While many studies have focused on the spread of COVID-19 in large urban centers in Brazil, few have evaluated medium or small cities in the Northeast region. The aims of this study were: (i) to identify risk factors for mortality from SARS-CoV-2 infection, (ii) to evaluate the gene expression patterns of key immune response pathways using nasopharyngeal swabs of COVID-19 patients, and (iii) to identify the circulating SARS-CoV-2 variants in the residents of a medium-sized city in Northeast Brazil. A total of 783 patients infected with SARS-CoV-2 between May 2020 and August 2021 were included in this study. Clinical-epidemiological data from patients who died and those who survived were compared. Patients were also retrospectively divided into three groups based on disease severity: asymptomatic, mild, and moderate/severe. Samples were added to a qPCR array for analyses of 84 genes involved with immune response pathways and sequenced using the Oxford Nanopore MinION technology. Having pre-existing comorbidity; being male; having cardiovascular disease, diabetes, and/or chronic obstructive pulmonary disease; and PCR cycle threshold (Ct) values under 22 were identified as risk factors for mortality. Analysis of the expression profiles of inflammatory pathway genes showed that the greater the infection severity, the greater the activation of inflammatory pathways, triggering the cytokine storm and downregulating anti-inflammatory pathways. Viral genome analysis revealed the circulation of multiple lineages, such as B.1, B.1.1.28, Alpha, and Gamma, suggesting that multiple introduction events had occurred over time. This study's findings help identify the specific strains and increase our understanding of the true state of local health. In addition, our data demonstrate that epidemiological and genomic surveillance together can help formulate public health strategies to guide governmental actions.
Collapse
Affiliation(s)
- Anna Carolina S. Dantas
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
- Municipal Central Laboratory, Vitória da Conquista City Hall, Avenida Macaúbas, 100, Kadija, Vitória da Conquista 45065-060, BA, Brazil
| | - Hellen B. M. Oliveira
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Camila P. Gomes
- State University of Santa Cruz, Rod. Jorge Amado, Km 6, Salobrinho, Ilhéus 55662-900, BA, Brazil
| | - Daniele L. Alves
- Municipal Central Laboratory, Vitória da Conquista City Hall, Avenida Macaúbas, 100, Kadija, Vitória da Conquista 45065-060, BA, Brazil
| | - Priscilla D. B. Infante
- Municipal Central Laboratory, Vitória da Conquista City Hall, Avenida Macaúbas, 100, Kadija, Vitória da Conquista 45065-060, BA, Brazil
| | - Rosimara de J. A. Caitité
- Municipal Central Laboratory, Vitória da Conquista City Hall, Avenida Macaúbas, 100, Kadija, Vitória da Conquista 45065-060, BA, Brazil
| | - Hegger M. Fritsch
- Oswaldo Cruz Institute—Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, RJ, Brazil
| | - Marina S. Cucco
- Medical School, Federal University of Bahia, Largo Terreiro de Jesus, s/n, Pelourinho, Salvador 40026-010, BA, Brazil
| | - Lucas S. C. Silva
- State University of Santa Cruz, Rod. Jorge Amado, Km 6, Salobrinho, Ilhéus 55662-900, BA, Brazil
| | - Caline N. T. Oliveira
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Rafaela de S. Bittencourt
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Aline T. Amorim
- Northeast Independent College, Avenida Luís Eduardo Magalhães, 1305, Candeias, Vitória da Conquista 45055-030, BA, Brazil
| | - Ana Luisa P. Nascimento
- Northeast Independent College, Avenida Luís Eduardo Magalhães, 1305, Candeias, Vitória da Conquista 45055-030, BA, Brazil
| | - Francely A. G. C. Marinho
- Northeast Independent College, Avenida Luís Eduardo Magalhães, 1305, Candeias, Vitória da Conquista 45055-030, BA, Brazil
| | - Danielle S. de Medeiros
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Márcio G. G. de Oliveira
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Sostenes Mistro
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Fabricio F. de Melo
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Taiana T. S. Pereira
- Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 2415, Butantã, São Paulo 05508-900, SP, Brazil
| | - Ana M. S. Guimarães
- Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 2415, Butantã, São Paulo 05508-900, SP, Brazil
| | - Jorge Timenetsky
- Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 2415, Butantã, São Paulo 05508-900, SP, Brazil
| | - Pablo Maciel B. Moreira
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
- Vitória da Conquista City Hall, Rua Rotary Club, 69-Centro, Vitória da Conquista 45040-150, BA, Brazil
| | - Sandra Helena P. de Oliveira
- Campus Universitário, Júlio de Mesquita Filho State University of São Paulo, Rodovia Marechal Rondon km 527/528, Araçatuba 16015-050, SP, Brazil
| | - Luiz C. J. Alcantara
- Oswaldo Cruz Institute—Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, RJ, Brazil
| | - Marta Giovanetti
- Oswaldo Cruz Institute—Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, RJ, Brazil
- University of Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Luciane A. Santos
- Medical School, Federal University of Bahia, Largo Terreiro de Jesus, s/n, Pelourinho, Salvador 40026-010, BA, Brazil
- Bahia School of Medicine and Public Heath, Av. Dom João VI, 275, Brotas, Salvador 40290-000, BA, Brazil
| | - Vagner Fonseca
- Pan American Health Organization, Lote 19-Avenida das Nações, SEN-Asa Norte, Brasilia 70312-970, GO, Brazil
| | - Fernanda K. Barreto
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Guilherme B. Campos
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
| | - Lucas M. Marques
- Institute of Multidisciplinary Health, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista 45029-094, BA, Brazil
- Municipal Central Laboratory, Vitória da Conquista City Hall, Avenida Macaúbas, 100, Kadija, Vitória da Conquista 45065-060, BA, Brazil
| |
Collapse
|
87
|
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
|
88
|
Sun Y, Zou Y, Wang H, Cui G, Yu Z, Ren Z. Immune response induced by novel coronavirus infection. Front Cell Infect Microbiol 2022; 12:988604. [PMID: 36389144 PMCID: PMC9641212 DOI: 10.3389/fcimb.2022.988604] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2 has been prominent around the world since it was first discovered, affecting more than 100 million people. Although the symptoms of most infected patients are not serious, there is still a considerable proportion of patients who need hospitalization and even develop fatal symptoms such as cytokine storms, acute respiratory distress syndrome and so on. Cytokine storm is usually described as a collection of clinical manifestations caused by overactivation of the immune system, which plays an important role in tissue injury and multiorgan failure. The immune system of healthy individuals is composed of two interrelated parts, the innate immune system and the adaptive immune system. Innate immunity is the body's first line of defense against viruses; it can quickly perceive viruses through pattern recognition receptors and activate related inflammatory pathways to clear pathogens. The adaptive immune system is activated by specific antigens and is mainly composed of CD4+ T cells, CD8+ T cells and B cells, which play different roles in viral infection. Here, we discuss the immune response after SARS-CoV-2 infection. In-depth study of the recognition of and response of innate immunity and adaptive immunity to SARS-CoV-2 will help to prevent the development of critical cases and aid the exploration of more targeted treatments.
Collapse
Affiliation(s)
- Ying Sun
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yawen Zou
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haiyu Wang
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangying Cui
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zujiang Yu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Ren
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
89
|
Savic G, Stevanovic I, Mihajlovic D, Jurisevic M, Gajovic N, Jovanovic I, Ninkovic M. MMP-9/BDNF ratio predicts more severe COVID-19 outcomes. Int J Med Sci 2022; 19:1903-1911. [PMID: 36438922 PMCID: PMC9682503 DOI: 10.7150/ijms.75337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/16/2022] [Indexed: 01/24/2023] Open
Abstract
COVID-19 clinically manifests from asymptomatic to the critical range. Immune response provokes the pro-inflammatory interactions, which lead to the cytokines, reactive oxygen/nitrogen species, peptidases, and arachidonic acid metabolites enlargement and activation of coagulation components. Matrix metalloproteinases (MMPs) contribute to tissue destruction in the development of COVID-19. Due to the endothelial, systemic course of the disease, VEGF A participates actively in COVID-19 development, while neurotrophic and metabolic effects of BDNF recommends for the prediction of complications in COVID-19 patients. Searching for a marker that would improve and simplify the ranking in COVID-19, the study intended to evaluate the relationship of MMP-9 with VEGF A, BDNF, and MMP-8 with the COVID-19 severity. Upon admission to the hospital and before the therapy administration, 77 patients were classified into a mild, moderate, severe, or critical group. Due to the inflammatory stage in COVID-19, a comparison between groups showed related differences in leukocytes, neutrophils, lymphocytes, and platelets counts as anticipated. Only in seriously ill patients, there is a significant increase in the serum concentration of MMP-9, MMP-8, and VEGF A, while BDNF values did not show significant variations between groups. However, all those parameters positively correlated with each other. The ratio of MMP-9/BDNF markedly decreased in the severe and critically patients compared to the mild group. Testing the capability of this ratio to predict the COVID-19 stage by ROC curves, we found the MMP-9/BDNF could be a suitable marker for differentiating stages I/II (AUC 0.7597), stage I/III (AUC 0.9011), and stage I/IV (AUC 0.7727). Presented data describe for the first time the high-level systemic MMP-9/BDNF ratio in patients with COVID-19. This parameter could contribute to a more precise determination of the phase of the disease.
Collapse
Affiliation(s)
- Goran Savic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
| | - Ivana Stevanovic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
- Institute of Medical Research, Military Medical Academy, Crnotravska 17, Belgrade, Serbia
| | - Dusan Mihajlovic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
| | - Milena Jurisevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Nevena Gajovic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Milica Ninkovic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
- Institute of Medical Research, Military Medical Academy, Crnotravska 17, Belgrade, Serbia
| |
Collapse
|
90
|
Muhammad M, Hassan TM, Baba SS, Radda MI, Mutawakkil MM, Musa MA, AbuBakar S, Loong SK, Yusuf I. Exploring NF κB pathway as a potent strategy to mitigate COVID-19 severe morbidity and mortality. J Public Health Afr 2022; 13:1679. [PMID: 36313924 PMCID: PMC9614690 DOI: 10.4081/jphia.2022.1679] [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] [Received: 10/07/2020] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19), for which there does not appear to be an approved cure, the primary treatment options consist of non-pharmacological preventive measures and supportive treatment that are aimed at halting the progression of the disease. Nuclear factor kappa B (NFkB) presents a promising therapeutic opportunity to mitigate COVID-19-induced cytokine storm and reduce the risk of severe morbidity and mortality resulting from the disease. However, the effective clinical application of NFkB modulators in COVID-19 is hampered by a number of factors that must be taken into consideration. This paper therefore explored the modulation of the NFB pathway as a potential strategy to mitigate the severe morbidity and mortality caused by COVID-19. The paper also discusses the factors that form the barrier, and it offers potential solutions to the various limitations that may impede the clinical use of NFkB modulators against COVID-19. This paper revealed and identified three key potential solutions for the future clinical use of NFkB modulators against COVID-19. These solutions are pulmonary tissue-specific NFkB blockade, agents that target common regulatory proteins of both canonical and non-canonical NFkB pathways, and monitoring clinical indicators of hyperinflammation and cytokine storm in COVID-19 prior to using NFkB modulators.
Collapse
Affiliation(s)
- Mubarak Muhammad
- Department of Physiology, College of Medicine, University of Ibadan, Nigeria,Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Tasneem M. Hassan
- Department of Physiotherapy, Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Sani S. Baba
- Department of Human Physiology, College of Health Sciences, Bayero University Kano, Nigeria
| | - Mustapha I. Radda
- Department of Human Physiology, College of Health Sciences, Bayero University Kano, Nigeria
| | - Mubarak M. Mutawakkil
- Pharmacology and Therapeutics, College of Health Sciences, Bayero University Kano, Nigeria
| | - Majida A. Musa
- Pharmacology and Therapeutics, College of Health Sciences, Bayero University Kano, Nigeria
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Centre, Higher Institution Centre of Excellence, Universiti of Malaya, Kuala Lumpur, Malaysia
| | - Shih Keng Loong
- Tropical Infectious Diseases Research and Education Centre, Higher Institution Centre of Excellence, Universiti of Malaya, Kuala Lumpur, Malaysia
| | - Ibrahim Yusuf
- Department of Pathology, Aminu Kano Teaching Hospital, Kano, Nigeria
| |
Collapse
|
91
|
Al-Mazedi MS, Rajan R, Al-Jarallah M, Dashti R, Al Saber A, Pan J, Zhanna KD, Abdelnaby H, Aboelhassan W, Almutairi F, Alotaibi N, Al Saleh M, AlNasrallah N, Al-Bader B, Malhas H, Ramadhan M, Brady PA, Al-Zakwani I, Setiya P, Abdullah M, Alroomi M, Tse G. Neutrophil to lymphocyte ratio and in-hospital mortality among patients with SARS-CoV-2: A retrospective study. Ann Med Surg (Lond) 2022; 82:104748. [PMID: 36212733 PMCID: PMC9525242 DOI: 10.1016/j.amsu.2022.104748] [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: 07/30/2022] [Revised: 09/18/2022] [Accepted: 09/18/2022] [Indexed: 11/06/2022] Open
Abstract
The goal of this study was to investigate in-hospital mortality in patients suffering from acute respiratory syndrome coronavirus 2 (SARS-CoV-2) relative to the neutrophil to lymphocyte ratio (NLR) and to determine if there are gender disparities in outcome. Between February 26 and September 8, 2020, patients having SARS-CoV-2 infection were enrolled in this retrospective cohort research, which was categorized by NLR levels ≥9 and < 9. In total, 6893 patients were involved included of whom6591 had NLR <9, and 302 had NLR ≥9. The age of most of the patients in the NLR<9 group was 50 years, on the other hand, the age of most of the NLR ≥9 group patients was between 50 and 70 years. The majority of patients in both groups were male 2211 (66.1%). The ICU admission time and mortality rate for the patients with NLR ≥9 was significantly higher compared to patients with NLR <9. Logistic regression's outcome indicated that NLR ≥9 (odds ratio (OR), 24.9; 95% confidence interval (CI): 15.5–40.0; p < 0.001), male sex (OR, 3.5; 95% CI: 2.0–5.9; p < 0.001) and haemoglobin (HB) (OR, 0.95; 95% CI; 0.94–0.96; p < 0.001) predicted in-hospital mortality significantly. Additionally, Cox proportional hazards analysis (B = 4.04, SE = 0.18, HR = 56.89, p < 0.001) and Kaplan–Meier survival probability plots also indicated that NLR>9 had a significant effect on mortality. NLR ≥9 is an independent predictor of mortality(in-hospital) among SARS-CoV-2 patients. The main finding of this study is that NLR is an autonomous predictor of in-hospital mortality in patients with SARS-CoV-2. Fatality in SARS-CoV-2 patients with NLR >9 was 25 times higher than that in patients with NLR <9. Patients with NLR >9, the average length of ICU stay was higher. Mortality rate in males was high compared to females with NLR>9.
Collapse
Affiliation(s)
- Maryam Salah Al-Mazedi
- Dept. of Medical Laboratory Technology, Public Authority for Applied Education and Training, Kuwait
| | - Rajesh Rajan
- Department of Cardiology, Sabah Al Ahmed Cardiac Centre, Al Amiri Hospital, Kuwait City, Kuwait
- Corresponding author.
| | - Mohammed Al-Jarallah
- Department of Cardiology, Sabah Al Ahmed Cardiac Centre, Al Amiri Hospital, Kuwait City, Kuwait
| | - Raja Dashti
- Department of Cardiology, Sabah Al Ahmed Cardiac Centre, Al Amiri Hospital, Kuwait City, Kuwait
| | - Ahmad Al Saber
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, G1 1XH, UK
| | - Jiazhu Pan
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, G1 1XH, UK
| | - Kobalava D. Zhanna
- Department of Internal Medicine with the Subspecialty of Cardiology and Functional Diagnostics Named After V.S. Moiseev, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russian Federation
| | - Hassan Abdelnaby
- Department of Endemic and Infectious Diseases, Faculty of Medicine, Suez Canal University, Ismailia, Egypt. Department of Medicine, Division of Gastroenterology, Al Sabah Hospital, Kuwait
| | - Wael Aboelhassan
- Department of Medicine, Division of Gastroenterology, Jaber Al Ahmed Hospital, South Surra, Kuwait
| | - Farah Almutairi
- Department of Medicine, Farwaniya Hospital, Farwaniya, Kuwait
| | - Naser Alotaibi
- Department of Medicine, Al Adan Hospital, Hadiya, Kuwait
| | | | | | - Bader Al-Bader
- Department of Medicine, Farwaniya Hospital, Farwaniya, Kuwait
| | - Haya Malhas
- Department of Emergency Medicine, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| | - Maryam Ramadhan
- Department of Obstetrics and Gynaecology, Maternity Hospital, Shuwaikh Medical Area, Kuwait
| | - Peter A. Brady
- Department of Cardiology, Illinois Masonic Medical Center, Chicago, IL, USA
| | - Ibrahim Al-Zakwani
- Department of Pharmacology & Clinical Pharmacy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman & Gulf Health Research, Muscat, Oman
| | - Parul Setiya
- Department of Agrometeorology, College of Agriculture, G.B.Pant University of Agriculture & Technology, Pantnagar, Uttarakhand, India
| | - Mohammed Abdullah
- Department of Infectious Diseases, Infectious Diseases Hospital, Shuwaikh Medical Area, Kuwait
| | - Moudhi Alroomi
- Department of Infectious Diseases, Infectious Diseases Hospital, Shuwaikh Medical Area, Kuwait
| | - Gary Tse
- Cardiovascular Analytics Group, Hong Kong, China; Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| |
Collapse
|
92
|
Lu LW, Gao Y, Quek SY, Foster M, Eason CT, Liu M, Wang M, Chen JH, Chen F. The landscape of potential health benefits of carotenoids as natural supportive therapeutics in protecting against Coronavirus infection. Biomed Pharmacother 2022; 154:113625. [PMID: 36058151 PMCID: PMC9428603 DOI: 10.1016/j.biopha.2022.113625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 01/08/2023] Open
Abstract
The Coronavirus Disease-2019 (COVID-19) pandemic urges researching possibilities for prevention and management of the effects of the virus. Carotenoids are natural phytochemicals of anti-oxidant, anti-inflammatory and immunomodulatory properties and may exert potential in aiding in combatting the pandemic. This review presents the direct and indirect evidence of the health benefits of carotenoids and derivatives based on in vitro and in vivo studies, human clinical trials and epidemiological studies and proposes possible mechanisms of action via which carotenoids may have the capacity to protect against COVID-19 effects. The current evidence provides a rationale for considering carotenoids as natural supportive nutrients via antioxidant activities, including scavenging lipid-soluble radicals, reducing hypoxia-associated superoxide by activating antioxidant enzymes, or suppressing enzymes that produce reactive oxygen species (ROS). Carotenoids may regulate COVID-19 induced over-production of pro-inflammatory cytokines, chemokines, pro-inflammatory enzymes and adhesion molecules by nuclear factor kappa B (NF-κB), renin-angiotensin-aldosterone system (RAS) and interleukins-6- Janus kinase-signal transducer and activator of transcription (IL-6-JAK/STAT) pathways and suppress the polarization of pro-inflammatory M1 macrophage. Moreover, carotenoids may modulate the peroxisome proliferator-activated receptors γ by acting as agonists to alleviate COVID-19 symptoms. They also may potentially block the cellular receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), human angiotensin-converting enzyme 2 (ACE2). These activities may reduce the severity of COVID-19 and flu-like diseases. Thus, carotenoid supplementation may aid in combatting the pandemic, as well as seasonal flu. However, further in vitro, in vivo and in particular long-term clinical trials in COVID-19 patients are needed to evaluate this hypothesis.
Collapse
|
93
|
Li F, Eteleeb AM, Buchser W, Sohn C, Wang G, Xiong C, Payne PR, McDade E, Karch CM, Harari O, Cruchaga C. Weakly activated core neuroinflammation pathways were identified as a central signaling mechanism contributing to the chronic neurodegeneration in Alzheimer's disease. Front Aging Neurosci 2022; 14:935279. [PMID: 36238934 PMCID: PMC9551568 DOI: 10.3389/fnagi.2022.935279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Objectives Neuroinflammation signaling has been identified as an important hallmark of Alzheimer's disease (AD) in addition to amyloid β plaques (Aβ) and neurofibrillary tangles (NFTs). However, the molecular mechanisms and biological processes of neuroinflammation remain unclear and have not well delineated using transcriptomics data available. Our objectives are to uncover the core neuroinflammation signaling pathways in AD using integrative network analysis on the transcriptomics data. Materials and methods From a novel perspective, i.e., investigating weakly activated molecular signals (rather than the strongly activated molecular signals), we developed integrative and systems biology network analysis to uncover potential core neuroinflammation signaling targets and pathways in AD using the two large-scale transcriptomics datasets, i.e., Mayo Clinic (77 controls and 81 AD samples) and ROSMAP (97 controls and 260 AD samples). Results Our analysis identified interesting core neuroinflammation signaling pathways, which are not systematically reported in the previous studies of AD. Specifically, we identified 7 categories of signaling pathways implicated on AD and related to virus infection: immune response, x-core signaling, apoptosis, lipid dysfunctional, biosynthesis and metabolism, and mineral absorption signaling pathways. More interestingly, most of the genes in the virus infection, immune response, and x-core signaling pathways are associated with inflammation molecular functions. The x-core signaling pathways were defined as a group of 9 signaling proteins: MAPK, Rap1, NF-kappa B, HIF-1, PI3K-Akt, Wnt, TGF-beta, Hippo, and TNF, which indicated the core neuroinflammation signaling pathways responding to the low-level and weakly activated inflammation and hypoxia and leading to the chronic neurodegeneration. It is interesting to investigate the detailed signaling cascades of these weakly activated neuroinflammation signaling pathways causing neurodegeneration in a chronic process, and consequently uncover novel therapeutic targets for effective AD treatment and prevention. Conclusions The potential core neuroinflammation and associated signaling targets and pathways were identified using integrative network analysis on two large-scale transcriptomics datasets of AD.
Collapse
Affiliation(s)
- Fuhai Li
- Institute for Informatics (I2), Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- NeuroGenomics and Informatics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Abdallah M. Eteleeb
- NeuroGenomics and Informatics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - William Buchser
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Christopher Sohn
- NeuroGenomics and Informatics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Guoqiao Wang
- Division of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Chengjie Xiong
- Division of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Philip R. Payne
- Institute for Informatics (I2), Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Eric McDade
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Celeste M. Karch
- NeuroGenomics and Informatics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Oscar Harari
- NeuroGenomics and Informatics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Carlos Cruchaga
- NeuroGenomics and Informatics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
94
|
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
|
95
|
Duan L, Reisch B, Mach P, Kimmig R, Gellhaus A, Iannaccone A. The immunological role of b7-h4 in pregnant women with sars-cov2 infection. Am J Reprod Immunol 2022; 88:e13626. [PMID: 36121927 PMCID: PMC9538547 DOI: 10.1111/aji.13626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022] Open
Abstract
Problem T‐cells are key players in fighting the coronavirus disease 2019 (COVID‐19). The checkpoint molecule B7‐H4, a member of the B7 family, can inhibit T‐cell activation and proliferation by inhibiting NF‐kb expression. We aimed to elucidate the immunological role of soluble B7‐H4 (sB7‐H4) and B7‐H4 in pregnant women suffered from an acute Sars‐Cov2 infection. Methods Expression levels of sB7‐H4 and cytokines were detected by enzyme linked immunosorbent assay. B7‐H4 and cytokines mRNA expression was analyzed by qPCR, and B7‐H4 and NF‐κb (p65) protein levels were investigated by western blot and immunofluorescence staining in placenta chorionic villous and decidual basalis tissues of COVID‐19 affected women and healthy controls. Results Fibrinoid necrosis in the periphery of placental villi was increased in the COVID‐19‐affected patients. sB7‐H4 protein in maternal and cord blood serum and IL‐6/IL‐10 were increased while leukocytes were decreased during SARS‐CoV‐2 infection. Serum sB7‐H4 level was increased according to the severity of SARS‐Cov‐2 infection. Cytokines (IL‐6, IL‐18, IL‐1β, TNF‐α), B7‐H4 mRNA and protein in the decidual basalis tissues of COVID‐19‐infected pregnant women were significantly increased compared to healthy controls. IL‐18 and IL‐1β were significantly increased in the placenta chorionic villous samples of COVID‐19 affected patients, while NF‐κb (p65) expression was decreased. Conclusions The expression of the immunological marker sB7‐H4 correlated with the severity of COVID‐19 disease in pregnant women. sB7‐H4 and B7‐H4 can be used to monitor the progression of COVID‐19 infection during pregnancy, and for evaluating of the maternal immune status.
Collapse
Affiliation(s)
- Liyan Duan
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany
| | - Beatrix Reisch
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany
| | - Pawel Mach
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany
| | - Antonella Iannaccone
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
96
|
Renalase Challenges the Oxidative Stress and Fibroproliferative Response in COVID-19. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4032704. [PMID: 36132227 PMCID: PMC9484957 DOI: 10.1155/2022/4032704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/03/2022] [Accepted: 08/24/2022] [Indexed: 01/08/2023]
Abstract
The hallmark of the coronavirus disease 2019 (COVID-19) pathophysiology was reported to be an inappropriate and uncontrolled immune response, evidenced by activated macrophages, and a robust surge of proinflammatory cytokines, followed by the release of reactive oxygen species, that synergistically result in acute respiratory distress syndrome, fibroproliferative lung response, and possibly even death. For these reasons, all identified risk factors and pathophysiological processes of COVID-19, which are feasible for the prevention and treatment, should be addressed in a timely manner. Accordingly, the evolving anti-inflammatory and antifibrotic therapy for severe COVID-19 and hindering post-COVID-19 fibrosis development should be comprehensively investigated. Experimental evidence indicates that renalase, a novel amino-oxidase, derived from the kidneys, exhibits remarkable organ protection, robustly addressing the most powerful pathways of cell trauma: inflammation and oxidative stress, necrosis, and apoptosis. As demonstrated, systemic renalase administration also significantly alleviates experimentally induced organ fibrosis and prevents adverse remodeling. The recognition that renalase exerts cytoprotection via sirtuins activation, by raising their NAD+ levels, provides a “proof of principle” for renalase being a biologically impressive molecule that favors cell protection and survival and maybe involved in the pathogenesis of COVID-19. This premise supports the rationale that renalase's timely supplementation may prove valuable for pathologic conditions, such as cytokine storm and related acute respiratory distress syndrome. Therefore, the aim for this review is to acknowledge the scientific rationale for renalase employment in the experimental model of COVID-19, targeting the acute phase mechanisms and halting fibrosis progression, based on its proposed molecular pathways. Novel therapies for COVID-19 seek to exploit renalase's multiple and distinctive cytoprotective mechanisms; therefore, this review should be acknowledged as the thorough groundwork for subsequent research of renalase's employment in the experimental models of COVID-19.
Collapse
|
97
|
Pandey A, Madan R, Singh S. Immunology to Immunotherapeutics of SARS-CoV-2: Identification of Immunogenic Epitopes for Vaccine Development. Curr Microbiol 2022; 79:306. [PMID: 36064873 PMCID: PMC9444117 DOI: 10.1007/s00284-022-03003-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 08/16/2022] [Indexed: 11/21/2022]
Abstract
The emergence of COVID19 pandemic caused by SARS-CoV-2 virus has created a global public health and socio-economic crisis. Immunoinformatics-based approaches to investigate the potential antigens is the fastest way to move towards a multiepitope-based vaccine development. This review encompasses the underlying mechanisms of pathogenesis, innate and adaptive immune signaling along with evasion pathways of SARS-CoV-2. Furthermore, it compiles the promiscuous peptides from in silico studies which are subjected to prediction of cytokine milieu using web-based servers. Out of the 434 peptides retrieved from all studies, we have identified 33 most promising T cell vaccine candidates. This review presents a list of the most potential epitopes from several proteins of the virus based on their immunogenicity, homology, conservancy and population coverage studies. These epitopes can form a basis of second generation of vaccine development as the first generation vaccines in various stages of trials mostly focus only on Spike protein. We therefore, propose them as most potential candidates which can be taken up immediately for confirmation by experimental studies.
Collapse
Affiliation(s)
- Apoorva Pandey
- Indian Council of Medical Research, V. Ramalingaswami Bhawan, Ansari Nagar, P.O. Box No. 4911, New Delhi, 110029 India
| | - Riya Madan
- Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Sector 81, Sahibzada Ajit Singh Nagar, Punjab 140306 India
| | - Swati Singh
- Department of Zoology, University of Delhi, Delhi, 110007 India
| |
Collapse
|
98
|
Mirmohammadi S, Kianmehr A, Sabbaghian A, Mohebbi A, Shahbazmohammadi H, Sheykharabi M, Bazzi Z. In silico drug repurposing against SARS-CoV-2 using an integrative transcriptomic profiling approach: Hydrocortisone and Benzhydrocodone as potential drug candidates against COVID-19. INFECTION, GENETICS AND EVOLUTION 2022; 103:105318. [PMID: 35718334 PMCID: PMC9212771 DOI: 10.1016/j.meegid.2022.105318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022]
Abstract
COVID-19 pathogenesis is mainly attributed to dysregulated antiviral immune response, the prominent hallmark of COVID-19. As no established drugs are available against SARS-CoV-2 and developing new ones would be a big challenge, repurposing of existing drugs holds promise against COVID-19. Here, we used a signature-based strategy to delve into cellular responses to SARS-CoV-2 infection in order to identify potential host contributors in COVID-19 pathogenesis and to find repurposable drugs using in silico approaches. We scrutinized transcriptomic profile of various human alveolar cell sources infected with SARS-CoV-2 to determine up-regulated genes specific to COVID-19. Enrichment analysis revealed that the up-regulated genes were involved mainly in viral infectious disease, immune system, and signal transduction pathways. Analysis of protein-protein interaction network and COVID-19 molecular pathway resulted in identifying several anti-viral proteins as well as 11 host pro-viral proteins, ADAR, HBEGF, MMP9, USP18, JUN, FOS, IRF2, ICAM1, IFI35, CASP1, and STAT3. Finally, molecular docking of up-regulated proteins and all FDA-approved drugs revealed that both Hydrocortisone and Benzhydrocodone possess high binding affinity for all pro-viral proteins. The suggested repurposed drugs should be subject to complementary in vitro and in vivo experiments in order to be evaluated in detail prior to clinical studies in potential management of COVID-19.
Collapse
Affiliation(s)
- SeyedehMozhdeh Mirmohammadi
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Anvarsadat Kianmehr
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Amir Sabbaghian
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Hamid Shahbazmohammadi
- Enzyme Technology Laboratory, Department of Biochemistry, Genetic and Metabolism Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Sheykharabi
- Department of Medical Nanotechnology, School of Advanced technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zahra Bazzi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
99
|
Tangos M, Budde H, Kolijn D, Sieme M, Zhazykbayeva S, Lódi M, Herwig M, Gömöri K, Hassoun R, Robinson EL, Meister TL, Jaquet K, Kovács Á, Mustroph J, Evert K, Babel N, Fagyas M, Lindner D, Püschel K, Westermann D, Mannherz HG, Paneni F, Pfaender S, Tóth A, Mügge A, Sossalla S, Hamdani N. SARS-CoV-2 infects human cardiomyocytes promoted by inflammation and oxidative stress. Int J Cardiol 2022; 362:196-205. [PMID: 35643215 PMCID: PMC9132721 DOI: 10.1016/j.ijcard.2022.05.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/11/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022]
Abstract
Introduction The respiratory illness triggered by severe acute respiratory syndrome virus-2 (SARS-CoV-2) is often particularly serious or fatal amongst patients with pre-existing heart conditions. Although the mechanisms underlying SARS-CoV-2-related cardiac damage remain elusive, inflammation (i.e. ‘cytokine storm’) and oxidative stress are likely involved. Methods and results Here we sought to determine: 1) if cardiomyocytes are targeted by SARS-CoV-2 and 2) how inflammation and oxidative stress promote the viral entry into cardiac cells. We analysed pro-inflammatory and oxidative stress and its impact on virus entry and virus-associated cardiac damage from SARS-CoV-2 infected patients and compared it to left ventricular myocardial tissues obtained from non-infected transplanted hearts either from end stage heart failure or non-failing hearts (donor group). We found that neuropilin-1 potentiates SARS-CoV-2 entry into human cardiomyocytes, a phenomenon driven by inflammatory and oxidant signals. These changes accounted for increased proteases activity and apoptotic markers thus leading to cell damage and apoptosis. Conclusion This study provides new insights into the mechanisms of SARS-CoV-2 entry into the heart and defines promising targets for antiviral interventions for COVID-19 patients with pre-existing heart conditions or patients with co-morbidities.
Collapse
Affiliation(s)
- Melina Tangos
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Detmar Kolijn
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Marcel Sieme
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Saltanat Zhazykbayeva
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Mária Lódi
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Melissa Herwig
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Kamilla Gömöri
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Emma Louise Robinson
- School of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| | - Toni Luise Meister
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Kornelia Jaquet
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Árpád Kovács
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Julian Mustroph
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Katja Evert
- Institute of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Nina Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Bochum, Germany
| | - Miklós Fagyas
- Center for Molecular Cardiology, University of Zürich, University Heart Center, Cardiology, University Hospital Zurich, Zürich, Switzerland
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Germany
| | - Klaus Püschel
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Germany
| | - Hans Georg Mannherz
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, University Heart Center, Cardiology, University Hospital Zurich, Zürich, Switzerland; University Heart Center, Cardiology, Department of Research and Education, University Hospital Zurich, Zürich, Switzerland
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, Debrecen, Hungary
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany; Clinic for Cardiology & Pneumology, Georg-August University Goettingen, DZHK (German Centre for Cardiovascular Research), partner site Goettingen, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
100
|
Ravichandran S, Manickam N, Kandasamy M. Liposome encapsulated clodronate mediated elimination of pathogenic macrophages and microglia: A promising pharmacological regime to defuse cytokine storm in COVID-19. MEDICINE IN DRUG DISCOVERY 2022; 15:100136. [PMID: 35721801 PMCID: PMC9190184 DOI: 10.1016/j.medidd.2022.100136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022] Open
Abstract
The emergence of new SARS-CoV-2 variants continues to pose an enormous public health concern. The SARS-CoV-2 infection disrupted host immune response accounting for cytokine storm has been linked to multiorgan failure and mortality in a significant portion of positive cases. Abruptly activated macrophages have been identified as the key pathogenic determinant of cytokine storm in COVID-19. Besides, reactive microglia have been known to discharge a surplus amount of proinflammatory factors leading to neuropathogenic events in the brains of SARS-CoV-2 infected individuals. Considering the fact, depletion of activated macrophages and microglia could be proposed to eradicate the life-threatening cytokine storm in COVID-19. Clodronate, a non-nitrogenous bisphosphonate drug has been identified as a potent macrophage and microglial depleting agent. While recent advancement in the field of liposome encapsulation technology offers the most promising biological tool for drug delivery, liposome encapsulated clodronate has been reported to effectively target and induce prominent phagocytic cell death in activated macrophages and microglia compared to free clodronate molecules. Thus, in this review article, we emphasize that depletion of activated macrophages and microglial cells by administration of liposome encapsulated clodronate can be a potential therapeutic strategy to diminish the pathogenic cytokine storm and alleviate multiorgan failure in COVID-19. Moreover, recently developed COVID-19 vaccines appear to render the chronic activation of macrophages accounting for immunological dysregulation in some cases. Therefore, the use of liposome encapsulated clodronate can also be extended to the clinical management of unforeseen immunogenic reactions resulting from activated macrophages associated adverse effects of COVID-19 vaccines.
Collapse
Affiliation(s)
- Sowbarnika Ravichandran
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Nivethitha Manickam
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
- University Grants Commission, Faculty Recharge Programme (UGC-FRP), New Delhi 110002, India
| |
Collapse
|