Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
mBio ; 13(3): e0040122, 2022 06 28.
Article in English | MEDLINE | ID: covidwho-1854237

ABSTRACT

Influenza viruses cause respiratory tract infections, which lead to human disease outbreaks and pandemics. Influenza A virus (IAV) circulates in diverse animal species, predominantly aquatic birds. This often results in the emergence of novel viral strains causing severe human disease upon zoonotic transmission. Innate immune sensing of the IAV infection promotes host cell death and inflammatory responses to confer antiviral host defense. Dysregulated respiratory epithelial cell death and excessive proinflammatory responses drive immunopathology in highly pathogenic influenza infections. Here, we discuss the critical mechanisms regulating IAV-induced cell death and proinflammatory responses. We further describe the essential role of the Z-form nucleic acid sensor ZBP1/DAI and RIPK3 in triggering apoptosis, necroptosis, and pyroptosis during IAV infection and their impact on host defense and pathogenicity in vivo. We also discuss the functional importance of ZBP1-RIPK3 signaling in recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other viral infections. Understanding these mechanisms of RNA virus-induced cytopathic and pathogenic inflammatory responses is crucial for targeting pathogenic lung infections and human respiratory illness.


Subject(s)
COVID-19 , Influenza A virus , Influenza, Human , Pneumonia , Animals , Cell Death , Humans , Influenza A virus/physiology , RNA , RNA-Binding Proteins/genetics , SARS-CoV-2
2.
Crit Rev Immunol ; 41(3): 43-56, 2021.
Article in English | MEDLINE | ID: covidwho-1753247

ABSTRACT

Pathogenic coronaviruses (CoVs) have caused human respiratory infections and severe disease outbreaks in the past two decades. Recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans shows high transmissibility causing a wide range of clinical outcomes, named coronavirus disease-2019 (COVID-19), which emerged into an ongoing pandemic. Innate immune sensing of SARS-CoV-2 infection is critical for mounting antiviral and inflammatory responses to restrict the viral spread and initiate lung tissue repair processes. However, excessive cytokine and chemokine levels and dysregulated inflammatory immune cell function in the lungs are associated with respiratory failure and severe COVID-19. Thus, there is a tremendous need for understanding SARS-CoV-2-host interactions determining the aberrant inflammatory responses and loss of respiratory function. In this article, we discuss host innate immune responses determining dysregulated inflammation and immunopathology during SARS-CoV-2 infection. We also provide the perspective for the inflammatory cell death contribution for this immunopathology. Virus-induced acute host responses are complex, and elucidating this complex mechanism facilitates safe therapeutic interventions to alleviate inflammation-mediated immunopathology during pathogenic virus infections.


Subject(s)
COVID-19 , Immunity, Innate , Inflammation , SARS-CoV-2 , Cell Death , Cytokines/metabolism , Humans , Lung , SARS-CoV-2/pathogenicity
3.
J Leukoc Biol ; 111(2): 497-508, 2022 02.
Article in English | MEDLINE | ID: covidwho-1669515

ABSTRACT

Coronaviruses (CoVs) are RNA viruses that cause human respiratory infections. Zoonotic transmission of the SARS-CoV-2 virus caused the recent COVID-19 pandemic, which led to over 2 million deaths worldwide. Elevated inflammatory responses and cytotoxicity in the lungs are associated with COVID-19 severity in SARS-CoV-2-infected individuals. Bats, which host pathogenic CoVs, operate dampened inflammatory responses and show tolerance to these viruses with mild clinical symptoms. Delineating the mechanisms governing these host-specific inflammatory responses is essential to understand host-virus interactions determining the outcome of pathogenic CoV infections. Here, we describe the essential role of inflammasome activation in determining COVID-19 severity in humans and innate immune tolerance in bats that host several pathogenic CoVs. We further discuss mechanisms leading to inflammasome activation in human SARS-CoV-2 infection and how bats are molecularly adapted to suppress these inflammasome responses. We also report an analysis of functionally important residues of inflammasome components that provide new clues of bat strategies to suppress inflammasome signaling and innate immune responses. As spillover of bat viruses may cause the emergence of new human disease outbreaks, the inflammasome regulation in bats and humans likely provides specific strategies to combat the pathogenic CoV infections.


Subject(s)
COVID-19/pathology , Immune Tolerance , Immunity, Innate , Inflammasomes/immunology , SARS-CoV-2/immunology , Animals , COVID-19/immunology , COVID-19/virology , Chiroptera , Humans , Inflammasomes/metabolism , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL