1
|
Al-Naimi MS, Abu-Raghif AR. Potential therapeutic and ameliorative effects of ramipril alone and in combination with methylprednisolone for the cytokine releasing syndrome in mice: An in vivo study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5845-5865. [PMID: 39614897 DOI: 10.1007/s00210-024-03659-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/19/2024] [Indexed: 04/11/2025]
Abstract
Cytokine-releasing syndrome (CRS) is a special form of systemic inflammatory response syndrome provoked by factors like viral infections and certain immunomodulatory drugs like monoclonal antibodies and adoptive T therapy. To elucidate the potential role of ramipril (RM) and its combination with methylprednisolone (MP) against the development and progression of CRS in mice. This experiment consists of two parts: protective and therapeutic interventions. The protective experiment: in the induction group, mice received an intraperitoneal injection (IP) of 5mg/kg lipopolysaccharide (LPS) without intervention. The other groups received various drugs before the induction by three days, then observed for an additional two days (50 mg/kg MP, 3 mg/kg RM, and a combination of 1.5 mg/kg RM with 25 mg/kg MP). The second part of the study involves the therapeutic potential; all groups received similar doses of drugs in the prevention groups, except LPS induction was given first, and after one hour, the mice received daily doses of the drugs for five days. At the end of the experiment, blood and tissue samples were obtained. Mice treated with RM and its combination with MP showed improved serum TNF-α, IL-6, IL-8, IL-1β, INF-γ, MDA, and GSH in both prevention and therapeutic groups. Histopathologically, mice treated with ramipril and its combination with MP ameliorate the tissue damage in both lung and liver tissues following LPS induction. Ramipril showed protective and therapeutic effects in LPS-induced cytokine storms in mice through anti-inflammatory and antioxidant mechanisms.
Collapse
Affiliation(s)
- Marwa Salih Al-Naimi
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq.
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Farahidi University, Baghdad, Iraq.
| | - Ahmed R Abu-Raghif
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| |
Collapse
|
2
|
Goleij P, Amini A, Tabari MAK, Hadipour M, Rezaee A, Daglia M, Aschner M, Sanaye PM, Kumar AP, Khan H. Unraveling the role of the IL-20 cytokine family in neurodegenerative diseases: Mechanisms and therapeutic insights. Int Immunopharmacol 2025; 152:114399. [PMID: 40068518 DOI: 10.1016/j.intimp.2025.114399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/03/2025] [Accepted: 02/27/2025] [Indexed: 03/24/2025]
Abstract
The IL-20 cytokine family, comprising IL-19, IL-20, IL-22, IL-24, and IL-26, has emerged as a critical player in the pathogenesis of neurodegenerative diseases due to its multiple roles in inflammation, tissue repair, and immune modulation. These cytokines signal through IL-20 receptor complexes (IL-20RA/IL-20RB and IL-22RA1/IL-20RB), triggering diverse immune processes. Recent evidence highlights their significant contributions to neuroinflammation and neurodegeneration in central nervous system disorders. IL-20 family cytokines impact microglial activation, which, when dysregulated, exacerbates neuronal damage. Specifically, IL-20 and IL-24 are linked to elevated pro-inflammatory markers in glial cells, promoting neurodegeneration. In contrast, IL-22 exhibits dual functionality, exerting protective and pathological roles depending on the inflammatory milieu. Key mechanisms involve the regulation of blood-brain barrier integrity, oxidative stress, and autophagy. IL-22 and IL-24 also protect neurons by enhancing antioxidant defenses and maintaining epithelial barrier function, while their dysregulation contributes to blood-brain barrier disruption and protein aggregate accumulation, hallmark features of Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Elevated IL-22 levels in Alzheimer's disease and IL-19's regulatory role in multiple sclerosis suggest they may serve as potential biomarkers and therapeutic targets. IL-26's role in amplifying inflammatory cascades further underscores the complexity of this cytokine family in neurodegenerative pathology. Therapeutically, strategies targeting IL-20 cytokines include monoclonal antibodies, receptor modulation, and recombinant cytokine administration. These approaches aim to mitigate neuroinflammation, restore immune balance, and protect neuronal integrity. This review underscores the IL-20 family's emerging relevance in neurodegenerative diseases, highlighting its potential for novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran; Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Alireza Amini
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran 4815733971, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran 4815733971, Iran
| | - Mahboube Hadipour
- Department of Biochemistry, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas 7919693116, Iran
| | - Aryan Rezaee
- Medical Doctor, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | - Pantea Majma Sanaye
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan 4513956184, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| |
Collapse
|
3
|
Tang X, Lu L, Li X, Huang P. Bridging Cancer and COVID-19: The Complex Interplay of ACE2 and TMPRSS2. Cancer Med 2025; 14:e70829. [PMID: 40145441 PMCID: PMC11947763 DOI: 10.1002/cam4.70829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic presents heightened risks for cancer patients, who are more susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and severe outcomes due to immunosuppression from both the malignancy and anticancer therapies. This review investigates the dual roles of angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) in SARS-CoV-2 infection among cancer patients. ACE2, the vital entry receptor for SARS-CoV-2, is overexpressed in certain tumors such as colon adenocarcinoma, renal carcinomas, pancreatic adenocarcinoma, and lung adenocarcinoma, potentially increasing viral susceptibility. Paradoxically, ACE2 also exhibits tumor-suppressive properties by inhibiting angiogenesis and modulating the tumor microenvironment, leading to improved patient prognoses in some cancers like breast cancer. TMPRSS2, essential for viral entry, shows decreased expression in several tumors but acts as a prognostic biomarker in prostate and lung cancers. This review illustrates the complexity of therapeutically targeting ACE2 and TMPRSS2 due to their contrasting roles in cancer progression and viral entry. We analyze the expression levels of ACE2 and TMPRSS2 in relation to immune cell infiltration and patient outcomes, and propose personalized therapeutic strategies. Furthermore, we underscore the necessity for multidisciplinary approaches, integrating antiviral treatments with cancer therapies and tailoring interventions based on individual molecular profiles. This approach to personalized medicine seeks to enhance treatment results and better manage cancer patients who have contracted SARS-CoV-2.
Collapse
Affiliation(s)
- Xuerui Tang
- School of Basic MedicineGannan Medical UniversityGanzhouJiangxiChina
| | - Liuzhi Lu
- School of Basic MedicineGannan Medical UniversityGanzhouJiangxiChina
| | - Xiaoping Li
- Clinical LaboratoryTongxiang First People's HospitalZhejiangChina
| | - Panpan Huang
- School of Basic MedicineGannan Medical UniversityGanzhouJiangxiChina
| |
Collapse
|
4
|
Ulrich-Merzenich GS, Shcherbakova A, Pizarro C, Skowasch D. Dexamethasone, Remdesivir and Azithromycin modulate ACE2 and IL-6 in Lung Epithelial Cells. Pneumologie 2025; 79:134-140. [PMID: 39284346 DOI: 10.1055/a-2372-3632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
BACKGROUND The optimal use of steroids in COVID-19 patients remains challenging. Current S3-guidelines "Recommendations for patients with COVID-19" recommend dexamethasone (DEX) for patients requiring respiratory support, remdesivir (RD) in the early disease phase and azythromycin (AZ) is no longer recommended. We investigated effects of DEX, RD and AZ in a lipopolysaccharide induced inflammation in lung cells in vitro and analyzed publicly available datasets with a focus on the Angiotensin-converting enzyme 2 (ACE2) to better understand drugs' mechanisms of action. METHODS human bronchial (Calu) and alveolar (A549) lung epithelial cells were treated with DEX, AZ or RDV in the presence of lipopolysaccharides (LPS). Gene expression (GE) of ACE2, IL-6 and the IL-6 protein release were measured. Publicly available GE data from lung tissues of COVID-19 patients and from lung cells treated with DEX were analyzed for the GE of ACE2. RESULTS DEX increased and RDV and AZ reduced the GE of ACE2 in LPS-stimulated bronchial and alveolar epithelial cells. Only DEX significantly reduced LPS-induced IL-6 releases in alveolar cells substantially. The database analyses showed an, albeit not always significant, increase in ACE2 for lung tissue or cell lines treated with DEX. Lung tissue from patients after COVID-19 infection as well as bronchial cell cultures after COVID-19 infection showed lower GEs of ACE2. DISCUSSION AND CONCLUSION DEX can increase ACE2 expression in vitro and thereby the portal of entry of SARS-CoV-2 into lung cells during an LPS induced inflammation. Simultaneously the inflammatory marker IL-6 is reduced. Comparative database analyses indicate that these processes can also take place in vivo.
Collapse
Affiliation(s)
| | - Anastasiia Shcherbakova
- Medical Clinic III, AG Synergy Research and Experimental Medicine, University Hospital Bonn, Bonn, Germany
| | - Carmen Pizarro
- Department Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Dirk Skowasch
- Department Internal Medicine II, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
5
|
Li Y, Dong C, Xing Y, Ma X, Ma Z, Zhang L, Du X, Feng L, Huo R, Wu QN, Li P, Hu F, Liu D, Dong Y, Cheng E, Tian X, Tian X. A Retrospective Data Audit of Outcome of Moderate and Severe Covid-19 Patients Who Had Received MP and Dex: A Single Center Study. Infect Drug Resist 2024; 17:5491-5505. [PMID: 39676848 PMCID: PMC11639968 DOI: 10.2147/idr.s418788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose To evaluate the necessity of the application of glucocorticoid (GC) in moderate COVID-19 patients, and which is the optimal choice between methylprednisolone (MP) and dexamethasone (DEX) in the clinical use of GC in different types of COVID-19 patients. Patients and Methods The study included patients with COVID-19 in Shanxi, China, from December 18, 2022, to March 1, 2023. The main clinical outcomes were 30-day mortality, disease exacerbations, and hospitalization days. Secondary outcomes included the demand for non-invasive ventilator-assisted ventilation (NIPPV)/invasive mechanical ventilation (IMV), the need for GC regimen escalation in follow-up treatment, duration of GC treatment, and complications including hyperglycemia and fungal infection. Results In moderate patients (N = 351), the rate of exacerbation and the need for GC regimen escalation in follow-up treatment was highest in the no-use GC group (P = 0.025, P = 0.01), the rate of fungal infections was highest in the DEX group (P = 0.038), and MP 40 mg/day or DEX 5 mg/day reduced exacerbations with consistent effects. In severe patients (N = 371), the two GC regimens do not affect their 30-day mortality and exacerbation rate, but the number of hospital days was significantly lower in the MP group compared with the DEX group (P < 0.001). Conclusion GC use is beneficial in mitigating exacerbations in moderate patients and in patients with moderate COVID-19. In severe patients, MP reduces the number of hospitalization days compared with DEX and may be a superior choice.
Collapse
Affiliation(s)
- Yupeng Li
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Chuanchuan Dong
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yanqing Xing
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xinkai Ma
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Zhen Ma
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Lulu Zhang
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xianglin Du
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Liting Feng
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Rujie Huo
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Qian Nan Wu
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Peiqi Li
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Fei Hu
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Dai Liu
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yanting Dong
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Erjing Cheng
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xinrui Tian
- The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xinli Tian
- Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Al-Naimi MS, Abu-Raghif AR, Fawzi HA. Novel therapeutic effects of rifaximin in combination with methylprednisolone for LPS-induced oxidative stress and inflammation in mice: An in vivo study. Toxicol Rep 2024; 13:101808. [PMID: 39640902 PMCID: PMC11617758 DOI: 10.1016/j.toxrep.2024.101808] [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: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 12/07/2024] Open
Abstract
Cytokine-releasing syndrome (CRS) is a special form of systemic inflammatory response syndrome provoked by factors like viral infections and certain immunomodulatory drugs. To elucidate the potential role of rifaximin (RIF) and its combination with methylprednisolone (MP) against the development and progression of CRS in mice. This experiment consists of two parts: protective and therapeutic interventions. The protective experiment: in the induction group, mice received an intraperitoneal injection (IP) of 5 mg/kg lipopolysaccharide (LPS) without intervention. The other group received various drugs before the induction by three days, then observed for an additional two days (50 mg/kg MP, 50 mg/kg RIF, and a combination of 25 mg/kg RIF with 25 mg/kg MP. The second part of the study involves the therapeutic potential; all groups received similar doses of drugs to that received in the prevention groups, except LPS induction was given first, and after one hour, the mice received daily doses of the drugs for five days. At the end of the experiment, blood and tissue samples were obtained. Mice treated with RIF and its combination with MP showed improved serum TNF-α, IL-6, IL-8, IL-1β, INF-γ, MDA, and GSH in both prevention and therapeutic groups. Histopathologically, mice treated with rifaximin and its combination with MP ameliorates the tissue damage in both lung and liver tissues following LPS induction. In conclusion, rifaximin showed protective and therapeutic effects in LPS-induced cytokine storms in mice through anti-inflammatory and antioxidant mechanisms, and its combination with methylprednisolone showed additive/ synergistic action.
Collapse
Affiliation(s)
- Marwa Salih Al-Naimi
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Farahidi University, Baghdad, Iraq
| | - Ahmed R. Abu-Raghif
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | | |
Collapse
|
7
|
Zhuang R, Xia H, Xu L, Liu Z, Zong K, Peng H, Liu B, Wu H, Huang L, Yang H, Luo C, Yin Y, Guo S. Corticosteroids for hospitalized patients with severe/critical COVID-19: a retrospective study in Chongqing, China. Sci Rep 2024; 14:24317. [PMID: 39414922 PMCID: PMC11484943 DOI: 10.1038/s41598-024-75926-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024] Open
Abstract
Corticosteroids have always been recommended for severe cases of COVID-19. However, the efficacy of treatment with corticosteroids for COVID-19 during the SARS-CoV-2 omicron outbreak in China has not been reported. Clinical data from 406 patients hospitalized for severe/critical COVID-19 from December 2022 to January 2023 at six hospitals in Chongqing were retrospectively analyzed. The primary outcome was all-cause mortality at 28 days in the groups with and without corticosteroids treatment after propensity score matching (PSM). Secondary outcomes were to compare in-hospital mortality and length of survival time with corticosteroids and those without corticosteroids. This study included 406 patients with severe or critical COVID-19, divided into the corticosteroids group (231, 56.9%) and non-corticosteroids group (175, 43.1%). After PSM, the use of corticosteroids did not reduce all-cause mortality at 28 days (42.5% vs. 39.1%). Univariate analysis showed that corticosteroids were not associated with improved all-cause mortality at 28 days [hazard ratio (HR), 1.019; 95% confidence interval (CI), 0.639-1.623; p = 0.938]. Multivariate analysis showed similar results (HR, 1.047; 95% CI, 0.633-1.732; p = 0.858). Among non-survivors, the survival time was significantly larger in those who received corticosteroids compared with the non-corticosteroid users [median 13 (IQR 6.5-15.5) vs. 6 (4-11.25), p = 0.007]. The use of systemic corticosteroids in severe/critical COVID-19 may provide certain potential survival benefits but does not improve prognosis.
Collapse
Affiliation(s)
- Rongjuan Zhuang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hongli Xia
- Department of General Practice, People's Hospital of Chongqing Heuchan, Chongqing, 401520, People's Republic of China
| | - Li Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Zhiqiang Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Kaican Zong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hailang Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Bin Liu
- Department of Respiratory and Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Zhuzhou Central Hospital, Central South University, Zhuzhou, 412007, People's Republic of China
| | - Huizi Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lan Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hongwei Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Chun Luo
- Department of Respiratory and Critical Care Medicine, Affiliated University Town Hospital of Chongqing Medical University, Chongqing, 401331, People's Republic of China
| | - Yuting Yin
- Department of Infectious Diseases, People's Hospital of Shapingba District, Chongqing, 400030, People's Republic of China
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
8
|
Lesur O, Segal ED, Rego K, Mercat A, Asfar P, Chagnon F. Process-Specific Blood Biomarkers and Outcomes in COVID-19 Versus Non-COVID-19 ARDS (APEL-COVID Study): A Prospective, Observational Cohort Study. J Clin Med 2024; 13:5919. [PMID: 39407979 PMCID: PMC11477790 DOI: 10.3390/jcm13195919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Severe acute respiratory syndrome (SARS) and acute respiratory distress syndrome (ARDS) are often considered separate clinico-radiological entities. Whether these conditions also present a single process-specific systemic biomolecular phenotype and how this relates to patient outcomes remains unknown. A prospective cohort study was conducted, including adult patients admitted to the ICU and general floors for COVID-19-related (COVID+) or non-COVID-19-related (COVID-) acute respiratory failure during the main phase of the pandemic. The primary objective was to study blood biomarkers and outcomes among different groups and severity subsets. Results: A total of 132 patients were included, as follows: 67 COVID+, 54 COVID- (with 11 matched control subjects for biomarker reference), and 58 of these patients allowed for further pre- and post-analysis. The baseline apelin (APL) levels were higher in COVID+ patients (p < 0.0001 vs. COVID- patients) and in SARS COVID+ patients (p ≤ 0.02 vs. ARDS), while the IL-6 levels were higher in ARDS COVID- patients (p ≤ 0.0001 vs. SARS). Multivariable logistic regression analyses with cohort biomarkers and outcome parameters revealed the following: (i) log-transformed neprilysin (NEP) activity was significantly higher in COVID+ patients (1.11 [95% CI: 0.4-1.9] vs. 0.37 [95% CI: 0.1-0.8], fold change (FC): 1.43 [95% CI: 1.04-1.97], p = 0.029) and in SARS patients (FC: 1.65 [95% CI: 1.05-2.6], p = 0.032 vs. non-SARS COVID+ patients, and 1.73 [95% CI: 1.19-2.5], p = 0.005 vs. ARDS COVID- patients) and (ii) higher lysyl oxidase (LOX) activity and APL levels were respectively associated with death and a shorter length of hospital stay in SARS COVID+ patients (Odds Ratios (OR): 1.01 [1.00-1.02], p = 0.05, and OR: -0.007 [-0.013-0.0001], p = 0.048). Conclusion: Process-specific blood biomarkers exhibited distinct profiles between COVID+ and COVID- patients, and across stages of severity. NEP and LOX activities, as well as APL levels, are particularly linked to COVID+ patients and their outcomes (ClinicalTrials.gov Identifier: NCT04632732).
Collapse
Affiliation(s)
- Olivier Lesur
- Centre de Recherche Clinique du CHU Sherbrooke (CRCHUS), Department of Intensive Care Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue Nord, Sherbrooke, QC J1H 5N4, Canada;
- Départements de Soins Intensifs et Service de Pneumologie, CHU Sherbrooke, 3001, 12th Avenue Nord, Sherbrooke QC J1H 5N4, Canada
- Département de Médecine, CHU Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Eric David Segal
- Département de Médecine, CHU Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Kevin Rego
- Centre de Recherche Clinique du CHU Sherbrooke (CRCHUS), Department of Intensive Care Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue Nord, Sherbrooke, QC J1H 5N4, Canada;
- Départements de Soins Intensifs et Service de Pneumologie, CHU Sherbrooke, 3001, 12th Avenue Nord, Sherbrooke QC J1H 5N4, Canada
- Département de Médecine, CHU Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Alain Mercat
- Département de Médecine Intensive-Réanimation, CHU Angers, 49000 Angers, France; (A.M.); (P.A.)
| | - Pierre Asfar
- Département de Médecine Intensive-Réanimation, CHU Angers, 49000 Angers, France; (A.M.); (P.A.)
| | - Frédéric Chagnon
- Centre de Recherche Clinique du CHU Sherbrooke (CRCHUS), Department of Intensive Care Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue Nord, Sherbrooke, QC J1H 5N4, Canada;
| |
Collapse
|
9
|
Dey D, Katso B, Nyame D, Issaka S, Adjei P. Management and associated outcomes of COVID-19 infection among Ghanaian autoimmune rheumatic disease patients. Ghana Med J 2024; 58:184-191. [PMID: 39398093 PMCID: PMC11465720 DOI: 10.4314/gmj.v58i3.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Objective This study assessed the prevalence of infection, management strategies and associated disease outcomes of COVID-19 among Autoimmune Rheumatic Disease (AIRD) patients in a teaching hospital in Ghana. Design This was a retrospective cross-sectional study. Setting Rheumatology Unit, Korle Bu Teaching Hospital. Participants Autoimmune Rheumatic Disease patients. Results Thirty-one (31) out of approximately 1700 AIRD patients in the unit tested positive for COVID-19, registering a COVID-19 prevalence of 1.82%. The majority, 25(80.6%), were females with a mean ± SD age of 41.7 ± 12.8 years. Systemic lupus erythematosus was the most affected autoimmune rheumatic condition, reporting fever as the commonest COVID-19-related symptom. Most participants, 22(71%), were managed by the "self-isolation"/home management" strategy. In comparison, 7(22.5%) were monitored at the hospital, with both strategies having resulted in complete recovery. The remaining 2(6.5%) patients who managed under "intensive care unit" strategy resulted in mortality. Conclusion These findings highlight the relatively low frequency of COVID-19 infection among AIRD patients, the encouraging recovery, and the low severe disease rates observed within this cohort. Additionally, the outcome of self-isolation and home management strategies underscore the importance of personalised approaches to COVID-19 management in this population. Funding None.
Collapse
Affiliation(s)
- Dzifa Dey
- Department of Medicine and Therapeutics, University of Ghana Medical School, Legon, Accra, Ghana
- Rheumatology Unit, Department of Medicine and Therapeutics, Korle-Bu Teaching Hospital, Accra, Ghana
| | - Bright Katso
- Rheumatology Unit, Department of Medicine and Therapeutics, Korle-Bu Teaching Hospital, Accra, Ghana
| | - Derrick Nyame
- Department of Medicine and Therapeutics, University of Ghana Medical School, Legon, Accra, Ghana
| | - Saudatu Issaka
- Rheumatology Unit, Department of Medicine and Therapeutics, Korle-Bu Teaching Hospital, Accra, Ghana
| | - Partrick Adjei
- Department of Medicine and Therapeutics, University of Ghana Medical School, Legon, Accra, Ghana
| |
Collapse
|
10
|
Xu L, Xu G, Sun N, Yao J, Wang C, Zhang W, Tian K, Liu M, Sun H. Catalpol ameliorates dexamethasone-induced osteoporosis by promoting osteogenic differentiation of bone marrow mesenchymal stem cells via the activation of PKD1 promoter. J Pharmacol Sci 2023; 153:221-231. [PMID: 37973220 DOI: 10.1016/j.jphs.2023.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVE To investigate the effects of CA on glucocorticoid-induced osteoporosis (GIOP) and lucubrate the underlying mechanism of CA via the activation of polycystic kidney disease-1(PKD1) in bone marrow mesenchymal stem cells (BMSCs). METHODS In vivo, a GIOP model in mice treated with dexamethasone (Dex) was established. Biomechanical, micro-CT, immunofluorescence staining of OCN, ALP and PKD1 and others were severally determined. qRT-PCR and Western blot methods were adopted to elucidate the particular mechanisms of CA on GIOP. In addition, BMSCs cultured in vitro were also induced by Dex to verify the effects of CA. Finally, siRNA and luciferase activity assays were performed to confirm the mechanisms. RESULTS We found that CA could restore the destroyed bone microarchitecture and increase the bone mass in GIOP mice. CA could also upregulate PKD1 protein expression, reduce oxidative stress, and promote mRNA expression of bone formation-associated markers in GIOP mice. Furthermore, it was also observed that CA reduced oxidative stress and promoted osteogenic differentiation in Dex-induced BMSCs. Mechanically, CA could promote protein expression via increasing the activity of PKD1 promoter. CONCLUSION This study provides important evidences for CA in the further clinical treatment of GIOP, reveals the activation of PKD1 promoter as the underlying mechanism.
Collapse
Affiliation(s)
- Lei Xu
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Office of Ethics Committee, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Gang Xu
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Na Sun
- Department of Pharmacy, The Third People's Hospital of Dalian, Dalian, China
| | - Jialin Yao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Wanhao Zhang
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Kang Tian
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Mozhen Liu
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Huijun Sun
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.
| |
Collapse
|
11
|
Xiao Y, Chang L, Ji H, Sun H, Song S, Feng K, Nuermaimaiti A, Halemubieke S, Mei L, Lu Z, Yan Y, Wang L. Posttranslational modifications of ACE2 protein: Implications for SARS-CoV-2 infection and beyond. J Med Virol 2023; 95:e29304. [PMID: 38063421 DOI: 10.1002/jmv.29304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/21/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023]
Abstract
The present worldwide pandemic of coronavirus disease 2019 (COVID-19) has highlighted the important function of angiotensin-converting enzyme 2 (ACE2) as a receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry. A deeper understanding of ACE2 could offer insights into the mechanisms of SARS-CoV-2 infection. While ACE2 is subject to regulation by various factors in vivo, current research in this area is insufficient to fully elucidate the corresponding pathways of control. Posttranslational modification (PTM) is a powerful tool for broadening the variety of proteins. The PTM study of ACE2 will help us to make up for the deficiency in the regulation of protein synthesis and translation. However, research on PTM-related aspects of ACE2 remains limited, mostly focused on glycosylation. Accordingly, a comprehensive review of ACE2 PTMs could help us better understand the infection process and provide a basis for the treatment of COVID-19 and beyond.
Collapse
Affiliation(s)
- Yingzi Xiao
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Le Chang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Huimin Ji
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Huizhen Sun
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Shi Song
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Kaihao Feng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Abudulimutailipu Nuermaimaiti
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Shana Halemubieke
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Ling Mei
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Zhuoqun Lu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Ying Yan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| | - Lunan Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, People's Republic of China
| |
Collapse
|
12
|
Liu X, Guo Y, Pan W, Xue Q, Fu J, Qu G, Zhang A. Exogenous Chemicals Impact Virus Receptor Gene Transcription: Insights from Deep Learning. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:18038-18047. [PMID: 37186679 DOI: 10.1021/acs.est.2c09837] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Despite the fact that coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been disrupting human life and health worldwide since the outbreak in late 2019, the impact of exogenous substance exposure on the viral infection remains unclear. It is well-known that, during viral infection, organism receptors play a significant role in mediating the entry of viruses to enter host cells. A major receptor of SARS-CoV-2 is the angiotensin-converting enzyme 2 (ACE2). This study proposes a deep learning model based on the graph convolutional network (GCN) that enables, for the first time, the prediction of exogenous substances that affect the transcriptional expression of the ACE2 gene. It outperforms other machine learning models, achieving an area under receiver operating characteristic curve (AUROC) of 0.712 and 0.703 on the validation and internal test set, respectively. In addition, quantitative polymerase chain reaction (qPCR) experiments provided additional supporting evidence for indoor air pollutants identified by the GCN model. More broadly, the proposed methodology can be applied to predict the effect of environmental chemicals on the gene transcription of other virus receptors as well. In contrast to typical deep learning models that are of black box nature, we further highlight the interpretability of the proposed GCN model and how it facilitates deeper understanding of gene change at the structural level.
Collapse
Affiliation(s)
- Xian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Yunhe Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Wenxiao Pan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Qiao Xue
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Jianjie Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100190, P. R. China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, P.R. China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Aiqian Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100190, P. R. China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, P.R. China
| |
Collapse
|
13
|
Ogarek N, Oboza P, Olszanecka-Glinianowicz M, Kocelak P. SARS-CoV-2 infection as a potential risk factor for the development of cancer. Front Mol Biosci 2023; 10:1260776. [PMID: 37753372 PMCID: PMC10518417 DOI: 10.3389/fmolb.2023.1260776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
The COVID-19 pandemic has a significant impact on public health and the estimated number of excess deaths may be more than three times higher than documented in official statistics. Numerous studies have shown an increased risk of severe COVID-19 and death in patients with cancer. In addition, the role of SARS-CoV-2 as a potential risk factor for the development of cancer has been considered. Therefore, in this review, we summarise the available data on the potential effects of SARS-CoV-2 infection on oncogenesis, including but not limited to effects on host signal transduction pathways, immune surveillance, chronic inflammation, oxidative stress, cell cycle dysregulation, potential viral genome integration, epigenetic alterations and genetic mutations, oncolytic effects and reactivation of dormant cancer cells. We also investigated the potential long-term effects and impact of the antiviral therapy used in COVID-19 on cancer development and its progression.
Collapse
Affiliation(s)
- Natalia Ogarek
- Pathophysiology Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
| | - Paulina Oboza
- Students’ Scientific Society at the Pathophysiology Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
| | - Magdalena Olszanecka-Glinianowicz
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
| | - Piotr Kocelak
- Pathophysiology Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
| |
Collapse
|
14
|
Nayak SS, Naidu A, Sudhakaran SL, Vino S, Selvaraj G. Prospects of Novel and Repurposed Immunomodulatory Drugs against Acute Respiratory Distress Syndrome (ARDS) Associated with COVID-19 Disease. J Pers Med 2023; 13:664. [PMID: 37109050 PMCID: PMC10142859 DOI: 10.3390/jpm13040664] [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: 02/13/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is intricately linked with SARS-CoV-2-associated disease severity and mortality, especially in patients with co-morbidities. Lung tissue injury caused as a consequence of ARDS leads to fluid build-up in the alveolar sacs, which in turn affects oxygen supply from the capillaries. ARDS is a result of a hyperinflammatory, non-specific local immune response (cytokine storm), which is aggravated as the virus evades and meddles with protective anti-viral innate immune responses. Treatment and management of ARDS remain a major challenge, first, because the condition develops as the virus keeps replicating and, therefore, immunomodulatory drugs are required to be used with caution. Second, the hyperinflammatory responses observed during ARDS are quite heterogeneous and dependent on the stage of the disease and the clinical history of the patients. In this review, we present different anti-rheumatic drugs, natural compounds, monoclonal antibodies, and RNA therapeutics and discuss their application in the management of ARDS. We also discuss on the suitability of each of these drug classes at different stages of the disease. In the last section, we discuss the potential applications of advanced computational approaches in identifying reliable drug targets and in screening out credible lead compounds against ARDS.
Collapse
Affiliation(s)
- Smruti Sudha Nayak
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Akshayata Naidu
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Sajitha Lulu Sudhakaran
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Sundararajan Vino
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Gurudeeban Selvaraj
- Centre for Research in Molecular Modeling, Department of Chemistry and Biochemistry, Concordia University-Loyola Campus, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
15
|
Sharma G, Song LF, Merz KM. Effect of an Inhibitor on the ACE2-Receptor-Binding Domain of SARS-CoV-2. J Chem Inf Model 2022; 62:6574-6585. [PMID: 35118864 PMCID: PMC8848506 DOI: 10.1021/acs.jcim.1c01283] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Indexed: 01/07/2023]
Abstract
The recent outbreak of COVID-19 infection started in Wuhan, China, and spread across China and beyond. Since the WHO declared COVID-19 a pandemic (March 11, 2020), three vaccines and only one antiviral drug (remdesivir) have been approved (Oct 22, 2020) by the FDA. The coronavirus enters human epithelial cells by the binding of the densely glycosylated fusion spike protein (S protein) to a receptor (angiotensin-converting enzyme 2, ACE2) on the host cell surface. Therefore, inhibiting the viral entry is a promising treatment pathway for preventing or ameliorating the effects of COVID-19 infection. In the current work, we have used all-atom molecular dynamics (MD) simulations to investigate the influence of the MLN-4760 inhibitor on the conformational properties of ACE2 and its interaction with the receptor-binding domain (RBD) of SARS-CoV-2. We have found that the presence of an inhibitor tends to completely/partially open the ACE2 receptor where the two subdomains (I and II) move away from each other, while the absence results in partial or complete closure. The current study increases our understanding of ACE inhibition by MLN-4760 and how it modulates the conformational properties of ACE2.
Collapse
Affiliation(s)
- Gaurav Sharma
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Lin Frank Song
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kenneth M. Merz
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
16
|
The effectiveness of dexamethasone as a combination therapy for COVID-19. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2022; 72:345-358. [PMID: 36651541 DOI: 10.2478/acph-2022-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/28/2022] [Indexed: 01/26/2023]
Abstract
Coronavirus disease 2019 (COVID-19) was reported as a global pandemic in March 2020 after invading many countries and leaving behind tens of thousands of infected patients in a brief time span. Approval of a few vaccines has been obtained and their efficacy of varying degrees established. Still, there is no effective pharmaceutical agent for the treatment of COVID-19 though several drugs are undergoing clinical trials. Recent studies have shown that dexamethasone, a corticosteroid, can reduce the rate of COVID-19-related mortality in the intensive care unit by 35 % for patients who are on mechanical ventilation. Although variable efficacy of other combination therapies has been reported for treating COVID-19 associated with acute respiratory distress syndrome (ARDS), dexamethasone is an extensively used drug in many treatment regimens against COVID-19. The current review aims to explore the role of dexamethasone as an efficient combination treatment for COVID-19.
Collapse
|
17
|
Liang Y, Mao X, Kuang M, Zhi J, Zhang Z, Bo M, Zhang G, Lin P, Wang W, Shen Z. IL-6 affects the severity of olfactory disorder: A cross-sectional survey of 148 patients who recovered from Omicron infection using the Sniffin' Sticks test in Tianjin, China. Int J Infect Dis 2022; 123:17-24. [PMID: 35931372 DOI: 10.1016/j.ijid.2022.07.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The mechanism of olfactory disorder (OD) in patients with COVID-19 is unclear. Our study aimed to elucidate the relationships between inflammatory factors and OD in a sample of Omicron-infected patients with a high rate of vaccination in China. METHODS The Sniffin' Sticks 12-item test (SST-12) was performed in a cross-sectional study of 148 recovered Omicron-infected patients to evaluate OD severity. We compared demographic, laboratory, and clinical data. RESULTS One hundred forty-eight Omicron-infected patients were enrolled. One hundred twenty-nine cases of OD were detected. Increased inflammation contributed to OD severity, especially in the adult group. OD was shown to be aggravated by an increase in IL-6 levels. The adjusted odds ratio was 2.22 (95% CI: 0.98-5.05, P=0.056) after adjustment for age, sex and vaccine characteristics. CONCLUSIONS These findings indicated that the prevalence of OD remains high in vaccinated Omicron-infected patients and that SST-12 might be a feasible method to screen for OD. IL-6 may play a role in the biochemical and pathological processes underlying OD.
Collapse
Affiliation(s)
- Yibo Liang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Xiang Mao
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Manbao Kuang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Jingtai Zhi
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Ziyue Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Mingyu Bo
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Guimin Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Peng Lin
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China; Institute of Otolaryngology of Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China; Key Medical Discipline of Tianjin (Otolaryngology), China; Quality Control Centre of Otolaryngology, Tianjin, China.
| | - Zhongyang Shen
- Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China.
| |
Collapse
|
18
|
Faist A, Janowski J, Kumar S, Hinse S, Çalışkan DM, Lange J, Ludwig S, Brunotte L. Virus Infection and Systemic Inflammation: Lessons Learnt from COVID-19 and Beyond. Cells 2022; 11:2198. [PMID: 35883640 PMCID: PMC9316821 DOI: 10.3390/cells11142198] [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: 06/02/2022] [Revised: 06/28/2022] [Accepted: 07/09/2022] [Indexed: 02/06/2023] Open
Abstract
Respiratory infections with newly emerging zoonotic viruses such as SARS-CoV-2, the etiological agent of COVID-19, often lead to the perturbation of the human innate and adaptive immune responses causing severe disease with high mortality. The responsible mechanisms are commonly virus-specific and often include either over-activated or delayed local interferon responses, which facilitate efficient viral replication in the primary target organ, systemic viral spread, and rapid onset of organ-specific and harmful inflammatory responses. Despite the distinct replication strategies, human infections with SARS-CoV-2 and highly pathogenic avian influenza viruses demonstrate remarkable similarities and differences regarding the mechanisms of immune induction, disease dynamics, as well as the long-term sequelae, which will be discussed in this review. In addition, we will highlight some important lessons about the effectiveness of antiviral and immunomodulatory therapeutic strategies that this pandemic has taught us.
Collapse
Affiliation(s)
- Aileen Faist
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- CiM-IMPRS, International Max Planck Research School—Molecular Biomedicine, Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany
| | - Josua Janowski
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- SP BioSciences Graduate Program, University of Muenster, 48149 Muenster, Germany
| | - Sriram Kumar
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- EvoPAD Research Training Group 2220, University of Muenster, 48149 Muenster, Germany
| | - Saskia Hinse
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
| | - Duygu Merve Çalışkan
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- EvoPAD Research Training Group 2220, University of Muenster, 48149 Muenster, Germany
| | - Julius Lange
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
| | - Stephan Ludwig
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- CiM-IMPRS, International Max Planck Research School—Molecular Biomedicine, Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany
- EvoPAD Research Training Group 2220, University of Muenster, 48149 Muenster, Germany
- Interdisciplinary Center for Clinical Research, University of Muenster, 48149 Muenster, Germany
| | - Linda Brunotte
- Institute of Virology, University of Muenster, 48149 Muenster, Germany; (A.F.); (J.J.); (S.K.); (S.H.); (D.M.Ç.); (J.L.); (S.L.)
- Interdisciplinary Center for Clinical Research, University of Muenster, 48149 Muenster, Germany
| |
Collapse
|
19
|
Thakur A, Sharma G, Badavath VN, Jayaprakash V, Merz KM, Blum G, Acevedo O. Primer for Designing Main Protease (M pro) Inhibitors of SARS-CoV-2. J Phys Chem Lett 2022; 13:5776-5786. [PMID: 35726889 PMCID: PMC9235046 DOI: 10.1021/acs.jpclett.2c01193] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/13/2022] [Indexed: 05/08/2023]
Abstract
The COVID-19 outbreak has been devastating, with hundreds of millions of infections and millions of deaths reported worldwide. In response, the application of structure-activity relationships (SAR) upon experimentally validated inhibitors of SARS-CoV-2 main protease (Mpro) may provide an avenue for the identification of new lead compounds active against COVID-19. Upon the basis of information gleaned from a combination of reported crystal structures and the docking of experimentally validated inhibitors, four "rules" for designing potent Mpro inhibitors have been proposed. The aim here is to guide medicinal chemists toward the most probable hits and to provide guidance on repurposing available structures as Mpro inhibitors. Experimental examination of our own previously reported inhibitors using the four "rules" identified a potential lead compound, the cathepsin inhibitor GB111-NH2, that was 2.3 times more potent than SARS-CoV-2 Mpro inhibitor N3.
Collapse
Affiliation(s)
- Abhishek Thakur
- Department
of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Gaurav Sharma
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Vishnu Nayak Badavath
- School
of Pharmacy & Technology Management, SVKM’s Narsee Monjee Institute of Management Studies (NMIMS), Hyderabad 509301, India
- Department
of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835 215, India
| | - Venkatesan Jayaprakash
- Department
of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835 215, India
| | - Kenneth M. Merz
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Galia Blum
- Institute
for Drug Research, The Hebrew University
of Jerusalem, Jerusalem, 9112001, Israel
| | - Orlando Acevedo
- Department
of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| |
Collapse
|
20
|
Pouremamali A, Babaei A, Malekshahi SS, Abbasi A, Rafiee N. Understanding the pivotal roles of ACE2 in SARS-CoV-2 infection: from structure/function to therapeutic implication. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022; 23:103. [PMID: 37521846 PMCID: PMC9206724 DOI: 10.1186/s43042-022-00314-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/12/2022] [Indexed: 01/08/2023] Open
Abstract
In December 2019, a novel respiratory tract infection, from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in China that rapidly spread around the world. This virus possesses spike (S) glycoproteins on the surface of mature virions, like other members of coronaviridae. The S glycoprotein is a crucial viral protein for binding, fusion, and entry into the target cells. Binding the receptor-binding domain (RBD) of S protein to angiotensin-converting enzyme 2 (ACE 2), a cell-surface receptor, mediates virus entry into cells; thus, understanding the basics of ACE2 and S protein, their interactions, and ACE2 targeting could be a potent priority for inhibition of virus infection. This review presents current knowledge of the SARS-CoV-2 basics and entry mechanism, structure and organ distribution of ACE2, and also its function in SARS-CoV-2 entry and pathogenesis. Furthermore, it highlights ACE2 targeting by recombinant ACE2 (rACE2), ACE2 activators, ACE inhibitor, and angiotensin II (Ang II) receptor blocker to control the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Amir Pouremamali
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abouzar Babaei
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nastaran Rafiee
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
21
|
Yeh JJ, Lai M, Lin CL, Lu KH, Kao CH. Effects of statins on the risks of ischemic stroke and heart disease in human immunodeficiency virus infection, influenza and severe acute respiratory syndrome-associated coronavirus: respiratory virus infection with steroid use. Postgrad Med 2022; 134:589-597. [PMID: 35590450 DOI: 10.1080/00325481.2022.2080359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES We sought to fill the research gap on the effects of statins on the risks of ischemic stroke and heart disease among individuals with human immunodeficiency virus infection, influenza, and severe acute respiratory syndrome associated-coronavirus (HIS) disorders. METHODS We enrolled a HIS cohort treated with statins (n = 4921) and a HIS cohort not treated with statins (n = 4921). The cumulative incidence of ischemic stroke and heart disease was analyzed using a time-dependent Cox proportional regression analysis. We analyzed the adjusted hazard ratio (aHR) and 95% confidence interval (CI) of ischemic stroke and heart disease for statins users relative to nonusers based on sex, age, comorbidities and medications. RESULTS The aHR (95% CI) was 0.38 (0.22-0.65) for ischemic stroke. The aHR (95% CI) of heart disease was 0.50 (0.46-0.55). The aHRs (95% CI) of statin users with low, medium, and high adherence (statin use covering <33%, 33%-66%, and >66%, respectively, of the study period) for the risks of ischemic stroke were 0.50 (0.27-0.92), 0.31 (0.10-1.01), and 0.16 (0.04-0.68) and for heart disease were 0.56 (0.51-0.61), 0.40 (0.33-0.48), and 0.44 (0.38-0.51), respectively, compared with statin nonusers. CONCLUSION Statin use was associated with lower aHRs for ischemic stroke and heart disease in those with HIS disorders with comorbidities.
Collapse
Affiliation(s)
- Jun-Jun Yeh
- Department of Family Medicine, Geriatric Medicine, and Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Meichu Lai
- Department of Laboratory Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Cheng-Li Lin
- College of Medicine, China Medical University, Taichung, Taiwan.,Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Kuan-Hua Lu
- Department of Family Medicine, Geriatric Medicine, and Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Chia-Hung Kao
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan.,Center of Augmented Intelligence in Healthcare, China Medical University Hospital, Taichung, Taiwan.,Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
22
|
Wang SC, Zhang F, Zhu H, Yang H, Liu Y, Wang P, Parpura V, Wang YF. Potential of Endogenous Oxytocin in Endocrine Treatment and Prevention of COVID-19. Front Endocrinol (Lausanne) 2022; 13:799521. [PMID: 35592777 PMCID: PMC9110836 DOI: 10.3389/fendo.2022.799521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/14/2022] [Indexed: 01/09/2023] Open
Abstract
Coronavirus disease 2019 or COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a significant threat to the health of human beings. While wearing mask, maintaining social distance and performing self-quarantine can reduce virus spreading passively, vaccination actively enhances immune defense against COVID-19. However, mutations of SARS-CoV-2 and presence of asymptomatic carriers frustrate the effort of completely conquering COVID-19. A strategy that can reduce the susceptibility and thus prevent COVID-19 while blocking viral invasion and pathogenesis independent of viral antigen stability is highly desirable. In the pathogenesis of COVID-19, endocrine disorders have been implicated. Correspondingly, many hormones have been identified to possess therapeutic potential of treating COVID-19, such as estrogen, melatonin, corticosteroids, thyroid hormone and oxytocin. Among them, oxytocin has the potential of both treatment and prevention of COVID-19. This is based on oxytocin promotion of immune-metabolic homeostasis, suppression of inflammation and pre-existing comorbidities, acceleration of damage repair, and reduction of individuals' susceptibility to pathogen infection. Oxytocin may specifically inactivate SARS-COV-2 spike protein and block viral entry into cells via angiotensin-converting enzyme 2 by suppressing serine protease and increasing interferon levels and number of T-lymphocytes. In addition, oxytocin can promote parasympathetic outflow and the secretion of body fluids that could dilute and even inactivate SARS-CoV-2 on the surface of cornea, oral cavity and gastrointestinal tract. What we need to do now is clinical trials. Such trials should fully balance the advantages and disadvantages of oxytocin application, consider the time- and dose-dependency of oxytocin effects, optimize the dosage form and administration approach, combine oxytocin with inhibitors of SARS-CoV-2 replication, apply specific passive immunization, and timely utilize efficient vaccines. Meanwhile, blocking COVID-19 transmission chain and developing other efficient anti-SARS-CoV-2 drugs are also important. In addition, relative to the complex issues with drug applications over a long term, oxytocin can be mobilized through many physiological stimuli, and thus used as a general prevention measure. In this review, we explore the potential of oxytocin for treatment and prevention of COVID-19 and perhaps other similar pathogens.
Collapse
Affiliation(s)
- Stephani C. Wang
- Division of Cardiology, Department of Medicine, University of California-Irvine, Irvine, CA, United States
| | - Fengmin Zhang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hui Zhu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Haipeng Yang
- Neonatal Division of the Department of Pediatrics, Harbin Medical University The Fourth Affiliated Hospital, Harbin, China
| | - Yang Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Ping Wang
- Department of Genetics, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
23
|
Dai J, Wang H, Liao Y, Tan L, Sun Y, Song C, Liu W, Qiu X, Ding C. Coronavirus Infection and Cholesterol Metabolism. Front Immunol 2022; 13:791267. [PMID: 35529872 PMCID: PMC9069556 DOI: 10.3389/fimmu.2022.791267] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/21/2022] [Indexed: 12/19/2022] Open
Abstract
Host cholesterol metabolism remodeling is significantly associated with the spread of human pathogenic coronaviruses, suggesting virus-host relationships could be affected by cholesterol-modifying drugs. Cholesterol has an important role in coronavirus entry, membrane fusion, and pathological syncytia formation, therefore cholesterol metabolic mechanisms may be promising drug targets for coronavirus infections. Moreover, cholesterol and its metabolizing enzymes or corresponding natural products exert antiviral effects which are closely associated with individual viral steps during coronavirus replication. Furthermore, the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 infections are associated with clinically significant low cholesterol levels, suggesting cholesterol could function as a potential marker for monitoring viral infection status. Therefore, weaponizing cholesterol dysregulation against viral infection could be an effective antiviral strategy. In this review, we comprehensively review the literature to clarify how coronaviruses exploit host cholesterol metabolism to accommodate viral replication requirements and interfere with host immune responses. We also focus on targeting cholesterol homeostasis to interfere with critical steps during coronavirus infection.
Collapse
Affiliation(s)
- Jun Dai
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Experimental Animal Center, Zunyi Medical University, Zunyi City, China
| | - Huan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lei Tan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yingjie Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Cuiping Song
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Weiwei Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- *Correspondence: Xusheng Qiu, ; Chan Ding,
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- *Correspondence: Xusheng Qiu, ; Chan Ding,
| |
Collapse
|
24
|
Simon M, Le Borgne P, Lefevbre F, Chabrier S, Cipolat L, Remillon A, Baicry F, Bilbault P, Lavoignet CE, Abensur Vuillaume L. Lymphopenia and Early Variation of Lymphocytes to Predict In-Hospital Mortality and Severity in ED Patients with SARS-CoV-2 Infection. J Clin Med 2022; 11:1803. [PMID: 35407409 PMCID: PMC8999889 DOI: 10.3390/jcm11071803] [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: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 01/27/2023] Open
Abstract
(1) Introduction: Multiple studies have demonstrated that lymphocyte count monitoring is a valuable prognostic tool for clinicians during inflammation. The aim of our study was to determine the prognostic value of delta lymphocyte H24 from admission from the emergency department for mortality and severity of SARS-CoV-2 infection. (2) Methods: We have made a retrospective and multicentric study in six major hospitals of northeastern France. The patients were hospitalized and had a confirmed diagnosis of SARS-CoV-2 infection. (3): Results: A total of 1035 patients were included in this study. Factors associated with infection severity were CRP > 100 mg/L (OR: 2.51, CI 95%: (1.40−3.71), p < 0.001) and lymphopenia < 800/mm3 (OR: 2.15, CI 95%: (1.42−3.27), p < 0.001). In multivariate analysis, delta lymphocytes H24 (i.e., the difference between lymphocytes values at H24 and upon admission to the ED) < 135 was one of the most significant biochemical factors associated with mortality (OR: 2.23, CI 95%: (1.23−4.05), p = 0.009). The most accurate threshold for delta lymphocytes H24 was 75 to predict severity and 135 for mortality. (4) Conclusion: Delta lymphocytes H24 could be a helpful early screening prognostic biomarker to predict severity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Maxence Simon
- Emergency Department, University Hospital of Strasbourg, 67000 Strasbourg, France; (M.S.); (P.L.B.); (S.C.); (F.B.); (P.B.)
| | - Pierrick Le Borgne
- Emergency Department, University Hospital of Strasbourg, 67000 Strasbourg, France; (M.S.); (P.L.B.); (S.C.); (F.B.); (P.B.)
- Regenerative NanoMedicine (RNM), Fédération de Médecine Translationnelle (FMTS), INSERM (French National Institute of Health and Medical Research), UMR 1260, Strasbourg University, 67000 Strasbourg, France
| | - François Lefevbre
- Department of Public Health, University Hospital of Strasbourg, 67000 Strasbourg, France;
| | - Sylvie Chabrier
- Emergency Department, University Hospital of Strasbourg, 67000 Strasbourg, France; (M.S.); (P.L.B.); (S.C.); (F.B.); (P.B.)
| | - Lauriane Cipolat
- Service d’Accueil des Urgences SAMU 57, CHR Metz-Thionville, 57085 Metz, France; (L.C.); (A.R.)
| | - Aline Remillon
- Service d’Accueil des Urgences SAMU 57, CHR Metz-Thionville, 57085 Metz, France; (L.C.); (A.R.)
| | - Florent Baicry
- Emergency Department, University Hospital of Strasbourg, 67000 Strasbourg, France; (M.S.); (P.L.B.); (S.C.); (F.B.); (P.B.)
| | - Pascal Bilbault
- Emergency Department, University Hospital of Strasbourg, 67000 Strasbourg, France; (M.S.); (P.L.B.); (S.C.); (F.B.); (P.B.)
| | | | - Laure Abensur Vuillaume
- Service d’Accueil des Urgences SAMU 57, CHR Metz-Thionville, 57085 Metz, France; (L.C.); (A.R.)
| |
Collapse
|
25
|
Iwabuchi S, Miyamoto K, Hatai M, Mikasa Y, Katsuda M, Murata SI, Kondo T, Yamaue H, Hashimoto S. Immune Cells Profiles in the Different Sites of COVID-19-Affected Lung Lobes in a Single Patient. Front Med (Lausanne) 2022; 9:841170. [PMID: 35252273 PMCID: PMC8888686 DOI: 10.3389/fmed.2022.841170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Whereas the COVID-19 disease pathophysiology is under investigation, it is important to identify the pathways of viral transmission and inflammation from the pre-illness to the disease-onset stages. We analyzed five lung lobes from a patient with COVID-19 who finally died after prolonged lung protective ventilation. Pathological examination revealed moderate inflammation in upper lung lobes and uneven yet severe inflammation and diffuse alveolar damage in lower lung lobes. SARS-CoV-2 was detected at higher levels not in severely, but rather moderately inflamed middle lung lobes, and immunohistochemistry and bulk RNA-sequencing results showed that immune cells were detected at higher levels in lower lung lobes. The mRNA expression of cytokine families varied. We found an increase in keratin 5- or aquaporin 3-expressing basal cells in the severely inflamed lower lung lobes, and the alveolar stromal tissues were filled with them. Thus, this analysis of lung samples from a patient helps to determine the COVID-19 pathophysiology at a specific time point, and the virus localization and inflammatory responses at each site of the lungs provide various important indications.
Collapse
Affiliation(s)
- Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kyohei Miyamoto
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mayuko Hatai
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yurina Mikasa
- Departments of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | - Masahiro Katsuda
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shin-Ichi Murata
- Departments of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
26
|
Gelzo M, Scialò F, Cacciapuoti S, Pinchera B, De Rosa A, Cernera G, Comegna M, Tripodi L, Schiano Moriello N, Mormile M, Fabbrocini G, Parrella R, Corso G, Gentile I, Castaldo G. Inducible Nitric Oxide Synthase (iNOS): Why a Different Production in COVID-19 Patients of the Two Waves? Viruses 2022; 14:534. [PMID: 35336941 PMCID: PMC8948744 DOI: 10.3390/v14030534] [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: 01/11/2022] [Revised: 02/17/2022] [Accepted: 03/04/2022] [Indexed: 12/18/2022] Open
Abstract
Profound clinical differences between the first and second waves of COVID-19 were observed in Europe. Nitric oxide (NO) may positively impact patients with Severe Acute Respiratory Syndrome CoronaVirus-2 (SARS-CoV-2) infection. It is mainly generated by inducible nitric oxide synthase (iNOS). We studied serum iNOS levels together with serum interleukin (IL)-6 and IL-10 in patients with SARS-CoV-2 infection in the first wave (n = 35) and second wave (n = 153). In the first wave, serum iNOS, IL-6, IL-10 levels increased significantly, in line with the World Health Organization (WHO) score severity, while in the second wave, iNOS did not change with the severity. The patients of the second wave showed lower levels of iNOS, IL-6, and IL-10, as compared to the corresponding subgroup of the first wave, suggesting a less severe outcome of COVID-19 in these patients. However, in the severe patients of the second wave, iNOS levels were significantly lower in patients treated with steroids or azithromycin before the hospitalization, as compared to the untreated patients. This suggests an impairment of the defense mechanism against the virus and NO-based therapies as a potential therapy in patients with low iNOS levels.
Collapse
Affiliation(s)
- Monica Gelzo
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., 80145 Naples, Italy; (M.G.); (F.S.); (G.C.); (M.C.); (L.T.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Filippo Scialò
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., 80145 Naples, Italy; (M.G.); (F.S.); (G.C.); (M.C.); (L.T.); (G.C.)
- Dipartimento di Medicina Traslazionale, Università della Campania L. Vanvitelli, 80131 Naples, Italy
| | - Sara Cacciapuoti
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (S.C.); (B.P.); (N.S.M.); (M.M.); (G.F.); (I.G.)
| | - Biagio Pinchera
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (S.C.); (B.P.); (N.S.M.); (M.M.); (G.F.); (I.G.)
| | - Annunziata De Rosa
- Dipartimento di Malattie Infettive e Emergenze Infettive, Divisione di Malattie Infettive Respiratorie, Ospedale Cotugno, AORN dei Colli, 80131 Naples, Italy; (A.D.R.); (R.P.)
| | - Gustavo Cernera
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., 80145 Naples, Italy; (M.G.); (F.S.); (G.C.); (M.C.); (L.T.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Marika Comegna
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., 80145 Naples, Italy; (M.G.); (F.S.); (G.C.); (M.C.); (L.T.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Lorella Tripodi
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., 80145 Naples, Italy; (M.G.); (F.S.); (G.C.); (M.C.); (L.T.); (G.C.)
| | - Nicola Schiano Moriello
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (S.C.); (B.P.); (N.S.M.); (M.M.); (G.F.); (I.G.)
| | - Mauro Mormile
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (S.C.); (B.P.); (N.S.M.); (M.M.); (G.F.); (I.G.)
| | - Gabriella Fabbrocini
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (S.C.); (B.P.); (N.S.M.); (M.M.); (G.F.); (I.G.)
| | - Roberto Parrella
- Dipartimento di Malattie Infettive e Emergenze Infettive, Divisione di Malattie Infettive Respiratorie, Ospedale Cotugno, AORN dei Colli, 80131 Naples, Italy; (A.D.R.); (R.P.)
| | - Gaetano Corso
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., 80145 Naples, Italy; (M.G.); (F.S.); (G.C.); (M.C.); (L.T.); (G.C.)
- Dipartimento di Medicina Clinica e Sperimentale, Università di Foggia, 71122 Foggia, Italy
| | - Ivan Gentile
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (S.C.); (B.P.); (N.S.M.); (M.M.); (G.F.); (I.G.)
| | - Giuseppe Castaldo
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., 80145 Naples, Italy; (M.G.); (F.S.); (G.C.); (M.C.); (L.T.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| |
Collapse
|
27
|
Shokri-Mashhadi N, Aliyari A, Hajhashemy Z, Saadat S, Rouhani MH. Is it time to reconsider the administration of thiamine alone or in combination with vitamin C in critically ill patients? A meta-analysis of clinical trial studies. J Intensive Care 2022; 10:8. [PMID: 35177121 PMCID: PMC8851730 DOI: 10.1186/s40560-022-00594-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 01/04/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Although the effect of thiamine alone or in combination with vitamin C has been studied in multiple trials (RCT and interventional studies), their results are inconsistent. This meta-analysis aimed to assess impact of thiamine administration alone, thiamine in combination with vitamin C, and co-administration of low-dose hydrocortisone, vitamin C and thiamine (HVT) on clinical outcomes in critically ill patients. METHODS AND MATERIALS After electronic searches on PubMed, Scopus, Cochrane Library, and Web of Science databases, initially 3367 papers were found, and 20 interventional studies were included in our analysis. We assessed the risk-difference between treatment and control (standard treatment) groups by pooling available data on ICU length of stay, number of ventilator free days, mortality, and changes in Sequential Organ Failure Assessment (SOFA) scores. RESULTS The results of present studies revealed no significant effect of thiamine in combination with vitamin C, and HVT on number of free days of ventilation. Thiamine alone supplementation was associated with high mortality percentage (WMD: 5.17%; 95% CI: 2.67, 7.67). Thiamine in combination with vitamin C had no significant impact on mortality rate. In contrast, HVT could decrease mortality rate (WMD: - 7.23%; 95% CI: - 10.31, - 4.16; I-square: 0.0%). There was no significant effect of thiamine alone, co-administration of thiamine and vitamin C, and HVT on ICU length of stay. The results of the meta-analysis showed that thiamine alone and HVT supplementation had no significant effect on SOFA score. Interestingly, co-supplementation of thiamine and vitamin C had a significant decreasing effect on SOFA score (WMD: - 0.73; 95% CI: - 1.29, - 0.17; I-square: 0.0%). CONCLUSION In contrast to HVT, thiamine supplementation alone was associated with increased mortality rate in ICU. However, co-supplementation of thiamine and vitamin C had a significant decreasing effect on SOFA score.
Collapse
Affiliation(s)
- Nafiseh Shokri-Mashhadi
- Food Security Research Center and Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Aliyari
- Food Security Research Center and Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Hajhashemy
- Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Saadat
- Faculty of Mathematics and Natural Sciences, Department of Computer Sciences, Heinrich Heine Universität, Düsseldorf, Germany
| | - Mohammad Hossein Rouhani
- Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
28
|
Jin X, Zhang J, Li Y, Zhang Z, Cui T, Wang Y, Yao L, Yang X, Qu G, Zheng Y, Jiang G. Exogenous Chemical Exposure Increased Transcription Levels of the Host Virus Receptor Involving Coronavirus Infection. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:1854-1863. [PMID: 35049283 PMCID: PMC8790821 DOI: 10.1021/acs.est.1c07172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 05/28/2023]
Abstract
Virus receptors are highly involved in mediating the entrance of infectious viruses into host cells. Here, we found that typical chemical exposure caused the upregulation of virus receptor mRNA levels. Chemicals with the same structural characteristics can affect the transcription of angiotensin-converting enzyme 2 (ACE2), a dominant receptor of SARS-CoV-2. Some chemicals can also regulate the transcription of ACE2 by similar regulatory mechanisms, such as multilayer biological responses and the crucial role of TATA-box binding protein associated factor 6. The abovementioned finding suggested that chemical mixtures may have a joint effect on the ACE2 mRNA level in the real scenario, where humans are exposed to numerous chemicals simultaneously in daily life. Chemically regulated virus receptor transcription was in a tissue-dependent manner, with the highest sensitivity in pulmonary epithelial cells. Therefore, in addition to genetic factors, exogenous chemical exposure can be an emerging nongenetic factor that stimulates the transcription of virus receptor abundance and may elevate the protein expression. These alterations could ultimately give rise to the susceptibility to virus infection and disease severity. This finding highlights new requirements for sufficient epidemiological data about exposomes on pathogen receptors in the host.
Collapse
Affiliation(s)
- Xiaoting Jin
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, P. R. China
- Department
of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Jingxu Zhang
- Department
of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Yanting Li
- Department
of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Ze Zhang
- Department
of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Tenglong Cui
- Department
of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Yuanyuan Wang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, P. R. China
| | - Linlin Yao
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, P. R. China
| | - Xiaoxi Yang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, P. R. China
| | - Guangbo Qu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, P. R. China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, P. R.
China
- School
of Environment, Hangzhou Institute for Advanced
Study, UCAS, Hangzhou 310000, P. R. China
| | - Yuxin Zheng
- Department
of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Guibin Jiang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, P. R. China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, P. R.
China
- School
of Environment, Hangzhou Institute for Advanced
Study, UCAS, Hangzhou 310000, P. R. China
| |
Collapse
|
29
|
Malbeteau L, Pham HT, Eve L, Stallcup MR, Poulard C, Le Romancer M. How Protein Methylation Regulates Steroid Receptor Function. Endocr Rev 2022; 43:160-197. [PMID: 33955470 PMCID: PMC8755998 DOI: 10.1210/endrev/bnab014] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Indexed: 02/06/2023]
Abstract
Steroid receptors (SRs) are members of the nuclear hormonal receptor family, many of which are transcription factors regulated by ligand binding. SRs regulate various human physiological functions essential for maintenance of vital biological pathways, including development, reproduction, and metabolic homeostasis. In addition, aberrant expression of SRs or dysregulation of their signaling has been observed in a wide variety of pathologies. SR activity is tightly and finely controlled by post-translational modifications (PTMs) targeting the receptors and/or their coregulators. Whereas major attention has been focused on phosphorylation, growing evidence shows that methylation is also an important regulator of SRs. Interestingly, the protein methyltransferases depositing methyl marks are involved in many functions, from development to adult life. They have also been associated with pathologies such as inflammation, as well as cardiovascular and neuronal disorders, and cancer. This article provides an overview of SR methylation/demethylation events, along with their functional effects and biological consequences. An in-depth understanding of the landscape of these methylation events could provide new information on SR regulation in physiology, as well as promising perspectives for the development of new therapeutic strategies, illustrated by the specific inhibitors of protein methyltransferases that are currently available.
Collapse
Affiliation(s)
- Lucie Malbeteau
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Ha Thuy Pham
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Louisane Eve
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Michael R Stallcup
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Coralie Poulard
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Muriel Le Romancer
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| |
Collapse
|
30
|
Deng F, Zhang LQ, Wu H, Chen Y, Yu WQ, Han RH, Han Y, Zhang XQ, Sun QS, Lin ZB, Wang Y, Liu YP, Chen JY, Liu KX, Hu JJ. Propionate alleviates myocardial ischemia-reperfusion injury aggravated by Angiotensin II dependent on caveolin-1/ACE2 axis through GPR41. Int J Biol Sci 2022; 18:858-872. [PMID: 35002530 PMCID: PMC8741842 DOI: 10.7150/ijbs.67724] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/27/2021] [Indexed: 02/06/2023] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury is still a lack of effective therapeutic drugs, and its molecular mechanism is urgently needed. Studies have shown that the intestinal flora plays an important regulatory role in cardiovascular injury, but the specific mechanism has not been fully elucidated. In this study, we found that an increase in Ang II in plasma was accompanied by an increase in the levels of myocardial injury during myocardial reperfusion in patients with cardiopulmonary bypass. Furthermore, Ang II treatment enhanced mice myocardial I/R injury, which was reversed by caveolin-1 (CAV-1)-shRNA or strengthened by angiotensin-converting enzyme 2 (ACE2)-shRNA. The results showed that CAV-1 and ACE2 have protein interactions and inhibit each other's expression. In addition, propionate, a bacterial metabolite, inhibited the elevation of Ang II and myocardial injury, while GPR41-shRNA abolished the protective effects of propionate on myocardial I/R injury. Clinically, the propionate content in the patient's preoperative stool was related to Ang II levels and myocardial I/R injury levels during myocardial reperfusion. Taken together, propionate alleviates myocardial I/R injury aggravated by Ang II dependent on CAV-1/ACE2 axis through GPR41, which provides a new direction that diet to regulate the intestinal flora for treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China.,Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Liang-Qing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Han Wu
- Department of Dermatology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Yu Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wen-Qian Yu
- The First Ward of Pain Department, Hubei NO. 3 People's Hospital of Jianghan University, Wuhan 430000, China
| | - Rong-Hui Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yuan Han
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Qi Zhang
- Major of Clinical Medicine, Nanshan College, Guangzhou Medical University, Guangzhou 510515, China
| | - Qi-Shun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ze-Bin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yong-Pan Liu
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Jing-Yi Chen
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jing-Juan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
31
|
Grau-Expósito J, Perea D, Suppi M, Massana N, Vergara A, Soler MJ, Trinite B, Blanco J, García-Pérez J, Alcamí J, Serrano-Mollar A, Rosado J, Falcó V, Genescà M, Buzon MJ. Evaluation of SARS-CoV-2 entry, inflammation and new therapeutics in human lung tissue cells. PLoS Pathog 2022; 18:e1010171. [PMID: 35025963 PMCID: PMC8791477 DOI: 10.1371/journal.ppat.1010171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/26/2022] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
The development of physiological models that reproduce SARS-CoV-2 infection in primary human cells will be instrumental to identify host-pathogen interactions and potential therapeutics. Here, using cell suspensions directly from primary human lung tissues (HLT), we have developed a rapid platform for the identification of viral targets and the expression of viral entry factors, as well as for the screening of viral entry inhibitors and anti-inflammatory compounds. The direct use of HLT cells, without long-term cell culture and in vitro differentiation approaches, preserves main immune and structural cell populations, including the most susceptible cell targets for SARS-CoV-2; alveolar type II (AT-II) cells, while maintaining the expression of proteins involved in viral infection, such as ACE2, TMPRSS2, CD147 and AXL. Further, antiviral testing of 39 drug candidates reveals a highly reproducible method, suitable for different SARS-CoV-2 variants, and provides the identification of new compounds missed by conventional systems, such as VeroE6. Using this method, we also show that interferons do not modulate ACE2 expression, and that stimulation of local inflammatory responses can be modulated by different compounds with antiviral activity. Overall, we present a relevant and rapid method for the study of SARS-CoV-2.
Collapse
Affiliation(s)
- Judith Grau-Expósito
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - David Perea
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Marina Suppi
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Núria Massana
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Ander Vergara
- Nephrology Research Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Maria José Soler
- Nephrology Research Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Benjamin Trinite
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Autonomous University of Barcelona (UAB), Badalona, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Autonomous University of Barcelona (UAB), Badalona, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Javier García-Pérez
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Alcamí
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Clinic HIV Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Anna Serrano-Mollar
- Experimental Pathology Department, Institut d’Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Joel Rosado
- Thoracic Surgery and Lung Transplantation Department, Vall d’Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d’Hebron, VHIR Task Force COVID-19, Barcelona, Spain
| | - Vicenç Falcó
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Maria J. Buzon
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| |
Collapse
|
32
|
Law MF, Ho R, Law KWT, Cheung CKM. Gastrointestinal and hepatic side effects of potential treatment for COVID-19 and vaccination in patients with chronic liver diseases. World J Hepatol 2021; 13:1850-1874. [PMID: 35069994 PMCID: PMC8727202 DOI: 10.4254/wjh.v13.i12.1850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/20/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) is a global pandemic. Many clinical trials have been performed to investigate potential treatments or vaccines for this disease to reduce the high morbidity and mortality. The drugs of higher interest include umifenovir, bromhexine, remdesivir, lopinavir/ritonavir, steroid, tocilizumab, interferon alpha or beta, ribavirin, fivapiravir, nitazoxanide, ivermectin, molnupiravir, hydroxychloroquine/chloroquine alone or in combination with azithromycin, and baricitinib. Gastrointestinal (GI) symptoms and liver dysfunction are frequently seen in patients with COVID-19, which can make it difficult to differentiate disease manifestations from treatment adverse effects. GI symptoms of COVID-19 include anorexia, dyspepsia, nausea, vomiting, diarrhea and abdominal pain. Liver injury can be a result of systemic inflammation or cytokine storm, or due to the adverse drug effects in patients who have been receiving different treatments. Regular monitoring of liver function should be performed. COVID-19 vaccines have been rapidly developed with different technologies including mRNA, viral vectors, inactivated viruses, recombinant DNA, protein subunits and live attenuated viruses. Patients with chronic liver disease or inflammatory bowel disease and liver transplant recipients are encouraged to receive vaccination as the benefits outweigh the risks. Vaccination against COVID-19 is also recommended to family members and healthcare professionals caring for these patients to reduce exposure to the severe acute respiratory syndrome coronavirus 2 virus.
Collapse
Affiliation(s)
- Man Fai Law
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| | - Rita Ho
- Department of Medicine, North District Hospital, Hong Kong, China
| | | | - Carmen Ka Man Cheung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
33
|
Camelo S, Latil M, Agus S, Dioh W, Veillet S, Lafont R, Dilda PJ. A comparison between virus- versus patients-centred therapeutic attempts to reduce COVID-19 mortality. Emerg Microbes Infect 2021; 10:2256-2263. [PMID: 34783636 PMCID: PMC8648031 DOI: 10.1080/22221751.2021.2006579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 01/08/2023]
Abstract
Since December 2019, coronavirus disease 2019 (COVID-19), caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has changed our lives. Elderly and those with comorbidities represent the vast majority of patients hospitalized with severe COVID-19 symptoms, including acute respiratory disease syndrome and cardiac dysfunction. Despite a huge effort of the scientific community, improved treatment modalities limiting the severity and mortality of hospitalized COVID-19 patients are still required. Here, we compare the effectiveness of virus- and patients-centred strategies to reduce COVID-19 mortality. We also discuss the therapeutic options that might further reduce death rates associated with the disease in the future. Unexpectedly, extensive review of the literature suggests that SARS-CoV-2 viral load seems to be associated neither with the severity of symptoms nor with mortality of hospitalized patients with COVID-19. This may explain why, so far, virus-centred strategies using antivirals aiming to inhibit the viral replicative machinery have failed to reduce COVID-19 mortality in patients with respiratory failure. By contrast, anti-inflammatory treatments without antiviral capacities but centred on patients, such as dexamethasone or Tocilizumab®, reduce COVID-19 mortality. Finally, since the spike protein of SARS-CoV-2 binds to angiotensin converting enzyme 2 and inhibits its function, we explore the different treatment options focussing on rebalancing the renin-angiotensin system. This new therapeutic strategy could hopefully further reduce the severity of respiratory failure and limit COVID-19 mortality in elderly patients.
Collapse
Affiliation(s)
| | | | - Sam Agus
- Biophytis Inc., Cambridge, MA, USA
| | - Waly Dioh
- Biophytis, Sorbonne Université, Paris, France
| | | | - René Lafont
- Biophytis, Sorbonne Université, Paris, France
- CNRS – Institut de Biologie Paris Seine (BIOSIPE), Sorbonne Université, Paris, France
| | | |
Collapse
|
34
|
Zhang MM, Chen LN, Qian JM. Gastrointestinal manifestations and possible mechanisms of COVID-19 in different periods. J Dig Dis 2021; 22:683-694. [PMID: 34738727 PMCID: PMC8652439 DOI: 10.1111/1751-2980.13065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, has become a pandemic worldwide. Although COVID-19 mainly affects the respiratory system, gastrointestinal (GI) manifestations have been frequently reported in such cases, even as initial symptoms. There have been several studies on different GI manifestations in patients with mild and severe disease or in remission. In this review article we summarized different GI manifestations of COVID-19 at various disease stages and the possible mechanisms based on published literatures, as well as the significance of GI manifestations in systemic inflammatory injury.
Collapse
Affiliation(s)
- Meng Meng Zhang
- Department of GastroenterologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lu Ni Chen
- Department of Microbiology and Tumor and Cell BiologyKarolinska InstituteSolnaSweden
| | - Jia Ming Qian
- Department of GastroenterologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
35
|
Choi Y, Jeon H, Brännström M, Akin JW, Curry TE, Jo M. Ovulatory upregulation of angiotensin-converting enzyme 2, a receptor for SARS-CoV-2, in dominant follicles of the human ovary. Fertil Steril 2021; 116:1631-1640. [PMID: 34538460 PMCID: PMC8354803 DOI: 10.1016/j.fertnstert.2021.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/25/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To determine the temporal expression of angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV-2, in dominant follicles throughout the periovulatory period in women and the regulatory mechanisms underlying ACE2 expression in human granulosa/lutein cells (hGLC). DESIGN Experimental prospective clinical study and laboratory-based investigation. SETTING University Medical Center and private in vitro fertilization center. PATIENT(S) Thirty premenopausal women undergoing surgery for tubal ligation and 16 premenopausal women undergoing in vitro fertilization. INTERVENTION(S) Administration of human chorionic gonadotropin (hCG) and harvesting of preovulatory/ovulatory follicles by timed laparoscopy, and collection of granulosa/lutein cells and cumulus cells at the time of oocyte retrieval. MAIN OUTCOME MEASURE(S) Expression and localization of ACE2 in granulosa cells and dominant follicles collected throughout the periovulatory period of the menstrual cycle and in hGLC using quantitative polymerase chain reaction, immunoblotting, and immunohistochemistry. RESULT(S) ACE2 expression (mRNA and protein) is up-regulated in human ovulatory follicles after administration of hCG. ACE2 expression was higher in cumulus cells than in granulosa cells. hCG increased the expression of ACE2 in primary hGLC cultures; the increase was inhibited by RU486 (an antagonist for progesterone receptor and glucocorticoid receptor) and CORT125281 (a selective glucocorticoid receptor antagonist), but not by AG1478 (an EGF receptor tyrosine kinase inhibitor) or by dexamethasone. CONCLUSION(S) The hormone-regulated expression of ACE2 in granulosa cells suggests a potential role of ACE2 in the ovulatory process. These data also imply the possible impact of COVID-19 on a vital cyclic event of ovarian function and thus on women's overall reproductive health. However, SAR-CoV-2 infection in ovarian cells in vivo or in vitro has yet to be determined.
Collapse
Affiliation(s)
- Yohan Choi
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Hayce Jeon
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden, and Stockholm IVF-EUGIN, Stockholm, Sweden
| | | | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky.
| |
Collapse
|
36
|
Reichardt SD, Amouret A, Muzzi C, Vettorazzi S, Tuckermann JP, Lühder F, Reichardt HM. The Role of Glucocorticoids in Inflammatory Diseases. Cells 2021; 10:cells10112921. [PMID: 34831143 PMCID: PMC8616489 DOI: 10.3390/cells10112921] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
For more than 70 years, glucocorticoids (GCs) have been a powerful and affordable treatment option for inflammatory diseases. However, their benefits do not come without a cost, since GCs also cause side effects. Therefore, strong efforts are being made to improve their therapeutic index. In this review, we illustrate the mechanisms and target cells of GCs in the pathogenesis and treatment of some of the most frequent inflammatory disorders affecting the central nervous system, the gastrointestinal tract, the lung, and the joints, as well as graft-versus-host disease, which often develops after hematopoietic stem cell transplantation. In addition, an overview is provided of novel approaches aimed at improving GC therapy based on chemical modifications or GC delivery using nanoformulations. GCs remain a topic of highly active scientific research despite being one of the oldest class of drugs in medical use.
Collapse
Affiliation(s)
- Sybille D. Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
| | - Agathe Amouret
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
| | - Chiara Muzzi
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
| | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm, Germany; (S.V.); (J.P.T.)
| | - Jan P. Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm, Germany; (S.V.); (J.P.T.)
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Holger M. Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany; (S.D.R.); (A.A.); (C.M.)
- Correspondence: ; Tel.: +49-551-3963365
| |
Collapse
|
37
|
Duan H, Wang W, Li Y, Jilany Khan G, Chen Y, Shen T, Bao N, Hua J, Xue Z, Zhai K, Wei Z. Identification of phytochemicals and antioxidant activity of Premna microphylla Turcz. stem through UPLC-LTQ-Orbitrap-MS. Food Chem 2021; 373:131482. [PMID: 34731817 DOI: 10.1016/j.foodchem.2021.131482] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/15/2021] [Accepted: 10/24/2021] [Indexed: 12/17/2022]
Abstract
Premna microphylla Turcz. is a commonly used traditional Chinese medicine totreatdysentery and appendicitis. Present study is focused to explore antioxidants and other compounds in the Premna microphylla Turcz. stem. Assessment of chemical composition was done with high sensitivity UPLC-LTQ-Orbitrap-MS and for Separation Thermo Hypersil Gold (100 mm × 2.1 mm, 1.9 µm) was used while electrospray ionization (ESI) was used for the mass spectrometry. 18 compounds were identified including Vitexin (1), Schaftoside (2), Vicenin-2 (3), Apigenin-6, 8-di-C-arabinoside (4), Apigenin-7-O-β-d-glucoside (5), Carnosic acid (6), Apigenin-8-C-β-d-xylopyranoside (7), Prostratin (8), Aurantio-obtusin-β-d-glucoside (9), Royleanone (10), 5-hydroxy-7,3',4'-Trimethoxy flavonols (11), 6-Hydroxy-5,6-dehydrosugiol (12), 14-deoxycoleon (13), Arucadiol (14), Obtusinone-B (15), Trehalose (16), Citric acid (17) and Betaine (18). Among these, 6 compounds including (6), (8), (9), (16), (17) and (18) were identified first time within this genus and plant. Study highlights the importance of Premna microphylla Turcz. stem extract for strong therapeutic potential against oxidation-related diseases.
Collapse
Affiliation(s)
- Hong Duan
- Suzhou Engineering and Technological Research Center of Natural Medicine and Functional Food, School of Biological and Food Engineering, Suzhou University, Suzhou 234000, PR China
| | - Wei Wang
- Suzhou Engineering and Technological Research Center of Natural Medicine and Functional Food, School of Biological and Food Engineering, Suzhou University, Suzhou 234000, PR China; School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Yongxiang Li
- Suzhou Engineering and Technological Research Center of Natural Medicine and Functional Food, School of Biological and Food Engineering, Suzhou University, Suzhou 234000, PR China
| | - Ghulam Jilany Khan
- Department of Pharmacology, Faculty of Pharmacy (FOP), University of Central Punjab, Lahore, Pakistan; National Drug Screening Center of Pharmacokinetics and Pharmacodynamics, School of Pharmacy, China Pharmaceutical University, Nanjing, PR China
| | - Yuan Chen
- Suzhou Engineering and Technological Research Center of Natural Medicine and Functional Food, School of Biological and Food Engineering, Suzhou University, Suzhou 234000, PR China; School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Tianci Shen
- Suzhou Engineering and Technological Research Center of Natural Medicine and Functional Food, School of Biological and Food Engineering, Suzhou University, Suzhou 234000, PR China
| | - Nina Bao
- Suzhou Engineering and Technological Research Center of Natural Medicine and Functional Food, School of Biological and Food Engineering, Suzhou University, Suzhou 234000, PR China
| | - Jing Hua
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Guangxi Normal University), Guilin 541004, PR China
| | - Zhenglian Xue
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Kefeng Zhai
- Suzhou Engineering and Technological Research Center of Natural Medicine and Functional Food, School of Biological and Food Engineering, Suzhou University, Suzhou 234000, PR China; School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China; Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Guangxi Normal University), Guilin 541004, PR China.
| | - Zhaojun Wei
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, PR China.
| |
Collapse
|
38
|
Fang L, Zhou L, Tamm M, Roth M. OM-85 Broncho-Vaxom ®, a Bacterial Lysate, Reduces SARS-CoV-2 Binding Proteins on Human Bronchial Epithelial Cells. Biomedicines 2021; 9:1544. [PMID: 34829773 PMCID: PMC8615539 DOI: 10.3390/biomedicines9111544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 12/21/2022] Open
Abstract
In clinical studies, OM-85 Broncho-Vaxom®, a bacterial lysate, reduced viral respiratory tract infection. Infection of epithelial cells by SARS-CoV-2 depends on the interaction of its spike-protein (S-protein) with host cell membrane proteins. In this study, we investigated the effect of OM-85 on the expression of S-protein binding proteins by human bronchial epithelial cells. Human bronchial epithelial cells were treated with OM-85 over 5 days. The expression of SARS-CoV-2 receptor angiotensin converting enzyme 2 (ACE2), transmembrane protease serine subtype 2 (TMPRSS2), dipeptidyl peptidase-4 (DPP4), and a disintegrin and metalloprotease 17 (ADAM17) were determined by Western blotting and quantitative RT-PCR. Soluble (s)ACE2, heparan sulfate, heparanase, and hyaluronic acid were assessed by ELISA. OM-85 significantly reduced the expression of ACE2 (p < 0.001), TMPRSS2 (p < 0.001), DPP4 (p < 0.005), and cellular heparan sulfate (p < 0.01), while ADAM17 (p < 0.02) expression was significantly upregulated. Furthermore, OM-85 increased the level of sACE2 (p < 0.05), hyaluronic acid (p < 0.002), and hyaluronan synthase 1 (p < 0.01). Consequently, the infection by a SARS-CoV-2 spike protein pseudo-typed lentivirus was reduced in cells pretreated with OM-85. All effects of OM-85 were concentration- and time-dependent. The results suggest that OM-85 might reduce the binding of SARS-CoV-2 S-protein to epithelial cells by modification of host cell membrane proteins and specific glycosaminoglycans. Thus, OM-85 might be considered as an add-on for COVID-19 therapy.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research, Department of Biomedicine & Internal Medicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Liang Zhou
- Pulmonary Cell Research, Department of Biomedicine & Internal Medicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Michael Tamm
- Pulmonary Cell Research, Department of Biomedicine & Internal Medicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Michael Roth
- Pulmonary Cell Research, Department of Biomedicine & Internal Medicine, University Hospital Basel, CH-4031 Basel, Switzerland
| |
Collapse
|
39
|
Yu Y. Repurposing glucocorticoids as adjuvant reagents for immune checkpoint inhibitors in solid cancers. Cancer Biol Med 2021; 18:j.issn.2095-3941.2021.0491. [PMID: 34697935 PMCID: PMC8610151 DOI: 10.20892/j.issn.2095-3941.2021.0491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 11/11/2022] Open
Affiliation(s)
- Yingyan Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
40
|
Moes DJAR, van Westerloo DJ, Arend SM, Swen JJ, de Vries A, Guchelaar HJ, Joosten SA, de Boer MGJ, van Gelder T, van Paassen J. Towards Fixed Dosing of Tocilizumab in ICU-Admitted COVID-19 Patients: Results of an Observational Population Pharmacokinetic and Descriptive Pharmacodynamic Study. Clin Pharmacokinet 2021; 61:231-247. [PMID: 34633645 PMCID: PMC8502793 DOI: 10.1007/s40262-021-01074-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVE In the randomized controlled trial REMAP-CAP, it was shown that next to dexamethasone, the interleukin (IL)-6 receptor antagonist tocilizumab improves outcome, including survival in intensive care unit (ICU)-admitted coronavirus disease 2019 (COVID)-19 patients. Therefore tocilizumab has been added to many COVID-19 treatment guidelines. Because obesity is a risk factor for the development of severe COVID-19, concerns have been raised about overtreatment, as well as undertreatment, through weight-based dosing of tocilizumab. The currently applied dose of 8 mg/kg is based on the use of this drug for other indications, however it has not formally been investigated for COVID-19. In this study, the pharmacokinetics and pharmacodynamics of tocilizumab were investigated in ICU-admitted COVID-19 patients. METHODS This was an open-label, single-centre, observational population pharmacokinetic and descriptive pharmacodynamic evaluation study. Enrolled patients, with polymerase chain reaction-confirmed COVID-19 were admitted to the ICU for mechanical ventilation or high flow nasal canula oxygen support. All patients were 18 years of age or older and received intravenous tocilizumab 8 mg/kg (maximum 800 mg) within 24 h after admission to the ICU and received dexamethasone 6 mg daily as concomitant therapy. For evaluation of the pharmacokinetics and pharmacodynamics of tocilizumab, all time points from day 0 to 20 days after dose administration were eligible for collection. A nonlinear mixed-effects model was developed to characterize the population pharmacokinetic parameters of tocilizumab in ICU-admitted COVID-19 patients. Covariate analysis was performed to identify potential covariates for dose individualization. For the development of alternative dosing schedules, Monte Carlo simulations using the final model were performed. RESULTS Overall, 29 patients were enrolled between 15 December 2020 and 15 March 2021. A total of 139 tocilizumab plasma samples were obtained covering the pharmacokinetic curve of day 0 to day 20 after tocilizumab initiation. A population pharmacokinetic model with parallel linear and nonlinear clearance (CL) was developed and validated. Average CL was estimated to be 0.725 L/day, average volume of distribution (Vd) was 4.34 L, maximum elimination rate (Vmax) was 4.19 μg/day, and concentration at which the elimination pathway is half saturated (Km) was 0.22 μg/mL. Interindividual variability was identified for CL (18.9%) and Vd (21%). Average area under the concentration versus time curve from time zero to infinity of the first dose (AUCinf 1st DOSE) was 938 [±190] μg/mL*days. All patients had tocilizumab exposure above 1 μg/mL for at least 15 days. Bodyweight-based dosing increases variability in exposure compared with fixed dosing. CONCLUSIONS This study provides evidence to support a fixed dose of tocilizumab 600 mg in COVID-19 patients. Fixed dosing is a safe, logistically attractive, and drug expenses saving alternative compared with the current 8 mg/kg recommendation.
Collapse
Affiliation(s)
- Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
- Leiden Network for Personalised Therapeutics, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - David J van Westerloo
- Department of Intensive Care, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sandra M Arend
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Leiden Network for Personalised Therapeutics, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Annick de Vries
- Biologics Lab, Sanquin Diagnostic Services, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Leiden Network for Personalised Therapeutics, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Mark G J de Boer
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Teun van Gelder
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Leiden Network for Personalised Therapeutics, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Judith van Paassen
- Department of Intensive Care, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
41
|
Scalia G, Raia M, Gelzo M, Cacciapuoti S, De Rosa A, Pinchera B, Scotto R, Giaccone A, Mormile M, Fabbrocini G, Gentile I, Parrella R, Castaldo G. Cytometric analysis of patients with COVID-19: what is changed in the second wave? J Transl Med 2021; 19:403. [PMID: 34556132 PMCID: PMC8460184 DOI: 10.1186/s12967-021-03072-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) pandemic had a 1st wave in Europe from March to May 2020 and a 2nd wave since September 2020. We previously studied 35 hospitalized COVID-19 patients of the 1st wave demonstrating a cytokine storm and the exhaustion of most lymphocyte subpopulations. Herein, we describe the results obtained from COVID-19 patients of the 2nd wave. Methods We analyzed interleukin (IL)-6 by human-specific enzyme-linked immunosorbent assay and a large set of lymphocyte subpopulations by flow cytometry in 274 COVID-19 patients hospitalized from September 2020 to May 2021. Results Patients of 2nd wave compared with those of 1st wave showed lower serum IL-6 levels and a higher number of B and most T lymphocyte subpopulations in advanced stages, in relation with the age and the gender. On the other hand, we observed in 2nd wave patients: (i) a reduction of most lymphocyte subpopulations at mild and moderate stages; (ii) a reduction of natural killer cells and T regulatory cells together with a higher number of activated T helper (TH) 17 lymphocytes in all stages, which were mainly related to steroid and azithromycin therapies before hospitalization. Conclusions COVID-19 had a less severe impact in patients of the 2nd wave in advanced stages, while the impact appeared more severe in patients of mild and moderate stages, as compared with 1st wave patients. This finding suggests that in COVID-19 patients with milder expression at diagnosis, steroid and azithromycin therapies appear to worsen the immune response against the virus. Furthermore, the cytometric profile may help to drive targeted therapies by monoclonal antibodies to modulate specific IL/lymphocyte inhibition or activation in COVID-19 patients.
Collapse
Affiliation(s)
- Giulia Scalia
- CEINGE-Biotecnologie Avanzate, via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Maddalena Raia
- CEINGE-Biotecnologie Avanzate, via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Monica Gelzo
- CEINGE-Biotecnologie Avanzate, via Gaetano Salvatore 486, 80145, Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Sara Cacciapuoti
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Annunziata De Rosa
- Dipartimento di Malattie Infettive e Emergenze Infettive, Divisione di malattie infettive respiratorie, Ospedale Cotugno, AORN dei Colli, Naples, Italy
| | - Biagio Pinchera
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Riccardo Scotto
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Agnese Giaccone
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Mauro Mormile
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Gabriella Fabbrocini
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Ivan Gentile
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Roberto Parrella
- Dipartimento di Malattie Infettive e Emergenze Infettive, Divisione di malattie infettive respiratorie, Ospedale Cotugno, AORN dei Colli, Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie Avanzate, via Gaetano Salvatore 486, 80145, Naples, Italy. .,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy.
| |
Collapse
|
42
|
Abstract
The pathophysiology of COVID comprises an exaggerated pro-inflammatory response. Hypothalamic-pituitary-adrenal (HPA) axis has a crucial role in various inflammatory conditions and modulated immunological response. Limited evidence is available regarding the incidence and the effect of HPA dysfunction in COVID-19. Although the cortisol levels have only been estimated in a few studies, the dehydroepiandrosterone sulfate (DHEAS) release from the adrenal gland has not been explored yet. In this mini review, the authors discuss the role of dehydroepiandrosterone (DHEA) and DHEAS in the acute stress response and immunological modulation. Various effects of DHEAS have been demonstrated in different diseases. The specific inhibitory effect of DHEA on interleukin 6 (IL-6) could be of paramount importance in COVID-19. Further, DHEA supplementation has already been proposed in inflammatory conditions, like rheumatoid arthritis. DHEAS levels in COVID-19 may help to understand the HPA axis dysfunction as well as the possibility of repurposing DHEA as a drug for mitigating the pro-inflammatory COVID-19.
Collapse
|
43
|
Xu S, He X, Shi J, Li Z, Song J, Wang J, Wang G, Brand-Saberi B, Cheng X, Yang X. Interaction between retinoic acid and FGF/ERK signals are involved in Dexamethasone-induced abnormal myogenesis during embryonic development. Toxicology 2021; 461:152917. [PMID: 34464682 DOI: 10.1016/j.tox.2021.152917] [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: 04/17/2021] [Revised: 08/01/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
Despite the common application in pregnancy at clinical practice, it remains ambiguous whether dexamethasone (Dex) exposure can affect embryonic myogenesis. In this study, firstly we showed that 10-6 M Dex (Cheng et al., 2016; 2017) treatment resulted in abnormal myogenesis in chicken embryos. Secondly, we demonstrated that 10-6 M Dex-induced abnormality of myogenesis resulted from aberrant cell proliferation, as well as from alteration of the differentiation process from the early stage of somitogenesis up to the late stage of myogenesis. The above-mentioned results caused by Dex exposure might be due to the aberrant gene expressions of somite formation (Raldh2, Fgf8, Wnt3a, β-catenin, Slug, Paraxis, N-cadherin) and differentiation (Pax3, MyoD, Wnt3a, Msx1, Shh). Thirdly, RNA sequencing implied the statistically significant differential gene expressions in regulating the myofibril and systemic development, as well as a dramatical alteration of retinoic acid (RA) signaling during somite development in the chicken embryos exposed to Dex. The subsequent validation experiments verified that Dex treatment indeed led to a metabolic change of RA signaling, which was up-regulated and principally mediated by FGF-ERK signaling revealed by means of the combination of chicken embryos and in vitro C2C12 cells. These findings highlight that 10-6 M Dex exposure enhances the risk of abnormal myogenesis through interfering with RA signaling during development.
Collapse
Affiliation(s)
- Shujie Xu
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Xiangyue He
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China; Department of Pathology, Medical School, Jinan University, Guangzhou, 510632, China
| | - Junzhu Shi
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Ziguang Li
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Jinhuan Song
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Jingyun Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Guang Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstrasse 150, 44801, Bochum, Germany
| | - Xin Cheng
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, 510632, China.
| | - Xuesong Yang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
44
|
Piticchio T, Le Moli R, Tumino D, Frasca F. Relationship between betacoronaviruses and the endocrine system: a new key to understand the COVID-19 pandemic-A comprehensive review. J Endocrinol Invest 2021; 44:1553-1570. [PMID: 33583003 PMCID: PMC7882054 DOI: 10.1007/s40618-020-01486-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND A new harmful respiratory disease, called COVID-19 emerged in China in December 2019 due to the infection of a novel coronavirus, called SARS-Coronavirus 2 (SARS-CoV-2), which belongs to the betacoronavirus genus, including SARS-CoV-1 and MERS-CoV. SARS-CoV-2 shares almost 80% of the genome with SARS-CoV-1 and 50% with MERS-CoV. Moreover, SARS-CoV-2 proteins share a high degree of homology (approximately 95%) with SARS-CoV-1 proteins. Hence, the mechanisms of SARS-Cov-1 and SARS-Cov-2 infection are similar and occur via binding to ACE2 protein, which is widely distributed in the human body, with a predominant expression in endocrine tissues including testis, thyroid, adrenal and pituitary. PURPOSE On the basis of expression pattern of the ACE2 protein among different tissues, similarity between SARS-Cov-1 and SARS-Cov-2 and the pathophysiology of COVID-19 disease, we aimed at discussing, after almost one-year pandemic, about the relationships between COVID-19 infection and the endocrine system. First, we discussed the potential effect of hormones on the susceptibility to COVID-19 infection; second, we examined the evidences regarding the effect of COVID-19 on the endocrine system. When data were available, a comparative discussion between SARS and COVID-19 effects was also performed. METHODS A comprehensive literature search within Pubmed was performed. This review has been conducted according to the PRISMA statements. RESULTS Among 450, 100 articles were selected. Tissue and vascular damages have been shown on thyroid, adrenal, testis and pituitary glands, with multiple alterations of endocrine function. CONCLUSION Hormones may affect patient susceptibility to COVID-19 infection but evidences regarding therapeutic implication of these findings are still missing. SARS and COVID-19 may affect endocrine glands and their dense vascularization, impairing endocrine system function. A possible damage of endocrine system in COVID-19 patients should be investigated in both COVID-19 acute phase and recovery to identify both early and late endocrine complications that may be important for patient's prognosis and well-being after COVID-19 infection.
Collapse
Affiliation(s)
- T Piticchio
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - R Le Moli
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - D Tumino
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - F Frasca
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy.
| |
Collapse
|
45
|
Ricciotti E, Laudanski K, FitzGerald GA. Nonsteroidal anti-inflammatory drugs and glucocorticoids in COVID-19. Adv Biol Regul 2021; 81:100818. [PMID: 34303107 PMCID: PMC8280659 DOI: 10.1016/j.jbior.2021.100818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by a wide spectrum of symptom severity, which is manifested at different phases of infection and demands different levels of care. Viral load, host innate-immune response to SARS-CoV-2, and comorbidities have a direct impact on the clinical outcomes of COVID-19 patients and determine the diverse disease trajectories. The initial SARS-CoV-2 penetrance and replication in the host causes death of infected cells, determining the viral response. SARS-CoV-2 replication in the host triggers the activation of host antiviral immune mechanisms, determining the inflammatory response. While a healthy immune response is essential to eliminate infected cells and prevent spread of the virus, a dysfunctional immune response can result in a cytokine storm and hyperinflammation, contributing to disease progression. Current therapies for COVID-19 target the virus and/or the host immune system and may be complicated in their efficacy by comorbidities. Here we review the evidence for use of two classes of anti-inflammatory drugs, glucocorticoids and nonsteroidal anti-inflammatory drugs (NSAIDs) for the treatment of COVID-19. We consider the clinical evidence regarding the timing and efficacy of their use, their potential limitations, current recommendations and the prospect of future studies by these and related therapies.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Krzysztof Laudanski
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA; Leonard Davis Institute of Healthcare Economics, University of Pennsylvania, Philadelphia, PA, USA
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Li HP, He X, Zhang L, Li CX, Li SQ, Li QY. Therapeutic Agents Rounding Up the Immunopathology of COVID-19. Ther Clin Risk Manag 2021; 17:657-668. [PMID: 34234442 PMCID: PMC8254585 DOI: 10.2147/tcrm.s313003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 pandemic has caused more than 3 million deaths globally during the past year. The direct attack from SARS-CoV-2 and hyperactivated immune response contribute to the progress and deterioration of COVID-19. After the virus invades, the activation and release of cytokines/chemokines cause "cytokine storm", leading to acute respiratory distress syndrome (ARDS) and multiple organs dysfunction syndrome (MODS). Eliminating virus and blocking cytokines are important checkpoints of COVID-19 therapy, and several agents targeting immunopathology, including interferons, thymosin, glucocorticoids and immunoglobulin, have shown therapeutic effects in severe patients with COVID-19. Herein, we reviewed the practice evidences and concluded that several agents rounding up the immunopathology of COVID-19 may be the alternative approaches under the scenario of the lacking of effective antiviral drugs.
Collapse
Affiliation(s)
- Hong Peng Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Xuan He
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Liu Zhang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Chuan Xiang Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Department of Respiratory Medicine, Wuhan No.3 Hospital, Wuhan, 430000, People’s Republic of China
| | - Shi Qi Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Qing Yun Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
47
|
Kostorz-Nosal S, Jastrzębski D, Chyra M, Kubicki P, Zieliński M, Ziora D. A prolonged steroid therapy may be beneficial in some patients after the COVID-19 pneumonia. Eur Clin Respir J 2021; 8:1945186. [PMID: 34221256 PMCID: PMC8231404 DOI: 10.1080/20018525.2021.1945186] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION: This report introduces two cases presenting absorption of considerable radiological changes in the course of the coronavirus pneumonia in patients treated with prolonged oral steroids.CASES: The first case concerns a male receiving steroids only during hospitalisation in the Infectious Disease Hospital. After discharge, the patient experienced increasing dyspnoea resulting in hospitalisation in our Department of Lung Diseases. HRCT revealed progression of a bilateral, middle, and basal ground-glass opacity when compared to the examination performed at the early stage of the disease. The supplementary oxygen therapy and steroids were administered, followed by extended prednisone consumption up to 2 months after discharge. Follow-up HRCT revealed an almost complete absorption of the ground-glass opacity. The second case concerns a male treated with steroids only during hospitalisation in the Infectious Disease Hospital. Chest CT revealed widespread bilateral ground-glass opacities with consolidations. After discharge with no treatment, he suffered from severe dyspnoea and exercise intolerance, resulting in hospitalisation on the 7th day of home stay. Since then, a continued steroid treatment was administered resulting in a clinical, spirometric, and radiological improvement.CONCLUSIONS: Based on these observations, patients after the COVID-pneumonia may derive benefits from a prolonged steroid treatment. Therefore, this class of medications should be considered in SARS-CoV-2 patients, especially in patients with persistent radiological changes and dyspnoea requiring the supplementary oxygen therapy. However, randomised controlled trials are required to establish guidelines for the steroid treatment in this group of patients.
Collapse
Affiliation(s)
- Sabina Kostorz-Nosal
- Department of Lung Diseases and Tuberculosis, Faculty of Medical Sciences in Zabrze, Silesia Medical University in Katowice, Poland
| | - Dariusz Jastrzębski
- Department of Lung Diseases and Tuberculosis, Faculty of Medical Sciences in Zabrze, Silesia Medical University in Katowice, Poland
| | - Michał Chyra
- Pulmonology Department, Silesia Medical University in Katowice, Poland
| | - Piotr Kubicki
- Department of Lung Diseases and Tuberculosis, Faculty of Medical Sciences in Zabrze, Silesia Medical University in Katowice, Poland
| | - Michał Zieliński
- Department of Lung Diseases and Tuberculosis, Faculty of Medical Sciences in Zabrze, Silesia Medical University in Katowice, Poland
| | - Dariusz Ziora
- Department of Lung Diseases and Tuberculosis, Faculty of Medical Sciences in Zabrze, Silesia Medical University in Katowice, Poland
| |
Collapse
|
48
|
Pantazi I, Al-Qahtani AA, Alhamlan FS, Alothaid H, Matou-Nasri S, Sourvinos G, Vergadi E, Tsatsanis C. SARS-CoV-2/ACE2 Interaction Suppresses IRAK-M Expression and Promotes Pro-Inflammatory Cytokine Production in Macrophages. Front Immunol 2021; 12:683800. [PMID: 34248968 PMCID: PMC8261299 DOI: 10.3389/fimmu.2021.683800] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/31/2021] [Indexed: 01/08/2023] Open
Abstract
The major cause of death in SARS-CoV-2 infected patients is due to de-regulation of the innate immune system and development of cytokine storm. SARS-CoV-2 infects multiple cell types in the lung, including macrophages, by engagement of its spike (S) protein on angiotensin converting enzyme 2 (ACE2) receptor. ACE2 receptor initiates signals in macrophages that modulate their activation, including production of cytokines and chemokines. IL-1R-associated kinase (IRAK)-M is a central regulator of inflammatory responses regulating the magnitude of TLR responsiveness. Aim of the work was to investigate whether SARS-CoV-2 S protein-initiated signals modulate pro-inflammatory cytokine production in macrophages. For this purpose, we treated PMA-differentiated THP-1 human macrophages with SARS-CoV-2 S protein and measured the induction of inflammatory mediators including IL6, TNFα, IL8, CXCL5, and MIP1a. The results showed that SARS-CoV-2 S protein induced IL6, MIP1a and TNFα mRNA expression, while it had no effect on IL8 and CXCL5 mRNA levels. We further examined whether SARS-CoV-2 S protein altered the responsiveness of macrophages to TLR signals. Treatment of LPS-activated macrophages with SARS-CoV-2 S protein augmented IL6 and MIP1a mRNA, an effect that was evident at the protein level only for IL6. Similarly, treatment of PAM3csk4 stimulated macrophages with SARS-CoV-2 S protein resulted in increased mRNA of IL6, while TNFα and MIP1a were unaffected. The results were confirmed in primary human peripheral monocytic cells (PBMCs) and isolated CD14+ monocytes. Macrophage responsiveness to TLR ligands is regulated by IRAK-M, an inactive IRAK kinase isoform. Indeed, we found that SARS-CoV-2 S protein suppressed IRAK-M mRNA and protein expression both in THP1 macrophages and primary human PBMCs and CD14+ monocytes. Engagement of SARS-CoV-2 S protein with ACE2 results in internalization of ACE2 and suppression of its activity. Activation of ACE2 has been previously shown to induce anti-inflammatory responses in macrophages. Treatment of macrophages with the ACE2 activator DIZE suppressed the pro-inflammatory action of SARS-CoV-2. Our results demonstrated that SARS-CoV-2/ACE2 interaction rendered macrophages hyper-responsive to TLR signals, suppressed IRAK-M and promoted pro-inflammatory cytokine expression. Thus, activation of ACE2 may be a potential anti-inflammatory therapeutic strategy to eliminate the development of cytokine storm observed in COVID-19 patients.
Collapse
Affiliation(s)
- Ioanna Pantazi
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece.,Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Fatimah S Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Sabine Matou-Nasri
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - George Sourvinos
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Greece
| | - Eleni Vergadi
- Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| |
Collapse
|
49
|
Implications of Oxidative Stress in Glioblastoma Multiforme Following Treatment with Purine Derivatives. Antioxidants (Basel) 2021; 10:antiox10060950. [PMID: 34204594 PMCID: PMC8231124 DOI: 10.3390/antiox10060950] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
Recently, small compound-based therapies have provided new insights into the treatment of glioblastoma multiforme (GBM) by inducing oxidative impairment. Kinetin riboside (KR) and newly designed derivatives (8-azaKR, 7-deazaKR) selectively affect the molecular pathways crucial for cell growth by interfering with the redox status of cancer cells. Thus, these compounds might serve as potential alternatives in the oxidative therapy of GBM. The increased basal levels of reactive oxygen species (ROS) in GBM support the survival of cancer cells and cause drug resistance. The simplest approach to induce cell death is to achieve the redox threshold and circumvent the antioxidant defense mechanisms. Consequently, cells become more sensitive to oxidative stress (OS) caused by exogenous agents. Here, we investigated the effect of KR and its derivatives on the redox status of T98G cells in 2D and 3D cell culture. The use of spheroids of T98G cells enabled the selection of one derivative-7-deazaKR-with comparable antitumor activity to KR. Both compounds induced ROS generation and genotoxic OS, resulting in lipid peroxidation and leading to apoptosis. Taken together, these results demonstrated that KR and 7-deazaKR modulate the cellular redox environment of T98G cells, and vulnerability of these cells is dependent on their antioxidant capacity.
Collapse
|
50
|
Watson A, Öberg L, Angermann B, Spalluto CM, Hühn M, Burke H, Cellura D, Freeman A, Muthas D, Etal D, Belfield G, Karlsson F, Nordström K, Ostridge K, Staples KJ, Wilkinson T. Dysregulation of COVID-19 related gene expression in the COPD lung. Respir Res 2021; 22:164. [PMID: 34051791 PMCID: PMC8164067 DOI: 10.1186/s12931-021-01755-3] [Citation(s) in RCA: 12] [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: 01/28/2021] [Accepted: 05/17/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) patients are at increased risk of poor outcome from Coronavirus disease (COVID-19). Early data suggest elevated Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) receptor angiotensin converting enzyme 2 (ACE2) expression, but relationships to disease phenotype and downstream regulators of inflammation in the Renin-Angiotensin system (RAS) are unknown. We aimed to determine the relationship between RAS gene expression relevant to SARS-CoV-2 infection in the lung with disease characteristics in COPD, and the regulation of newly identified SARS-CoV-2 receptors and spike-cleaving proteases, important for SARS-CoV-2 infection. METHODS We quantified gene expression using RNA sequencing of epithelial brushings and bronchial biopsies from 31 COPD and 37 control subjects. RESULTS ACE2 gene expression (log2-fold change (FC)) was increased in COPD compared to ex-smoking (HV-ES) controls in epithelial brushings (0.25, p = 0.042) and bronchial biopsies (0.23, p = 0.050), and correlated with worse lung function (r = - 0.28, p = 0.0090). ACE2 was further increased in frequent exacerbators compared to infrequent exacerbators (0.51, p = 0.00045) and associated with use of ACE inhibitors (ACEi) (0.50, p = 0.0034), having cardiovascular disease (0.23, p = 0.048) or hypertension (0.34, p = 0.0089), and inhaled corticosteroid use in COPD subjects in bronchial biopsies (0.33, p = 0.049). Angiotensin II receptor type (AGTR)1 and 2 expression was decreased in COPD bronchial biopsies compared to HV-ES controls with log2FC of -0.26 (p = 0.033) and - 0.40, (p = 0.0010), respectively. However, the AGTR1:2 ratio was increased in COPD subjects compared with HV-ES controls, log2FC of 0.57 (p = 0.0051). Basigin, a newly identified potential SARS-CoV-2 receptor was also upregulated in both brushes, log2FC of 0.17 (p = 0.0040), and bronchial biopsies, (log2FC of 0.18 (p = 0.017), in COPD vs HV-ES. Transmembrane protease, serine (TMPRSS)2 was not differentially regulated between control and COPD. However, various other spike-cleaving proteases were, including TMPRSS4 and Cathepsin B, in both epithelial brushes (log2FC of 0.25 (p = 0.0012) and log2FC of 0.56 (p = 5.49E-06), respectively) and bronchial biopsies (log2FC of 0.49 (p = 0.00021) and log2FC of 0.246 (p = 0.028), respectively). CONCLUSION This study identifies key differences in expression of genes related to susceptibility and aetiology of COVID-19 within the COPD lung. Further studies to understand the impact on clinical course of disease are now required.
Collapse
MESH Headings
- Aged
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/metabolism
- Basigin/genetics
- Basigin/metabolism
- COVID-19/diagnosis
- COVID-19/genetics
- COVID-19/metabolism
- COVID-19/physiopathology
- Case-Control Studies
- Female
- Forced Expiratory Volume
- Gene Expression Regulation
- Humans
- Lung/metabolism
- Lung/physiopathology
- Male
- Middle Aged
- Prognosis
- Pulmonary Disease, Chronic Obstructive/diagnosis
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Transcriptome
- Vital Capacity
Collapse
Affiliation(s)
- Alastair Watson
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Lisa Öberg
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Bastian Angermann
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - C Mirella Spalluto
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Michael Hühn
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Hannah Burke
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Doriana Cellura
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Anna Freeman
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Daniel Muthas
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Damla Etal
- Translational Genomics, Discovery Biology, Discovery Sciences, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Graham Belfield
- Translational Genomics, Discovery Biology, Discovery Sciences, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Fredrik Karlsson
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Karl Nordström
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Kris Ostridge
- Faculty of Medicine, University of Southampton, Southampton, UK
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
- Clinical Development, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karl J Staples
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Tom Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, UK.
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK.
| |
Collapse
|