Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 78
Filtrer
1.
J Exp Med ; 221(9)2024 09 02.
Article de Anglais | MEDLINE | ID: mdl-39058386

RÉSUMÉ

Autoantibody-mediated glomerulonephritis (AGN) arises from dysregulated renal inflammation, with urgent need for improved treatments. IL-17 is implicated in AGN and drives pathology in a kidney-intrinsic manner via renal tubular epithelial cells (RTECs). Nonetheless, downstream signaling mechanisms provoking kidney pathology are poorly understood. A noncanonical RNA binding protein (RBP), Arid5a, was upregulated in human and mouse AGN. Arid5a-/- mice were refractory to AGN, with attenuated myeloid infiltration and impaired expression of IL-17-dependent cytokines and transcription factors (C/EBPß, C/EBPδ). Transcriptome-wide RIP-Seq revealed that Arid5a inducibly interacts with conventional IL-17 target mRNAs, including CEBPB and CEBPD. Unexpectedly, many Arid5a RNA targets corresponded to translational regulation and RNA processing pathways, including rRNAs. Indeed, global protein synthesis was repressed in Arid5a-deficient cells, and C/EBPs were controlled at the level of protein rather than RNA accumulation. IL-17 prompted Arid5a nuclear export and association with 18S rRNA, a 40S ribosome constituent. Accordingly, IL-17-dependent renal autoimmunity is driven by Arid5a at the level of ribosome interactions and translation.


Sujet(s)
Autoanticorps , Protéines de liaison à l'ADN , Glomérulonéphrite , Interleukine-17 , Souris knockout , Facteurs de transcription , Animaux , Interleukine-17/métabolisme , Glomérulonéphrite/immunologie , Glomérulonéphrite/génétique , Glomérulonéphrite/métabolisme , Glomérulonéphrite/anatomopathologie , Humains , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Autoanticorps/immunologie , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Protéine delta liant les séquences stimulatrices de type CCAAT/métabolisme , Protéine delta liant les séquences stimulatrices de type CCAAT/génétique , Souris de lignée C57BL , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/immunologie , ARN messager/génétique , ARN messager/métabolisme
2.
Immunity ; 57(8): 1812-1827.e7, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-38955184

RÉSUMÉ

An important property of the host innate immune response during microbial infection is its ability to control the expression of antimicrobial effector proteins, but how this occurs post-transcriptionally is not well defined. Here, we describe a critical antibacterial role for the classic antiviral gene 2'-5'-oligoadenylate synthetase 1 (OAS1). Human OAS1 and its mouse ortholog, Oas1b, are induced by interferon-γ and protect against cytosolic bacterial pathogens such as Francisella novicida and Listeria monocytogenes in vitro and in vivo. Proteomic and transcriptomic analysis showed reduced IRF1 protein expression in OAS1-deficient cells. Mechanistically, OAS1 binds and localizes IRF1 mRNA to the rough endoplasmic reticulum (ER)-Golgi endomembranes, licensing effective translation of IRF1 mRNA without affecting its transcription or decay. OAS1-dependent translation of IRF1 leads to the enhanced expression of antibacterial effectors, such as GBPs, which restrict intracellular bacteria. These findings uncover a noncanonical function of OAS1 in antibacterial innate immunity.


Sujet(s)
2',5'-Oligoadenylate synthetase , Immunité innée , Facteur-1 de régulation d'interféron , 2',5'-Oligoadenylate synthetase/métabolisme , 2',5'-Oligoadenylate synthetase/génétique , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Animaux , Humains , Souris , Biosynthèse des protéines/immunologie , Listeria monocytogenes/immunologie , Souris knockout , Souris de lignée C57BL , Infections à Listeria/immunologie , Interféron gamma/métabolisme , Interféron gamma/immunologie
3.
Immunity ; 57(3): 446-461.e7, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38423012

RÉSUMÉ

In response to viral infection, how cells balance translational shutdown to limit viral replication and the induction of antiviral components like interferons (IFNs) is not well understood. Moreover, how distinct isoforms of IFN-induced oligoadenylate synthetase 1 (OAS1) contribute to this antiviral response also requires further elucidation. Here, we show that human, but not mouse, OAS1 inhibits SARS-CoV-2 replication through its canonical enzyme activity via RNase L. In contrast, both mouse and human OAS1 protect against West Nile virus infection by a mechanism distinct from canonical RNase L activation. OAS1 binds AU-rich elements (AREs) of specific mRNAs, including IFNß. This binding leads to the sequestration of IFNß mRNA to the endomembrane regions, resulting in prolonged half-life and continued translation. Thus, OAS1 is an ARE-binding protein with two mechanisms of antiviral activity: driving inhibition of translation but also a broader, non-canonical function of protecting IFN expression from translational shutdown.


Sujet(s)
2',5'-Oligoadenylate synthetase , Interférons , Oligoribonucléotides , Maladies virales , Fièvre à virus West Nile , Animaux , Humains , Souris , 2',5'-Oligoadenylate synthetase/génétique , 2',5'-Oligoadenylate synthetase/métabolisme , Nucléotides adényliques , Antiviraux/pharmacologie , Fièvre à virus West Nile/génétique , Fièvre à virus West Nile/métabolisme , Virus du Nil occidental/métabolisme , Virus du Nil occidental/pathogénicité
4.
bioRxiv ; 2023 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-37808873

RÉSUMÉ

Immune signaling needs to be well-regulated to promote clearance of pathogens, while preventing aberrant inflammation. Interferons (IFNs) and antiviral genes are activated by the detection of viral RNA by RIG-I-like receptors (RLRs). Signal transduction downstream of RLRs proceeds through a multi-protein complex organized around the central adaptor protein MAVS. Recent work has shown that protein complex function can be modulated by RNA molecules providing allosteric regulation or acting as molecular guides or scaffolds. Thus, we hypothesized that RNA plays a role in organizing MAVS signaling platforms. Here, we show that MAVS, through its central intrinsically disordered domain, directly interacts with the 3' untranslated regions of cellular mRNAs. Importantly, elimination of RNA by RNase treatment disrupts the MAVS signalosome, including newly identified regulators of RLR signaling, and inhibits phosphorylation of the transcription factor IRF3. This supports the hypothesis that RNA molecules scaffold proteins in the MAVS signalosome to induce IFNs. Together, this work uncovers a function for cellular RNA in promoting signaling through MAVS and highlights a generalizable principle of RNA regulatory control of cytoplasmic immune signaling complexes.

5.
Cell Rep ; 42(8): 112805, 2023 08 29.
Article de Anglais | MEDLINE | ID: mdl-37467105

RÉSUMÉ

Cellular stress in the form of disrupted transcription, loss of organelle integrity, or damage to nucleic acids can elicit inflammatory responses by activating signaling cascades canonically tasked with controlling pathogen infections. These stressors must be kept in check to prevent unscheduled activation of interferon, which contributes to autoinflammation. This study examines the role of the transcription factor myocyte enhancing factor 2A (MEF2A) in setting the threshold of transcriptional stress responses to prevent R-loop accumulation. Increases in R-loops lead to the induction of interferon and inflammatory responses in a DEAD-box helicase 41 (DDX41)-, cyclic GMP-AMP synthase (cGAS)-, and stimulator of interferon genes (STING)-dependent manner. The loss of MEF2A results in the activation of ATM and RAD3-related (ATR) kinase, which is also necessary for the activation of STING. This study identifies the role of MEF2A in sustaining transcriptional homeostasis and highlights the role of ATR in positively regulating R-loop-associated inflammatory responses.


Sujet(s)
Nucleotidyltransferases , Transduction du signal , Nucleotidyltransferases/métabolisme , RNA helicases , Interférons , Immunité innée
6.
Immunity ; 56(7): 1443-1450, 2023 07 11.
Article de Anglais | MEDLINE | ID: mdl-37437537

RÉSUMÉ

Innate immunity and the actions of type I and III interferons (IFNs) are essential for protection from SARS-CoV-2 and COVID-19. Each is induced in response to infection and serves to restrict viral replication and spread while directing the polarization and modulation of the adaptive immune response. Owing to the distribution of their specific receptors, type I and III IFNs, respectively, impart systemic and local actions. Therapeutic IFN has been administered to combat COVID-19 but with differential outcomes when given early or late in infection. In this perspective, we sort out the role of innate immunity and complex actions of IFNs in the context of SARS-CoV-2 infection and COVID-19. We conclude that IFNs are a beneficial component of innate immunity that has mediated natural clearance of infection in over 700 million people. Therapeutic induction of innate immunity and use of IFN should be featured in strategies to treat acute SARS-CoV-2 infection in people at risk for severe COVID-19.


Sujet(s)
COVID-19 , Interférons , Humains , Interférons/usage thérapeutique , SARS-CoV-2 , Immunité innée , Mouvement cellulaire
7.
PLoS Biol ; 21(6): e3002144, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37289745

RÉSUMÉ

Hosts have evolved diverse strategies to respond to microbial infections, including the detection of pathogen-encoded proteases by inflammasome-forming sensors such as NLRP1 and CARD8. Here, we find that the 3CL protease (3CLpro) encoded by diverse coronaviruses, including Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), cleaves a rapidly evolving region of human CARD8 and activates a robust inflammasome response. CARD8 is required for cell death and the release of pro-inflammatory cytokines during SARS-CoV-2 infection. We further find that natural variation alters CARD8 sensing of 3CLpro, including 3CLpro-mediated antagonism rather than activation of megabat CARD8. Likewise, we find that a single nucleotide polymorphism (SNP) in humans reduces CARD8's ability to sense coronavirus 3CLpros and, instead, enables sensing of 3C proteases (3Cpro) from select picornaviruses. Our findings demonstrate that CARD8 is a broad sensor of viral protease activities and suggests that CARD8 diversity contributes to inter- and intraspecies variation in inflammasome-mediated viral sensing and immunopathology.


Sujet(s)
COVID-19 , Picornaviridae , Humains , Inflammasomes/métabolisme , Picornaviridae/génétique , Picornaviridae/métabolisme , SARS-CoV-2/métabolisme , Inhibiteurs de protéases , Protéines régulatrices de l'apoptose/métabolisme , Protéines tumorales/métabolisme , Protéines adaptatrices de signalisation CARD/métabolisme
8.
Cytokine ; 164: 156159, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36809715

RÉSUMÉ

Interleukin (IL)-4 and IL-13 are related cytokines with well-known specific roles in type 2 immune response. However, their effects on neutrophils are not completely understood. For this, we studied human primary neutrophil responses to IL-4 and IL-13. Neutrophils are dose-dependently responsive to both IL-4 and IL-13 as indicated by signal transducer and activator of transcription 6 (STAT6) phosphorylation upon stimulation, with IL-4 being more potent inducer of STAT6. IL-4-, IL-13- and Interferon (IFN)-γ-stimulated gene expression in highly purified human neutrophils induced both overlapping and unique gene expression in highly purified human neutrophils. IL-4 and IL-13 specifically regulate several immune-related genes, including IL-10, tumor necrosis factor (TNF) and leukemia inhibitory factor (LIF), while type1 immune response-related IFN-γ induced gene expression related for example, to intracellular infections. In analysis of neutrophil metabolic responses, oxygen independent glycolysis was specifically regulated by IL-4, but not by IL-13 or IFN-γ, suggesting specific role for type I IL-4 receptor in this process. Our results provide a comprehensive analysis of IL-4, IL-13 and IFN-γ -induced gene expression in neutrophils while also addressing cytokine-mediated metabolic changes in neutrophils.


Sujet(s)
Interleukine-13 , Interleukine-4 , Humains , Cytokines/métabolisme , Interféron gamma/métabolisme , Interleukine-13/pharmacologie , Interleukine-13/métabolisme , Interleukine-4/pharmacologie , Interleukine-4/métabolisme , Granulocytes neutrophiles/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
9.
J Exp Med ; 220(3)2023 03 06.
Article de Anglais | MEDLINE | ID: mdl-36598533

RÉSUMÉ

Regulatory T cells (Tregs) suppress the activation and subsequent effector functions of CD4 effector T cells (Teffs). However, molecular mechanisms that enforce Treg-mediated suppression in CD4 Teff are unclear. We found that Tregs suppressed activation-induced global protein synthesis in CD4 Teffs prior to cell division. We analyzed genome-wide changes in the transcriptome and translatome of activated CD4 Teffs. We show that mRNAs encoding for the protein synthesis machinery are regulated at the level of translation in activated CD4 Teffs by Tregs. Tregs suppressed global protein synthesis of CD4 Teffs by specifically inhibiting mRNAs of the translation machinery at the level of mTORC1-mediated translation control through concerted action of immunosuppressive cytokines IL-10 and TGFß. Lastly, we found that the therapeutic targeting of protein synthesis with the RNA helicase eIF4A inhibitor rocaglamide A can alleviate inflammatory CD4 Teff activation caused by acute Treg depletion in vivo. These data show that peripheral tolerance is enforced by Tregs through mRNA translational control in CD4 Teffs.


Sujet(s)
Lymphocytes T CD4+ , Lymphocytes T régulateurs , Activation des lymphocytes , Cytokines/métabolisme , Facteur de croissance transformant bêta/métabolisme , ARN messager/génétique , ARN messager/métabolisme
10.
bioRxiv ; 2022 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-36172130

RÉSUMÉ

Hosts have evolved diverse strategies to respond to microbial infections, including the detection of pathogen-encoded proteases by inflammasome-forming sensors such as NLRP1 and CARD8. Here, we find that the 3CL protease (3CL pro ) encoded by diverse coronaviruses, including SARS-CoV-2, cleaves a rapidly evolving region of human CARD8 and activates a robust inflammasome response. CARD8 is required for cell death and the release of pro-inflammatory cytokines during SARS-CoV-2 infection. We further find that natural variation alters CARD8 sensing of 3CL pro , including 3CL pro -mediated antagonism rather than activation of megabat CARD8. Likewise, we find that a single nucleotide polymorphism (SNP) in humans reduces CARD8’s ability to sense coronavirus 3CL pros , and instead enables sensing of 3C proteases (3C pro ) from select picornaviruses. Our findings demonstrate that CARD8 is a broad sensor of viral protease activities and suggests that CARD8 diversity contributes to inter- and intra-species variation in inflammasome-mediated viral sensing and immunopathology.

11.
Nature ; 607(7920): 769-775, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35859177

RÉSUMÉ

The RNA-editing enzyme ADAR1 is essential for the suppression of innate immune activation and pathology caused by aberrant recognition of self-RNA, a role it carries out by disrupting the duplex structure of endogenous double-stranded RNA species1,2. A point mutation in the sequence encoding the Z-DNA-binding domain (ZBD) of ADAR1 is associated with severe autoinflammatory disease3-5. ZBP1 is the only other ZBD-containing mammalian protein6, and its activation can trigger both cell death and transcriptional responses through the kinases RIPK1 and RIPK3, and the protease caspase 8 (refs. 7-9). Here we show that the pathology caused by alteration of the ZBD of ADAR1 is driven by activation of ZBP1. We found that ablation of ZBP1 fully rescued the overt pathology caused by ADAR1 alteration, without fully reversing the underlying inflammatory program caused by this alteration. Whereas loss of RIPK3 partially phenocopied the protective effects of ZBP1 ablation, combined deletion of caspase 8 and RIPK3, or of caspase 8 and MLKL, unexpectedly exacerbated the pathogenic effects of ADAR1 alteration. These findings indicate that ADAR1 is a negative regulator of sterile ZBP1 activation, and that ZBP1-dependent signalling underlies the autoinflammatory pathology caused by alteration of ADAR1.


Sujet(s)
Adenosine deaminase , Maladies du système immunitaire , Inflammation , Mutation , Protéines de liaison à l'ARN , Adenosine deaminase/génétique , Adenosine deaminase/métabolisme , Animaux , Caspase 8/génétique , Caspase 8/métabolisme , Mort cellulaire , Délétion de gène , Maladies du système immunitaire/génétique , Maladies du système immunitaire/métabolisme , Maladies du système immunitaire/anatomopathologie , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Mammifères/génétique , Protein kinases/déficit , Protein kinases/génétique , ARN double brin/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/déficit , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Transduction du signal
12.
Blood ; 138(8): 722-737, 2021 08 26.
Article de Anglais | MEDLINE | ID: mdl-34436524

RÉSUMÉ

Immunopathology and intestinal stem cell (ISC) loss in the gastrointestinal (GI) tract is the prima facie manifestation of graft-versus-host disease (GVHD) and is responsible for significant mortality after allogeneic bone marrow transplantation (BMT). Approaches to prevent GVHD to date focus on immune suppression. Here, we identify interferon-λ (IFN-λ; interleukin-28 [IL-28]/IL-29) as a key protector of GI GVHD immunopathology, notably within the ISC compartment. Ifnlr1-/- mice displayed exaggerated GI GVHD and mortality independent of Paneth cells and alterations to the microbiome. Ifnlr1-/- intestinal organoid growth was significantly impaired, and targeted Ifnlr1 deficiency exhibited effects intrinsic to recipient Lgr5+ ISCs and natural killer cells. PEGylated recombinant IL-29 (PEG-rIL-29) treatment of naive mice enhanced Lgr5+ ISC numbers and organoid growth independent of both IL-22 and type I IFN and modulated proliferative and apoptosis gene sets in Lgr5+ ISCs. PEG-rIL-29 treatment improved survival, reduced GVHD severity, and enhanced epithelial proliferation and ISC-derived organoid growth after BMT. The preservation of ISC numbers in response to PEG-rIL-29 after BMT occurred both in the presence and absence of IFN-λ-signaling in recipient natural killer cells. IFN-λ is therefore an attractive and rapidly testable approach to prevent ISC loss and immunopathology during GVHD.


Sujet(s)
Transplantation de moelle osseuse , Cytokines/pharmacologie , Maladies gastro-intestinales , Maladie du greffon contre l'hôte , Interleukines/pharmacocinétique , Transduction du signal , Animaux , Cytokines/immunologie , Maladies gastro-intestinales/traitement médicamenteux , Maladies gastro-intestinales/génétique , Maladies gastro-intestinales/immunologie , Maladie du greffon contre l'hôte/traitement médicamenteux , Maladie du greffon contre l'hôte/génétique , Maladie du greffon contre l'hôte/immunologie , Interleukines/immunologie , Souris , Souris knockout , Récepteur interféron/génétique , Récepteur interféron/immunologie , Indice de gravité de la maladie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Transduction du signal/immunologie , Transplantation homologue
13.
Elife ; 102021 08 03.
Article de Anglais | MEDLINE | ID: mdl-34342578

RÉSUMÉ

Many host RNA sensors are positioned in the cytosol to detect viral RNA during infection. However, most positive-strand RNA viruses replicate within a modified organelle co-opted from intracellular membranes of the endomembrane system, which shields viral products from cellular innate immune sensors. Targeting innate RNA sensors to the endomembrane system may enhance their ability to sense RNA generated by viruses that use these compartments for replication. Here, we reveal that an isoform of oligoadenylate synthetase 1, OAS1 p46, is prenylated and targeted to the endomembrane system. Membrane localization of OAS1 p46 confers enhanced access to viral replication sites and results in increased antiviral activity against a subset of RNA viruses including flaviviruses, picornaviruses, and SARS-CoV-2. Finally, our human genetic analysis shows that the OAS1 splice-site SNP responsible for production of the OAS1 p46 isoform correlates with protection from severe COVID-19. This study highlights the importance of endomembrane targeting for the antiviral specificity of OAS1 and suggests that early control of SARS-CoV-2 replication through OAS1 p46 is an important determinant of COVID-19 severity.


Sujet(s)
2',5'-Oligoadenylate synthetase/métabolisme , COVID-19/virologie , SARS-CoV-2/métabolisme , Animaux , COVID-19/immunologie , Systèmes CRISPR-Cas , Lignée cellulaire , Édition de gène , Humains , Polymorphisme de nucléotide simple , SARS-CoV-2/isolement et purification
14.
Immunol Rev ; 304(1): 77-96, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34405416

RÉSUMÉ

From the initial sensing of viral nucleotides by pattern recognition receptors, through the induction of type I and III interferons (IFN), upregulation of antiviral effector proteins, and resolution of the inflammatory response, each step of innate immune signaling is under tight control. Though innate immunity is often associated with broad regulation at the level of gene transcription, RNA-centric post-transcriptional processes have emerged as critical mechanisms for ensuring a proper antiviral response. Here, we explore the diverse RNA regulatory mechanisms that modulate the innate antiviral immune response, with a focus on RNA sensing by RIG-I-like receptors (RLR), interferon (IFN) and IFN signaling pathways, viral pathogenesis, and host genetic variation that contributes to these processes. We address the post-transcriptional interactions with RNA-binding proteins, non-coding RNAs, transcript elements, and modifications that control mRNA stability, as well as alternative splicing events that modulate the innate immune antiviral response.


Sujet(s)
Facteurs de restriction antiviraux/immunologie , Immunité innée , ARN viral , Maladies virales/immunologie , Humains , Interférons , ARN viral/génétique , Récepteurs de reconnaissance de motifs moléculaires/génétique
17.
J Exp Med ; 218(5)2021 05 03.
Article de Anglais | MEDLINE | ID: mdl-33724365

RÉSUMÉ

SAMD9L is an interferon-induced tumor suppressor implicated in a spectrum of multisystem disorders, including risk for myeloid malignancies and immune deficiency. We identified a heterozygous de novo frameshift variant in SAMD9L in an infant with B cell aplasia and clinical autoinflammatory features who died from respiratory failure with chronic rhinovirus infection. Autopsy demonstrated absent bone marrow and peripheral B cells as well as selective loss of Langerhans and Purkinje cells. The frameshift variant led to expression of a truncated protein with interferon treatment. This protein exhibited a gain-of-function phenotype, resulting in interference in global protein synthesis via inhibition of translational elongation. Using a mutational scan, we identified a region within SAMD9L where stop-gain variants trigger a similar translational arrest. SAMD9L variants that globally suppress translation had no effect or increased mRNA transcription. The complex-reported phenotype likely reflects lineage-dominant sensitivities to this translation block. Taken together, our findings indicate that interferon-triggered SAMD9L gain-of-function variants globally suppress translation.


Sujet(s)
Mutation avec décalage du cadre de lecture , Régulation de l'expression des gènes/génétique , Mutation germinale , Biosynthèse des protéines/génétique , Protéines suppresseurs de tumeurs/génétique , Cellules A549 , Lymphocytes B/métabolisme , Lymphocytes B/anatomopathologie , Issue fatale , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules HEK293 , Hétérozygote , Humains , Nouveau-né , Interférons/pharmacologie , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Séquençage du génome entier
18.
Article de Anglais | MEDLINE | ID: mdl-32341066

RÉSUMÉ

Activation and viral control of the innate immune response are hallmarks of hepatitis C virus (HCV) infection and are major determinants of spontaneous clearance or progression to chronic infection and liver disease. In this review, we provide a contemporary overview of how HCV is sensed by the host cell to trigger innate immune activation and the mechanisms deployed by the virus to evade this response. Type I and III interferons (IFNs) are crucial mediators of antiviral innate immunity against HCV, and we specifically highlight the importance of IFN-λ host genetics for the outcome of HCV infection. Last, we focus on the proinflammatory responses elicited by HCV infection and describe our current understanding of how interleukin (IL)-1ß signaling and cross talk between the IL-1ß and IFN signaling pathways lead to sustained inflammation and increased risk of liver pathology.


Sujet(s)
Antiviraux/usage thérapeutique , Évolution de la maladie , Hépatite C chronique/traitement médicamenteux , Antiviraux/pharmacologie , Hepacivirus , Hépatite C chronique/immunologie , Humains , Immunité innée , Interféron de type I/immunologie , Interférons/immunologie , Foie/virologie , Polymorphisme de nucléotide simple , Transduction du signal , Interféron lambda
19.
JCI Insight ; 6(1)2021 01 11.
Article de Anglais | MEDLINE | ID: mdl-33232305

RÉSUMÉ

Cardiopulmonary bypass (CPB) is required during most cardiac surgeries. CBP drives systemic inflammation and multiorgan dysfunction that is especially severe in neonatal patients. Limited understanding of molecular mechanisms underlying CPB-associated inflammation presents a significant barrier to improve clinical outcomes. To better understand these clinical issues, we performed mRNA sequencing on total circulating leukocytes from neonatal patients undergoing CPB. Our data identify myeloid cells, particularly monocytes, as the major cell type driving transcriptional responses to CPB. Furthermore, IL-8 and TNF-α were inflammatory cytokines robustly upregulated in leukocytes from both patients and piglets exposed to CPB. To delineate the molecular mechanism, we exposed THP-1 human monocytic cells to CPB-like conditions, including artificial surfaces, high shear stress, and cooling/rewarming. Shear stress was found to drive cytokine upregulation via calcium-dependent signaling pathways. We also observed that a subpopulation of THP-1 cells died via TNF-α-mediated necroptosis, which we hypothesize contributes to post-CPB inflammation. Our study identifies a shear stress-modulated molecular mechanism that drives systemic inflammation in pediatric CPB patients. These are also the first data to our knowledge to demonstrate that shear stress causes necroptosis. Finally, we observe that calcium and TNF-α signaling are potentially novel targets to ameliorate post-CPB inflammation.


Sujet(s)
Pontage cardiopulmonaire/effets indésirables , Cytokines/génétique , Monocytes/immunologie , Monocytes/anatomopathologie , Animaux , Animaux nouveau-nés , Signalisation calcique , Cytokines/biosynthèse , Femelle , Cardiopathies congénitales/chirurgie , Humains , Nourrisson , Nouveau-né , Médiateurs de l'inflammation/métabolisme , Interleukine-8/biosynthèse , Interleukine-8/génétique , Mâle , Modèles animaux , Monocytes/physiologie , Nécroptose/génétique , Nécroptose/physiologie , RNA-Seq , Contrainte mécanique , Sus scrofa , Syndrome de réponse inflammatoire généralisée/étiologie , Syndrome de réponse inflammatoire généralisée/génétique , Syndrome de réponse inflammatoire généralisée/immunologie , Cellules THP-1 , Facteur de nécrose tumorale alpha/biosynthèse , Facteur de nécrose tumorale alpha/génétique , Régulation positive
20.
Sci Signal ; 13(661)2020 12 08.
Article de Anglais | MEDLINE | ID: mdl-33293463

RÉSUMÉ

Small, genetically determined differences in transcription [expression quantitative trait loci (eQTLs)] are implicated in complex diseases through unknown molecular mechanisms. Here, we showed that a small, persistent increase in the abundance of the innate pathogen sensor NOD1 precipitated large changes in the transcriptional state of monocytes. A ~1.2- to 1.3-fold increase in NOD1 protein abundance resulting from loss of regulation by the microRNA cluster miR-15b/16 lowered the threshold for ligand-induced activation of the transcription factor NF-κB and the MAPK p38. An additional sustained increase in NOD1 abundance to 1.5-fold over basal amounts bypassed this low ligand concentration requirement, resulting in robust ligand-independent induction of proinflammatory genes and oncogenes. These findings reveal that tight regulation of NOD1 abundance prevents this sensor from exceeding a physiological switching checkpoint that promotes persistent inflammation and oncogene expression. Furthermore, our data provide insight into how a quantitatively small change in protein abundance can produce marked changes in cell state that can serve as the initiator of disease.


Sujet(s)
Régulation de l'expression des gènes , Monocytes/métabolisme , Protéine adaptatrice de signalisation NOD1/biosynthèse , Protéines oncogènes/biosynthèse , Transduction du signal , Transcription génétique , Humains , Inflammation/métabolisme , Cellules THP-1
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE