1
|
Zhang W, Dai W, Xie Y, Chen X, Zhang P, Cui W. Retinoic Acid Regulates Allergic Inflammation via Limiting Mast Cell Activation. Food Sci Nutr 2025; 13:e4727. [PMID: 39803223 PMCID: PMC11717043 DOI: 10.1002/fsn3.4727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
BackgroundAllergic diseases have become one of the major public health problems to be addressed in the world today. As a tissue resident cell, mast cells are crucial in the pathogenesis of allergic diseases. Vitamin A is an important fat-soluble vitamin with immunomodulatory functions. Vitamin A deficiency has been shown to be associated with allergic disease states, including asthma; however, no studies have been reported on whether vitamin A deficiency has an effect on the activation of mast cells in allergic reactions. ObjectiveTo explore whether blocking retinoic acid receptors has an effect on mast cell degranulation. Methods Flow cytometry was used to analyze the expression of FCεRIα and CD117 on the cell surface, toluidine blue staining was used to visualize cellular features and morphological changes. ELISA was used to detect histamine release. High-throughput transcriptome sequencing and qRT-PCR were used to detect the expression of relevant signaling pathways and cytokine genes. Western blot was used to detect the expression of relevant signaling pathway proteins. ResultsIn the present study, we found that antagonism of the retinoic acid receptor (RAR) resulted in overactive mast cells and increased their degranulation. Furthermore, inflammatory signaling pathways such as MyD88-IKK-NF-κB and PI3K-Akt-m-TOR were involved in the effect of retinoic acid (RA) on the activation state of mast cells. ConclusionsIn this paper, we demonstrated that blocking RAR can exacerbate its activation state in IgE-mediated mast cells. This study provided new insights into the possibility that vitamin A deficiency exacerbated mast cell activation and thus affectd allergic diseases and their mechanisms.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of PathologyThe First Hospital of Jilin UniversityChangchunChina
| | - Wenwen Dai
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
| | - Yingdong Xie
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
| | - Xingyang Chen
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
| | - Peng Zhang
- Department of Thoracic SurgeryThe First Hospital of Jilin UniversityChangchunChina
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
| |
Collapse
|
2
|
Yan K, Li J, Li Y, Zhu P, Tang X, Yuan D, Yang Y, Gao R, Yuan J, Zhao X. Inflammation modifies the platelet reactivity among thrombocytopenia patients undergoing percutaneous coronary intervention. Platelets 2024; 35:2327835. [PMID: 38655673 DOI: 10.1080/09537104.2024.2327835] [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: 11/16/2023] [Accepted: 02/28/2024] [Indexed: 04/26/2024]
Abstract
Percutaneous coronary intervention (PCI) patients combined with thrombocytopenia (TP) are usually considered to be at low ischemic risk, receiving less proper antiplatelet therapy. However, recent studies reported a paradoxical phenomenon that PCI patients with TP were prone to experience thrombotic events, while the mechanisms and future treatment remain unclear. We aim to investigate whether inflammation modifies platelet reactivity among these patients. Consecutive 10 724 patients undergoing PCI in Fuwai Hospital were enrolled throughout 2013. High-sensitivity C-reactive protein (hsCRP) ≥2 mg/L was considered inflammatory status. TP was defined as platelet count <150×109/L. High on-treatment platelet reactivity (HTPR) was defined as adenosine diphosphate-induced platelet maximum amplitude of thromboelastogram >47mm. Among 6617 patients finally included, 879 (13.3%) presented with TP. Multivariate logistic regression demonstrated that patients with TP were associated with a lower risk of HTPR (odds ratio [OR] 0.64, 95% confidence interval [CI] 0.53-0.76) than those without TP in the overall cohort. In further analysis, among hsCRP <2 mg/L group, patients with TP exhibited a decreased risk of HTPR (OR 0.53, 95% CI 0.41-0.68); however, in hsCRP ≥2mg/L group, TP patients had a similar risk of HTPR as those without TP (OR 0.83, 95% CI 0.63-1.08). Additionally, these results remain consistent across subgroups, including patients presenting with acute coronary syndrome and chronic coronary syndrome. Inflammation modified the platelet reactivity of PCI patients with TP, providing new insights into the mechanisms of the increased thrombotic risk. Future management for this special population should pay more attention to inflammation status and timely adjustment of antiplatelet therapy in TP patients with inflammation.
Collapse
Affiliation(s)
- Kailun Yan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiawen Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yulong Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deshan Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runlin Gao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueyan Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Demopoulos CA, Antonopoulou S, Theoharides TC. Is Platelet-Activating Factor the Missing Link Between COVID-19 and Atherosclerosis? JACC. ADVANCES 2024; 3:101397. [PMID: 39583869 PMCID: PMC11582732 DOI: 10.1016/j.jacadv.2024.101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
|
4
|
Papadopoulou P, Polissidis A, Kythreoti G, Sagnou M, Stefanatou A, Theoharides TC. Anti-Inflammatory and Neuroprotective Polyphenols Derived from the European Olive Tree, Olea europaea L., in Long COVID and Other Conditions Involving Cognitive Impairment. Int J Mol Sci 2024; 25:11040. [PMID: 39456822 PMCID: PMC11507169 DOI: 10.3390/ijms252011040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The European olive tree, Olea europaea L., and its polyphenols hold great therapeutic potential to treat neuroinflammation and cognitive impairment. This review examines the evidence for the anti-inflammatory and neuroprotective actions of olive polyphenols and their potential in the treatment of long COVID and neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Key findings suggest that olive polyphenols exhibit antioxidant, anti-inflammatory, neuroprotective, and antiviral properties, making them promising candidates for therapeutic intervention, especially when formulated in unique combinations. Recommendations for future research directions include elucidating molecular pathways through mechanistic studies, exploring the therapeutic implications of olive polyphenol supplementation, and conducting clinical trials to assess efficacy and safety. Investigating potential synergistic effects with other agents addressing different targets is suggested for further exploration. The evidence reviewed strengthens the translational value of olive polyphenols in conditions involving cognitive dysfunction and emphasizes the novelty of new formulations.
Collapse
Affiliation(s)
- Paraskevi Papadopoulou
- Department of Science and Mathematics, Deree-The American College of Greece, 15342 Athens, Greece; (P.P.)
| | - Alexia Polissidis
- Department of Science and Mathematics, Deree-The American College of Greece, 15342 Athens, Greece; (P.P.)
| | - Georgia Kythreoti
- Department of Science and Mathematics, Deree-The American College of Greece, 15342 Athens, Greece; (P.P.)
| | - Marina Sagnou
- Institute of Biosciences and Applications, National Centre for Scientific Research Demokritos, 15310 Athens, Greece;
| | - Athena Stefanatou
- School of Graduate & Professional Education, Deree–The American College of Greece, 15342 Athens, Greece
| | - Theoharis C. Theoharides
- Institute for Neuro-Immune Medicine-Clearwater, Clearwater, FL 33759, USA
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
5
|
Shavelle DM, Bosson N, French WJ, Thomas JL, Niemann JT, Gausche-Hill M, Rollman JE, Rafique AM, Klomhaus AM, Kloner RA. Association of the COVID-19 Pandemic on Treatment Times for ST-Elevation Myocardial Infarction: Observations from the Los Angeles County Regional System. Am J Cardiol 2024; 213:93-98. [PMID: 38016494 DOI: 10.1016/j.amjcard.2023.11.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/03/2023] [Accepted: 11/11/2023] [Indexed: 11/30/2023]
Abstract
Previous studies have documented longer treatment times and worse outcomes for patients with ST-elevation myocardial infarction (STEMI) who underwent primary percutaneous coronary intervention (PCI) during the COVID-19 pandemic. The objective of the present study was to evaluate the impact of the COVID-19 pandemic on treatment times and outcomes for patients with STEMI who underwent primary PCI within a regional system of care. This was a retrospective study using data from the Los Angeles County Emergency Medical Services Agency. Data on the emergency medical service activations were abstracted for patients with STEMI from March 19, 2020 to January 31, 2021, during the COVID-19 pandemic and for the same interval the previous year. All adult patients (≥18 years) with STEMI who underwent emergent coronary angiography were included. The primary end point was the first medical contact (FMC) to device time. The secondary end points included treatment time intervals, vascular complications, need for emergent coronary artery bypass surgery, length of hospital stay, and in-hospital mortality. During the study period, 3,017 patients underwent coronary angiography for STEMI, 1,893 patients pre-COVID-19 and 1,124 patients during COVID-19 (40% lower). A total of 2,334 patients (77%) underwent PCI. During the COVID-19 period, rates of PCI were significantly lower compared with the control period (75.1% vs 78.7%, p = 0.02). FMC to device time was shorter during the COVID-19 period compared with the control period (median 77.0 vs 81.0 minutes, p = 0.004). For patients with STEMI complicated by out-of-hospital cardiac arrest, FMC to device time was similar during the COVID-19 period compared with the control period (median 95.0 [33.0] vs 100.0 [40.0] minutes, p = 0.34). Vascular complications, the need for emergent bypass surgery, length of hospital stay, and in-hospital mortality were similar between the periods. In conclusion, in this large regional system of care, we found a relatively small but significant decrease in treatment times, yet overall, similar clinical outcomes for patients with STEMI who underwent primary PCI and were treated during the COVID-19 period compared with a control period. These findings suggest that mature cardiac systems of care were able to maintain efficient care despite the challenges of the COVID-19 pandemic.
Collapse
Affiliation(s)
- David M Shavelle
- Memorial Care Heart and Vascular Institute, Long Beach Medical Center, Long Beach, California.
| | - Nichole Bosson
- Los Angeles County EMS Agency, Sante Fe Springs, California; David Geffen School of Medicine, University of California, Los Angeles, California; Department of Emergency Medicine, Harbor UCLA Medical Center, Torrance, California
| | - William J French
- David Geffen School of Medicine, University of California, Los Angeles, California; Division of Cardiology, Harbor UCLA Medical Center, Torrance, CA
| | - Joseph L Thomas
- David Geffen School of Medicine, University of California, Los Angeles, California; Division of Cardiology, Harbor UCLA Medical Center, Torrance, CA
| | - James T Niemann
- David Geffen School of Medicine, University of California, Los Angeles, California; Department of Emergency Medicine, Harbor UCLA Medical Center, Torrance, California
| | - Marianne Gausche-Hill
- Los Angeles County EMS Agency, Sante Fe Springs, California; David Geffen School of Medicine, University of California, Los Angeles, California; Department of Emergency Medicine, Harbor UCLA Medical Center, Torrance, California
| | - Jeffrey Eric Rollman
- Department of Health Policy and Management, UCLA Fielding School of Public Health
| | - Asim M Rafique
- David Geffen School of Medicine, University of California, Los Angeles, California; Division of Cardiology, Department of Medicine, and
| | - Alexandra M Klomhaus
- Department of Medicine Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Robert A Kloner
- Keck School of Medicine, University of Southern California, Los Angeles, California; Huntington Medical Research Institutes, Pasadena, California
| |
Collapse
|
6
|
Li D, Cao W, Zhou Q, Wu X, Song X, Qin H. COVID-19 and primary wound healing: A new insights and advance. Int Wound J 2023; 20:4422-4428. [PMID: 37488776 PMCID: PMC10681437 DOI: 10.1111/iwj.14324] [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: 06/11/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
With the outbreak and pandemic of coronavirus disease-2019 (COVID-19), a huge number of people died of it. Apart from lung injuries, multiple organs have been confirmed to be impaired. In COVID-19 time, primary wound healing processes always prolong, however, its possible underlying mechanisms are still unclear. Therefore, to overcome this clinical problem, clarifying its underlying mechanisms clearly is necessary and urgently needed. In this review, we summarized that COVID-19 can prolong primary wound healing by inducing excessive inflammation and oxidative stress, disturbing immune system and haematological system, as well as influencing the functions and viability of epidermal stem cells (ESCs). Otherwise, we summarized that the strict control measures of blocking up COVID-19 pandemic can also have side effects on primary wound healing process.
Collapse
Affiliation(s)
- Danyi Li
- Department of OphthalmologyJiading District Central Hospital Affiliated Shanghai University of Medicine & Health SciencesShanghaiChina
| | - Wenjie Cao
- Department of OphthalmologyJiading District Central Hospital Affiliated Shanghai University of Medicine & Health SciencesShanghaiChina
| | - Qun Zhou
- Department of OphthalmologyJiading District Central Hospital Affiliated Shanghai University of Medicine & Health SciencesShanghaiChina
| | - Xiaomin Wu
- Department of OphthalmologyJiading District Central Hospital Affiliated Shanghai University of Medicine & Health SciencesShanghaiChina
| | - Xiayun Song
- Department of OphthalmologyJiading District Central Hospital Affiliated Shanghai University of Medicine & Health SciencesShanghaiChina
| | - Haofang Qin
- Department of OphthalmologyJiading District Central Hospital Affiliated Shanghai University of Medicine & Health SciencesShanghaiChina
| |
Collapse
|
7
|
Doaei S, Mardi A, Zare M. Role of micronutrients in the modulation of immune system and platelet activating factor in patients with COVID-19; a narrative review. Front Nutr 2023; 10:1207237. [PMID: 37781112 PMCID: PMC10540693 DOI: 10.3389/fnut.2023.1207237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Background Dietary micronutrients may play important roles in the improvement of the immune responses against SARS-CoV-2. This study aimed to assess the effect of micronutrients on platelet activating factor (PAF) and immunity with a special focus on the coronavirus disease 2019 (COVID-19). Methods All paper published in English on the effects of micronutrients including fat soluble vitamins, water soluble vitamins, and minerals on PAF, immunity, and COVID-19 were collected from online valid databases. Results Vitamin A may modulate the expression of PAF-receptor gene in patients with COVID-19. Vitamin D may modulate inflammatory response through influencing PAF pathway. Vitamin E may improve COVID-19 related heart injuries by exert anti-PAF activities. Vitamin C status may have PAF related anti-inflammatory and micro-thrombotic effects in SARS-CoV-2 patients. Furthermore, some trace elements such as copper, selenium, and iron may have key roles in strengthens immunity by inactivate PAF acetyl hydrolase. Conclusion This narrative review study highlighted the importance of micronutrients in the improvement of immune function through modulation of PAF in patients with COVID-19. Further longitudinal studies are warranted.
Collapse
Affiliation(s)
- Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afrouz Mardi
- Department of Public Health, School of Health, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Zare
- Department of Nutrition, Khalkhal University of Medical Sciences, Khalkhal, Iran
| |
Collapse
|
8
|
Arvinte OM, Senila L, Becze A, Amariei S. Rowanberry-A Source of Bioactive Compounds and Their Biopharmaceutical Properties. PLANTS (BASEL, SWITZERLAND) 2023; 12:3225. [PMID: 37765389 PMCID: PMC10536293 DOI: 10.3390/plants12183225] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/26/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
After a period of intense development in the synthesis pharmaceutical industry, plants are making a comeback in the public focus as remedies or therapeutic adjuvants and in disease prevention and ensuring the wellbeing and equilibrium of the human body. Plants are being recommended more and more in alimentation, in their natural form, or as extracts, supplements or functional aliments. People, in general, are in search of new sources of nutrients and phytochemicals. As a result, scientific research turns to lesser known and used plants, among them being rowanberries, a species of fruit very rich in nutrients and underused due to their bitter astringent taste and a lack of knowledge regarding the beneficial effects of these fruit. Rowan fruits (rowanberries) are a rich source of vitamins, polysaccharides, organic acids and minerals. They are also a source of natural polyphenols, which are often correlated with the prevention and treatment of modern world diseases. This article presents the existing data regarding the chemical composition, active principles and biopharmaceutical properties of rowan fruits and the different opportunities for their usage.
Collapse
Affiliation(s)
- Ofelia Marioara Arvinte
- Faculty of Food Engineering, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
| | - Lăcrimioara Senila
- INCDO-INOE 2000, Research Institute for Analytical Instrumentation, 67 Donath Street, 400293 Cluj-Napoca, Romania; (L.S.); (A.B.)
| | - Anca Becze
- INCDO-INOE 2000, Research Institute for Analytical Instrumentation, 67 Donath Street, 400293 Cluj-Napoca, Romania; (L.S.); (A.B.)
| | - Sonia Amariei
- Faculty of Food Engineering, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
| |
Collapse
|
9
|
Zhu J, Yan H, Shi M, Zhang M, Lu J, Wang J, Chen L, Wang Y, Li L, Miao L, Zhang H. Luteolin inhibits spike protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) binding to angiotensin-converting enzyme 2. Phytother Res 2023; 37:3508-3521. [PMID: 37166054 DOI: 10.1002/ptr.7826] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 05/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a respiratory illness that poses a serious threat to global public health. In an essential step during infection, SARS-CoV-2 uses the receptor-binding domain (RBD) of the spike (S) protein to engage with angiotensin-converting enzyme 2 (ACE2) in host cells. Chinese herbal medicines and their active components exhibit antiviral activity, with luteolin being a flavonoid that can significantly inhibit SARS-CoV infection. However, whether it can block the interaction between the S-protein RBD of SARS-CoV-2 and ACE2 has not yet been elucidated. Here, we investigated the effects of luteolin on the binding of the S-protein RBD to ACE2. By employing a competitive binding assay in vitro, we found that luteolin significantly blocked the binding of S-protein RBD to ACE2 with IC50 values of 0.61 mM, which was confirmed by the neutralized infection with SARS-CoV-2 pseudovirus in vivo. A surface plasmon resonance-based competition assay revealed that luteolin significantly affects the binding of the S-protein RBD to the ACE2 receptor. Molecular docking was performed to predict the binding sites of luteolin to the S-protein RBD-ACE2 complex. The active binding sites were defined based on published literature, and we found that luteolin might interfere with the mixture at residues including LYS353, ASP30, and TYR83 in the cellular ACE2 receptor and GLY496, GLN498, TYR505, LEU455, GLN493, and GLU484 in the S-protein RBD. These residues may together form attractive charges and destroy the stable interaction of S-protein RBD-ACE2. Luteolin also inhibits SARS-CoV-2 spike protein-induced platelet spreading, thereby inhibiting the binding of the spike protein to ACE2. Our results are the first to provide evidence that luteolin is an anti-SARS-CoV-2 agent associated with interference between viral S-protein RBD-ACE2 interactions.
Collapse
Affiliation(s)
- Junjie Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huimin Yan
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengyao Shi
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Min Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Lu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiabao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Miao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
10
|
Beheshti M, Neisi N, Parsanahad M, Rasti M, Nashibi R, Cheraghian B. Correlation of vitamin D levels with serum parameters in Covid-19 patients. Clin Nutr ESPEN 2023; 55:325-331. [PMID: 37202065 DOI: 10.1016/j.clnesp.2023.04.012] [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: 01/26/2023] [Revised: 04/05/2023] [Accepted: 04/17/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND It is well-established that vitamins have many beneficial roles and protect humans against inflammatory diseases. Vitamin D, a lipid-soluble vitamin, plays a crucial role in viral infections. Therefore, this study aimed to investigate if serum 25(OH)D levels affect morbidity, mortality, and levels of inflammatory parameters in COVID-19 patients. METHODS 140 COVID-19 patients participated in this study (65 outpatients and 75 inpatients). Their blood samples were collected to determine TNFα, IL-6, D-dimer, zinc, Ca2+, and 25(OH)D levels. Patients with O2 saturation <93% were admitted and hospitalized in the infectious disease ward (inpatient group). Patients with O2 saturation >93% received routine treatment and were discharged (Outpatient group). RESULTS The serum levels of 25(OH)D in the inpatient group were significantly lower than those in the outpatient group (p < 0.001). Serum TNF-α, IL-6, and D-dimer levels in the inpatient group were significantly higher than those in the outpatient group (p < 0.001). Serum TNF-α, IL-6, and D-dimer levels were inversely correlated with 25(OH)D levels. No significant differences were observed in the serum levels of zinc and Ca2+ between the studied groups (p = 0.96, p = 0.41 respectively). Ten out of 75 patients in the inpatient group were admitted to ICU (intubated). Nine out of them lost their lives (the mortality rate in ICU-admitted patients was 90%). CONCLUSIONS The lower mortality and severity of COVID-19 patients with higher 25(OH)D levels represented that this vitamin alleviates the severity of COVID-19.
Collapse
Affiliation(s)
- Masoud Beheshti
- Infectious and Tropical Diseases Research Center, Health Research Institute, Department of Medical Virology, The School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Niloofar Neisi
- Infectious and Tropical Diseases Research Center, Health Research Institute, Department of Medical Virology, The School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mehdi Parsanahad
- Infectious and Tropical Diseases Research Center, Health Research Institute, Department of Medical Virology, The School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Rasti
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Roohangiz Nashibi
- Infectious and Tropical Diseases Research Center, Health Research Institute, Infectious Diseases and Tropical Medicine Ward, Razi Teaching Hospital, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Bahman Cheraghian
- Alimentary Tract Research Center, Clinical Sciences Research Institute, Department of Biostatistics and Epidemiology, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
11
|
Farooqui AA, Farooqui T, Sun GY, Lin TN, Teh DBL, Ong WY. COVID-19, Blood Lipid Changes, and Thrombosis. Biomedicines 2023; 11:biomedicines11041181. [PMID: 37189799 DOI: 10.3390/biomedicines11041181] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Although there is increasing evidence that oxidative stress and inflammation induced by COVID-19 may contribute to increased risk and severity of thromboses, the underlying mechanism(s) remain to be understood. The purpose of this review is to highlight the role of blood lipids in association with thrombosis events observed in COVID-19 patients. Among different types of phospholipases A2 that target cell membrane phospholipids, there is increasing focus on the inflammatory secretory phospholipase A2 IIA (sPLA2-IIA), which is associated with the severity of COVID-19. Analysis indicates increased sPLA2-IIA levels together with eicosanoids in the sera of COVID patients. sPLA2 could metabolise phospholipids in platelets, erythrocytes, and endothelial cells to produce arachidonic acid (ARA) and lysophospholipids. Arachidonic acid in platelets is metabolised to prostaglandin H2 and thromboxane A2, known for their pro-coagulation and vasoconstrictive properties. Lysophospholipids, such as lysophosphatidylcholine, could be metabolised by autotaxin (ATX) and further converted to lysophosphatidic acid (LPA). Increased ATX has been found in the serum of patients with COVID-19, and LPA has recently been found to induce NETosis, a clotting mechanism triggered by the release of extracellular fibres from neutrophils and a key feature of the COVID-19 hypercoagulable state. PLA2 could also catalyse the formation of platelet activating factor (PAF) from membrane ether phospholipids. Many of the above lipid mediators are increased in the blood of patients with COVID-19. Together, findings from analyses of blood lipids in COVID-19 patients suggest an important role for metabolites of sPLA2-IIA in COVID-19-associated coagulopathy (CAC).
Collapse
Affiliation(s)
- Akhlaq A Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Tahira Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11929, Taiwan
| | - Daniel B L Teh
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Neurobiology Research Programme, Life Sciences Institute, National University of Singapore, Singapore 119260, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Neurobiology Research Programme, Life Sciences Institute, National University of Singapore, Singapore 119260, Singapore
| |
Collapse
|
12
|
Müller L, Di Benedetto S. Aged brain and neuroimmune responses to COVID-19: post-acute sequelae and modulatory effects of behavioral and nutritional interventions. Immun Ageing 2023; 20:17. [PMID: 37046272 PMCID: PMC10090758 DOI: 10.1186/s12979-023-00341-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
Advanced age is one of the significant risk determinants for coronavirus disease 2019 (COVID-19)-related mortality and for long COVID complications. The contributing factors may include the age-related dynamical remodeling of the immune system, known as immunosenescence and chronic low-grade systemic inflammation. Both of these factors may induce an inflammatory milieu in the aged brain and drive the changes in the microenvironment of neurons and microglia, which are characterized by a general condition of chronic inflammation, so-called neuroinflammation. Emerging evidence reveals that the immune privilege in the aging brain may be compromised. Resident brain cells, such as astrocytes, neurons, oligodendrocytes and microglia, but also infiltrating immune cells, such as monocytes, T cells and macrophages participate in the complex intercellular networks and multiple reciprocal interactions. Especially changes in microglia playing a regulatory role in inflammation, contribute to disturbing of the brain homeostasis and to impairments of the neuroimmune responses. Neuroinflammation may trigger structural damage, diminish regeneration, induce neuronal cell death, modulate synaptic remodeling and in this manner negatively interfere with the brain functions.In this review article, we give insights into neuroimmune interactions in the aged brain and highlight the impact of COVID-19 on the functional systems already modulated by immunosenescence and neuroinflammation. We discuss the potential ways of these interactions with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and review proposed neuroimmune mechanisms and biological factors that may contribute to the development of persisting long COVID conditions. We summarize the potential mechanisms responsible for long COVID, including inflammation, autoimmunity, direct virus-mediated cytotoxicity, hypercoagulation, mitochondrial failure, dysbiosis, and the reactivation of other persisting viruses, such as the Cytomegalovirus (CMV). Finally, we discuss the effects of various interventional options that can decrease the propagation of biological, physiological, and psychosocial stressors that are responsible for neuroimmune activation and which may inhibit the triggering of unbalanced inflammatory responses. We highlight the modulatory effects of bioactive nutritional compounds along with the multimodal benefits of behavioral interventions and moderate exercise, which can be applied as postinfectious interventions in order to improve brain health.
Collapse
Affiliation(s)
- Ludmila Müller
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - Svetlana Di Benedetto
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| |
Collapse
|
13
|
de Carvalho JCS, da Silva-Neto PV, Toro DM, Fuzo CA, Nardini V, Pimentel VE, Pérez MM, Fraga-Silva TFC, Oliveira CNS, Degiovani AM, Ostini FM, Feitosa MR, Parra RS, da Rocha JJR, Feres O, Vilar FC, Gaspar GG, Santos IKFM, Fernandes APM, Maruyama SR, Russo EMS, Bonato VLD, Cardoso CRB, Dias-Baruffi M, Faccioli LH, Sorgi CA. The Interplay among Glucocorticoid Therapy, Platelet-Activating Factor and Endocannabinoid Release Influences the Inflammatory Response to COVID-19. Viruses 2023; 15:v15020573. [PMID: 36851787 PMCID: PMC9959303 DOI: 10.3390/v15020573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
COVID-19 is associated with a dysregulated immune response. Currently, several medicines are licensed for the treatment of this disease. Due to their significant role in inhibiting pro-inflammatory cytokines and lipid mediators, glucocorticoids (GCs) have attracted a great deal of attention. Similarly, the endocannabinoid (eCB) system regulates various physiological processes including the immunological response. Additionally, during inflammatory and thrombotic processes, phospholipids from cell membranes are cleaved to produce platelet-activating factor (PAF), another lipid mediator. Nonetheless, the effect of GCs on this lipid pathway during COVID-19 therapy is still unknown. This is a cross-sectional study involving COVID-19 patients (n = 200) and healthy controls (n = 35). Target tandem mass spectrometry of plasma lipid mediators demonstrated that COVID-19 severity affected eCBs and PAF synthesis. This increased synthesis of eCB was adversely linked with systemic inflammatory markers IL-6 and sTREM-1 levels and neutrophil counts. The use of GCs altered these lipid pathways by reducing PAF and increasing 2-AG production. Corroborating this, transcriptome analysis of GC-treated patients blood leukocytes showed differential modulation of monoacylglycerol lipase and phospholipase A2 gene expression. Altogether, these findings offer a breakthrough in our understanding of COVID-19 pathophysiology, indicating that GCs may promote additional protective pharmacological effects by influencing the eCB and PAF pathways involved in the disease course.
Collapse
Affiliation(s)
- Jonatan C. S. de Carvalho
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-901, SP, Brazil
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Pedro V. da Silva-Neto
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada-PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas-UFAM, Manaus 69080-900, AM, Brazil
| | - Diana M. Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada-PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas-UFAM, Manaus 69080-900, AM, Brazil
| | - Carlos A. Fuzo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Viviani Nardini
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Vinícius E. Pimentel
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-900, SP, Brazil
| | - Malena M. Pérez
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Thais F. C. Fraga-Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-900, SP, Brazil
| | - Camilla N. S. Oliveira
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-900, SP, Brazil
| | - Augusto M. Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirao Preto 14085-000, SP, Brazil
| | - Fátima M. Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirao Preto 14085-000, SP, Brazil
| | - Marley R. Feitosa
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14048-900, SP, Brazil
| | - Rogerio S. Parra
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14048-900, SP, Brazil
| | - José J. R. da Rocha
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14048-900, SP, Brazil
| | - Omar Feres
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14048-900, SP, Brazil
| | - Fernando C. Vilar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14049-900, SP, Brazil
| | - Gilberto G. Gaspar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14049-900, SP, Brazil
| | - Isabel K. F. M. Santos
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-900, SP, Brazil
| | - Ana P. M. Fernandes
- Departamento de Enfermagem Geral e Especializada, Escola de Enfermagem de Ribeirão Preto-EERP, Universidade de São Paulo-USP, Ribeirao Preto 14040-902, SP, Brazil
| | - Sandra R. Maruyama
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos-UFSCar, Sao Carlos 13565-905, SP, Brazil
| | - Elisa M. S. Russo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Vânia L. D. Bonato
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-900, SP, Brazil
| | - Cristina R. B. Cardoso
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Lúcia H. Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-903, SP, Brazil
| | - Carlos A. Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-901, SP, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada-PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas-UFAM, Manaus 69080-900, AM, Brazil
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirao Preto 14040-900, SP, Brazil
- Correspondence: ; Tel.: +55-(16)-3315-9176
| | | |
Collapse
|
14
|
Zhang K, Wang L, Peng J, Sangji K, Luo Y, Zeng Y, Zeweng Y, Fan G. Traditional Tibetan medicine to fight against COVID-19: Basic theory and therapeutic drugs. Front Pharmacol 2023; 14:1098253. [PMID: 36874035 PMCID: PMC9978713 DOI: 10.3389/fphar.2023.1098253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
The Coronavirus Diseases 2019 (COVID-19) has been rapidly spreading globally and has caused severe harm to the health of people and a substantial social burden. In response to this situation, experts around the world have considered various treatments, including the use of traditional medicine. Traditional Tibetan medicine (TTM), one of the traditional medicines in China, has played an important role in the treatment of infectious diseases in history. It has formed a solid theoretical foundation and accumulated rich experience in the treatment of infectious diseases. In this review, we provide a comprehensive introduction to the basic theory, treatment strategies, and commonly used drugs of TTM for the treatment of COVID-19. In addition, the efficacies and potential mechanisms of these TTM drugs against COVID-19 are discussed based on available experimental data. This review may provide important information for the basic research, clinical application and drug development of traditional medicines for the treatment of COVID-19 or other infectious diseases. More pharmacological studies are needed to reveal the therapeutic mechanisms and active ingredients of TTM drugs in the treatment of COVID-19.
Collapse
Affiliation(s)
- Kun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijie Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayan Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kangzhuo Sangji
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuting Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yujiao Zeng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongzhong Zeweng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gang Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Varga I, Michalka P, Mištinová JP. Complications after administration of mRNA vaccine against COVID-19 - case report and short review. VNITRNI LEKARSTVI 2023; 69:20-27. [PMID: 37468319 DOI: 10.36290/vnl.2023.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
The pandemic of the disease COVID-19 (COronaVIrus Disease 2019) caused by the SARS-CoV-2 coronavirus (severe acute respiratory syndrome coronavirus 2) resulted in millions of deaths and many patients have chronic consequences after overcoming the acute condition. Several vaccines have been developed in an effort to stop the spread of the virus, but they have potentially serious adverse effects. We present a case report of a patient with acute (myocarditis, exacerbation of bronchial asthma) and long-term (postural orthostatic tachycardia syndrome - POTS) complications after vaccination with the second dose of mRNA vaccine BNT162b2 (Comirnaty®). Treatment consists of regimen measures, numerous pharmacotherapy (metoprolol, ivabradine, corticosteroids, antihistamines, antiphlogistics, bronchodilators) and several nutraceuticals (maritime pine bark extract, quercetin, vitamins, magnesium, phosphatidylcholine). In the discussion, we analyze post-vaccination injury and present a short review of the current literature.
Collapse
|
16
|
Tremblay MÈ, Almsherqi ZA, Deng Y. Plasmalogens and platelet-activating factor roles in chronic inflammatory diseases. Biofactors 2022; 48:1203-1216. [PMID: 36370412 DOI: 10.1002/biof.1916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022]
Abstract
Fatty acids and phospholipid molecules are essential for determining the structure and function of cell membranes, and they hence participate in many biological processes. Platelet activating factor (PAF) and its precursor plasmalogen, which represent two subclasses of ether phospholipids, have attracted increasing research attention recently due to their association with multiple chronic inflammatory, neurodegenerative, and metabolic disorders. These pathophysiological conditions commonly involve inflammatory processes linked to an excess presence of PAF and/or decreased levels of plasmalogens. However, the molecular mechanisms underlying the roles of plasmalogens in inflammation have remained largely elusive. While anti-inflammatory responses most likely involve the plasmalogen signal pathway; pro-inflammatory responses recruit arachidonic acid, a precursor of pro-inflammatory lipid mediators which is released from membrane phospholipids, notably derived from the hydrolysis of plasmalogens. Plasmalogens per se are vital membrane phospholipids in humans. Changes in their homeostatic levels may alter cell membrane properties, thus affecting key signaling pathways that mediate inflammatory cascades and immune responses. The plasmalogen analogs of PAF are also potentially important, considering that anti-PAF activity has strong anti-inflammatory effects. Plasmalogen replacement therapy was further identified as a promising anti-inflammatory strategy allowing for the relief of pathological hallmarks in patients affected by chronic diseases with an inflammatory component. The aim of this Short Review is to highlight the emerging roles and implications of plasmalogens in chronic inflammatory disorders, along with the promising outcomes of plasmalogen replacement therapy for the treatment of various PAF-related chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, Canada
- Department of Molecular Medicine, Université de Laval, Québec City, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
| | - Zakaria A Almsherqi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| |
Collapse
|
17
|
Pantazi D, Tellis C, Tselepis AD. Oxidized phospholipids and lipoprotein-associated phospholipase A 2 (Lp-PLA 2 ) in atherosclerotic cardiovascular disease: An update. Biofactors 2022; 48:1257-1270. [PMID: 36192834 DOI: 10.1002/biof.1890] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022]
Abstract
Inflammation and oxidative stress conditions lead to a variety of oxidative modifications of lipoprotein phospholipids implicated in the occurrence and development of atherosclerotic lesions. Lipoprotein-associated phospholipase A2 (Lp-PLA2 ) is established as an independent risk biomarker of atherosclerosis-related cardiovascular disease (ASCVD) and mediates vascular inflammation through the regulation of lipid metabolism in the blood and in atherosclerotic lesions. Lp-PLA2 is associated with low- and high-density lipoproteins and Lipoprotein (a) in human plasma and specifically hydrolyzes oxidized phospholipids involved in oxidative stress modification. Several oxidized phospholipids (OxPLs) subspecies can be detoxified through enzymatic degradation by Lp-PLA2 activation, forming lysophospholipids and oxidized non-esterified fatty acids (OxNEFAs). Lysophospholipids promote the expression of adhesion molecules, stimulate cytokines production (TNF-α, IL-6), and attract macrophages to the arterial intima. The present review article discusses new data on the functional roles of OxPLs and Lp-PLA2 associated with lipoproteins.
Collapse
Affiliation(s)
- Despoina Pantazi
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Constantinos Tellis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
18
|
Thyagarajan A, Awasthi K, Rapp CM, Johnson RM, Chen Y, Miller KL, Travers JB, Sahu RP. Topical application of gemcitabine generates microvesicle particles in human and murine skin. Biofactors 2022; 48:1295-1304. [PMID: 36504167 PMCID: PMC9789190 DOI: 10.1002/biof.1924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022]
Abstract
Chemotherapy has remained the mainstay for the treatment of multiple types of cancers. In particular, topical use of chemotherapy has been used for skin cancers. Though effective, topical chemotherapy has been limited due to adverse effects such as local and even systemic toxicities. Our recent studies demonstrated that exposure to pro-oxidative stressors, including therapeutic agents induces the generation of extracellular vesicles known as microvesicle particles (MVP) which are dependent on activation of the Platelet-activating factor-receptor (PAFR), a G-protein coupled receptor present on various cell types, and acid sphingomyelinase (aSMase), an enzyme required for MVP biogenesis. Based upon this premise, we tested the hypothesis that topical application of gemcitabine will induce MVP generation in human and murine skin. Our ex vivo studies using human skin explants demonstrate that gemcitabine treatment results in MVP generation in a dose-dependent manner in a process blocked by PAFR antagonist and aSMase inhibitor. Importantly, gemcitabine-induced MVPs carry PAFR agonists. To confirm the mechanisms, we employed PAFR-expressing and deficient (Ptafr-/- ) mouse models as well as mice deficient in aSMase enzyme (Spmd1-/- ). Similar to the findings using pharmacologic tools, genetic-based approaches demonstrate that gemcitabine-induced MVP release in WT mice was blunted in Ptafr-/- and Spmd1-/- mice. These findings demonstrate a novel mechanism by which local chemotherapy can generate bioactive components as a bystander effect in a process that is dependent upon the PAFR-aSMase pathway.
Collapse
Affiliation(s)
- Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
- Correspondence to: (AT) and (RPS), 230 Health Sciences Bldg, 3640 Colonel Glenn Hwy, Dayton, OH45435
| | - Krishna Awasthi
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
| | - Christine M. Rapp
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
| | - R. Michael Johnson
- Department of Orthopedics and Plastic Surgery, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
| | - Yanfang Chen
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
| | - Kelly L.R. Miller
- Department of Internal Medicine, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
| | - Jeffrey B. Travers
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
- Department of Dermatology, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
- Dayton VA Medical Center, Dayton, OH 45428
| | - Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45435
- Correspondence to: (AT) and (RPS), 230 Health Sciences Bldg, 3640 Colonel Glenn Hwy, Dayton, OH45435
| |
Collapse
|
19
|
Antonopoulou S, Petsini F, Detopoulou M, Theoharides TC, Demopoulos CA. Is there an interplay between the SARS-CoV-2 spike protein and Platelet-Activating factor? Biofactors 2022; 48:1271-1283. [PMID: 35852257 PMCID: PMC9349578 DOI: 10.1002/biof.1877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022]
Abstract
Previous publications have reported a potent effect of COVID-19 on platelet function and that the Spike protein enhances washed human platelet aggregation induced by various agonists. This study aims to evaluate whether mRNA vaccination for COVID-19 affects human platelet-rich plasma (hPRP) aggregation response, whether a recombinant Spike protein modulates PAF-induced aggregation in hPRP and in washed rabbit platelets (WRP), and to investigate the effect of recombinant Spike protein on the PAF production in the U-937 cell line. Our results showed that PRP from vaccinated individuals exhibited ex vivo lower EC50 values in response to PAF, ADP, and collagen. Platelet incubation with the Spike protein alone did not induce aggregation either in hPRP or in WRP, but resulted in augmentation of in vitro PAF-induced aggregation in hPRP from non-vaccinated individuals and in WRP. When PRP from vaccinated individuals was incubated with the Spike protein and PAF was subsequently added, elimination of the secondary wave of the biphasic aggregation curve was recorded compared with the aggregation induced by PAF alone. Collagen-induced in vitro aggregation was dose-dependently reduced when platelets were pre-incubated with the Spike protein in all tested aggregation experiments. Stimulation of U-937 by the Spike protein induced an increase in intracellular PAF production accompanied by elevation of the activities of all three PAF biosynthetic enzymes. In conclusion, since the Spike protein appears to modulate PAF production and activity, the use of compounds that act as PAF inhibitors, could be considered at least in mild cases of patients infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Smaragdi Antonopoulou
- Laboratory of Biology, Biochemistry and Microbiology, Department of Nutrition‐Dietetics, School of Health Sciences and EducationHarokopio UniversityAthensGreece
| | - Filio Petsini
- Laboratory of Biology, Biochemistry and Microbiology, Department of Nutrition‐Dietetics, School of Health Sciences and EducationHarokopio UniversityAthensGreece
| | - Maria Detopoulou
- Laboratory of Biology, Biochemistry and Microbiology, Department of Nutrition‐Dietetics, School of Health Sciences and EducationHarokopio UniversityAthensGreece
| | - Theoharis C. Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of ImmunologyTufts University School of MedicineBostonMassachusettsUnited States
- School of Graduate Biomedical SciencesTufts University School of MedicineBostonMassachusettsUnited States
- Department of Internal MedicineTufts University School of Medicine and Tufts Medical CenterBostonMassachusettsUnited States
| | | |
Collapse
|
20
|
Theoharides TC, Antonopoulou S, Demopoulos CA. Platelet activating factor: Have we been missing the forest for the trees? Biofactors 2022; 48:1184-1188. [PMID: 36300767 DOI: 10.1002/biof.1908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 01/19/2023]
Affiliation(s)
- Theoharis C Theoharides
- Institute of Neuro-Immune Medicine, Nova Southeastern University, Clearwater, Florida, USA
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Smaragdi Antonopoulou
- Laboratory of Biology, Biochemistry and Microbiology, Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Constantinos A Demopoulos
- Laboratory of Biochemistry, Faculty of Chemistry, National & Kapodistrian University, Athens, Greece
| |
Collapse
|
21
|
Upton JEM, Grunebaum E, Sussman G, Vadas P. Platelet Activating Factor (PAF): A Mediator of Inflammation. Biofactors 2022; 48:1189-1202. [PMID: 36029481 DOI: 10.1002/biof.1883] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022]
Abstract
Platelet-activating factor (PAF) is a phospholipid-derived mediator with an established role in multiple inflammatory states. PAF is synthesized and secreted by multiple cell types and is then rapidly hydrolyzed and degraded to an inactive metabolite, lyso-PAF, by the enzyme PAF acetylhydrolase. In addition to its role in platelet aggregation and activation, PAF contributes to allergic and nonallergic inflammatory diseases such as anaphylaxis, sepsis, cardiovascular disease, neurological disease, and malignancy as demonstrated in multiple animal models and, increasingly, in human disease states. Recent research has demonstrated the importance of the PAF pathway in multiple conditions including the prediction of severe pediatric anaphylaxis, effects on blood-brain barrier permeability, effects on reproduction, ocular diseases, and further understanding of its role in cardiovascular risk. Investigation of PAF as both a biomarker and a therapeutic target continues because of the need for directed management of inflammation. Collectively, studies have shown that therapies focused on the PAF pathway have the potential to provide targeted and effective treatments for multiple inflammatory conditions.
Collapse
Affiliation(s)
- Julia E M Upton
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Gordon Sussman
- Division of Clinical Immunology and Allergy, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Peter Vadas
- Division of Clinical Immunology and Allergy, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Kell DB, Pretorius E. The potential role of ischaemia-reperfusion injury in chronic, relapsing diseases such as rheumatoid arthritis, Long COVID, and ME/CFS: evidence, mechanisms, and therapeutic implications. Biochem J 2022; 479:1653-1708. [PMID: 36043493 PMCID: PMC9484810 DOI: 10.1042/bcj20220154] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Ischaemia-reperfusion (I-R) injury, initiated via bursts of reactive oxygen species produced during the reoxygenation phase following hypoxia, is well known in a variety of acute circumstances. We argue here that I-R injury also underpins elements of the pathology of a variety of chronic, inflammatory diseases, including rheumatoid arthritis, ME/CFS and, our chief focus and most proximally, Long COVID. Ischaemia may be initiated via fibrin amyloid microclot blockage of capillaries, for instance as exercise is started; reperfusion is a necessary corollary when it finishes. We rehearse the mechanistic evidence for these occurrences here, in terms of their manifestation as oxidative stress, hyperinflammation, mast cell activation, the production of marker metabolites and related activities. Such microclot-based phenomena can explain both the breathlessness/fatigue and the post-exertional malaise that may be observed in these conditions, as well as many other observables. The recognition of these processes implies, mechanistically, that therapeutic benefit is potentially to be had from antioxidants, from anti-inflammatories, from iron chelators, and via suitable, safe fibrinolytics, and/or anti-clotting agents. We review the considerable existing evidence that is consistent with this, and with the biochemical mechanisms involved.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| |
Collapse
|
23
|
Abstract
Medical nutrition therapy may have a key role in the COVID-19 pandemic. Given the spread of misinformation, the present review organizes and summarizes nutrition recommendations regarding COVID-19, serving as a reference guide for health professionals. Nineteen official recommendations were included of international, US, Asian, European, Canadian, and Australian origin on (i) lactation, (ii) nutrition during quarantine, (iii) nutrition in high-risk groups, (iv) nutrition for recovery at home, and (v) nutrition in hospital. Breastfeeding is encouraged, and the role of hydration and the adoption of a healthy diet during quarantine are emphasized. Older people and/or people with comorbidities should be checked for malnutrition and follow a healthy diet. For patients recovering at home, hydration, protein, and energy intake should be ensured. For hospitalized patients, early feeding with a priority on enteral route is recommended.
Collapse
Affiliation(s)
- Paraskevi Detopoulou
- Department of Clinical Nutrition, General Hospital Korgialenio-Benakio (Red Cross Hospital), Athens, Greece (Dr Detopoulou, Ms Tsouma, and Mr Papamikos); and Department of Dietetics and Nutrition, University of Peloponnese, Kalamata, Greece (Dr Detopoulou)
| | - Christina Tsouma
- Department of Clinical Nutrition, General Hospital Korgialenio-Benakio (Red Cross Hospital), Athens, Greece (Dr Detopoulou, Ms Tsouma, and Mr Papamikos); and Department of Dietetics and Nutrition, University of Peloponnese, Kalamata, Greece (Dr Detopoulou)
| | - Vassilios Papamikos
- Department of Clinical Nutrition, General Hospital Korgialenio-Benakio (Red Cross Hospital), Athens, Greece (Dr Detopoulou, Ms Tsouma, and Mr Papamikos); and Department of Dietetics and Nutrition, University of Peloponnese, Kalamata, Greece (Dr Detopoulou)
| |
Collapse
|
24
|
Szukiewicz D, Wojdasiewicz P, Watroba M, Szewczyk G. Mast Cell Activation Syndrome in COVID-19 and Female Reproductive Function: Theoretical Background vs. Accumulating Clinical Evidence. J Immunol Res 2022; 2022:9534163. [PMID: 35785029 PMCID: PMC9242765 DOI: 10.1155/2022/9534163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), a pandemic disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, can affect almost all systems and organs of the human body, including those responsible for reproductive function in women. The multisystem inflammatory response in COVID-19 shows many analogies with mast cell activation syndrome (MCAS), and MCAS may be an important component in the course of COVID-19. Of note, the female sex hormones estradiol (E2) and progesterone (P4) significantly influence mast cell (MC) behavior. This review presents the importance of MCs and the mediators from their granules in the female reproductive system, including pregnancy, and discusses the mechanism of potential disorders related to MCAS. Then, the available data on COVID-19 in the context of hormonal disorders, the course of endometriosis, female fertility, and the course of pregnancy were compiled to verify intuitively predicted threats. Surprisingly, although COVID-19 hyperinflammation and post-COVID-19 illness may be rooted in MCAS, the available clinical data do not provide grounds for treating this mechanism as significantly increasing the risk of abnormal female reproductive function, including pregnancy. Further studies in the context of post COVID-19 condition (long COVID), where inflammation and a procoagulative state resemble many aspects of MCAS, are needed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Wojdasiewicz
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Watroba
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Szewczyk
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
25
|
Detopoulou P, Al-Khelefawi ZH, Kalonarchi G, Papamikos V. Formulation of the Menu of a General Hospital After Its Conversion to a "COVID Hospital": A Nutrient Analysis of 28-Day Menus. Front Nutr 2022; 9:833628. [PMID: 35495923 PMCID: PMC9043649 DOI: 10.3389/fnut.2022.833628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/10/2022] [Indexed: 12/19/2022] Open
Abstract
Aim The aim of the present study was to modify the hospital menu to increase energy and protein provision in COVID-19 patients. Methods After the conversion of our hospital to a COVID-19 hospital, eggs, and comfort foods such as vanilla pudding were added to the menu to boost energy and protein intake of patients. All meals of the standard menu of the hospital, i.e., breakfast, lunch, and dinner were recorded for 14 consecutive days during two periods: pre-COVID-19 and after being converted to a "COVID hospital." The menus were analyzed with the use of the USDA database. Results The total content of energy (1,873 ± 87 vs. 2,489 ± 137 Kcal), protein (97 ± 11 vs. 126.4 ± 18.7 g), fat (55 ± 9 vs. 74.1 ± 12.8 g), and carbohydrate (241.0 ± 16.0 vs. 323.0 ± 16.0 g) of the provided menus was increased in the COVID-19 period compared to the pre-COVID period. The leucine provision was also increased (4.8 ± 1.08 vs. 7.2 ± 1.3 g). Changes in protein and leucine were greater for breakfast (10 vs. 21 g protein and 0.8 vs. 1.7 g of leucine). The menu during COVID-29 provided more vitamin C (69.5 vs. 109.4 mg), thiamine (1.5 vs. 1.6 mg), riboflavin (2.1 vs. 2.6 mg), niacin (20.6 vs. 27.2 mg), pantothenic Acid (5.7 vs. 7.9 mg), vitamin B6 (2 vs. 2.6 mg), folate (274 vs. 334 μg), B 12 (4.8 vs. 6.2 μg), choline (296 vs. 458 mg) as well as vitamins A (8,564 vs. 21,258 IU), D (3.9 vs. 4.7 μg), and K (59.3-111.5 μg). As far as micronutrients are concerned, the provisions of calcium (972 vs. 1375 mg), iron (10.2-12.8 mg), magnesium (236 vs. 294 mg), phosphorus (1,325 vs. 1,807 mg), copper (1.0 vs. 1.3 mg), manganese (2.1 vs. 2.4 mg) and selenium (148 vs. 183 μg) were increased during the COVID-19 period. Conclusion Simple menu changes and addition of comfort foods can substantially boost the nutrient content of a hospital diet, which in concert with provision of oral nutritional supplements could have an impact on patients' nutritional status.
Collapse
Affiliation(s)
- Paraskevi Detopoulou
- Department of Nutrition, “Korgialenio-Benakio” Hellenic Red Cross Hospital, Athens, Greece
- Department of Nutrition and Dietetics, University of Peloponnese, Kalamata, Greece
| | | | - Garifallia Kalonarchi
- Department of Nutrition, “Korgialenio-Benakio” Hellenic Red Cross Hospital, Athens, Greece
| | - Vasilios Papamikos
- Department of Nutrition, “Korgialenio-Benakio” Hellenic Red Cross Hospital, Athens, Greece
| |
Collapse
|
26
|
Rong P, Wang JL, Angelova A, Almsherqi ZA, Deng Y. Plasmalogenic Lipid Analogs as Platelet-Activating Factor Antagonists: A Potential Novel Class of Anti-inflammatory Compounds. Front Cell Dev Biol 2022; 10:859421. [PMID: 35493091 PMCID: PMC9048793 DOI: 10.3389/fcell.2022.859421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/24/2022] [Indexed: 12/31/2022] Open
Abstract
Plasmalogens and Platelet-Activating Factor (PAF) are both bioactive ether phospholipids. Whereas plasmalogens are recognized for their important antioxidant function and modulatory role in cell membrane structure and dynamics, PAF is a potent pro-inflammatory lipid mediator known to have messenger functions in cell signaling and inflammatory response. The relationship between these two types of lipids has been rarely studied in terms of their metabolic interconversion and reciprocal modulation of the pro-inflammation/anti-inflammation balance. The vinyl-ether bonded plasmalogen lipid can be the lipid sources for the precursor of the biosynthesis of ether-bonded PAF. In this opinion paper, we suggest a potential role of plasmalogenic analogs of PAF as modulators and PAF antagonists (anti-PAF). We discuss that the metabolic interconversion of these two lipid kinds may be explored towards the development of efficient preventive and relief strategies against PAF-mediated pro-inflammation. We propose that plasmalogen analogs, acting as anti-PAF, may be considered as a new class of bioactive anti-inflammatory drugs. Despite of the scarcity of available experimental data, the competition between PAF and its natural plasmalogenic analogs for binding to the PAF receptor (PAF-R) can be proposed as a mechanistic model and potential therapeutic perspective against multiple inflammatory diseases (e.g., cardiovascular and neurodegenerative disorders, diabetes, cancers, and various manifestations in coronavirus infections such as COVID-19).
Collapse
Affiliation(s)
- Pu Rong
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Jie-Li Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Angelina Angelova
- CNRS, Institut Galien Paris-Saclay, Université Paris-Saclay, Châtenay-Malabry, France
| | - Zakaria A. Almsherqi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- *Correspondence: Zakaria A. Almsherqi, ; Yuru Deng,
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
- *Correspondence: Zakaria A. Almsherqi, ; Yuru Deng,
| |
Collapse
|
27
|
Dagla I, Iliou A, Benaki D, Gikas E, Mikros E, Bagratuni T, Kastritis E, Dimopoulos MA, Terpos E, Tsarbopoulos A. Plasma Metabolomic Alterations Induced by COVID-19 Vaccination Reveal Putative Biomarkers Reflecting the Immune Response. Cells 2022; 11:1241. [PMID: 35406806 PMCID: PMC8997405 DOI: 10.3390/cells11071241] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 12/12/2022] Open
Abstract
Vaccination is currently the most effective strategy for the mitigation of the COVID-19 pandemic. mRNA vaccines trigger the immune system to produce neutralizing antibodies (NAbs) against SARS-CoV-2 spike proteins. However, the underlying molecular processes affecting immune response after vaccination remain poorly understood, while there is significant heterogeneity in the immune response among individuals. Metabolomics have often been used to provide a deeper understanding of immune cell responses, but in the context of COVID-19 vaccination such data are scarce. Mass spectrometry (LC-MS) and nuclear magnetic resonance (NMR)-based metabolomics were used to provide insights based on the baseline metabolic profile and metabolic alterations induced after mRNA vaccination in paired blood plasma samples collected and analysed before the first and second vaccination and at 3 months post first dose. Based on the level of NAbs just before the second dose, two groups, "low" and "high" responders, were defined. Distinct plasma metabolic profiles were observed in relation to the level of immune response, highlighting the role of amino acid metabolism and the lipid profile as predictive markers of response to vaccination. Furthermore, levels of plasma ceramides along with certain amino acids could emerge as predictive biomarkers of response and severity of inflammation.
Collapse
Affiliation(s)
- Ioanna Dagla
- The Goulandris Natural History Museum, Bioanalytical Laboratory, GAIA Research Center, 145 62 Kifissia, Greece;
| | - Aikaterini Iliou
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 157 71 Athens, Greece; (A.I.); (D.B.)
| | - Dimitra Benaki
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 157 71 Athens, Greece; (A.I.); (D.B.)
| | - Evagelos Gikas
- Laboratory of Analytical Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 157 71 Athens, Greece;
| | - Emmanuel Mikros
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 157 71 Athens, Greece; (A.I.); (D.B.)
| | - Tina Bagratuni
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 115 28 Athens, Greece; (T.B.); (E.K.); (M.A.D.); (E.T.)
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 115 28 Athens, Greece; (T.B.); (E.K.); (M.A.D.); (E.T.)
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 115 28 Athens, Greece; (T.B.); (E.K.); (M.A.D.); (E.T.)
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 115 28 Athens, Greece; (T.B.); (E.K.); (M.A.D.); (E.T.)
| | - Anthony Tsarbopoulos
- The Goulandris Natural History Museum, Bioanalytical Laboratory, GAIA Research Center, 145 62 Kifissia, Greece;
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 115 27 Athens, Greece
| |
Collapse
|
28
|
Zhang Y, Zhang S. Prognostic value of glucose-to-lymphocyte ratio in critically ill patients with acute respiratory distress syndrome: A retrospective cohort study. J Clin Lab Anal 2022; 36:e24397. [PMID: 35358348 PMCID: PMC9102764 DOI: 10.1002/jcla.24397] [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: 02/14/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 11/11/2022] Open
Abstract
Background There is need to identify biomarkers for prognosis of acute respiratory distress syndrome (ADRS). This may allow early and accurate identification of patients with high‐risk ARDS to guide adjustment of clinical treatment and nursing intervention, which would ultimately improve prognosis of patients with ARDS. Biomarkers based on a combination of fasting glucose and lymphocyte counts to predict prognosis in critically ill patients with ARDS remain undefined. In this study, we investigated the association between glucose‐to‐lymphocyte ratio (GLR) and in‐hospital mortality. Methods The study obtained data from Medical Information Mart for Intensive Care‐IV (MIMIC‐IV Version 1.0) database. We defined the GLR as fasting glucose/lymphocyte count and the patient in‐hospital mortality was considered as the outcome. In addition, we employed linear and logistic regression models for analysis. Results In total, 1,085 patients with ARDS were included in this study. The eligible participants included 498 female and 587 males, with a mean age of 64.2 ± 17.5 years. Logistic regression analysis demonstrated that higher GLR was an independent risk factor for all‐cause mortality (OR =1.67, 95% CI: 1.26–2.22) after adjusting for age, sex, anion gap, white blood cell count, congestive heart failure, sequential organ failure assessment (SOFA), SBP, DBP, and respiratory rate in both the dichotomized group and subgroups. We also analyzed the in‐hospital mortality to ROC curves by comparing the value between SOFA + GLR and SOFA. The area under the curve (AUC) was 0.6991 for the SOFA + GLR (95% CI: 0.6634–0.7348), and 0.6613 for the SOFA (95% CI: 0.6238–0.6988). Conclusion Our data showed that the GLR was an independent predictor of in‐hospital mortality for patients with ARDS. The GLR is an integrated, readily available clinical biomarker for mortality in patients with ARDS.
Collapse
Affiliation(s)
- Yi Zhang
- Emergency department, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shuo Zhang
- Emergency department, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
29
|
Arancibia-Hernández YL, Aranda-Rivera AK, Cruz-Gregorio A, Pedraza-Chaverri J. Antioxidant/anti-inflammatory effect of Mg 2+ in coronavirus disease 2019 (COVID-19). Rev Med Virol 2022; 32:e2348. [PMID: 35357063 PMCID: PMC9111052 DOI: 10.1002/rmv.2348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 12/26/2022]
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), characterised by high levels of inflammation and oxidative stress (OS). Oxidative stress induces oxidative damage to lipids, proteins, and DNA, causing tissue damage. Both inflammation and OS contribute to multi-organ failure in severe cases. Magnesium (Mg2+ ) regulates many processes, including antioxidant and anti-inflammatory responses, as well as the proper functioning of other micronutrients such as vitamin D. In addition, Mg2+ participates as a second signalling messenger in the activation of T cells. Therefore, Mg2+ deficiency can cause immunodeficiency, exaggerated acute inflammatory response, decreased antioxidant response, and OS. Supplementation with Mg2+ has an anti-inflammatory response by reducing the levels of nuclear factor kappa B (NF-κB), interleukin (IL) -6, and tumor necrosis factor alpha. Furthermore, Mg2+ supplementation improves mitochondrial function and increases the antioxidant glutathione (GSH) content, reducing OS. Therefore, Mg2+ supplementation is a potential way to reduce inflammation and OS, strengthening the immune system to manage COVID-19. This narrative review will address Mg2+ deficiency associated with a worse disease prognosis, Mg2+ supplementation as a potent antioxidant and anti-inflammatory therapy during and after COVID-19 disease, and suggest that randomised controlled trials are indicated.
Collapse
Affiliation(s)
| | - Ana Karina Aranda-Rivera
- Facultad de Química, Departamento de Biología, Laboratorio F-315, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alfredo Cruz-Gregorio
- Facultad de Química, Departamento de Biología, Laboratorio F-315, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Pedraza-Chaverri
- Facultad de Química, Departamento de Biología, Laboratorio F-315, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
30
|
Theoharides TC, Guerra L, Patel K. Successful Treatment of a Patient With Severe COVID-19 Using an Integrated Approach Addressing Mast Cells and Their Mediators. Int J Infect Dis 2022; 118:164-166. [PMID: 35227867 PMCID: PMC8881225 DOI: 10.1016/j.ijid.2022.02.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA; School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA; Institute of Neuro-Immune Medicine, Nova Southeastern University, Tampa FL, USA.
| | - Lucy Guerra
- Department of Medicine, University of South Florida Morsani College of Medicine, Tampa General Hospital, Tampa, FL, USA
| | - Kapilkumar Patel
- Center for Advanced Lung Disease and Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, University of South Florida, Morsani College of Medicine, Tampa General Hospital, Center, Tampa, FL, USA
| |
Collapse
|
31
|
Ibrahim MA, Abdelmonaem AA, Abdel-Gaber SA, Hafez HM, Abdel Hafez SMN, Yehia Abdelzaher W. Rupatadine ameliorated ulcerative colitis in rats via modulation of platelet-activatiweng factor/interleukin-6/vascular endothelial growth factor signalling pathway. J Pharm Pharmacol 2022; 74:537-546. [PMID: 35134225 DOI: 10.1093/jpp/rgab170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVES This study aimed to analyse the potential effect of rupatadine (RUP) on ulcerative colitis (UC) induced by acetic acid (AA). METHODS Forty male adult Wistar rats were divided into five groups: Control group: received vehicles for 14 days; AA model group: received AA at the 13th day; Sulfasalazine (SLZ) + AA group: received SLZ (250 mg/kg) for 14 days and AA at the 13th day; RUP-3 + AA group: received RUP (3 mg/kg/day) for 14 days and AA at the 13th day; and RUP-6 + AA group: received RUP (6 mg/kg/day) for 14 days and AA at the 13th day. Evidence of UC was assessed both macroscopically and microscopically. Oxidative stress markers (total antioxidant capacity and malondialdehyde), antioxidant enzyme (superoxide dismutase), histamine and platelet-activating factor (PAF) were determined. Immunohistochemical estimations of vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6) were done. KEY FINDINGS The AA group showed evidence of UC that was associated with a significant increase in oxidative stress, histamine and PAF levels with significant elevation in colonic VEGF and IL-6 immuno-expressions. RUP, in a dose-dependent manner, significantly ameliorated UC. CONCLUSION RUP protects against UC by reducing oxidative stress and by regulating the PAF/IL-6/VEGF pathway.
Collapse
Affiliation(s)
- Mohamed A Ibrahim
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | | | - Seham A Abdel-Gaber
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Heba M Hafez
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | | | | |
Collapse
|
32
|
Mohamed MZ, Mohammed HH, Khalaf HM. Therapeutic effect of rupatadine against l-arginine-induced acute pancreatitis in rats: role of inflammation. Can J Physiol Pharmacol 2022; 100:176-183. [PMID: 35050802 DOI: 10.1139/cjpp-2021-0330] [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: 11/22/2022]
Abstract
Acute pancreatitis (AP) is an abrupt inflammatory disorder causing high morbidity and mortality. As AP is an insidious medical emergency, a curative modality is required instead of a preventive measure. Thus, we investigated the possible curative effect of rupatadine on a rat model of AP. Rupatadine is a potent histamine receptor 1 (H1R) and platelet-activating factor (PAF) blocker. We used four groups of six Wistar rats as follows: the control group received vehicle; the rupatadine control group received rupatadine as 6 mg/kg orally; the AP group received l-arginine intraperitoneally, and the treatment group received rupatadine at 1, 6, and 24 h after l-arginine injection. The levels of serum amylase, pancreatic oxidative parameters, and pancreatic cytokines were measured. PAF, histamine, and myeloperoxidase levels were determined in the pancreas. Histopathological and immunohistochemical examinations were performed to determine nuclear factor kappa-B (NF-κB) and caspase 3 expressions. Oxidative damage and severe inflammation were detected in the pancreas of the AP group. Rupatadine reduced the oxidative damage and the levels of proinflammatory cytokines, PAF, histamine, myeloperoxidase, NF-κB, and caspase 3 expressions. It restored the pancreatic acini to almost normal condition. Rupatadine induced important anti-inflammatory and antiapoptotic effects against l-arginine-induced AP.
Collapse
Affiliation(s)
- Mervat Z Mohamed
- Department of Pharmacology, Faculty of Medicine, Minia University, 61511 Minia, Egypt
| | - Hanaa H Mohammed
- Department of Histology, Faculty of Medicine, Minia University, 61511 Minia, Egypt
| | - Hanaa M Khalaf
- Department of Pharmacology, Faculty of Medicine, Minia University, 61511 Minia, Egypt
| |
Collapse
|
33
|
Theoharides TC. Could SARS-CoV-2 Spike Protein Be Responsible for Long-COVID Syndrome? Mol Neurobiol 2022; 59:1850-1861. [PMID: 35028901 PMCID: PMC8757925 DOI: 10.1007/s12035-021-02696-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2 infects cells via its spike protein binding to its surface receptor on target cells and results in acute symptoms involving especially the lungs known as COVID-19. However, increasing evidence indicates that many patients develop a chronic condition characterized by fatigue and neuropsychiatric symptoms, termed long-COVID. Most of the vaccines produced so far for COVID-19 direct mammalian cells via either mRNA or an adenovirus vector to express the spike protein, or administer recombinant spike protein, which is recognized by the immune system leading to the production of neutralizing antibodies. Recent publications provide new findings that may help decipher the pathogenesis of long-COVID. One paper reported perivascular inflammation in brains of deceased patients with COVID-19, while others showed that the spike protein could damage the endothelium in an animal model, that it could disrupt an in vitro model of the blood-brain barrier (BBB), and that it can cross the BBB resulting in perivascular inflammation. Moreover, the spike protein appears to share antigenic epitopes with human molecular chaperons resulting in autoimmunity and can activate toll-like receptors (TLRs), leading to release of inflammatory cytokines. Moreover, some antibodies produced against the spike protein may not be neutralizing, but may change its conformation rendering it more likely to bind to its receptor. As a result, one wonders whether the spike protein entering the brain or being expressed by brain cells could activate microglia, alone or together with inflammatory cytokines, since protective antibodies could not cross the BBB, leading to neuro-inflammation and contributing to long-COVID. Hence, there is urgent need to better understand the neurotoxic effects of the spike protein and to consider possible interventions to mitigate spike protein-related detrimental effects to the brain, possibly via use of small natural molecules, especially the flavonoids luteolin and quercetin.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Suite 304, Boston, MA, 02111, USA.
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA.
- Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, 02111, USA.
- Institute of Neuro-Immune Medicine, Nova Southeastern University, Clearwater, FL, 33759, USA.
| |
Collapse
|
34
|
Megiorni F, Pontecorvi P, Gerini G, Anastasiadou E, Marchese C, Ceccarelli S. Sex-Related Factors in Cardiovascular Complications Associated to COVID-19. Biomolecules 2021; 12:biom12010021. [PMID: 35053169 PMCID: PMC8773922 DOI: 10.3390/biom12010021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), the pandemic infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents with an extremely heterogeneous spectrum of symptoms and signs. The clinical manifestations seem to be correlated with disease severity. COVID-19 susceptibility and mortality show a significant sex imbalance, with men being more prone to infection and showing a higher rate of hospitalization and mortality compared to women. Such variability can be ascribed to both sex-related biological factors and gender-related behavioral cues. This review will discuss the potential mechanisms accounting for sex/gender influence in vulnerability to COVID-19. Cardiovascular diseases play a central role in determining COVID-19 outcome, whether they are pre-existent or arose upon infection. We will pay particular attention to the impact of sex and gender on cardiovascular manifestations related to COVID-19. Finally, we will discuss the sex-dependent variability in some biomarkers for the evaluation of COVID-19 infection and prognosis. The aim of this work is to highlight the significance of gendered medicine in setting up personalized programs for COVID-19 prevention, clinical evaluation and treatment.
Collapse
|
35
|
Lam HY, Tergaonkar V, Kumar AP, Ahn KS. Mast cells: Therapeutic targets for COVID-19 and beyond. IUBMB Life 2021; 73:1278-1292. [PMID: 34467628 PMCID: PMC8652840 DOI: 10.1002/iub.2552] [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: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 01/22/2023]
Abstract
Mast cells (MCs) are innate immune cells that widely distribute throughout all tissues and express a variety of cell surface receptors. Upon activation, MCs can rapidly release a diverse array of preformed mediators residing within their secretory granules and newly synthesize a broad spectrum of inflammatory and immunomodulatory mediators. These unique features of MCs enable them to act as sentinels in response to rapid changes within their microenvironment. There is increasing evidence now that MCs play prominent roles in other pathophysiological processes besides allergic inflammation. In this review, we highlight the recent findings on the emerging roles of MCs in the pathogenesis of coronavirus disease-2019 (COVID-19) and discuss the potential of MCs as novel therapeutic targets for COVID-19 and other non-allergic inflammatory diseases.
Collapse
Affiliation(s)
- Hiu Yan Lam
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
- Laboratory of NF‐κB SignalingInstitute of Molecular and Cell Biology (IMCB)SingaporeSingapore
- Department of Biochemistry, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Vinay Tergaonkar
- Laboratory of NF‐κB SignalingInstitute of Molecular and Cell Biology (IMCB)SingaporeSingapore
- Department of Biochemistry, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Pathology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- National University Cancer InstituteNational University Health SystemSingaporeSingapore
| | - Kwang Seok Ahn
- Department of Science in Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
| |
Collapse
|
36
|
Trapani V, Rosanoff A, Baniasadi S, Barbagallo M, Castiglioni S, Guerrero-Romero F, Iotti S, Mazur A, Micke O, Pourdowlat G, Scarpati G, Wolf FI, Maier JA. The relevance of magnesium homeostasis in COVID-19. Eur J Nutr 2021; 61:625-636. [PMID: 34687321 PMCID: PMC8540865 DOI: 10.1007/s00394-021-02704-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
Purpose In less than one and a half year, the COVID-19 pandemic has nearly brought to a collapse our health care and economic systems. The scientific research community has concentrated all possible efforts to understand the pathogenesis of this complex disease, and several groups have recently emphasized recommendations for nutritional support in COVID-19 patients. In this scoping review, we aim at encouraging a deeper appreciation of magnesium in clinical nutrition, in view of the vital role of magnesium and the numerous links between the pathophysiology of SARS-CoV-2 infection and magnesium-dependent functions. Methods By searching PubMed and Google Scholar from 1990 to date, we review existing evidence from experimental and clinical studies on the role of magnesium in chronic non-communicable diseases and infectious diseases, and we focus on recent reports of alterations of magnesium homeostasis in COVID-19 patients and their association with disease outcomes. Importantly, we conduct a census on ongoing clinical trials specifically dedicated to disclosing the role of magnesium in COVID-19. Results Despite many methodological limitations, existing data seem to corroborate an association between deranged magnesium homeostasis and COVID-19, and call for further and better studies to explore the prophylactic or therapeutic potential of magnesium supplementation. Conclusion We propose to reconsider the relevance of magnesium, frequently overlooked in clinical practice. Therefore, magnesemia should be monitored and, in case of imbalanced magnesium homeostasis, an appropriate nutritional regimen or supplementation might contribute to protect against SARS-CoV-2 infection, reduce severity of COVID-19 symptoms and facilitate the recovery after the acute phase.
Collapse
Affiliation(s)
- Valentina Trapani
- Sezione di Patologia Generale, Dipartimento di Medicina e Chirurgia Traslazionale, Fondazione Policlinico Universitario A. Gemelli IRCCS-Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy.,Alleanza Contro Il Cancro, Rome, Italy
| | - Andrea Rosanoff
- CMER Center for Magnesium Education and Research, Pahoa, Hawaii, USA
| | - Shadi Baniasadi
- Tracheal Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mario Barbagallo
- Geriatric Unit, Department of Medicine, University of Palermo, Palermo, Italy
| | - Sara Castiglioni
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Via G.B. Grassi 74, 20157, Milan, Italy
| | | | - Stefano Iotti
- Department of Pharmacy and Biotechnology (FaBit) National Institute of Biostructures and Biosystems, Università di Bologna, Bologna, Italy
| | - André Mazur
- Unité de Nutrition Humaine, INRAE, UNH, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Oliver Micke
- Department of Radiotherapy and Radiation Oncology, Franziskus Hospital, Bielefeld, Germany
| | - Guitti Pourdowlat
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Giuliana Scarpati
- Anestesiologia e Rianimazione, Dipartimento di Medicina e Chirurgia, Università Degli Studi di Salerno, Fisciano, Italy
| | - Federica I Wolf
- Sezione di Patologia Generale, Dipartimento di Medicina e Chirurgia Traslazionale, Fondazione Policlinico Universitario A. Gemelli IRCCS-Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy.
| | - Jeanette A Maier
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Via G.B. Grassi 74, 20157, Milan, Italy.
| |
Collapse
|
37
|
D’Amico R, Monaco F, Siracusa R, Cordaro M, Fusco R, Peritore AF, Gugliandolo E, Crupi R, Cuzzocrea S, Di Paola R, Impellizzeri D, Genovese T. Ultramicronized Palmitoylethanolamide in the Management of Sepsis-Induced Coagulopathy and Disseminated Intravascular Coagulation. Int J Mol Sci 2021; 22:ijms222111388. [PMID: 34768820 PMCID: PMC8583705 DOI: 10.3390/ijms222111388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Disseminated intravascular coagulation (DIC) is a severe condition characterized by the systemic formation of microthrombi complicated with bleeding tendency and organ dysfunction. In the last years, it represents one of the most frequent consequences of coronavirus disease 2019 (COVID-19). The pathogenesis of DIC is complex, with cross-talk between the coagulant and inflammatory pathways. The objective of this study is to investigate the anti-inflammatory action of ultramicronized palmitoylethanolamide (um-PEA) in a lipopolysaccharide (LPS)-induced DIC model in rats. Experimental DIC was induced by continual infusion of LPS (30 mg/kg) for 4 h through the tail vein. Um-PEA (30 mg/kg) was given orally 30 min before and 1 h after the start of intravenous infusion of LPS. Results showed that um-PEA reduced alteration of coagulation markers, as well as proinflammatory cytokine release in plasma and lung samples, induced by LPS infusion. Furthermore, um-PEA also has the effect of preventing the formation of fibrin deposition and lung damage. Moreover, um-PEA was able to reduce the number of mast cells (MCs) and the release of its serine proteases, which are also necessary for SARS-CoV-2 infection. These results suggest that um-PEA could be considered as a potential therapeutic approach in the management of DIC and in clinical implications associated to coagulopathy and lung dysfunction, such as COVID-19.
Collapse
Affiliation(s)
- Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
| | - Francesco Monaco
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy; (F.M.); (M.C.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy; (F.M.); (M.C.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
| | - Enrico Gugliandolo
- Department of Veterinary Science, University of Messina, 98166 Messina, Italy; (E.G.); (R.C.)
| | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, 98166 Messina, Italy; (E.G.); (R.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
- Correspondence: (S.C.); (R.D.P.); Tel.: +39-090-676-5208 (S.C. & R.D.P.)
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
- Correspondence: (S.C.); (R.D.P.); Tel.: +39-090-676-5208 (S.C. & R.D.P.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy; (R.D.); (R.S.); (R.F.); (A.F.P.); (D.I.); (T.G.)
| |
Collapse
|
38
|
Atila A, Alay H, Yaman ME, Akman TC, Cadirci E, Bayrak B, Celik S, Atila NE, Yaganoglu AM, Kadioglu Y, Halıcı Z, Parlak E, Bayraktutan Z. The serum amino acid profile in COVID-19. Amino Acids 2021; 53:1569-1588. [PMID: 34605988 PMCID: PMC8487804 DOI: 10.1007/s00726-021-03081-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
The pandemic of the coronavirus disease (COVID-19) caused by SARS-CoV-2 affects millions of people worldwide. There are still many unknown aspects to this infection which affects the whole world. In addition, the potential impacts caused by this infection are still unclear. Amino acid metabolism, in particular, contains significant clues in terms of the development and prevention of many diseases. Therefore, this study aimed to compare amino acid profile of COVID-19 and healthy subject. In this study, the amino acid profiles of patients with asymptomatic, mild, moderate, and severe/critical SARS-CoV-2 infection were scanned with LC–MS/MS. The amino acid profile encompassing 30 amino acids in 142 people including 30 control and 112 COVID-19 patients was examined. 20 amino acids showed significant differences when compared to the control group in COVID-19 patient groups with different levels of severity in the statistical analyses conducted. It was detected that the branched-chain amino acids (BCAAs) changed in correlation with one another, and l-2-aminobutyric acid and l-phenylalanine had biomarker potential for COVID-19. Moreover, it was concluded that l-2-aminobutyric acid could provide prognostic information about the course of the disease. We believe that a new viewpoint will develop regarding the diagnosis, treatment, and prognosis as a result of the evaluation of the serum amino acid profiles of COVID-19 patients. Determining l-phenylalanine and l-2-aminobutyric levels can be used in laboratories as a COVID-19-biomarker. Also, supplementing COVID patients with taurine and BCAAs can be beneficial for treatment protocols.
Collapse
Affiliation(s)
- Alptug Atila
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Handan Alay
- Department of Infectious Diseases and Clinical Microbiology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Mehmet Emrah Yaman
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Tugrul Cagri Akman
- Department of Analytical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, 25240 Erzurum, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Burak Bayrak
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Saffet Celik
- Technology Research and Development Application and Research Center, Trakya University, 22030 Edirne, Turkey
| | - Nihal Efe Atila
- Department of Otorhinolaryngology, Erzurum Regional Training and Research Hospital, 25240 Erzurum, Turkey
| | - Aycan Mutlu Yaganoglu
- Department of Animal Science, College of Agriculture, Ataturk University, 25240 Erzurum, Turkey
| | - Yucel Kadioglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Zekai Halıcı
- Department of Pharmacology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Emine Parlak
- Department of Infectious Diseases and Clinical Microbiology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Zafer Bayraktutan
- Department of Biochemistry, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| |
Collapse
|
39
|
Shirvaliloo M. The blood-gas barrier in COVID-19: an overview of the effects of SARS-CoV-2 infection on the alveolar epithelial and endothelial cells of the lung. Tissue Barriers 2021; 9:1937013. [PMID: 34232823 PMCID: PMC8794501 DOI: 10.1080/21688370.2021.1937013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 02/08/2023] Open
Abstract
Blood-gas barrier (BGB) or alveolar-capillary barrier is the primary tissue barrier affected by coronavirus disease 2019 (COVID-19). Comprising alveolar epithelial cells (AECs), endothelial cells (ECs) and the extracellular matrix (ECM) in between, the BGB is damaged following the action of multiple pro-inflammatory cytokines during acute inflammation. The infection of AECs and ECs with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen behind COVID-19, triggers an inflammatory response at the BGB, inducing the release of interleukin 1 (IL-1), IL-6, tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-β), high mobility group box 1 (HMGB1), matrix metalloproteinases (MMPs), intercellular adhesion molecule-1 (ICAM-1) and platelet activating factor (PAF). The end result is the disassembly of adherens junctions (AJs) and tight junctions (TJs) in both AECs and ECs, AEC hyperplasia, EC pyroptosis, ECM remodeling and deposition of fibrin clots in the alveolar capillaries, leading to disintegration and thickening of the BGB, and ultimately, hypoxia. This commentary seeks to provide a brief account of how the BGB might become affected in COVID-19.
Collapse
Affiliation(s)
- Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
Theoharides TC. Ways to Address Perinatal Mast Cell Activation and Focal Brain Inflammation, including Response to SARS-CoV-2, in Autism Spectrum Disorder. J Pers Med 2021; 11:860. [PMID: 34575637 PMCID: PMC8465360 DOI: 10.3390/jpm11090860] [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] [Received: 06/27/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/08/2023] Open
Abstract
The prevalence of autism spectrum disorder (ASD) continues to increase, but no distinct pathogenesis or effective treatment are known yet. The presence of many comorbidities further complicates matters, making a personalized approach necessary. An increasing number of reports indicate that inflammation of the brain leads to neurodegenerative changes, especially during perinatal life, "short-circuiting the electrical system" in the amygdala that is essential for our ability to feel emotions, but also regulates fear. Inflammation of the brain can result from the stimulation of mast cells-found in all tissues including the brain-by neuropeptides, stress, toxins, and viruses such as SARS-CoV-2, leading to the activation of microglia. These resident brain defenders then release even more inflammatory molecules and stop "pruning" nerve connections, disrupting neuronal connectivity, lowering the fear threshold, and derailing the expression of emotions, as seen in ASD. Many epidemiological studies have reported a strong association between ASD and atopic dermatitis (eczema), asthma, and food allergies/intolerance, all of which involve activated mast cells. Mast cells can be triggered by allergens, neuropeptides, stress, and toxins, leading to disruption of the blood-brain barrier (BBB) and activation of microglia. Moreover, many epidemiological studies have reported a strong association between stress and atopic dermatitis (eczema) during gestation, which involves activated mast cells. Both mast cells and microglia can also be activated by SARS-CoV-2 in affected mothers during pregnancy. We showed increased expression of the proinflammatory cytokine IL-18 and its receptor, but decreased expression of the anti-inflammatory cytokine IL-38 and its receptor IL-36R, only in the amygdala of deceased children with ASD. We further showed that the natural flavonoid luteolin is a potent inhibitor of the activation of both mast cells and microglia, but also blocks SARS-CoV-2 binding to its receptor angiotensin-converting enzyme 2 (ACE2). A treatment approach should be tailored to each individual patient and should address hyperactivity/stress, allergies, or food intolerance, with the introduction of natural molecules or drugs to inhibit mast cells and microglia, such as liposomal luteolin.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Suite 304, Boston, MA 02111, USA
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA 02111, USA
- Department of Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, MA 02111, USA
| |
Collapse
|
41
|
Chen F, Shi Q, Pei F, Vogt A, Porritt RA, Garcia G, Gomez AC, Cheng MH, Schurdak ME, Liu B, Chan SY, Arumugaswami V, Stern AM, Taylor DL, Arditi M, Bahar I. A systems-level study reveals host-targeted repurposable drugs against SARS-CoV-2 infection. Mol Syst Biol 2021; 17:e10239. [PMID: 34339582 PMCID: PMC8328275 DOI: 10.15252/msb.202110239] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanism of SARS-CoV-2 infection and identifying potential therapeutics are global imperatives. Using a quantitative systems pharmacology approach, we identified a set of repurposable and investigational drugs as potential therapeutics against COVID-19. These were deduced from the gene expression signature of SARS-CoV-2-infected A549 cells screened against Connectivity Map and prioritized by network proximity analysis with respect to disease modules in the viral-host interactome. We also identified immuno-modulating compounds aiming at suppressing hyperinflammatory responses in severe COVID-19 patients, based on the transcriptome of ACE2-overexpressing A549 cells. Experiments with Vero-E6 cells infected by SARS-CoV-2, as well as independent syncytia formation assays for probing ACE2/SARS-CoV-2 spike protein-mediated cell fusion using HEK293T and Calu-3 cells, showed that several predicted compounds had inhibitory activities. Among them, salmeterol, rottlerin, and mTOR inhibitors exhibited antiviral activities in Vero-E6 cells; imipramine, linsitinib, hexylresorcinol, ezetimibe, and brompheniramine impaired viral entry. These novel findings provide new paths for broadening the repertoire of compounds pursued as therapeutics against COVID-19.
Collapse
Affiliation(s)
- Fangyuan Chen
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- School of MedicineTsinghua UniversityBeijingChina
| | - Qingya Shi
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- School of MedicineTsinghua UniversityBeijingChina
| | - Fen Pei
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Andreas Vogt
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Rebecca A Porritt
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
- Biomedical Sciences, Infectious and Immunologic Diseases Research CenterCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Gustavo Garcia
- Department of Molecular and Medical PharmacologyDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of CaliforniaLos AngelesCAUSA
| | - Angela C Gomez
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Mary Hongying Cheng
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Mark E Schurdak
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Bing Liu
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Stephen Y Chan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine InstituteUniversity of Pittsburgh Medical CenterPittsburghPAUSA
- Division of CardiologyDepartment of MedicineUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical PharmacologyDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of CaliforniaLos AngelesCAUSA
| | - Andrew M Stern
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - D Lansing Taylor
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Moshe Arditi
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
- Biomedical Sciences, Infectious and Immunologic Diseases Research CenterCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Ivet Bahar
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| |
Collapse
|
42
|
Sibilio P, Bini S, Fiscon G, Sponziello M, Conte F, Pecce V, Durante C, Paci P, Falcone R, Norata GD, Farina L, Verrienti A. In silico drug repurposing in COVID-19: A network-based analysis. Biomed Pharmacother 2021; 142:111954. [PMID: 34358753 PMCID: PMC8316014 DOI: 10.1016/j.biopha.2021.111954] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022] Open
Abstract
The SARS-CoV-2 pandemic is a worldwide public health emergency. Despite the beginning of a vaccination campaign, the search for new drugs to appropriately treat COVID-19 patients remains a priority. Drug repurposing represents a faster and cheaper method than de novo drug discovery. In this study, we examined three different network-based approaches to identify potentially repurposable drugs to treat COVID-19. We analyzed transcriptomic data from whole blood cells of patients with COVID-19 and 21 other related conditions, as compared with those of healthy subjects. In addition to conventionally used drugs (e.g., anticoagulants, antihistaminics, anti-TNFα antibodies, corticosteroids), unconventional candidate compounds, such as SCN5A inhibitors and drugs active in the central nervous system, were identified. Clinical judgment and validation through clinical trials are always mandatory before use of the identified drugs in a clinical setting.
Collapse
Affiliation(s)
- Pasquale Sibilio
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Simone Bini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giulia Fiscon
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy; Fondazione per la Medicina Personalizzata, Via Goffredo Mameli, 3/1, Genova, Italy
| | - Marialuisa Sponziello
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Conte
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Valeria Pecce
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Cosimo Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Paola Paci
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy; Department of Computer, Control, and Management Engineering, Sapienza University of Rome, Rome, Italy.
| | - Rosa Falcone
- Phase 1 Unit-Clinical Trial Center Gemelli University Hospital, Rome, Italy
| | - Giuseppe Danilo Norata
- Department of Excellence in Pharmacological and Biomolecular Sciences, University of Milan and Center for the Study of Atherosclerosis, SISA Bassini Hospital, Milan, Italy
| | - Lorenzo Farina
- Department of Computer, Control, and Management Engineering, Sapienza University of Rome, Rome, Italy
| | - Antonella Verrienti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
43
|
Abstract
Since its emergence in Wuhan, China, covid-19 has spread and had a profound effect on the lives and health of people around the globe. As of 4 July 2021, more than 183 million confirmed cases of covid-19 had been recorded worldwide, and 3.97 million deaths. Recent evidence has shown that a range of persistent symptoms can remain long after the acute SARS-CoV-2 infection, and this condition is now coined long covid by recognized research institutes. Studies have shown that long covid can affect the whole spectrum of people with covid-19, from those with very mild acute disease to the most severe forms. Like acute covid-19, long covid can involve multiple organs and can affect many systems including, but not limited to, the respiratory, cardiovascular, neurological, gastrointestinal, and musculoskeletal systems. The symptoms of long covid include fatigue, dyspnea, cardiac abnormalities, cognitive impairment, sleep disturbances, symptoms of post-traumatic stress disorder, muscle pain, concentration problems, and headache. This review summarizes studies of the long term effects of covid-19 in hospitalized and non-hospitalized patients and describes the persistent symptoms they endure. Risk factors for acute covid-19 and long covid and possible therapeutic options are also discussed.
Collapse
Affiliation(s)
- Harry Crook
- Faculty of Medicine, Imperial College London, London, UK
| | - Sanara Raza
- Faculty of Medicine, Imperial College London, London, UK
| | - Joseph Nowell
- Faculty of Medicine, Imperial College London, London, UK
| | - Megan Young
- Faculty of Medicine, Imperial College London, London, UK
| | - Paul Edison
- Faculty of Medicine, Imperial College London, London, UK
- Cardiff University, Cardiff, UK
| |
Collapse
|
44
|
Han NR, Ko SG, Moon PD, Park HJ. Chloroquine attenuates thymic stromal lymphopoietin production via suppressing caspase-1 signaling in mast cells. Biomed Pharmacother 2021; 141:111835. [PMID: 34146852 DOI: 10.1016/j.biopha.2021.111835] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/21/2021] [Accepted: 06/14/2021] [Indexed: 01/14/2023] Open
Abstract
Thymic stromal lymphopoietin (TSLP) produced by mast cells is involved in allergic inflammation pathogenesis. Chloroquine (CQ) is known to be an anti-malarial drug; however, additional protective functions of CQ have been discovered. This study aims to clarify an anti-inflammatory effect of CQ through modulating TSLP levels using an in vitro model of phorbol myristate acetate (PMA) + A23187-activated human mast cell line (HMC-1) and an in vivo model of PMA-irritated ear edema. CQ treatment reduced the production and mRNA expression levels of TSLP in activated HMC-1 cells. CQ down-regulated caspase-1 (CASP1), MAPKs, and NF-κB levels enhanced by stimulation with PMA + A23187. Moreover, ear thickness in ear edema was suppressed following CQ treatment. CQ decreased CASP1 and NF-κB levels in the ear tissue. TSLP levels in the ear tissue and serum were reduced following CQ treatment. Collectively, the above findings elucidate that CQ inhibits the pro-inflammatory mechanisms of TSLP via the down-regulation of distinct intracellular signaling cascade in mast cells. Therefore, CQ may have protective roles against TSLP-mediated inflammatory disorders.
Collapse
Affiliation(s)
- Na-Ra Han
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seong-Gyu Ko
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Phil-Dong Moon
- Center for Converging Humanities, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Hi-Joon Park
- Department of Anatomy & Information Sciences, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
45
|
Bose S, Dasbiswas K, Gopinath A. Matrix Stiffness Modulates Mechanical Interactions and Promotes Contact between Motile Cells. Biomedicines 2021; 9:biomedicines9040428. [PMID: 33920918 PMCID: PMC8077938 DOI: 10.3390/biomedicines9040428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanical micro-environment of cells and tissues influences key aspects of cell structure and function, including cell motility. For proper tissue development, cells need to migrate, interact, and form contacts. Cells are known to exert contractile forces on underlying soft substrates and sense deformations in them. Here, we propose and analyze a minimal biophysical model for cell migration and long-range cell–cell interactions through mutual mechanical deformations of the substrate. We compute key metrics of cell motile behavior, such as the number of cell-cell contacts over a given time, the dispersion of cell trajectories, and the probability of permanent cell contact, and analyze how these depend on a cell motility parameter and substrate stiffness. Our results elucidate how cells may sense each other mechanically and generate coordinated movements and provide an extensible framework to further address both mechanical and short-range biophysical interactions.
Collapse
Affiliation(s)
- Subhaya Bose
- Department of Physics, University of California Merced, Merced, CA 95343, USA; (S.B.); (K.D.)
| | - Kinjal Dasbiswas
- Department of Physics, University of California Merced, Merced, CA 95343, USA; (S.B.); (K.D.)
| | - Arvind Gopinath
- Department of Bioengineering, University of California Merced, Merced, CA 95343, USA
- Correspondence:
| |
Collapse
|
46
|
Theoharides TC, Cholevas C, Polyzoidis K, Politis A. Long-COVID syndrome-associated brain fog and chemofog: Luteolin to the rescue. Biofactors 2021; 47:232-241. [PMID: 33847020 PMCID: PMC8250989 DOI: 10.1002/biof.1726] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/01/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 leads to severe respiratory problems, but also to long-COVID syndrome associated primarily with cognitive dysfunction and fatigue. Long-COVID syndrome symptoms, especially brain fog, are similar to those experienced by patients undertaking or following chemotherapy for cancer (chemofog or chemobrain), as well in patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) or mast cell activation syndrome (MCAS). The pathogenesis of brain fog in these illnesses is presently unknown but may involve neuroinflammation via mast cells stimulated by pathogenic and stress stimuli to release mediators that activate microglia and lead to inflammation in the hypothalamus. These processes could be mitigated by phytosomal formulation (in olive pomace oil) of the natural flavonoid luteolin.
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of ImmunologyTufts University School of MedicineBostonMassachusettsUSA
- School of Graduate Biomedical SciencesTufts University School of MedicineBostonMassachusettsUSA
- Department of Internal MedicineTufts University School of Medicine and Tufts Medical CenterBostonMassachusettsUSA
- Department of PsychiatryTufts University School of Medicine and Tufts Medical CenterBostonMassachusettsUSA
- BrainGateThessalonikiGreece
| | | | | | - Antonios Politis
- First Department of PsychiatryEginition Hospital, National and Kapodistrian UniversityAthensGreece
| |
Collapse
|
47
|
Detopoulou P, Demopoulos CA, Antonopoulou S. Micronutrients, Phytochemicals and Mediterranean Diet: A Potential Protective Role against COVID-19 through Modulation of PAF Actions and Metabolism. Nutrients 2021; 13:nu13020462. [PMID: 33573169 PMCID: PMC7911163 DOI: 10.3390/nu13020462] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 01/08/2023] Open
Abstract
The new coronavirus disease 2019 (COVID-19) pandemic is an emerging situation with high rates of morbidity and mortality, in the pathophysiology of which inflammation and thrombosis are implicated. The disease is directly connected to the nutritional status of patients and a well-balanced diet is recommended by official sources. Recently, the role of platelet activating factor (PAF) was suggested in the pathogenesis of COVID-19. In the present review several micronutrients (vitamin A, vitamin C, vitamin E, vitamin D, selenium, omega-3 fatty acids, and minerals), phytochemicals and Mediterranean diet compounds with potential anti-COVID activity are presented. We further underline that the well-known anti-inflammatory and anti-thrombotic actions of the investigated nutrients and/or holistic dietary schemes, such as the Mediterranean diet, are also mediated through PAF. In conclusion, there is no single food to prevent coronavirus Although the relationship between PAF and COVID-19 is not robust, a healthy diet containing PAF inhibitors may target both inflammation and thrombosis and prevent the deleterious effects of COVID-19. The next step is the experimental confirmation or not of the PAF-COVID-19 hypothesis.
Collapse
Affiliation(s)
- Paraskevi Detopoulou
- Department of Clinical Nutrition, General Hospital Korgialenio Benakio, 11526 Athens, Greece;
| | - Constantinos A. Demopoulos
- Laboratory of Biochemistry, Faculty of Chemistry, National & Kapodistrian University of Athens, 16121 Athens, Greece;
| | - Smaragdi Antonopoulou
- Laboratory of Biology, Biochemistry and Microbiology, Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 70 El. Venizelou Street, 17671 Athens, Greece
- Correspondence: ; Tel.: +30-210-954-9230; Fax: +30-210-957-7050
| |
Collapse
|