Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Front Immunol ; 14: 1185233, 2023.
Article in English | MEDLINE | ID: covidwho-20244458

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a contagious respiratory virus that is the cause of the coronavirus disease 2019 (COVID-19) pandemic which has posed a serious threat to public health. COVID-19 is characterized by a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild cold-like symptoms, severe pneumonia or even death. Inflammasomes are supramolecular signaling platforms that assemble in response to danger or microbial signals. Upon activation, inflammasomes mediate innate immune defense by favoring the release of proinflammatory cytokines and triggering pyroptotic cell death. Nevertheless, abnormalities in inflammasome functioning can result in a variety of human diseases such as autoimmune disorders and cancer. A growing body of evidence has showed that SARS-CoV-2 infection can induce inflammasome assembly. Dysregulated inflammasome activation and consequent cytokine burst have been associated with COVID-19 severity, alluding to the implication of inflammasomes in COVID-19 pathophysiology. Accordingly, an improved understanding of inflammasome-mediated inflammatory cascades in COVID-19 is essential to uncover the immunological mechanisms of COVID-19 pathology and identify effective therapeutic approaches for this devastating disease. In this review, we summarize the most recent findings on the interplay between SARS-CoV-2 and inflammasomes and the contribution of activated inflammasomes to COVID-19 progression. We dissect the mechanisms involving the inflammasome machinery in COVID-19 immunopathogenesis. In addition, we provide an overview of inflammasome-targeted therapies or antagonists that have potential clinical utility in COVID-19 treatment.


Subject(s)
COVID-19 , Humans , Inflammasomes/metabolism , SARS-CoV-2/physiology , COVID-19 Drug Treatment , Cytokines
2.
Viruses ; 15(5)2023 05 05.
Article in English | MEDLINE | ID: covidwho-20235103

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative pathogen of the coronavirus disease 2019 (COVID-19) pandemic, a fatal respiratory illness. The associated risk factors for COVID-19 are old age and medical comorbidities. In the current combined antiretroviral therapy (cART) era, a significant portion of people living with HIV-1 (PLWH) with controlled viremia is older and with comorbidities, making these people vulnerable to SARS-CoV-2 infection and COVID-19-associated severe outcomes. Additionally, SARS-CoV-2 is neurotropic and causes neurological complications, resulting in a health burden and an adverse impact on PLWH and exacerbating HIV-1-associated neurocognitive disorder (HAND). The impact of SARS-CoV-2 infection and COVID-19 severity on neuroinflammation, the development of HAND and preexisting HAND is poorly explored. In the present review, we compiled the current knowledge of differences and similarities between SARS-CoV-2 and HIV-1, the conditions of the SARS-CoV-2/COVID-19 and HIV-1/AIDS syndemic and their impact on the central nervous system (CNS). Risk factors of COVID-19 on PLWH and neurological manifestations, inflammatory mechanisms leading to the neurological syndrome, the development of HAND, and its influence on preexisting HAND are also discussed. Finally, we have reviewed the challenges of the present syndemic on the world population, with a particular emphasis on PLWH.


Subject(s)
COVID-19 , HIV Infections , HIV Seropositivity , HIV-1 , Nervous System Diseases , Humans , COVID-19/complications , SARS-CoV-2 , Nervous System Diseases/epidemiology , Nervous System Diseases/etiology , Central Nervous System , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/epidemiology
3.
Biomedicines ; 11(4)2023 Mar 23.
Article in English | MEDLINE | ID: covidwho-2293366

ABSTRACT

Increasingly prevalent acute and chronic human brain diseases are scourges for the elderly. Besides the lack of therapies, these ailments share a neuroinflammation that is triggered/sustained by different innate immunity-related protein oligomers called inflammasomes. Relevant neuroinflammation players such as microglia/monocytes typically exhibit a strong NLRP3 inflammasome activation. Hence the idea that NLRP3 suppression might solve neurodegenerative ailments. Here we review the recent Literature about this topic. First, we update conditions and mechanisms, including RNAs, extracellular vesicles/exosomes, endogenous compounds, and ethnic/pharmacological agents/extracts regulating NLRP3 function. Second, we pinpoint NLRP3-activating mechanisms and known NLRP3 inhibition effects in acute (ischemia, stroke, hemorrhage), chronic (Alzheimer's disease, Parkinson's disease, Huntington's disease, MS, ALS), and virus-induced (Zika, SARS-CoV-2, and others) human brain diseases. The available data show that (i) disease-specific divergent mechanisms activate the (mainly animal) brains NLRP3; (ii) no evidence proves that NLRP3 inhibition modifies human brain diseases (yet ad hoc trials are ongoing); and (iii) no findings exclude that concurrently activated other-than-NLRP3 inflammasomes might functionally replace the inhibited NLRP3. Finally, we highlight that among the causes of the persistent lack of therapies are the species difference problem in disease models and a preference for symptomatic over etiologic therapeutic approaches. Therefore, we posit that human neural cell-based disease models could drive etiological, pathogenetic, and therapeutic advances, including NLRP3's and other inflammasomes' regulation, while minimizing failure risks in candidate drug trials.

4.
Intensive Care Med Exp ; 11(1): 9, 2023 Feb 24.
Article in English | MEDLINE | ID: covidwho-2273755

ABSTRACT

BACKGROUND: Divergence between deterioration to life-threatening COVID-19 or clinical improvement occurs for most within the first 14 days of symptoms. Life-threatening COVID-19 shares clinical similarities with Macrophage Activation Syndrome, which can be driven by elevated Free Interleukin-18 (IL-18) due to failure of negative-feedback release of IL-18 binding protein (IL-18bp). We, therefore, designed a prospective, longitudinal cohort study to examine IL-18 negative-feedback control in relation to COVID-19 severity and mortality from symptom day 15 onwards. METHODS: 662 blood samples, matched to time from symptom onset, from 206 COVID-19 patients were analysed by enzyme-linked immunosorbent assay for IL-18 and IL-18bp, enabling calculation of free IL-18 (fIL-18) using the updated dissociation constant (Kd) of 0.05 nmol. Adjusted multivariate regression analysis was used to assess the relationship between highest fIL-18 and outcome measures of COVID-19 severity and mortality. Re-calculated fIL-18 values from a previously studied healthy cohort are also presented. RESULTS: Range of fIL-18 in COVID-19 cohort was 10.05-1157.7 pg/ml. Up to symptom day 14, mean fIL-18 levels increased in all patients. Levels in survivors declined thereafter, but remained elevated in non-survivors. Adjusted regression analysis from symptom day 15 onwards showed a 100 mmHg decrease in PaO2/FiO2 (primary outcome) for each 37.7 pg/ml increase in highest fIL-18 (p < 0.03). Per 50 pg/ml increase in highest fIL-18, adjusted logistic regression gave an odds-ratio (OR) for crude 60-day mortality of 1.41 (1.1-2.0) (p < 0.03), and an OR for death with hypoxaemic respiratory failure of 1.90 [1.3-3.1] (p < 0.01). Highest fIL-18 was associated also with organ failure in patients with hypoxaemic respiratory failure, with an increase of 63.67 pg/ml for every additional organ supported (p < 0.01). CONCLUSIONS: Elevated free IL-18 levels from symptom day 15 onwards are associated with COVID-19 severity and mortality. ISRCTN: #13450549; registration date: 30/12/2020.

5.
Arterioscler Thromb Vasc Biol ; 42(9): 1103-1112, 2022 09.
Article in English | MEDLINE | ID: covidwho-2285811

ABSTRACT

The activating interplay of thrombosis and inflammation (thromboinflammation) has been established as a major underlying pathway, driving not only cardiovascular disease but also autoimmune disease and most recently, COVID-19. Throughout the years, innate immune cells have emerged as important modulators of this process. As the most abundant white blood cell in humans, neutrophils are well-positioned to propel thromboinflammation. This includes their ability to trigger an organized cell death pathway with the release of decondensed chromatin structures called neutrophil extracellular traps. Decorated with histones and cytoplasmic and granular proteins, neutrophil extracellular traps exert cytotoxic, immunogenic, and prothrombotic effects accelerating disease progression. Distinct steps leading to extracellular DNA release (NETosis) require the activities of PAD4 (protein arginine deiminase 4) catalyzing citrullination of histones and are supported by neutrophil inflammasome. By linking the immunologic function of neutrophils with the procoagulant and proinflammatory activities of monocytes and platelets, PAD4 activity holds important implications for understanding the processes that fuel thromboinflammation. We will also discuss mechanisms whereby vascular occlusion in thromboinflammation depends on the interaction of neutrophil extracellular traps with ultra-large VWF (von Willebrand Factor) and speculate on the importance of PAD4 in neutrophil inflammasome assembly and neutrophil extracellular traps in thromboinflammatory diseases including atherosclerosis and COVID-19.


Subject(s)
Atherosclerosis , COVID-19 , Extracellular Traps , Thrombosis , Atherosclerosis/metabolism , Extracellular Traps/metabolism , Histones/metabolism , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Thromboinflammation , Thrombosis/etiology , Thrombosis/metabolism , von Willebrand Factor/metabolism
6.
J Interferon Cytokine Res ; 42(8): 406-420, 2022 08.
Article in English | MEDLINE | ID: covidwho-2282941

ABSTRACT

The coronavirus 2019 disease (COVID-19) pandemic has caused massive morbidity and mortality worldwide. In severe cases, it is mainly associated with acute pneumonia, cytokine storm, and multi-organ dysfunction. Inflammasomes play a primary role in various pathological processes such as infection, injury, and cancer. However, their role in COVID-19-related complications has not been explored. In addition, the role of underlying medical conditions on COVID-19 disease severity remains unclear. Therefore, this review expounds on the mechanisms of inflammasomes following COVID-19 infection and provides recent evidence on the potential double-edged sword effect of inflammasomes during COVID-19 pathogenesis. The assembly and activation of inflammasomes are critical for inducing effective antiviral immune responses and disease resolution. However, uncontrolled activation of inflammasomes causes excessive production of proinflammatory cytokines (cytokine storm), increased risk of acute respiratory distress syndrome, and death. Therefore, discoveries in the role of the inflammasome in mediating organ injury are key to identifying therapeutic targets and treatment modifications to prevent or reduce COVID-19-related complications.


Subject(s)
COVID-19 , Cytokine Release Syndrome , Cytokines , Humans , Inflammasomes , SARS-CoV-2
7.
Scandinavian Journal of Immunology ; 97(2), 2023.
Article in English | ProQuest Central | ID: covidwho-2230244

ABSTRACT

SARS‐CoV‐2 is the causative agent of coronavirus disease 2019 (COVID‐19). The disease presents different degrees of severity related to the antiviral response of the host. According to clinical manifestations, patients could show mild, moderate and severe COVID‐19. Regarding immunological aspects, an increased interferon (IFN) response in COVID‐19 patients with mild and moderate symptoms were observed;however, in severe COVID‐19, IFN response is decreased. Patients with severe COVID‐19 display a hyperinflammatory disorder that leads to acute respiratory distress syndrome. Interestingly, the expression and activation of AIM2, a receptor induced by IFN, play an important role in the onset of antiviral response. In this review, we discuss the possible role of AIM2 during SARS‐CoV‐2 infection. We summarize the studies reporting the expression and activity of proteins involved upstream and downstream of AIM2‐inflammasome activation, such as IFN, ASC, Caspase‐1, IL‐1β, IL‐18, free‐dsDNA, IFI16, as well as SARS‐CoV‐2 viral load, cell death in groups of COVID‐19 patients with different clinical outcomes to infer the possible contribution of AIM2 in antiviral response of SARS‐CoV‐2 infection.

8.
Anales de la Real Academia Nacional de Medicina ; 139(2):135-139, 2022.
Article in Spanish | ProQuest Central | ID: covidwho-2229960

ABSTRACT

La Melatonina es una hormona que actúa facilitando la aparición del sueño fisiológico. Además presenta potentes acciónes antiinflamatoria y antioxidante, con lo que ha demostrado ya ser capaz de ejercer efectos muy beneficioso sobre las alteraciones ligadas al envejecimiento que aparecen en el sistema cardiovascular y especialmente en pulmón, donde nuestro grupo ha podido constatar un efecto protector frente a procesos de estrés oxidativo , inflamatorios y de muerte celular programada ( apoptosis).. Aunque no es una sustancia antivírica, sin embargo ha demostrado tener efectos muy positivos en algunos modelos experimentales de infección por vírus disminuyendo la carga viral y también reduciendo la oxidación y la inflamación con lo que atenúa la gravedad de la enfermedad. En el COVID 19 es capaz también de interferir en el proceso infectivo que ocurre a través de los receptores de ACE2 y de EGF pues es capaz de bloquear dichas interacciones con lo que disminuye la viremia. Concretamente reduce la actividad del inflamasoma NLRP3 con lo que bloquea la liberación masiva de citoquinas disminuyendo el proceso inflamatorio lo que supone una mejoría de la evolución de la enfermedad. Por todo ello la melatonina puede desempeñar un importante papel en el tratamiento del COVID 19.Alternate : Melatonin is a hormone that acts facilitating the appearance of physiological sleep It has also a very evident antinflammatory and antioxidant capacities that result in beneficial actions on the aging processes in the cardiovascular system and in the lungs where our group has detected a protective action against oxidative stress , inflammation and apoptosis . Although melatonin is not viricidal by itself in some models of viral infections it has demonstrated its ability to reduce viral load and also inflammation and oxidation, reducing the severity of the disease. In COVID 19 melatonin has been shown to be able to interfere with the infectious process that takes place through ACE2 and EGF receptors being able to block these interactions thus reducing viremia .It is able to block the activation of the NLRP3 inflammasome thus dramatically reducing the massive secretion of cytokines and markedly reducing hyperinflammation and apoptosis leading to a better evolution of the disease .For all these reasons melatonin could play an important role in the treatment of COVID 19.

9.
Sci Afr ; 18: e01407, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2082654

ABSTRACT

Severe SARS-CoV-2 infection causes systemic inflammation, cytokine storm, and hypercytokinemia due to activation of the release of pro-inflammatory cytokines that have been associated with case-fatality rate. The immune overreaction and cytokine storm in the infection caused by SARS-CoV-2 may be linked to NLRP3 inflammasome activation which has supreme importance in human innate immune response mainly against viral infections. In SARS-CoV-2 infection, NLRP3 inflammasome activation results in the stimulation and synthesis of natural killer cells (NKs), NFκB, and interferon-gamma (INF-γ), while inhibiting IL-33 expression. Various efforts have identified selective inhibitors of NLRP3 inflammasome. To achieve this, studies are exploring the screening of natural compounds and/or repurposing of clinical drugs to identify potential NLRP3 inhibitors. NLRP3 inflammasome inhibitors are expected to suppress exaggerated immune reaction and cytokine storm-induced-organ damage in SARS-CoV-2 infection. Therefore, NLRP3 inflammasome inhibitors could mitigate the immune-overreaction and hypercytokinemia in Covid-19 infection.

10.
Int J Biol Sci ; 18(15): 5827-5848, 2022.
Article in English | MEDLINE | ID: covidwho-2056218

ABSTRACT

The rapid dissemination of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), remains a global public health emergency. The host immune response to SARS-CoV-2 plays a key role in COVID-19 pathogenesis. SARS-CoV-2 can induce aberrant and excessive immune responses, leading to cytokine storm syndrome, autoimmunity, lymphopenia, neutrophilia and dysfunction of monocytes and macrophages. Pyroptosis, a proinflammatory form of programmed cell death, acts as a host defense mechanism against infections. Pyroptosis deprives the replicative niche of SARS-CoV-2 by inducing the lysis of infected cells and exposing the virus to extracellular immune attack. Notably, SARS-CoV-2 has evolved sophisticated mechanisms to hijack this cell death mode for its own survival, propagation and shedding. SARS-CoV-2-encoded viral products act to modulate various key components in the pyroptosis pathways, including inflammasomes, caspases and gasdermins. SARS-CoV-2-induced pyroptosis contriubtes to the development of COVID-19-associated immunopathologies through leakage of intracellular contents, disruption of immune system homeostasis or exacerbation of inflammation. Therefore, pyroptosis has emerged as an important mechanism involved in COVID-19 immunopathogenesis. However, the entangled links between pyroptosis and SARS-CoV-2 pathogenesis lack systematic clarification. In this review, we briefly summarize the characteristics of SARS-CoV-2 and COVID-19-related immunopathologies. Moreover, we present an overview of the interplay between SARS-CoV-2 infection and pyroptosis and highlight recent research advances in the understanding of the mechanisms responsible for the implication of the pyroptosis pathways in COVID-19 pathogenesis, which will provide informative inspirations and new directions for further investigation and clinical practice. Finally, we discuss the potential value of pyroptosis as a therapeutic target in COVID-19. An in-depth discussion of the underlying mechanisms of COVID-19 pathogenesis will be conducive to the identification of potential therapeutic targets and the exploration of effective treatment measures aimed at conquering SARS-CoV-2-induced COVID-19.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Pyroptosis , Inflammasomes , Caspases
11.
Sci Afr ; : e01084, 2021 Dec 18.
Article in English | MEDLINE | ID: covidwho-1586561

ABSTRACT

Severe SARS-CoV-2 infection causes systemic inflammation, cytokine storm and hypercytokinemia due to activation the release of pro-inflammatory cytokines that have been associated with case-fatality rate. The immune overreaction and cytokine storm in the infection caused by SARS-CoV-2 may be linked to NLRP3 inflammasome activation which has supreme importance in human innate immune response mainly against viral infections. In SARS-CoV-2 infection, NLRP3 inflammasome activation results in the stimulation and synthesis of natural killer cells (NKs), NFκB, and interferon gamma (INF-γ), while inhibiting IL-33 expression. Various efforts have identified selective inhibitors of NLRP3 inflammasome. To achieve this, studies are exploring the screening of natural compounds and/or repurposing of clinical drugs to identify potential NLRP3 inhibitors. NLRP3 inflammasome inhibitors are expected to suppress exaggerated immune reaction and cytokine storm induced-organ damage in SARS-CoV-2 infection. Therefore, NLRP3 inflammasome inhibitors could mitigate the immune-overreaction and hypercytokinemia in Covid-19 infection.

12.
Viruses ; 13(12)2021 12 14.
Article in English | MEDLINE | ID: covidwho-1572670

ABSTRACT

SARS-CoV-2 is a new type of coronavirus that has caused worldwide pandemic. The disease induced by SARS-CoV-2 is called COVID-19. A majority of people with COVID-19 have relatively mild respiratory symptoms. However, a small percentage of COVID-19 patients develop a severe disease where multiple organs are affected. These severe forms of SARS-CoV-2 infections are associated with excessive production of pro-inflammatory cytokines, so called "cytokine storm". Inflammasomes, which are protein complexes of the innate immune system orchestrate development of local and systemic inflammation during virus infection. Recent data suggest involvement of inflammasomes in severe COVID-19. Activation of inflammasome exerts two major effects: it activates caspase-1-mediated processing and secretion of pro-inflammatory cytokines IL-1ß and IL-18, and induces inflammatory cell death, pyroptosis, via protein called gasdermin D. Here, we provide comprehensive review of current understanding of the activation and possible functions of different inflammasome structures during SARS-CoV-2 infection and compare that to response caused by influenza A virus. We also discuss how novel SARS-CoV-2 mRNA vaccines activate innate immune response, which is a prerequisite for the activation of protective adaptive immune response.


Subject(s)
COVID-19/immunology , Inflammasomes/immunology , Adaptive Immunity , COVID-19 Vaccines , Cell Death , Cytokine Release Syndrome , Cytokines/immunology , Humans , Immunity, Innate , Inflammation , Interleukin-18 , Interleukin-1beta , Neoplasm Proteins , Pyroptosis , SARS-CoV-2/immunology , mRNA Vaccines
13.
Microorganisms ; 9(11)2021 Nov 17.
Article in English | MEDLINE | ID: covidwho-1524079

ABSTRACT

Inflammasomes are cytoplasmic multiprotein complexes formed by the host's immune system as a response to microbial infection and cellular damage. Many studies have revealed various regulators of NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation, while it has been recently shown that NLRP3 is implicated in COVID-19 pathogenesis. At the same time, probiotics counteract the inflammatory process and modulate cytokine release, thus influencing both innate and adaptive immune systems. Herein, we review the immunomodulatory potential of probiotics on the assembly of NLRP3 inflammasome, as well as the pathophysiological mechanisms supporting the use of probiotic bacteria for SARS-CoV-2 infection management, presenting evidence from preclinical studies of the last decade: in vivo, ex vivo, and mixed trials. Data show that probiotics intake is related to NLRP3 inflammasome attenuation and lower levels of inflammation markers, highlighting the beneficial effects of probiotics on inflammatory conditions. Currently, none of the ongoing clinical trials evaluating the effectiveness of probiotics intake in humans with COVID-19 has been completed. However, evidence from preclinical studies indicates that probiotics may block virus invasion and replication through their metabolites, bacteriocins, and their ability to block Angiotensin-Converting Enzyme 2 (ACE2), and by stimulating the immune response through NLRP3 inflammasome regulation. In this review, the beneficial effects of probiotics in the inflammatory process through NLRP3 inflammasome attenuation are presented. Furthermore, probiotics may target SARS-CoV-2 both by blocking virus invasion and replication and by stimulating the immune response through NLRP3 inflammasome regulation. Heterogeneity of the results-due to, among others, different bacterial strains and their metabolites, forms, dosage, and experimental designs-indicates the need for more extensive research.

14.
J Reprod Immunol ; 148: 103427, 2021 11.
Article in English | MEDLINE | ID: covidwho-1433587

ABSTRACT

To overcome COVID-19 long-term consequences, one possible approach is to control inflammasomes activation, because SARS-CoV-2 can induce humoral and cellular immune responses. In this opinion article we hypothesized that if it is proven with convincing and unmistakable evidence that firstly, SARS-CoV-2 can enter cells and damage them through its common receptors in the reproductive tissues, and secondly, inflammasome pathway activation is responsible for the damages caused, then the inflammasome inhibitors might be considered as suitable candidates in preventing the pathological effects on the germ cells and reproductive tissues and subsequent fertility.


Subject(s)
COVID-19/complications , Infertility/virology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Angiotensin-Converting Enzyme 2 , COVID-19/immunology , Fertility , Humans
15.
Viral Immunol ; 34(3): 190-200, 2021 04.
Article in English | MEDLINE | ID: covidwho-1099573

ABSTRACT

The initial immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) includes an interferon-dependent antiviral response. A late and uncontrolled inflammatory response characterized by high activity of proinflammatory cytokines and the recruitment of neutrophils and macrophages develops in predisposed individuals and is potentially harmful in some cases. Interleukin (IL)-17 is one of the many cytokines released during coronavirus disease 2019 (COVID-19). IL-17 is crucial in recruiting and activating neutrophils, cells that can migrate to the lung, and are heavily involved in the pathogenesis of COVID-19. During the infection T helper 17 (Th17) cells and IL-17-related pathways are associated with a worse outcome of the disease. All these have practical consequences considering that some drugs with therapeutic targets related to the Th17 response may have a beneficial effect on patients with SARS-CoV-2 infection. Herein, we present the arguments underlying our assumption that blocking the IL-23/IL-17 axis using targeted biological therapies as well as drugs that act indirectly on this pathway such as convalescent plasma therapy and colchicine may be good therapeutic options.


Subject(s)
COVID-19/immunology , SARS-CoV-2/immunology , Th17 Cells/immunology , Adaptive Immunity , Adult , COVID-19/classification , Humans , Immunity, Innate , Interleukin-17/antagonists & inhibitors , Interleukin-17/physiology , Interleukin-23/antagonists & inhibitors , Middle Aged , COVID-19 Drug Treatment
16.
Nutrients ; 13(1)2020 Dec 25.
Article in English | MEDLINE | ID: covidwho-1016213

ABSTRACT

Inflammasomes are intracellular protein complexes that form in response to a variety of stress signals and that serve to catalyze the proteolytic conversion of pro-interleukin-1ß and pro-interleukin-18 to active interleukin-1ß and interleukin-18, central mediators of the inflammatory response; inflammasomes can also promote a type of cell death known as pyroptosis. The NLRP3 inflammasome has received the most study and plays an important pathogenic role in a vast range of pathologies associated with inflammation-including atherosclerosis, myocardial infarction, the complications of diabetes, neurological and autoimmune disorders, dry macular degeneration, gout, and the cytokine storm phase of COVID-19. A consideration of the molecular biology underlying inflammasome priming and activation enables the prediction that a range of nutraceuticals may have clinical potential for suppressing inflammasome activity-antioxidants including phycocyanobilin, phase 2 inducers, melatonin, and N-acetylcysteine, the AMPK activator berberine, glucosamine, zinc, and various nutraceuticals that support generation of hydrogen sulfide. Complex nutraceuticals or functional foods featuring a number of these agents may find utility in the prevention and control of a wide range of medical disorders.


Subject(s)
Antioxidants/therapeutic use , COVID-19 , Dietary Supplements , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , SARS-CoV-2/metabolism , Animals , COVID-19/diet therapy , COVID-19/metabolism , COVID-19/pathology , Humans
17.
Naunyn Schmiedebergs Arch Pharmacol ; 394(5): 997-1001, 2021 05.
Article in English | MEDLINE | ID: covidwho-1014115

ABSTRACT

Chloroquine and hydroxychloroquine belong to the aminoquinoline drugs. Studies revealed that chloroquine and hydroxychloroquine shows antagonism activity against COVID-19 under laboratory conditions. ARDS and ALI are conditions that occur in patients with COVID-19 as the main pathological complications of cytokine storm. Inflammasomes play a key role in the pathogenesis of many diseases associated with destructive inflammation. NLRP3 inflammasome has been shown to play a key role in the pathogenesis of viral diseases. The possible role of NLRP3 inflammasome inhibitors in the treatment of COVID-19 has been considered. We surveyed the potential inhibitory effect of chloroquine and hydroxychloroquine on inflammasome. Studies indicate that one of the possible anti-inflammatory mechanisms of chloroquine and hydroxychloroquine is inhibition of the activity of NLRP3 inflammasome.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19 Drug Treatment , Chloroquine/therapeutic use , Hydroxychloroquine/therapeutic use , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Cytokine Release Syndrome , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
18.
Biochem Pharmacol ; 183: 114316, 2021 01.
Article in English | MEDLINE | ID: covidwho-898506

ABSTRACT

Pattern recognition receptors (PRRs) and inflammasomes are a key part of the anti-viral innate immune system as they detect conserved viral pathogen-associated molecular patterns (PAMPs). A successful host response to viral infections critically depend on the initial activation of PRRs by viruses, mainly by viral DNA and RNA. The signalling pathways activated by PRRs leads to the expression of pro-inflammatory cytokines, to recruit immune cells, and type I and type III interferons which leads to the induction of interferon stimulated genes (ISG), powerful virus restriction factors that establish the "antiviral state". Inflammasomes contribute to anti-viral responses through the maturation of interleukin (IL)-1 and IL-18 and through triggering pyroptotic cell death. The activity of the innate immune system along with the adaptive immune response normally leads to successful virus elimination, although disproportionate innate responses contribute to viral pathology. In this review we will discuss recent insights into the influence of PRR activation and inflammasomes on viral infections and what this means for the mammalian host. We will also comment on how specific PRRs and inflammasomes may be relevant to how SARS-CoV-2, the virus responsible for the current COVID-19 pandemic, interacts with host innate immunity.


Subject(s)
Immunity, Innate/immunology , Inflammasomes/immunology , SARS-CoV-2/immunology , Virus Diseases/immunology , Animals , Humans , Inflammasomes/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , SARS-CoV-2/metabolism , Virus Diseases/diagnosis , Virus Diseases/metabolism
19.
Int J Biochem Cell Biol ; 122: 105738, 2020 05.
Article in English | MEDLINE | ID: covidwho-824867

ABSTRACT

Viroporins are virus encoded proteins that alter membrane permeability and can trigger subsequent cellular signals. Oligomerization of viroporin subunits results in formation of a hydrophilic pore which facilitates ion transport across host cell membranes. These viral channel proteins may be involved in different stages of the virus infection cycle. Inflammasomes are large multimolecular complexes best recognized for their ability to control activation of caspase-1, which in turn regulates the maturation of interleukin-1 ß (IL-1ß) and interleukin 18 (IL-18). IL-1ß was originally identified as a pro-inflammatory cytokine able to induce both local and systemic inflammation and a febrile reaction in response to infection or injury. Excessive production of IL-1ß is associated with autoimmune and inflammatory diseases. Microbial derivatives, bacterial pore-forming toxins, extracellular ATP and other pathogen-associated molecular patterns trigger activation of NLRP3 inflammasomes. Recent studies have reported that viroporin activity is capable of inducing inflammasome activity and production of IL-1ß, where NLRP3 is shown to be regulated by fluxes of K+, H+ and Ca2+ in addition to reactive oxygen species, autophagy and endoplasmic reticulum stress. The aim of this review is to present an overview of the key findings on viroporin activity with special emphasis on their role in virus immunity and as possible activators of inflammasomes.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , Inflammation/virology , Viral Proteins/immunology , Virus Diseases/immunology , Animals , Humans , Immunity, Innate , Inflammasomes/metabolism , Inflammation/metabolism , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Viral Proteins/metabolism , Virus Diseases/metabolism , Viruses/immunology , Viruses/metabolism
20.
Mult Scler Relat Disord ; 46: 102540, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-803007

ABSTRACT

A long-term neurologic sequela arising from COVID-19 infection in multiple sclerosis (MS) patients could be related both to the increase of cytokines and the activation of NLRP3 inflammasome by the Sars-CoV2. These two mechanisms may cause a worsening of MS several months after the resolution of the infection.


Subject(s)
COVID-19/virology , Inflammasomes/metabolism , Multiple Sclerosis/complications , SARS-CoV-2/pathogenicity , COVID-19/complications , COVID-19/metabolism , Disease Progression , Humans , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Multiple Sclerosis/virology
SELECTION OF CITATIONS
SEARCH DETAIL