Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Journal of Pediatric Neurology ; 2023.
Article in English | Web of Science | ID: covidwho-2309171

ABSTRACT

Myasthenia gravis (MG) is a rare, long-term neuromuscular disorder that can affect individuals of any age. In Japan, the Omicron variant of coronavirus disease 2019 (COVID-19) began spreading in 2022, and many cases of neurological symptoms caused by the virus have been reported. Although COVID-19 has been reported to exacerbate MG in adults, there are no reports on the effects of COVID-19 on the MG symptoms of pediatric patients. We report the case of a 6-year-old girl with a 3-year history of MG who presented to our hospital with symptom exacerbation after COVID-19 infection. Four days before admission, she developed fever with a runny nose and cough. Three days before admission, she developed severe bilateral blepharoptosis and progressive limb weakness, and 2 days before admission, she was diagnosed with COVID-19 by SARS-CoV-2 antigen test. Physical examination revealed moderate blepharoptosis and mild bilateral upper and lower limb weakness. We diagnosed her with worsening MG due to COVID-19, and she was administered 400 mg/kg intravenous immunoglobulin (IVIG) daily for 5 days with continued oral corticosteroids and tacrolimus. The patient's symptoms improved promptly after admission and, at discharge 7 days after admission, her symptoms had significantly improved. During the 1-month outpatient follow-up period, she remained stable and the anti-acetylcholine receptor (AchR) antibody level was reduced to 14.6 nmol/L (from 18.5 nmol/L on admission). Our case suggests that COVID-19 exacerbates MG in both children and adults.

2.
J Biol Chem ; 299(5): 104707, 2023 05.
Article in English | MEDLINE | ID: covidwho-2301085

ABSTRACT

Virus entry into animal cells is initiated by attachment to target macromolecules located on host cells. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) trimeric spike glycoprotein targets host angiotensin converting enzyme 2 to gain cellular access. The SARS-CoV-2 glycoprotein contains a neurotoxin-like region that has sequence similarities to the rabies virus and the HIV glycoproteins, as well as to snake neurotoxins, which interact with nicotinic acetylcholine receptor (nAChR) subtypes via this region. Using a peptide of the neurotoxin-like region of SARS-CoV-2 (SARS-CoV-2 glycoprotein peptide [SCoV2P]), we identified that this area moderately inhibits α3ß2, α3ß4, and α4ß2 subtypes, while potentiating and inhibiting α7 nAChRs. These nAChR subtypes are found in target tissues including the nose, lung, central nervous system, and immune cells. Importantly, SCoV2P potentiates and inhibits ACh-induced α7 nAChR responses by an allosteric mechanism, with nicotine enhancing these effects. Live-cell confocal microscopy was used to confirm that SCoV2P interacts with α7 nAChRs in transfected neuronal-like N2a and human embryonic kidney 293 cells. The SARS-CoV-2 ectodomain functionally potentiates and inhibits the α7 subtype with nanomolar potency. Our functional findings identify that the α7 nAChR is a target for the SARS-CoV-2 glycoprotein, providing a new aspect to our understanding of SARS-CoV-2 and host cell interactions, in addition to disease pathogenesis.


Subject(s)
Receptors, Nicotinic , SARS-CoV-2 , alpha7 Nicotinic Acetylcholine Receptor , Humans , alpha7 Nicotinic Acetylcholine Receptor/genetics , COVID-19 , Neurotoxins , Receptors, Nicotinic/genetics , Spike Glycoprotein, Coronavirus/genetics
3.
Pharmacol Res ; 191: 106758, 2023 05.
Article in English | MEDLINE | ID: covidwho-2297638

ABSTRACT

The α7-nicotinic acetylcholine receptor (α7nAChR) is a key protein in the cholinergic anti-inflammatory pathway (CAP) that links the nervous and immune systems. Initially, the pathway was discovered based on the observation that vagal nerve stimulation (VNS) reduced the systemic inflammatory response in septic animals. Subsequent studies form a foundation for the leading hypothesis about the central role of the spleen in CAP activation. VNS evokes noradrenergic stimulation of ACh release from T cells in the spleen, which in turn activates α7nAChRs on the surface of macrophages. α7nAChR-mediated signaling in macrophages reduces inflammatory cytokine secretion and modifies apoptosis, proliferation, and macrophage polarization, eventually reducing the systemic inflammatory response. A protective role of the CAP has been demonstrated in preclinical studies for multiple diseases including sepsis, metabolic disease, cardiovascular diseases, arthritis, Crohn's disease, ulcerative colitis, endometriosis, and potentially COVID-19, sparking interest in using bioelectronic and pharmacological approaches to target α7nAChRs for treating inflammatory conditions in patients. Despite a keen interest, many aspects of the cholinergic pathway are still unknown. α7nAChRs are expressed on many other subsets of immune cells that can affect the development of inflammation differently. There are also other sources of ACh that modify immune cell functions. How the interplay of ACh and α7nAChR on different cells and in various tissues contributes to the anti-inflammatory responses requires additional study. This review provides an update on basic and translational studies of the CAP in inflammatory diseases, the relevant pharmacology of α7nAChR-activated drugs and raises some questions that require further investigation.


Subject(s)
COVID-19 , Receptors, Nicotinic , Animals , Female , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Inflammation/metabolism , Macrophages/metabolism , Systemic Inflammatory Response Syndrome
4.
Comput Struct Biotechnol J ; 19: 1654-1660, 2021.
Article in English | MEDLINE | ID: covidwho-2261625

ABSTRACT

Susceptibility to severe illness from COVID-19 is anticipated to be associated with cigarette smoking as it aggravates the risk of cardiovascular and respiratory illness, including infections. This is particularly important with the advent of a new strain of coronaviruses, the severe acute respiratory syndrome coronavirus (SARS-CoV-2) that has led to the present pandemic, coronavirus disease 2019 (COVID-19). Although, the effects of smoking on COVID-19 are less described and controversial, we presume a link between smoking and COVID-19. Smoking has been shown to enhance the expression of the angiotensin-converting enzyme-2 (ACE-2) and transmembrane serine protease 2 (TMPRSS2) key entry genes utilized by SARS-CoV-2 to infect cells and induce a 'cytokine storm', which further increases the severity of COVID-19 clinical course. Nevertheless, the impact of smoking on ACE-2 and TMPRSS2 receptors expression remains paradoxical. Thus, further research is necessary to unravel the association between smoking and COVID-19 and to pursue the development of potential novel therapies that are able to constrain the morbidity and mortality provoked by this infectious disease. Herein we present a brief overview of the current knowledge on the correlation between smoking and the expression of SARS-CoV-2 key entry genes, clinical manifestations, and disease progression.

5.
Free Radic Biol Med ; 190: 247-263, 2022 09.
Article in English | MEDLINE | ID: covidwho-2269132

ABSTRACT

Clinical studies have shown a significant positive correlation between age and the likelihood of being infected with SARS-CoV-2. This increased susceptibility is positively correlated with chronic inflammation and compromised neurocognitive functions. Postmortem analyses suggest that acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), with systemic and lung hyperinflammation, can cause significant morbidity and mortality in COVID-19 patients. Supraphysiological supplemental oxygen, also known as hyperoxia, is commonly used to treat decreased blood oxygen saturation in COVID-19 patients. However, prolonged exposure to hyperoxia alone can cause oxygen toxicity, due to an excessive increase in the levels of reactive oxygen species (ROS), which can overwhelm the cellular antioxidant capacity. Subsequently, this causes oxidative cellular damage and increased levels of aging biomarkers, such as telomere shortening and inflammaging. The oxidative stress in the lungs and brain can compromise innate immunity, resulting in an increased susceptibility to secondary lung infections, impaired neurocognitive functions, and dysregulated hyperinflammation, which can lead to ALI/ARDS, and even death. Studies indicate that lung inflammation is regulated by the central nervous system, notably, the cholinergic anti-inflammatory pathway (CAIP), which is innervated by the vagus nerve and α7 nicotinic acetylcholine receptors (α7nAChRs) on lung cells, particularly lung macrophages. The activation of α7nAChRs attenuates oxygen toxicity in the lungs and improves clinical outcomes by restoring hyperoxia-compromised innate immunity. Mechanistically, α7nAChR agonist (e.g., GAT 107 and GTS-21) can regulate redox signaling by 1) activating Nrf2, a master regulator of the antioxidant response and a cytoprotective defense system, which can decrease cellular damage caused by ROS and 2) inhibiting the activation of the NF-κB-mediated inflammatory response. Notably, GTS-21 has been shown to be safe and it improves neurocognitive functions in humans. Therefore, targeting the α7nAChR may represent a viable therapeutic approach for attenuating dysregulated hyperinflammation-mediated ARDS and sepsis in COVID-19 patients receiving prolonged oxygen therapy.


Subject(s)
Acute Lung Injury , COVID-19 , Hyperoxia , Pneumonia , Respiratory Distress Syndrome , Acute Lung Injury/metabolism , Aging , Antioxidants/metabolism , COVID-19/therapy , Humans , Hyperoxia/complications , Hyperoxia/metabolism , Lung/metabolism , Oxygen/metabolism , Pneumonia/metabolism , Reactive Oxygen Species/metabolism , SARS-CoV-2 , alpha7 Nicotinic Acetylcholine Receptor/metabolism
6.
ACS Chem Neurosci ; 14(4): 689-698, 2023 02 15.
Article in English | MEDLINE | ID: covidwho-2236297

ABSTRACT

A deficiency of the functional α7 nicotinic acetylcholine receptor (α7nAChR) impairs neuronal and immune systems. The SARS-CoV-2 spike protein (S12) facilitates virus cell entry during COVID-19 infection and can also independently disrupt cellular functions. Here, we found that S12 expression significantly downregulated surface expression of α7nAChR in mammalian cells. A helical segment of S12 (L1145-L1152) in the spike neck was identified to be responsible for the downregulation of α7nAChR, as the mutant S12AAA (L1145A-F1148A-L1152A) had minimal effects on surface α7nAChR expression. This S12 segment is homologous to the α7nAChR intracellular helical motif known for binding chaperone proteins RIC3 and Bcl-2 to promote α7nAChR surface expression. Competition from S12 for binding these proteins likely underlies suppression of surface α7nAChR. Considering the critical roles of α7nAChR in cellular functions, these findings provide a new perspective for improving mRNA vaccines and developing treatment options for certain symptoms related to long COVID.


Subject(s)
COVID-19 , alpha7 Nicotinic Acetylcholine Receptor , Animals , Humans , alpha7 Nicotinic Acetylcholine Receptor/genetics , Spike Glycoprotein, Coronavirus , Post-Acute COVID-19 Syndrome , SARS-CoV-2 , Mammals
7.
Cureus ; 14(12): e33048, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-2226171

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disorder affecting the neuromuscular junction caused by a B-cell-mediated, T-cell-dependent immunologic attack at the end plate of the postsynaptic membrane. Attack on muscle acetylcholine receptors (AChR) of the postsynaptic membrane due to the AChR, muscle-specific tyrosine kinase, or lipoprotein receptor-related peptide 4 antibodies lead to symptoms of painless, fluctuating weakness of muscle groups and often begins with ocular signs and symptoms. Coronavirus disease 2019 (COVID-19) is an acute respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus closely related to SARS-CoV. Serious neurologic complications are infrequent and diverse with reported cases of stroke, encephalitis/meningitis, Guillain-Barré syndrome, acute disseminated encephalomyelitis, ataxia, and unspecified limb weakness. MG is a rarely reported sequela of COVID-19 infection. To date, there are 15 reported cases of post-COVID-19 MG. In this article, we present a case of post-COVID-19 MG and a concise review of other reported cases. An 83-year-old Caucasian male with a medical history of atrial fibrillation status post-ablation and non-ischemic cardiomyopathy was initially admitted for COVID-19 pneumonia. He was treated with remdesivir, convalescent plasma, and supplemental oxygen therapy but did not require invasive mechanical intubation. One month after discharge, he started experiencing fatigue with muscle weakness and progressive dyspnea. He progressed to develop dysphonia, especially at the end of the day. After extensive workup, he was diagnosed with MG with a positive antibody against the AChR. The chronological events of developing slowly worsening muscular weakness after recovering from COVID-19 infection and positive AChR antibody led to the diagnosis of post-COVID-19 new-onset MG. Post-COVID-19 fatigue, long-term use of steroids, and intensive care unit-related physical deconditioning can be confounders in the clinical presentation of post-COVID-19 new-onset MG. Careful history-taking and meticulous assessment of chronological events are needed to diagnose this rare entity.

8.
Molecules ; 28(3)2023 Jan 23.
Article in English | MEDLINE | ID: covidwho-2216645

ABSTRACT

As an indispensable component in human beings, the acetylcholine system regulates multiple physiological processes not only in neuronal tissues but also in nonneuronal tissues. However, since the concept of the "Nonneuronal cholinergic system (NNCS)" has been proposed, the role of the acetylcholine system in nonneuronal tissues has received increasing attention. A growing body of research shows that the acetylcholine system also participates in modulating inflammatory responses, regulating contraction and mucus secretion of respiratory tracts, and influencing the metastasis and invasion of lung cancer. In addition, the susceptibility and severity of respiratory tract infections caused by pathogens such as Mycobacterium Tuberculosis and the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) can also correlate with the regulation of the acetylcholine system. In this review, we summarized the major roles of the acetylcholine system in respiratory diseases. Despite existing achievements in the field of the acetylcholine system, we hope that more in-depth investigations on this topic will be conducted to unearth more possible pharmaceutical applications for the treatment of diverse respiratory diseases.


Subject(s)
COVID-19 , Respiratory Tract Infections , Humans , Acetylcholine , SARS-CoV-2 , Respiratory System
9.
Front Cardiovasc Med ; 9: 1037369, 2022.
Article in English | MEDLINE | ID: covidwho-2162989

ABSTRACT

Since early 2020, SARS-CoV-2-induced infection resulted in global pandemics with high morbidity, especially in the adult population. COVID-19 is a highly prothrombotic condition associated with subsequent multiorgan failure and lethal outcomes. The exact mechanism of the prothrombotic state is not well understood and might be multifactorial. Nevertheless, platelets are attributed to play a crucial role in COVID-19-associated thrombosis. To date, platelets' role was defined primarily in thrombosis and homeostasis. Currently, more focus has been set on their part in inflammation and immunity. Moreover, their ability to release various soluble factors under activation as well as internalize and degrade specific pathogens has been highly addressed in viral research. This review article will discuss platelet role in COVID-19-associated thrombosis and their role in the cholinergic anti-inflammatory pathway. Multiple studies confirmed that platelets display a hyperactivated phenotype in COVID-19 patients. Critically ill patients demonstrate increased platelet activation markers such as P-selectin, PF4, or serotonin. In addition, platelets contain acetylcholine and express α7 nicotinic acetylcholine receptors (α7nAchR). Thus, acetylcholine can be released under activation, and α7nAchR can be stimulated in an autocrine manner and support platelet function. α7 receptor is one of the most important mediators of the anti-inflammatory properties as it is associated with humoral and intrinsic immunity and was demonstrated to contribute to better outcomes in COVID-19 patients when under stimulation. Hematopoietic α7nAchR deficiency increases platelet activation and, in experimental studies, α7nAchR stimulation can diminish the pro-inflammatory state and modulate platelet reactiveness via increased levels of NO. NO has been described to inhibit platelet adhesion, activation, and aggregation. In addition, acetylcholine has been demonstrated to decrease platelet aggregation possibly by blocking the e p-38 pathway. SARS-CoV-2 proteins have been found to be similar to neurotoxins which can bind to nAChR and prevent the action of acetylcholine. Concluding, the platelet role in COVID-19 thrombotic events could be explained by their active function in the cholinergic anti-inflammatory pathway.

10.
BBA Adv ; 2: 100061, 2022.
Article in English | MEDLINE | ID: covidwho-2130127

ABSTRACT

Mammalian nicotinic acetylcholine receptors (nAChRs) were initially discovered as ligand-gated ion channels mediating fast synaptic transmission in the neuro-muscular junctions and autonomic ganglia. They were further found to be involved in a wide range of basic biological processes within the brain and in non-excitable tissues. The present review summarizes the data obtained in our laboratory during last two decades. Investigation of autonomic ganglia with the nAChR subunit-specific antibodies was followed by identification of nAChRs in B lymphocytes, discovery of mitochondrial nAChRs and their role in mitochondrial apoptosis pathway, and revealing the role of α7 nAChRs and α7-specific antibodies in neuroinflammation-related Alzheimer disease and COVID-19. The data obtained demonstrate the involvement of nAChRs in cell survival, proliferation, cell-to-cell communication and inflammatory reaction. Together with the ability of nAChRs to function in both ionotropic and metabotropic way, these data illustrate the universal nature of cholinergic regulation mediated by nAChRs.

11.
Curr Drug Targets ; 23(13): 1277-1287, 2022.
Article in English | MEDLINE | ID: covidwho-2098966

ABSTRACT

Covid-19 may be associated with various neurological disorders, including dysautonomia, a dysfunction of the autonomic nervous system (ANS). In Covid-19, hypoxia, immunoinflammatory abnormality, and deregulation of the renin-angiotensin system (RAS) may increase sympathetic discharge with dysautonomia development. Direct SARS-CoV-2 cytopathic effects and associated inflammatory reaction may lead to neuroinflammation, affecting different parts of the central nervous system (CNS), including the autonomic center in the hypothalamus, causing dysautonomia. High circulating AngII, hypoxia, oxidative stress, high pro-inflammatory cytokines, and emotional stress can also provoke autonomic deregulation and high sympathetic outflow with the development of the sympathetic storm. During SARS-CoV-2 infection with neuro-invasion, GABA-ergic neurons and nicotinic acetylcholine receptor (nAChR) are inhibited in the hypothalamic pre-sympathetic neurons leading to sympathetic storm and dysautonomia. Different therapeutic modalities are applied to treat SARS-CoV-2 infection, like antiviral and anti-inflammatory drugs. Ivermectin (IVM) is a robust repurposed drug widely used to prevent and manage mild-moderate Covid-19. IVM activates both GABA-ergic neurons and nAChRs to mitigate SARS-CoV-2 infection- induced dysautonomia. Therefore, in this brief report, we try to identify the potential role of IVM in managing Covid-19-induced dysautonomia.


Subject(s)
COVID-19 , Primary Dysautonomias , Humans , Animals , Bees , SARS-CoV-2 , Ivermectin , Hypoxia , gamma-Aminobutyric Acid
12.
Biochem Biophys Res Commun ; 622: 57-63, 2022 09 24.
Article in English | MEDLINE | ID: covidwho-1982609

ABSTRACT

COVID-19 is accompanied by strong inflammatory reaction and is often followed by long-term cognitive disorders. The fragment 674-685 of SARS-Cov-2 spike protein was shown to interact with α7 nicotinic acetylcholine receptor involved in regulating both inflammatory reactions and cognitive functions. Here we show that mice immunized with the peptide corresponding to 674-685 fragment of SARS-Cov-2 spike protein conjugated to hemocyanin (KLH-674-685) demonstrate decreased level of α7 nicotinic acetylcholine receptors, increased levels of IL-1ß and TNFα in the brain and impairment of episodic memory. Choline injections prevented α7 nicotinic receptor decline and memory loss. Mice injected with immunoglobulins obtained from the blood of (KLH-674-685)-immunized mice also demonstrated episodic memory decline. These data allow suggesting that post-COVID memory impairment in humans is related to SARS-Cov-2 spike protein-specific immune reaction. The mechanisms of such effect are being discussed.


Subject(s)
COVID-19 , Memory, Episodic , Animals , Humans , Immunization , Inflammation , Memory Disorders/etiology , Memory Disorders/metabolism , Mice , Neuroinflammatory Diseases , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/adverse effects , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
13.
Computation ; 10(4):25, 2022.
Article in English | Web of Science | ID: covidwho-1820189

ABSTRACT

Some clinical studies have indicated activity of ivermectin, a macrocyclic lactone, against COVID-19, but a biological mechanism initially proposed for this anti-viral effect is not applicable at physiological concentrations. This in silico investigation explores potential modes of action of ivermectin and 14 related compounds, by which the infectivity and morbidity of the SARS-CoV-2 virus may be limited. Binding affinity computations were performed for these agents on several docking sites each for models of (1) the spike glycoprotein of the virus, (2) the CD147 receptor, which has been identified as a secondary attachment point for the virus, and (3) the alpha-7 nicotinic acetylcholine receptor (alpha 7nAChr), an indicated point of viral penetration of neuronal tissue as well as an activation site for the cholinergic anti-inflammatory pathway controlled by the vagus nerve. Binding affinities were calculated for these multiple docking sites and binding modes of each compound. Our results indicate the high affinity of ivermectin, and even higher affinities for some of the other compounds evaluated, for all three of these molecular targets. These results suggest biological mechanisms by which ivermectin may limit the infectivity and morbidity of the SARS-CoV-2 virus and stimulate an alpha 7nAChr-mediated anti-inflammatory pathway that could limit cytokine production by immune cells.

14.
Bioimpacts ; 12(2): 171-174, 2022.
Article in English | MEDLINE | ID: covidwho-1668071

ABSTRACT

The cholinergic anti-inflammatory pathway (CAP) first described by Wang et al, 2003 has contemporary interest arising from the COVID-19 pandemic. While tobacco smoking has been considered an aggravating factor in the severity of COVID-19 infections, it has been suggested by some that the nicotine derived from tobacco could lessen the severity of COVID-19 infections. This spotlight briefly describes the CAP and its potential role as a therapeutic target for the treatment of COVID-19 infections using vagus nerve stimulation or selective alpha7 nicotinic acetylcholine receptor agonists.

15.
Int J Biochem Cell Biol ; 143: 106138, 2022 02.
Article in English | MEDLINE | ID: covidwho-1588223

ABSTRACT

Nicotinic acetylcholine receptors mediate fast synaptic transmission in neuro-muscular junctions and autonomic ganglia and modulate survival, proliferation and neurotransmitter or cytokine release in the brain and non-excitable cells. The neuronal-type nicotinic acetylcholine receptors are expressed in the outer mitochondria membrane to regulate the release of pro-apoptotic substances like cytochrome c or reactive oxygen species. In the intracellular environment, nicotinic acetylcholine receptor signaling is ion-independent and triggers intramitochondrial kinases, similar to those activated by plasma membrane nicotinic acetylcholine receptors. The present review will describe the data obtained during the last five years including, in particular, post-translational glycosylation as a targeting signal to mitochondria, mechanisms of mitochondrial nicotinic acetylcholine receptor signaling studied with subtype-specific agonists, antagonists, positive allosteric modulators and knockout mice lacking certain nicotinic acetylcholine receptor subunits, interaction of mitochondrial nicotinic acetylcholine receptors with Bcl-2 family proteins and their involvement in important pathologies like neuroinflammation, liver damage and SARS-CoV-2 infection.


Subject(s)
COVID-19/genetics , Chemical and Drug Induced Liver Injury/genetics , Mitochondria/genetics , Neuroinflammatory Diseases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Nicotinic/genetics , Allosteric Regulation , Animals , COVID-19/metabolism , COVID-19/pathology , COVID-19/virology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Humans , Mice , Mitochondria/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Nicotinic/metabolism , SARS-CoV-2/pathogenicity , Signal Transduction , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism
16.
Cells ; 10(12)2021 11 26.
Article in English | MEDLINE | ID: covidwho-1551567

ABSTRACT

High mobility group box 1 protein (HMGB1), a highly conserved nuclear DNA-binding protein, is a "damage-associated molecular pattern" molecule (DAMP) implicated in both stimulating and inhibiting innate immunity. As reviewed here, HMGB1 is an oxidation-reduction sensitive DAMP bearing three cysteines, and the post-translational modification of these residues establishes its proinflammatory and anti-inflammatory activities by binding to different extracellular cell surface receptors. The redox-sensitive signaling mechanisms of HMGB1 also occupy an important niche in innate immunity because HMGB1 may carry other DAMPs and pathogen-associated molecular pattern molecules (PAMPs). HMGB1 with DAMP/PAMP cofactors bind to the receptor for advanced glycation end products (RAGE) which internalizes the HMGB1 complexes by endocytosis for incorporation in lysosomal compartments. Intra-lysosomal HMGB1 disrupts lysosomal membranes thereby releasing the HMGB1-transported molecules to stimulate cytosolic sensors that mediate inflammation. This HMGB1-DAMP/PAMP cofactor pathway slowed the development of HMGB1-binding antagonists for diagnostic or therapeutic use. However, recent discoveries that HMGB1 released from neurons mediates inflammation via the TLR4 receptor system, and that cancer cells express fully oxidized HMGB1 as an immunosuppressive mechanism, offer new paths to targeting HMGB1 for inflammation, pain, and cancer.


Subject(s)
Disulfides/metabolism , HMGB1 Protein/metabolism , Inflammation/metabolism , Protein Processing, Post-Translational , Animals , COVID-19/metabolism , Humans , Sensory Receptor Cells/metabolism
17.
Inform Med Unlocked ; 26: 100714, 2021.
Article in English | MEDLINE | ID: covidwho-1492175

ABSTRACT

COVID-19 disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has posed a global health emergency. Repurposing of existing drugs can be a rapid and effective strategy to fight the infection. Clinical trials have reported reduction or elimination of viral load when patients were treated with the anti-malarial drug Hydroxychloroquine (HCQ). To understand the molecular mechanism of action for effective repurposing of this drug we have carried out in silico docking and dynamics studies on complexes between HCQ and target proteins, which were identified through both literature survey and structural similarity searches in databases of small molecule - protein complexes. The proteins identified as binding HCQ are: Angiotensin Converting Enzyme 2 (ACE2), α7 nicotinic AcetylCholine Receptor (α7 nAChR), α1D-adrenergic receptor (α1D-AR), Histamine N- Methyl Transferase (HNMT) and DNA gyrase/Topoisomerase III ß (Top3ß). The majority of these proteins are novel and have not been used before, in docking studies. Our docking and simulation results support action of HCQ both at the entry and post-entry stages of SARS-CoV2 infection. The mechanism of action at the entry stage is through blocking the virus-binding sites on the two receptors, ACE2 & α7 nAChR, by binding directly at those sites. Our computational studies also show that the action of HCQ at the post-entry stage is to prevent both viral replication and generation of 'cytokine storm' by inhibiting host Top3ß enzyme and α1D-AR, respectively. Binding of HCQ to HNMT is not a desired binding, and therefore this should be reduced during repurposing of HCQ.

18.
Biochem Biophys Res Commun ; 561: 14-18, 2021 07 05.
Article in English | MEDLINE | ID: covidwho-1225147

ABSTRACT

In spite of numerous studies, many details of SARS-Cov-2 interaction with human cells are still poorly understood. The 674-685 fragment of SARS-Cov-2 spike protein is homologous to the fragment of α-cobratoxin underlying its interaction with α7 nicotinic acetylcholine receptors (nAChRs). The interaction of 674-685 peptide with α7 nAChR has been predicted in silico. In the present paper we confirm this prediction experimentally and investigate the effect of SARS-Cov-2 spike protein peptide on mitochondria, which express α7 nAChRs to regulate apoptosis-related events. We demonstrate that SARS-Cov-2 spike protein peptide 674-685 competes with the antibody against 179-190 fragment of α7 nAChR subunit for the binding to α7-expressing cells and mitochondria and prevents the release of cytochrome c from isolated mitochondria in response to 0.5 mM H2O2 but does not protect intact U373 cells against apoptogenic effect of H2O2. Our data suggest that the α7 nAChR-binding portion of SARS-Cov-2 spike protein prevents mitochondria-driven apoptosis when the virus is uncoated inside the cell and, therefore, supports the infected cell viability before the virus replication cycle is complete.


Subject(s)
Apoptosis , Cytochromes c/metabolism , Mitochondria/metabolism , Peptide Fragments/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Computer Simulation , Female , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/growth & development , alpha7 Nicotinic Acetylcholine Receptor/metabolism
19.
Prev Med Rep ; 22: 101374, 2021 Jun.
Article in English | MEDLINE | ID: covidwho-1193451

ABSTRACT

JUUL is a groundbreaking electronic cigarette (e-cig) and the preeminent vaping product on the market. We present an overview of the rapid and spectacular rise of JUUL and its remarkable fall within the timespan of 2015 - 2020. We highlight JUUL's entering the market in June 2015, becoming the industry leader in mid 2017, and experiencing a litany of setbacks by late 2019 through to early 2020. We address the role played by JUUL in the ongoing epidemic of youth vaping. We also feature competing views on the public health impact of JUUL use (in particular), and e-cig vaping (in general). We further highlight the latest trends in youth vaping and sales records for JUUL and tobacco cigarettes. In view of the ongoing pandemic of COVID-19, we briefly summarize the existing evidence on the relationship between vaping and smoking and the prevalence, disease course, and clinical outcomes of COVID-19.

20.
Tob Use Insights ; 14: 1179173X20988674, 2021.
Article in English | MEDLINE | ID: covidwho-1153782

ABSTRACT

BACKGROUND: The recent global inclination for smoking during the Coronavirus Disease 2019 (COVID-19) pandemic has drawn attention to the impact of smoking on COVID-19. While smoking increases susceptibility to common respiratory pathogens including the closely related coronaviruses, COVID-19 causing Severe Acute Respiratory Coronavirus 2 (SARS-CoV-2) being a respiratory pathogen intrigues the possible association between smoking and viral pathogenicity. SMOKING AND COVID-19: The gender dependence of COVID-19 infection rates and a higher prevalence of smokers among males made the scientific world assume smoking to be a confounding variable in sex predisposition to COVID-19. Conversely, the controversial findings of discrepant morbidity and mortality rates of COVID-19 among smokers questioned the credibility of this hypothesis. More importantly, nicotine in smoking has been hypothesized to downregulate Interleukin-6 (IL-6) which plays a role in COVID-19 severity and to interfere with the Angiotensin-Converting Enzyme 2 (ACE2), the receptor of SARS-CoV-2 led the scientists to experiment nicotine patch prophylactically against COVID-19. Besides, interaction between spike protein and nicotinic acetylcholine receptors (nAChRs) supports the nicotinic cholinergic system dysregulation hypothesis in COVID-19 pathophysiology leading to its therapeutic use. However, despite the contradictions in the direct impact of smoking, it surely acts as fomites for viral transmission. CONCLUSION: Irrespective of the role nicotine in COVID-management, compassionate use of smoking against SARS-CoV-2 cannot be recommended until the therapeutic value gets proved and therapeutic form becomes available.

SELECTION OF CITATIONS
SEARCH DETAIL