Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Article in English | MEDLINE | ID: mdl-38696536

ABSTRACT

Major 5'-terminally deleted (5'TD) RNA forms of group-B coxsackievirus (CVB-5'TD) has been associated with myocarditis in both mice and humans. Although it is known that interferon-ß (IFN-ß) signaling is critical for an efficient innate immune response against CVB-induced myocarditis, the link between CVB-5'TD RNA forms and type I IFN signaling in cardiomyocytes remains to be explored. In a mouse model of CVB3/28-induced myocarditis, major early-emerging forms of CVB-5'TD RNA have been characterized as replicative viral populations that impair IFN-ß production in the heart. Synthetic CVB3/28 RNA forms mimicking each of these major 5'TD virus populations were transfected in mice and have been shown to modulate innate immune responses in the heart and to induce myocarditis in mice. Remarkably, transfection of synthetic viral RNA with deletions in the secondary structures of the 5'-terminal CVB3 RNA domain I, modifying stem-loops "b", "c" or "d", were found to impair IFN-ß production in human cardiomyocytes. In addition, the activation of innate immune response by Poly(I:C), was found to restore IFN-ß production and to reduce the burden of CVB-5'TD RNA-forms in cardiac tissues, thereby reducing the mortality rate of infected mice. Overall, our results indicate that major early-emerging CVB3 populations deleted in the domain I of genomic RNA, in the 5' noncoding region, modulate the activation of the type I IFN pathway in cardiomyocytes and induce myocarditis in mice. These findings shed new light on the role of replicative CVB-5'TD RNA forms as key pathophysiological factors in CVB-induced human myocarditis.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , Interferon Type I , Myocarditis , Myocytes, Cardiac , RNA, Viral , Myocarditis/virology , Myocarditis/immunology , Myocarditis/genetics , Animals , Myocytes, Cardiac/virology , Myocytes, Cardiac/metabolism , Mice , Enterovirus B, Human/immunology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Coxsackievirus Infections/genetics , Interferon Type I/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , Immunity, Innate , Signal Transduction , Interferon-beta/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Male , 5' Untranslated Regions
2.
J Med Virol ; 95(1): e28361, 2023 01.
Article in English | MEDLINE | ID: mdl-36451263

ABSTRACT

We assessed relationships between early peripheral blood type I interferons (IFN) levels, clinical new early warning scores (NEWS), and clinical outcomes in hospitalized coronavirus disease-19 (COVID-19) adult patients. Early IFN-ß levels were lower among patients who further required intensive care unit (ICU) admission than those measured in patients who did not require an ICU admission during severe acute respiratory syndrome coronavirus type 2 infection. IFN-ß levels were inversely correlated with NEWS only in the subgroup of patients who further required ICU admission. To assess whether peripheral blood IFN-ß levels could be a potential relevant biomarker to predict further need for ICU admission, we performed receiver operating characteristic (ROC) curve analyses that showed for all study patients an area under ROC curve of 0.77 growing to 0.86 (p = 0.003) when the analysis was restricted to a subset of patients with NEWS ≥5 at the time of hospital admission. Overall, our findings indicated that early peripheral blood IFN-ß levels might be a relevant predictive marker of further need for an ICU admission in hospitalized COVID-19 adult patients, specifically when clinical score (NEWS) was graded as upper than 5 at the time of hospital admission.


Subject(s)
COVID-19 , Early Warning Score , Interferon-beta , Adult , Humans , COVID-19/diagnosis , Hospitalization , Intensive Care Units , Interferon-beta/blood , Interferon-beta/chemistry , Retrospective Studies , ROC Curve , Prognosis , Biomarkers
3.
Viruses ; 14(12)2022 12 13.
Article in English | MEDLINE | ID: mdl-36560784

ABSTRACT

Emergence of 5' terminally deleted coxsackievirus-B RNA forms (CVB-TD) have been associated with the development of human diseases. These CVB-TD RNA forms have been detected in mouse pancreas during acute or persistent experimental infections. To date, the impact of the replication activities of CVB-TD RNA forms on insulin metabolism remains unexplored. Using an immunocompetent mouse model of CVB3/28 infection, acute and persistent infections of major CVB-TD populations were evidenced in the pancreas. The inoculation of mice with homogenized pancreases containing major CVB-TD populations induced acute and chronic pancreatic infections with pancreatitis. In the mouse pancreas, viral capsid protein 1 (VP1) expression colocalized with a decrease in beta cells insulin content. Moreover, in infected mouse pancreases, we showed a decrease in pro-hormone convertase 2 (PCSK2) mRNA, associated with a decrease in insulin plasmatic concentration. Finally, transfection of synthetic CVB-TD50 RNA forms into cultured rodent pancreatic beta cells demonstrated that viral replication with protein synthesis activities decreased the PCSK2 mRNA expression levels, impairing insulin secretion. In conclusion, our results show that the emergence and maintenance of major CVB-TD RNA replicative forms in pancreatic beta cells can play a direct, key role in the pathophysiological mechanisms leading to the development of type 1 diabetes.


Subject(s)
Coxsackievirus Infections , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Mice , Humans , Animals , Insulin/metabolism , RNA/metabolism , Enterovirus B, Human/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Virus Replication , Proprotein Convertase 2/metabolism
4.
Sci Rep ; 11(1): 20615, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663858

ABSTRACT

Differential kinetics of RNA loads and infectious viral levels in the upper respiratory tract between asymptomatic and symptomatic SARS-CoV-2 infected adult outpatients remain unclear limiting recommendations that may guide clinical management, infection control measures and occupational health decisions. In the present investigation, 496 (2.8%) of 17,911 French adult outpatients were positive for an upper respiratory tract SARS-CoV-2 RNA detection by a quantitative RT-PCR assay, of which 180 (36.3%) were COVID-19 asymptomatic. Of these adult asymptomatic viral shedders, 75% had mean to high RNA viral loads (Ct values < 30) which median value was significantly higher than that observed in symptomatic subjects (P = 0.029), and 50.6% were positive by cell culture assays of their upper respiratory tract specimens. Our findings indicate that COVID-19 asymptomatic adult outpatients are significant viable SARS-CoV-2 shedders in their upper respiratory tract playing a major potential role as SARS-CoV-2 transmitters in various epidemiological transmission chains, promoting COVID-19 resurgence in populations.


Subject(s)
COVID-19/therapy , COVID-19/virology , Outpatients , SARS-CoV-2 , Virus Shedding , Adolescent , Adult , Animals , Asymptomatic Infections , COVID-19 Nucleic Acid Testing , Chlorocebus aethiops , Female , France , Humans , Kinetics , Male , Middle Aged , RNA, Viral , Respiratory System/metabolism , Vero Cells , Viral Load , Young Adult
5.
Viruses ; 12(9)2020 08 21.
Article in English | MEDLINE | ID: mdl-32839386

ABSTRACT

Group-B enteroviruses (EV-B) are ubiquitous naked single-stranded positive RNA viral pathogens that are responsible for common acute or persistent human infections. Their genome is composed in the 5' end by a non-coding region, which is crucial for the initiation of the viral replication and translation processes. RNA domain-I secondary structures can interact with viral or cellular proteins to form viral ribonucleoprotein (RNP) complexes regulating viral genomic replication, whereas RNA domains-II to -VII (internal ribosome entry site, IRES) are known to interact with cellular ribosomal subunits to initiate the viral translation process. Natural 5' terminally deleted viral forms lacking some genomic RNA domain-I secondary structures have been described in EV-B induced murine or human infections. Recent in vitro studies have evidenced that the loss of some viral RNP complexes in the RNA domain-I can modulate the viral replication and infectivity levels in EV-B infections. Moreover, the disruption of secondary structures of RNA domain-I could impair viral RNA sensing by RIG-I (Retinoic acid inducible gene I) or MDA5 (melanoma differentiation-associated protein 5) receptors, a way to overcome antiviral innate immune response. Overall, natural 5' terminally deleted viral genomes resulting in the loss of various structures in the RNA domain-I could be major key players of host-cell interactions driving the development of acute or persistent EV-B infections.


Subject(s)
Enterovirus B, Human/genetics , Enterovirus Infections/virology , RNA, Untranslated/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Animals , Enterovirus B, Human/physiology , Genome, Viral , Host-Pathogen Interactions , Humans , Interferons/metabolism , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Untranslated/chemistry , RNA, Untranslated/metabolism , RNA, Viral/metabolism , Signal Transduction , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...