Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Immunology ; 169(2): 117-131, 2023 06.
Article in English | MEDLINE | ID: mdl-36571562

ABSTRACT

Hyperinflammation characterized by elevated proinflammatory cytokines known as 'cytokine storms' is the major cause of high severity and mortality seen in COVID-19 patients. The pathology behind the cytokine storms is currently unknown. Increased HMGB1 levels in serum/plasma of COVID-19 patients were reported by many studies, which positively correlated with the level of proinflammatory cytokines. Dead cells following SARS-CoV-2 infection might release a large amount of HMGB1 and RNA of SARS-CoV-2 into extracellular space. HMGB1 is a well-known inflammatory mediator. Additionally, extracellular HMGB1 might interact with SARS-CoV-2 RNA because of its high capability to bind with a wide variety of molecules including nucleic acids and could trigger massive proinflammatory immune responses. This review aimed to critically explore the many possible pathways by which HMGB1-SARS-CoV-2 RNA complexes mediate proinflammatory responses in COVID-19. The contribution of these pathways to impair host immune responses against SARS-CoV-2 infection leading to a cytokine storm was also evaluated. Moreover, since blocking the HMGB1-SARS-CoV-2 RNA interaction might have therapeutic value, some of the HMGB1 antagonists have been reviewed. The HMGB1- SARS-CoV-2 RNA complexes might trigger endocytosis via RAGE which is linked to lysosomal rupture, PRRs activation, and pyroptotic death. High levels of the proinflammatory cytokines produced might suppress many immune cells leading to uncontrolled viral infection and cell damage with more HMGB1 released. Altogether these mechanisms might initiate a proinflammatory cycle leading to a cytokine storm. HMGB1 antagonists could be considered to give benefit in alleviating cytokine storms and serve as a potential candidate for COVID-19 therapy.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , COVID-19 , Cytokine Release Syndrome , HMGB1 Protein , Molecular Targeted Therapy , RNA, Viral , SARS-CoV-2 , Humans , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , COVID-19/complications , COVID-19/immunology , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/metabolism , RNA, Viral/metabolism , Host Microbial Interactions/immunology , SARS-CoV-2/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
2.
Cell Rep Med ; 2(10): 100422, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34755134

ABSTRACT

Provoked by sterile/nonsterile insults, prolonged monocyte mobilization and uncontrolled monocyte/macrophage activation can pose imminent or impending harm to the affected organs. Curiously, folate receptor beta (FRß), with subnanomolar affinity for the vitamin folic acid (FA), is upregulated during immune activation in hematopoietic cells of the myeloid lineage. This phenomenon has inspired a strong interest in exploring FRß-directed diagnostics/therapeutics. Previously, we have reported that FA-targeted aminopterin (AMT) therapy can modulate macrophage function and effectively treat animal models of inflammation. Our current investigation of a lead compound (EC2319) leads to discovery of a highly FR-specific mechanism of action independent of the root causes against inflammatory monocytes. We further show that EC2319 suppresses interleukin-6/interleukin-1ß release by FRß+ monocytes in a triple co-culture leukemic model of cytokine release syndrome with anti-CD19 chimeric antigen receptor T cells. Because of its chemical stability and metabolically activated linker, EC2319 demonstrates favorable pharmacokinetic characteristics and cross-species translatability to support future pre-clinical and clinical development.


Subject(s)
Aminopterin/pharmacology , Cytokine Release Syndrome/prevention & control , Folate Receptor 2/genetics , Folic Acid Antagonists/pharmacology , Folic Acid/metabolism , Macrophages/drug effects , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , CHO Cells , Cricetulus , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Female , Folate Receptor 1/antagonists & inhibitors , Folate Receptor 1/genetics , Folate Receptor 1/immunology , Folate Receptor 2/antagonists & inhibitors , Folate Receptor 2/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Models, Biological , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , RAW 264.7 Cells , Rats , Rats, Inbred Lew , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
3.
Front Immunol ; 12: 648250, 2021.
Article in English | MEDLINE | ID: mdl-34248936

ABSTRACT

BACKGROUND: The newly identified betacoronavirus SARS-CoV-2 is the causative pathogen of the coronavirus disease of 2019 (COVID-19) that killed more than 3.5 million people till now. The cytokine storm induced in severe COVID-19 patients causes hyper-inflammation, is the primary reason for respiratory and multi-organ failure and fatality. This work uses a rational computational strategy to identify the existing drug molecules to target host pathways to reduce the cytokine storm. RESULTS: We used a "host response signature network" consist of 36 genes induced by SARS-CoV-2 infection and associated with cytokine storm. In order to attenuate the cytokine storm, potential drug molecules were searched against "host response signature network". Our study identified that drug molecule andrographolide, naturally present in a medicinal plant Andrographis paniculata, has the potential to bind with crucial proteins to block the TNF-induced NFkB1 signaling pathway responsible for cytokine storm in COVID-19 patients. The molecular docking method showed the binding of andrographolide with TNF and covalent binding with NFkB1 proteins of the TNF signaling pathway. CONCLUSION: We used a rational computational approach to repurpose existing drugs targeting host immunomodulating pathways. Our study suggests that andrographolide could bind with TNF and NFkB1 proteins, block TNF-induced cytokine storm in COVID-19 patients, and warrant further experimental validation.


Subject(s)
Antiviral Agents/pharmacology , COVID-19/immunology , Cytokine Release Syndrome/immunology , Diterpenes/pharmacology , Drug Development/methods , SARS-CoV-2/physiology , Andrographis/immunology , Cytokine Release Syndrome/drug therapy , Humans , Molecular Docking Simulation , NF-kappa B p50 Subunit/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , COVID-19 Drug Treatment
4.
Rheumatology (Oxford) ; 60(9): 3986-4000, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34117886

ABSTRACT

Adult-onset Still's disease (AOSD) is a rare, but characteristic non-familial, multi-genic systemic auto-inflammatory disorder, characterized by high spiking fever, salmon-like evanescent skin rash, polyarthritis, sore throat, hyperferritinemia and leucocytosis. The hallmark of AOSD is a cytokine storm triggered by dysregulation of inflammation. Nowadays, with advances in anti-cytokine biologic agents, the treatment of AOSD is no longer limited to NSAIDs, glucocorticoids or conventional synthetic DMARDs. In this review, we focussed on the roles of these cytokines in the pathogenesis of AOSD and summarized the current and emerging biological therapy.


Subject(s)
Biological Therapy/methods , Still's Disease, Adult-Onset/therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/therapy , Humans , Still's Disease, Adult-Onset/immunology
5.
Front Biosci (Landmark Ed) ; 26(5): 51-75, 2021 04 30.
Article in English | MEDLINE | ID: mdl-34027650

ABSTRACT

In 2020, a novel strain of coronavirus (COVID-19) has led to a significant morbidity and mortality worldwide. As of the date of this writing, a total of 116 M cases has been diagnosed worldwide leading to 2.5 M deaths. The number of mortalities is directly correlated with the rise of innate immune cells (especially macrophages) in the lungs that secrete inflammatory cytokines (IL-1ß and IL-6) leading to the development of "Cytokine Storm Syndrome" (CSS), multi-organ-failure and death. Given that currently the treatment of this condition is rare and release of effective vaccine might be months away, here, we review the plants and their pharmacologically active-compounds as potential phytopharmaceuticals for the virus induced inflammatory response. Experimental validation of the effectiveness of these natural compounds to prevent or reduce the cytokine storm might be beneficial as an adjunct treatment of SARS-CoV-2.


Subject(s)
COVID-19 Drug Treatment , Cytokine Release Syndrome/prevention & control , Phytotherapy/methods , Plant Extracts/therapeutic use , Plants, Medicinal/chemistry , SARS-CoV-2/drug effects , COVID-19/immunology , COVID-19/virology , Cytokine Release Syndrome/immunology , Cytokines/immunology , Cytokines/metabolism , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Plants, Medicinal/classification , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Virulence/drug effects , Virulence/immunology
6.
Clin Transl Sci ; 14(6): 2111-2116, 2021 11.
Article in English | MEDLINE | ID: mdl-34057814

ABSTRACT

This review describes the evidence for the potential benefit of vitamin D supplementation in people with respiratory diseases who may have a higher susceptibility to coronavirus disease 2019 (COVID-19) infection and its consequences. Clinical evidence indicates that vitamin D may reduce the risk of both upper and lower respiratory tract infections and offers benefit particularly in people with vitamin D deficiency. Some evidence exists for a higher incidence of active tuberculosis (TB) in patients who are deficient in vitamin D. An association between low levels of 25(OH)D (the active form of vitamin D) and COVID-19 severity of illness and mortality has also been reported. In addition, low 25(OH)D levels are associated with poor outcomes in acute respiratory distress syndrome (ARDS). The cytokine storm experienced in severe COVID-19 infections results from excessive release of pro-inflammatory cytokines. Due to its immunomodulatory effects, adequate vitamin D levels may cause a decrease in the pro-inflammatory cytokines and an increase in the anti-inflammatory cytokines during COVID-19 infections. Vitamin D deficiency was found in 82.2% of hospitalized COVID-19 cases and 47.2% of population-based controls (p < 0.0001). The available evidence warrants an evaluation of vitamin D supplementation in susceptible populations with respiratory diseases, such as TB, and particularly in those who are deficient in vitamin D. This may mitigate against serious complications of COVID-19 infections or reduce the impact of ARDS in those who have been infected.


Subject(s)
COVID-19/immunology , Dietary Supplements , Tuberculosis/immunology , Vitamin D Deficiency/diet therapy , Vitamin D/administration & dosage , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/prevention & control , Comorbidity , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/prevention & control , Cytokine Release Syndrome/virology , Disease Susceptibility/blood , Disease Susceptibility/immunology , Humans , Pandemics , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/prevention & control , Risk Factors , Severity of Illness Index , Tuberculosis/blood , Tuberculosis/epidemiology , Vitamin D/blood , Vitamin D Deficiency/complications , Vitamin D Deficiency/epidemiology , Vitamin D Deficiency/immunology
7.
Undersea Hyperb Med ; 48(1): 1-12, 2021.
Article in English | MEDLINE | ID: mdl-33648028

ABSTRACT

The SARS-Cov-2 (COVID-19) pandemic remains a major worldwide public health issue. Initially, improved supportive and anti-inflammatory intervention, often employing known drugs or technologies, provided measurable improvement in management. We have recently seen advances in specific therapeutic interventions and in vaccines. Nevertheless, it will be months before most of the world's population can be vaccinated to achieve herd immunity. In the interim, hyperbaric oxygen (HBO2) treatment offers several potentially beneficial therapeutic effects. Three small published series, one with a propensity-score-matched control group, have demonstrated safety and initial efficacy. Additional anecdotal reports are consistent with these publications. HBO2 delivers oxygen in extreme conditions of hypoxemia and tissue hypoxia, even in the presence of lung pathology. It provides anti-inflammatory and anti-proinflammatory effects likely to ameliorate the overexuberant immune response common to COVID-19. Unlike steroids, it exerts these effects without immune suppression. One study suggests HBO2 may reduce the hypercoagulability seen in COVID patients. Also, hyperbaric oxygen offers a likely successful intervention to address the oxygen debt expected to arise from a prolonged period of hypoxemia and tissue hypoxia. To date, 11 studies designed to investigate the impact of HBO2 on patients infected with SARS-Cov-2 have been posted on clinicaltrials.gov. This paper describes the promising physiologic and biochemical effects of hyperbaric oxygen in COVID-19 and potentially in other disorders with similar pathologic mechanisms.


Subject(s)
COVID-19/therapy , Hyperbaric Oxygenation/methods , COVID-19/blood , COVID-19/complications , COVID-19/immunology , Cell Hypoxia , Cytokine Release Syndrome/immunology , Cytokines/blood , Humans , Hypoxia/therapy , Inflammation/therapy , Mesenchymal Stem Cells , Oxygen/poisoning , Oxygen Consumption , Thrombophilia/etiology , Thrombophilia/therapy
8.
Med Hypotheses ; 146: 110473, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33385879

ABSTRACT

Severe forms of the Coronavirus disease 2019 (COVID-19) are characterized by an enhanced inflammatory syndrome called "cytokine storm" that produces an aberrant release of high amounts of cytokines, chemokines, and other proinflammatory mediators. The pathogenetic role of the "cytokine storm" has been confirmed by the efficacy of immunosuppressive drugs such as corticosteroids along with antiviral drugs in the treatment of the severe forms of this disease. Phenylmethimazole (C10) is a derivative of methimazole with anti-inflammatory properties. Studies performed both in vitro and in vivo have shown that C10 is able to block the production of multiple cytokines, chemokines, and other proinflammatory molecules involved in the pathogenesis of inflammation. Particularly, C10 is effective in reducing the increased secretion of cytokines in animal models of endotoxic shock. We hypothesize that these effects are not limited to the endotoxic shock, but can also be applied to any disease characterized by the presence of a "cytokine storm". Therefore, C10 may be a potential drug to be used alternatively or in association with the corticosteroids or other immunosuppressive agents in the severe forms of COVID-19 as well as other viral diseases that induce a "cytokine storm". Preclinical and clinical studies have to be performed to confirm this hypothesis.


Subject(s)
COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , Methimazole/analogs & derivatives , Thiones/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , COVID-19/immunology , Cytokine Release Syndrome/immunology , Cytokines/antagonists & inhibitors , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Methimazole/pharmacology , Mice , Pandemics , SARS-CoV-2 , Shock, Septic/drug therapy , Shock, Septic/immunology , Translational Research, Biomedical
9.
Front Immunol ; 12: 794780, 2021.
Article in English | MEDLINE | ID: mdl-35095870

ABSTRACT

Various metabolites identified with therapeutic mushrooms have been found from different sources and are known to have antibacterial, antiviral, and anticancer properties. Over thousands soil growth-based mushroom metabolites have been discovered, and utilized worldwide to combat malignancy. In this study, psilocybin-mushroom that contains the psychedelic compounds such as psilacetin, psilocin, and psilocybine were screened and found to be inhibitors of SARS-CoV-2 Mprotease. It has been found that psilacetin, psilocin, and psilocybine bind to Mprotease with -6.0, -5.4, and -5.8 kcal/mol, respectively. Additionally, the psilacetin was found to inhibit human interleukin-6 receptors to reduce cytokine storm. The binding of psilacetin to Mprotease of SARS-CoV-2 and human interleukin-6 receptors changes the structural dynamics and Gibbs free energy patterns of proteins. These results suggested that psilocybin-mushroom could be utilized as viable potential chemotherapeutic agents for SARS-CoV-2.


Subject(s)
COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , Psilocybin/therapeutic use , Receptors, Interleukin-6/immunology , SARS-CoV-2/drug effects , Agaricales/chemistry , Antiviral Agents/therapeutic use , COVID-19/immunology , Cytokine Release Syndrome/immunology , Humans , Interleukin-6/immunology , SARS-CoV-2/immunology , Virus Replication/drug effects
10.
Nutrition ; 81: 110900, 2021 01.
Article in English | MEDLINE | ID: mdl-32738510

ABSTRACT

The new coronavirus associated with severe acute respiratory syndrome (SARS-CoV-2), surprisingly, does not affect only the lungs. The severe response to SARS-CoV-2 appears to include a "cytokine storm," which indicates a state of hyperinflammation and subsequent dysfunction of multiple organs and tissues in the most severe cases. This could be the reason why populations at the highest risk for death from the SARS-CoV-2 infection-induced disease (coronavirus disease 2019 [COVID-19]) are those suffering from chronic low-grade inflammation, but prone to hyperinflammation. This includes individuals of advanced age and those with obesity, type 2 diabetes, hypertension, and metabolic syndrome. Inflammation resolution is strongly dependent on lipid mediators, the specialized pro-resolution mediators (SPMs). ω-3 polyunsaturated fatty acids (ω-3 PUFAs) are precursors of very potent SPMs, including resolvins, protectins, and maresins. Additionally, they are associated with a less aggressive inflammatory initiation, after competing with ω-6 fatty acids for eicosanoid synthesis. Therefore, it makes sense to consider the use of ω-3 PUFAs for clinical management of COVID-19 patients. ω-3 PUFAs may be given by oral, enteral, or parenteral routes; however, the parenteral route favors faster incorporation into plasma phospholipids, blood cells, and tissues. Here, we discuss these aspects to propose the parenteral infusion of ω-3 PUFAs as adjuvant immunopharmacotherapy for hospitalized patients with COVID-19.


Subject(s)
COVID-19 Drug Treatment , Fish Oils/administration & dosage , COVID-19/epidemiology , COVID-19/immunology , Chemotherapy, Adjuvant , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Fatty Acids, Omega-3/administration & dosage , Humans , Inflammation/diet therapy , Inflammation/immunology , Infusions, Parenteral , Models, Biological , Nutritional Physiological Phenomena , Pandemics , SARS-CoV-2
11.
Article in English | MEDLINE | ID: mdl-33100211

ABSTRACT

The complications of the SARS-CoV-2 infection and its COVID-19 disease on mothers and their offspring are less known. This review aimed to determine the transmission, severity, and complications of SARS- CoV-2 infection during pregnancy. This review showed the influence of COVID-19 disease on neonatal neurogenesis. Owing medicines that were reported for the treatment of COVID-19 disease, this review suggested some control strategies like treatments (medicinal plants, antiviral therapy, cellular therapy, and immunotherapy), nutrition uptake, prevention, and recommendations. This overview showed that severe infection of SARS-CoV-2 during the early stage of pregnancy might increase the risk of stress, panic, and anxiety. This disorder can disturb the maternal immune system, and thus causing a neurodevelopmental disturbance. This hypothesis may be depending on the severity and intensity of the SARS-CoV-2 infection during pregnancy. However, vertical transmission of SARS-CoV-2 from dams to their fetuses is absent until now. During this global pandemic disease, maintaining safety during pregnancy, vaginal delivery, and breastfeeding may play a vital role in a healthy life for the offspring. Thus, international, and national organizations should be continuing for perinatal management, particularly during the next pandemic or disaster time.


Subject(s)
COVID-19/therapy , COVID-19/transmission , Infectious Disease Transmission, Vertical/prevention & control , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/therapy , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19/immunology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/therapy , Female , Humans , Immunotherapy/methods , Pregnancy , SARS-CoV-2/drug effects , SARS-CoV-2/immunology
12.
Clin Transl Sci ; 14(1): 75-85, 2021 01.
Article in English | MEDLINE | ID: mdl-32882099

ABSTRACT

T-cell directing/engaging bispecifics (TDBs) enable a powerful mode of action by activating T-cells through the creation of artificial immune synapses. Their pharmacological response involves the dynamic inter-relationships among T-cells, tumor cells, and TDBs. This results in complex and challenging issues in understanding pharmacokinetics, tissue distribution, target engagement, and exposure-response relationship. Dosing strategy plays a crucial role in determining the therapeutic window of TDBs because of the desire to maximize therapeutic efficacy in the context of known mechanism-related adverse events, such as cytokine release syndrome and neurological adverse events. Such adverse events are commonly reported as the most prominent events during the initial treatment cycles and dissipate over time. Therefore, the kinetic characterization of the inter-relationships between exposure/target engagement and safety/efficacy outcomes is crucial in designing the optimal dosing regimen to maximize the benefit/risk of TDB agents. In this review, we discuss the key clinical pharmacological considerations in drug discovery and development for TDBs and provide a summary of TDBs currently in clinical development. We also propose forward-looking perspectives and opportunities to derive insights through quantitative clinical pharmacology approaches.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Lymphocyte Activation/drug effects , Neoplasms/drug therapy , T-Lymphocytes/drug effects , Animals , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Cytokine Release Syndrome/chemically induced , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/prevention & control , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Drug Development , Drug Discovery , Drug Evaluation, Preclinical , Humans , Macaca fascicularis , Models, Animal , Neoplasms/immunology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/prevention & control , T-Lymphocytes/immunology , Treatment Outcome
13.
Biomed Pharmacother ; 133: 110917, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33217688

ABSTRACT

Scutellaria baicalensis Georgi., a plant used in traditional Chinese medicine, has multiple biological activities, including anti-inflammatory, antiviral, antitumor, antioxidant, and antibacterial effects, and can be used to treat respiratory tract infections, pneumonia, colitis, hepatitis, and allergic diseases. The main active substances of S. baicalensis, baicalein, baicalin, wogonin, wogonoside, and oroxylin A, can act directly on immune cells such as lymphocytes, macrophages, mast cells, dendritic cells, monocytes, and neutrophils, and inhibit the production of the inflammatory cytokines IL-1ß, IL-6, IL-8, and TNF-α, and other inflammatory mediators such as nitric oxide, prostaglandins, leukotrienes, and reactive oxygen species. The molecular mechanisms underlying the immunomodulatory and anti-inflammatory effects of the active compounds of S. baicalensis include downregulation of toll-like receptors, activation of the Nrf2 and PPAR signaling pathways, and inhibition of the nuclear thioredoxin system and inflammation-associated pathways such as those of MAPK, Akt, NFκB, and JAK-STAT. Given that in addition to the downregulation of cytokine production, the active constituents of S. baicalensis also have antiviral and antibacterial effects, they may be more promising candidate therapeutics for the prevention of infection-related cytokine storms than are drugs having only antimicrobial or anti-inflammatory activities.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cytokine Release Syndrome/prevention & control , Cytokines/antagonists & inhibitors , Immune System/drug effects , Inflammation Mediators/antagonists & inhibitors , Inflammation/prevention & control , Phytochemicals/therapeutic use , Scutellaria , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/isolation & purification , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/metabolism , Cytokines/metabolism , Humans , Immune System/immunology , Immune System/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Phytochemicals/adverse effects , Phytochemicals/isolation & purification , Scutellaria/chemistry , Signal Transduction
14.
Molecules ; 25(22)2020 Nov 16.
Article in English | MEDLINE | ID: mdl-33207753

ABSTRACT

Low levels of micronutrients have been associated with adverse clinical outcomes during viral infections. Therefore, to maximize the nutritional defense against infections, a daily allowance of vitamins and trace elements for malnourished patients at risk of or diagnosed with coronavirus disease 2019 (COVID-19) may be beneficial. Recent studies on COVID-19 patients have shown that vitamin D and selenium deficiencies are evident in patients with acute respiratory tract infections. Vitamin D improves the physical barrier against viruses and stimulates the production of antimicrobial peptides. It may prevent cytokine storms by decreasing the production of inflammatory cytokines. Selenium enhances the function of cytotoxic effector cells. Furthermore, selenium is important for maintaining T cell maturation and functions, as well as for T cell-dependent antibody production. Vitamin C is considered an antiviral agent as it increases immunity. Administration of vitamin C increased the survival rate of COVID-19 patients by attenuating excessive activation of the immune response. Vitamin C increases antiviral cytokines and free radical formation, decreasing viral yield. It also attenuates excessive inflammatory responses and hyperactivation of immune cells. In this mini-review, the roles of vitamin C, vitamin D, and selenium in the immune system are discussed in relation to COVID-19.


Subject(s)
Ascorbic Acid/therapeutic use , Coronavirus Infections/prevention & control , Cytokine Release Syndrome/prevention & control , Dietary Supplements , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Selenium/therapeutic use , Vitamin D/therapeutic use , Antibodies, Viral/biosynthesis , Betacoronavirus/drug effects , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/diet therapy , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokine Release Syndrome/diet therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immune System/drug effects , Immunologic Factors/therapeutic use , Micronutrients/therapeutic use , Pneumonia, Viral/diet therapy , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , SARS-CoV-2 , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology
16.
Am J Chin Med ; 48(6): 1263-1277, 2020.
Article in English | MEDLINE | ID: mdl-32907358

ABSTRACT

In December 2019, a novel coronavirus SARS-CoV-2, causing the disease COVID-19, spread from Wuhan throughout China and has infected people over 200 countries. Thus far, more than 3,400,000 cases and 240,000 deaths have occurred worldwide, and the coronavirus pandemic continues to grip the globe. While numbers of cases in China have been steadying, the number of infections outside China is increasing at a worrying pace. We face an urgent need to control the spread of the COVID-19 epidemic, which is currently expanding to a global pandemic. Efforts have focused on testing antiviral drugs and vaccines, but there is currently no treatment specifically approved. Traditional Chinese medicine (TCM) is grounded in empirical observations and the Chinese people use TCM to overcome these sorts of plagues many times in thousands of years of history. Currently, the Chinese National Health Commission recommended a TCM prescription of Qing-Fei-Pai-Du-Tang (QFPDT) in the latest version of the "Diagnosis and Treatment guidelines of COVID-19" which has been reported to provide reliable effects for COVID-19. While doubts about TCM still exist today, this review paper will describe the rationalities that QFPDT is likely to bring a safe and effective treatment of COVID-19.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Coronavirus Infections/drug therapy , Drugs, Chinese Herbal/therapeutic use , Pneumonia, Viral/drug therapy , Adrenal Cortex Hormones/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Betacoronavirus , COVID-19 , Chloroquine/therapeutic use , Coronavirus Infections/immunology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Drug Combinations , Humans , Indoles/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Lopinavir/therapeutic use , Medicine, Chinese Traditional , Pandemics , Pneumonia, Viral/immunology , Ritonavir/therapeutic use , SARS-CoV-2 , COVID-19 Drug Treatment
17.
Exp Dermatol ; 29(9): 885-890, 2020 09.
Article in English | MEDLINE | ID: mdl-32779213

ABSTRACT

The negative outcomes of COVID-19 diseases respiratory distress (ARDS) and the damage to other organs are secondary to a "cytokine storm" and to the attendant oxidative stress. Active hydroxyl forms of vitamin D are anti-inflammatory, induce antioxidative responses, and stimulate innate immunity against infectious agents. These properties are shared by calcitriol and the CYP11A1-generated non-calcemic hydroxyderivatives. They inhibit the production of pro-inflammatory cytokines, downregulate NF-κΒ, show inverse agonism on RORγ and counteract oxidative stress through the activation of NRF-2. Therefore, a direct delivery of hydroxyderivatives of vitamin D deserves consideration in the treatment of COVID-19 or ARDS of different aetiology. We also recommend treatment of COVID-19 patients with high-dose vitamin D since populations most vulnerable to this disease are likely vitamin D deficient and patients are already under supervision in the clinics. We hypothesize that different routes of delivery (oral and parenteral) will have different impact on the final outcome.


Subject(s)
COVID-19 Drug Treatment , COVID-19/immunology , Pandemics , SARS-CoV-2 , Skin/drug effects , Skin/immunology , Vitamin D/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Antiviral Agents/therapeutic use , COVID-19/complications , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Humans , Immunity, Innate/drug effects , Models, Biological , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology , Vitamin D/administration & dosage , Vitamin D Deficiency/complications , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/immunology
18.
BMJ Open ; 10(7): e039519, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32641343

ABSTRACT

INTRODUCTION: The rapid worldwide spread of COVID-19 has caused a global health crisis. To date, symptomatic supportive care has been the most common treatment. It has been reported that the mechanism of COVID-19 is related to cytokine storms and subsequent immunogenic damage, especially damage to the endothelium and alveolar membrane. Vitamin C (VC), also known as L-ascorbic acid, has been shown to have antimicrobial and immunomodulatory properties. A high dose of intravenous VC (HIVC) was proven to block several key components of cytokine storms, and HIVC showed safety and varying degrees of efficacy in clinical trials conducted on patients with bacterial-induced sepsis and acute respiratory distress syndrome (ARDS). Therefore, we hypothesise that HIVC could be added to the treatment of ARDS and multiorgan dysfunction related to COVID-19. METHODS AND ANALYSIS: The investigators designed a multicentre prospective randomised placebo-controlled trial that is planned to recruit 308 adults diagnosed with COVID-19 and transferred into the intensive care unit. Participants will randomly receive HIVC diluted in sterile water or placebo for 7 days once enrolled. Patients with a history of VC allergy, end-stage pulmonary disease, advanced malignancy or glucose-6-phosphate dehydrogenase deficiency will be excluded. The primary outcome is ventilation-free days within 28 observational days. This is one of the first clinical trials applying HIVC to treat COVID-19, and it will provide credible efficacy and safety data. We predict that HIVC could suppress cytokine storms caused by COVID-19, help improve pulmonary function and reduce the risk of ARDS of COVID-19. ETHICS AND DISSEMINATION: The study protocol was approved by the Ethics Committee of Zhongnan Hospital of Wuhan University (identifiers: Clinical Ethical Approval No. 2020001). Findings of the trial will be disseminated through peer-reviewed journals and scientific conferences. TRIAL REGISTRATION NUMBER: NCT04264533.


Subject(s)
Ascorbic Acid/administration & dosage , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/drug therapy , Pneumonia, Viral/drug therapy , Vitamins/administration & dosage , Administration, Intravenous , Betacoronavirus , COVID-19 , China , Coronavirus Infections/complications , Coronavirus Infections/immunology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Hospital Mortality , Humans , Intensive Care Units , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Respiration, Artificial , SARS-CoV-2 , Severity of Illness Index , Treatment Outcome , COVID-19 Drug Treatment
19.
J Leukoc Biol ; 108(1): 17-41, 2020 07.
Article in English | MEDLINE | ID: mdl-32534467

ABSTRACT

Clinical evidence indicates that the fatal outcome observed with severe acute respiratory syndrome-coronavirus-2 infection often results from alveolar injury that impedes airway capacity and multi-organ failure-both of which are associated with the hyperproduction of cytokines, also known as a cytokine storm or cytokine release syndrome. Clinical reports show that both mild and severe forms of disease result in changes in circulating leukocyte subsets and cytokine secretion, particularly IL-6, IL-1ß, IL-10, TNF, GM-CSF, IP-10 (IFN-induced protein 10), IL-17, MCP-3, and IL-1ra. Not surprising, therapies that target the immune response and curtail the cytokine storm in coronavirus 2019 (COVID-19) patients have become a focus of recent clinical trials. Here we review reports on leukocyte and cytokine data associated with COVID-19 disease in 3939 patients in China and describe emerging data on immunopathology. With an emphasis on immune modulation, we also look at ongoing clinical studies aimed at blocking proinflammatory cytokines; transfer of immunosuppressive mesenchymal stem cells; use of convalescent plasma transfusion; as well as immunoregulatory therapy and traditional Chinese medicine regimes. In examining leukocyte and cytokine activity in COVID-19, we focus in particular on how these levels are altered as the disease progresses (neutrophil NETosis, macrophage, T cell response, etc.) and proposed consequences to organ pathology (coagulopathy, etc.). Viral and host interactions are described to gain further insight into leukocyte biology and how dysregulated cytokine responses lead to disease and/or organ damage. By better understanding the mechanisms that drive the intensity of a cytokine storm, we can tailor treatment strategies at specific disease stages and improve our response to this worldwide public health threat.


Subject(s)
Betacoronavirus , Coronavirus Infections/immunology , Cytokine Release Syndrome/immunology , Cytokines/immunology , Leukocytes/immunology , Pneumonia, Viral/immunology , COVID-19 , China , Humans , Pandemics , SARS-CoV-2
20.
Biomed Pharmacother ; 128: 110316, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32505821

ABSTRACT

BACKGROUND: Pudilan (PDL), a four-herb prescription with the traditional function of heat-clearing and detoxifying, has been clinically used as an anti-SARS-CoV-2 infectory agent in China. PDL might also have therapeutic potentials for COVID-19 while the underlying mechanisms remain to be clarified. METHODS: We used network pharmacology analysis and selected 68 co-targeted genes/proteins as targets of both PDL and COVID-19. These co-targeted genes/proteins were predicted by SwissDock Server for their high-precision docking simulation, and analyzed by STRING for proteins to protein interaction (PPI), pathway and GO (gene ontology) enrichment. The therapeutic effect for PDL treatment on COVID-19 was validated by the TCMATCOV (TCM Anti COVID-19) platform. RESULTS: PDL might prevent the entrance of SARS-CoV-2 entry into cells by blocking the angiotensin-converting enzyme 2 (ACE2). It might inhibit the cytokine storm by affecting C-reactive protein (CRP), interferon-γ (IFN-γ), interleukin- 6 (IL-6), interleukin- 10 (IL-10), tumor necrosis factor (TNF), epidermal growth factor receptor (EGFR), C-C motif chemokine ligand 5 (CCL5), transforming growth factor-ß1 (TGFß1), and other proteins. PDL might moderate the immune system to shorten the course of the disease, delay disease progression, and reduce the mortality rate. CONCLUSION: PDL might have a therapeutic effect on COVID-19 through three aspects, including the moderate immune system, anti-inflammation, and anti-virus entry into cells.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus , Coronavirus Infections , Cytokine Release Syndrome , Drugs, Chinese Herbal/pharmacology , Pandemics , Pneumonia, Viral , Virus Internalization/drug effects , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Anti-Inflammatory Agents/pharmacology , Betacoronavirus/drug effects , Betacoronavirus/isolation & purification , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Humans , Immunologic Factors/pharmacology , Interferon-gamma/immunology , Interleukins/immunology , Molecular Docking Simulation , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/drug therapy , Pneumonia, Viral/immunology , Protein Interaction Maps , SARS-CoV-2 , Transforming Growth Factor beta/immunology
SELECTION OF CITATIONS
SEARCH DETAIL