Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Nat Microbiol ; 9(2): 451-463, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38228858

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) human adaptation resulted in distinct lineages with enhanced transmissibility called variants of concern (VOCs). Omicron is the first VOC to evolve distinct globally dominant subvariants. Here we compared their replication in human cell lines and primary airway cultures and measured host responses to infection. We discovered that subvariants BA.4 and BA.5 have improved their suppression of innate immunity when compared with earlier subvariants BA.1 and BA.2. Similarly, more recent subvariants (BA.2.75 and XBB lineages) also triggered reduced innate immune activation. This correlated with increased expression of viral innate antagonists Orf6 and nucleocapsid, reminiscent of VOCs Alpha to Delta. Increased Orf6 levels suppressed host innate responses to infection by decreasing IRF3 and STAT1 signalling measured by transcription factor phosphorylation and nuclear translocation. Our data suggest that convergent evolution of enhanced innate immune antagonist expression is a common pathway of human adaptation and link Omicron subvariant dominance to improved innate immune evasion.


Sujet(s)
COVID-19 , Humains , SARS-CoV-2 , Lignée cellulaire , Échappement immunitaire , Immunité innée
2.
Cell ; 186(21): 4597-4614.e26, 2023 10 12.
Article de Anglais | MEDLINE | ID: mdl-37738970

RÉSUMÉ

SARS-CoV-2 variants of concern (VOCs) emerged during the COVID-19 pandemic. Here, we used unbiased systems approaches to study the host-selective forces driving VOC evolution. We discovered that VOCs evolved convergent strategies to remodel the host by modulating viral RNA and protein levels, altering viral and host protein phosphorylation, and rewiring virus-host protein-protein interactions. Integrative computational analyses revealed that although Alpha, Beta, Gamma, and Delta ultimately converged to suppress interferon-stimulated genes (ISGs), Omicron BA.1 did not. ISG suppression correlated with the expression of viral innate immune antagonist proteins, including Orf6, N, and Orf9b, which we mapped to specific mutations. Later Omicron subvariants BA.4 and BA.5 more potently suppressed innate immunity than early subvariant BA.1, which correlated with Orf6 levels, although muted in BA.4 by a mutation that disrupts the Orf6-nuclear pore interaction. Our findings suggest that SARS-CoV-2 convergent evolution overcame human adaptive and innate immune barriers, laying the groundwork to tackle future pandemics.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , COVID-19/virologie , Immunité innée/génétique , Pandémies , SARS-CoV-2/génétique
3.
Proc Natl Acad Sci U S A ; 120(5): e2212577120, 2023 01 31.
Article de Anglais | MEDLINE | ID: mdl-36693093

RÉSUMÉ

SARS-CoV-2 spike requires proteolytic processing for viral entry. A polybasic furin-cleavage site (FCS) in spike, and evolution toward an optimized FCS by dominant variants of concern (VOCs), are linked to enhanced infectivity and transmission. Here we show interferon-inducible restriction factors Guanylate-binding proteins (GBP) 2 and 5 interfere with furin-mediated spike cleavage and inhibit the infectivity of early-lineage isolates Wuhan-Hu-1 and VIC. By contrast, VOCs Alpha and Delta escape restriction by GBP2/5 that we map to the spike substitution D614G present in these VOCs. Despite inhibition of spike cleavage, these viruses remained sensitive to plasma membrane IFITM1, but not endosomal IFITM2 and 3, consistent with a preference for TMPRSS2-dependent plasma membrane entry. Strikingly, we find that Omicron is unique among VOCs, being sensitive to restriction factors GBP2/5, and also IFITM1, 2, and 3. Using chimeric spike mutants, we map the Omicron phenotype and show that the S1 domain determines Omicron's sensitivity to GBP2/5, whereas the S2' domain determines its sensitivity to endosomal IFITM2/3 and preferential use of TMPRSS2-independent entry. We propose that evolution of SARS-CoV-2 for the D614G substitution has allowed for escape from GBP restriction factors, but the selective pressures on Omicron for spike changes that mediate antibody escape, and altered tropism, have come at the expense of increased sensitivity to innate immune restriction factors that target virus entry.


Sujet(s)
COVID-19 , Furine , Humains , COVID-19/génétique , SARS-CoV-2/génétique , Anticorps , Membrane cellulaire , Proaccélérine , Glycoprotéine de spicule des coronavirus/génétique , Protéines membranaires/génétique
4.
EMBO Rep ; 23(10): e54322, 2022 10 06.
Article de Anglais | MEDLINE | ID: mdl-35999696

RÉSUMÉ

The emergence of SARS-CoV-2 variants has exacerbated the COVID-19 global health crisis. Thus far, all variants carry mutations in the spike glycoprotein, which is a critical determinant of viral transmission being responsible for attachment, receptor engagement and membrane fusion, and an important target of immunity. Variants frequently bear truncations of flexible loops in the N-terminal domain (NTD) of spike; the functional importance of these modifications has remained poorly characterised. We demonstrate that NTD deletions are important for efficient entry by the Alpha and Omicron variants and that this correlates with spike stability. Phylogenetic analysis reveals extensive NTD loop length polymorphisms across the sarbecoviruses, setting an evolutionary precedent for loop remodelling. Guided by these analyses, we demonstrate that variations in NTD loop length, alone, are sufficient to modulate virus entry. We propose that variations in NTD loop length act to fine-tune spike; this may provide a mechanism for SARS-CoV-2 to navigate a complex selection landscape encompassing optimisation of essential functionality, immune-driven antigenic variation and ongoing adaptation to a new host.


Sujet(s)
COVID-19 , SARS-CoV-2 , COVID-19/génétique , Humains , Phylogenèse , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique
5.
Cell Rep ; 39(2): 110650, 2022 04 12.
Article de Anglais | MEDLINE | ID: mdl-35417711

RÉSUMÉ

HIV-1 replicates in CD4+ T cells, leading to AIDS. Determining how HIV-1 shapes its niche to create a permissive environment is central to informing efforts to limit pathogenesis, disturb reservoirs, and achieve a cure. A key roadblock in understanding HIV-T cell interactions is the requirement to activate T cells in vitro to make them permissive to infection. This dramatically alters T cell biology and virus-host interactions. Here we show that HIV-1 cell-to-cell spread permits efficient, productive infection of resting memory T cells without prior activation. Strikingly, we find that HIV-1 infection primes resting T cells to gain characteristics of tissue-resident memory T cells (TRM), including upregulating key surface markers and the transcription factor Blimp-1 and inducing a transcriptional program overlapping the core TRM transcriptional signature. This reprogramming is driven by Vpr and requires Vpr packaging into virions and manipulation of STAT5. Thus, HIV-1 reprograms resting T cells, with implications for viral replication and persistence.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Lymphocytes T CD4+/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Cellules T mémoire , Phénotype , Réplication virale , Produits du gène vpr du virus de l'immunodéficience humaine/génétique
7.
NPJ Digit Med ; 5(1): 18, 2022 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-35165389

RÉSUMÉ

The COVID-19 pandemic has pushed healthcare systems globally to a breaking point. The urgent need for effective and affordable COVID-19 treatments calls for repurposing combinations of approved drugs. The challenge is to identify which combinations are likely to be most effective and at what stages of the disease. Here, we present the first disease-stage executable signalling network model of SARS-CoV-2-host interactions used to predict effective repurposed drug combinations for treating early- and late stage severe disease. Using our executable model, we performed in silico screening of 9870 pairs of 140 potential targets and have identified nine new drug combinations. Camostat and Apilimod were predicted to be the most promising combination in effectively supressing viral replication in the early stages of severe disease and were validated experimentally in human Caco-2 cells. Our study further demonstrates the power of executable mechanistic modelling to enable rapid pre-clinical evaluation of combination therapies tailored to disease progression. It also presents a novel resource and expandable model system that can respond to further needs in the pandemic.

8.
Life Sci Alliance ; 5(4)2022 04.
Article de Anglais | MEDLINE | ID: mdl-35012962

RÉSUMÉ

Plitidepsin, a marine-derived cyclic-peptide, inhibits SARS-CoV-2 replication at nanomolar concentrations by targeting the host protein eukaryotic translation elongation factor 1A. Here, we show that plitidepsin distributes preferentially to lung over plasma, with similar potency against across several SARS-CoV-2 variants in preclinical studies. Simultaneously, in this randomized, parallel, open-label, proof-of-concept study (NCT04382066) conducted in 10 Spanish hospitals between May and November 2020, 46 adult hospitalized patients with confirmed SARS-CoV-2 infection received either 1.5 mg (n = 15), 2.0 mg (n = 16), or 2.5 mg (n = 15) plitidepsin once daily for 3 d. The primary objective was safety; viral load kinetics, mortality, need for increased respiratory support, and dose selection were secondary end points. One patient withdrew consent before starting procedures; 45 initiated treatment; one withdrew because of hypersensitivity. Two Grade 3 treatment-related adverse events were observed (hypersensitivity and diarrhea). Treatment-related adverse events affecting more than 5% of patients were nausea (42.2%), vomiting (15.6%), and diarrhea (6.7%). Mean viral load reductions from baseline were 1.35, 2.35, 3.25, and 3.85 log10 at days 4, 7, 15, and 31. Nonmechanical invasive ventilation was required in 8 of 44 evaluable patients (16.0%); six patients required intensive care support (13.6%), and three patients (6.7%) died (COVID-19-related). Plitidepsin has a favorable safety profile in patients with COVID-19.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Depsipeptides/usage thérapeutique , Hospitalisation/statistiques et données numériques , Peptides cycliques/usage thérapeutique , SARS-CoV-2/effets des médicaments et des substances chimiques , Adulte , Sujet âgé , COVID-19/virologie , Lignée cellulaire tumorale , Depsipeptides/effets indésirables , Depsipeptides/pharmacologie , Évaluation préclinique de médicament/méthodes , Femelle , Humains , Estimation de Kaplan-Meier , Durée du séjour/statistiques et données numériques , Mâle , Adulte d'âge moyen , Neutropénie/induit chimiquement , Peptides cycliques/effets indésirables , Peptides cycliques/pharmacologie , SARS-CoV-2/physiologie , Résultat thérapeutique , Charge virale/effets des médicaments et des substances chimiques
9.
Nature ; 602(7897): 487-495, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34942634

RÉSUMÉ

The emergence of SARS-CoV-2 variants of concern suggests viral adaptation to enhance human-to-human transmission1,2. Although much effort has focused on the characterization of changes in the spike protein in variants of concern, mutations outside of spike are likely to contribute to adaptation. Here, using unbiased abundance proteomics, phosphoproteomics, RNA sequencing and viral replication assays, we show that isolates of the Alpha (B.1.1.7) variant3 suppress innate immune responses in airway epithelial cells more effectively than first-wave isolates. We found that the Alpha variant has markedly increased subgenomic RNA and protein levels of the nucleocapsid protein (N), Orf9b and Orf6-all known innate immune antagonists. Expression of Orf9b alone suppressed the innate immune response through interaction with TOM70, a mitochondrial protein that is required for activation of the RNA-sensing adaptor MAVS. Moreover, the activity of Orf9b and its association with TOM70 was regulated by phosphorylation. We propose that more effective innate immune suppression, through enhanced expression of specific viral antagonist proteins, increases the likelihood of successful transmission of the Alpha variant, and may increase in vivo replication and duration of infection4. The importance of mutations outside the spike coding region in the adaptation of SARS-CoV-2 to humans is underscored by the observation that similar mutations exist in the N and Orf9b regulatory regions of the Delta and Omicron variants.


Sujet(s)
COVID-19/immunologie , COVID-19/virologie , Évolution moléculaire , Échappement immunitaire , Immunité innée/immunologie , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , COVID-19/transmission , Protéines de la nucléocapside des coronavirus/composition chimique , Protéines de la nucléocapside des coronavirus/métabolisme , Humains , Immunité innée/génétique , Interférons/immunologie , Protéines du complexe d'import des protéines précurseurs mitochondriales/métabolisme , Phosphoprotéines/composition chimique , Phosphoprotéines/métabolisme , Phosphorylation , Protéomique , ARN viral/génétique , RNA-Seq , SARS-CoV-2/classification , SARS-CoV-2/croissance et développement
10.
Nature ; 602(7896): 321-327, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34937051

RÉSUMÉ

It is not fully understood why COVID-19 is typically milder in children1-3. Here, to examine the differences between children and adults in their response to SARS-CoV-2 infection, we analysed paediatric and adult patients with COVID-19 as well as healthy control individuals (total n = 93) using single-cell multi-omic profiling of matched nasal, tracheal, bronchial and blood samples. In the airways of healthy paediatric individuals, we observed cells that were already in an interferon-activated state, which after SARS-CoV-2 infection was further induced especially in airway immune cells. We postulate that higher paediatric innate interferon responses restrict viral replication and disease progression. The systemic response in children was characterized by increases in naive lymphocytes and a depletion of natural killer cells, whereas, in adults, cytotoxic T cells and interferon-stimulated subpopulations were significantly increased. We provide evidence that dendritic cells initiate interferon signalling in early infection, and identify epithelial cell states associated with COVID-19 and age. Our matching nasal and blood data show a strong interferon response in the airways with the induction of systemic interferon-stimulated populations, which were substantially reduced in paediatric patients. Together, we provide several mechanisms that explain the milder clinical syndrome observed in children.


Sujet(s)
COVID-19/sang , COVID-19/immunologie , Cellules dendritiques/immunologie , Interférons/immunologie , Cellules tueuses naturelles/immunologie , SARS-CoV-2/immunologie , Lymphocytes T cytotoxiques/immunologie , Adulte , Bronches/immunologie , Bronches/virologie , COVID-19/anatomopathologie , Chicago , Études de cohortes , Évolution de la maladie , Cellules épithéliales/cytologie , Cellules épithéliales/immunologie , Cellules épithéliales/virologie , Femelle , Humains , Immunité innée , Londres , Mâle , Muqueuse nasale/immunologie , Muqueuse nasale/virologie , SARS-CoV-2/croissance et développement , Analyse sur cellule unique , Trachée/virologie , Jeune adulte
11.
EMBO J ; 40(15): e107826, 2021 08 02.
Article de Anglais | MEDLINE | ID: mdl-34101213

RÉSUMÉ

SARS-CoV-2 infection causes broad-spectrum immunopathological disease, exacerbated by inflammatory co-morbidities. A better understanding of mechanisms underpinning virus-associated inflammation is required to develop effective therapeutics. Here, we discover that SARS-CoV-2 replicates rapidly in lung epithelial cells despite triggering a robust innate immune response through the activation of cytoplasmic RNA sensors RIG-I and MDA5. The inflammatory mediators produced during epithelial cell infection can stimulate primary human macrophages to enhance cytokine production and drive cellular activation. Critically, this can be limited by abrogating RNA sensing or by inhibiting downstream signalling pathways. SARS-CoV-2 further exacerbates the local inflammatory environment when macrophages or epithelial cells are primed with exogenous inflammatory stimuli. We propose that RNA sensing of SARS-CoV-2 in lung epithelium is a key driver of inflammation, the extent of which is influenced by the inflammatory state of the local environment, and that specific inhibition of innate immune pathways may beneficially mitigate inflammation-associated COVID-19.


Sujet(s)
COVID-19/immunologie , Protéine-58 à domaine DEAD/immunologie , Cellules épithéliales/immunologie , Hélicase IFIH1 inductrice de l'interféron/immunologie , Macrophages/immunologie , ARN viral/immunologie , Récepteurs immunologiques/immunologie , SARS-CoV-2 , COVID-19/génétique , COVID-19/virologie , Lignée cellulaire , Cytokines/génétique , Cytokines/immunologie , Cellules épithéliales/virologie , Interactions hôte-pathogène , Humains , Immunité innée , Inflammation/génétique , Inflammation/immunologie , Inflammation/virologie , Janus kinases/immunologie , Poumon/cytologie , Poumon/immunologie , Poumon/virologie , Activation des macrophages , Facteur de transcription NF-kappa B/immunologie , Muqueuse respiratoire/cytologie , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/virologie , SARS-CoV-2/génétique , SARS-CoV-2/physiologie , Facteurs de transcription STAT/immunologie , Réplication virale
12.
bioRxiv ; 2021 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-34127972

RÉSUMÉ

Emergence of SARS-CoV-2 variants, including the globally successful B.1.1.7 lineage, suggests viral adaptations to host selective pressures resulting in more efficient transmission. Although much effort has focused on Spike adaptation for viral entry and adaptive immune escape, B.1.1.7 mutations outside Spike likely contribute to enhance transmission. Here we used unbiased abundance proteomics, phosphoproteomics, mRNA sequencing and viral replication assays to show that B.1.1.7 isolates more effectively suppress host innate immune responses in airway epithelial cells. We found that B.1.1.7 isolates have dramatically increased subgenomic RNA and protein levels of Orf9b and Orf6, both known innate immune antagonists. Expression of Orf9b alone suppressed the innate immune response through interaction with TOM70, a mitochondrial protein required for RNA sensing adaptor MAVS activation, and Orf9b binding and activity was regulated via phosphorylation. We conclude that B.1.1.7 has evolved beyond the Spike coding region to more effectively antagonise host innate immune responses through upregulation of specific subgenomic RNA synthesis and increased protein expression of key innate immune antagonists. We propose that more effective innate immune antagonism increases the likelihood of successful B.1.1.7 transmission, and may increase in vivo replication and duration of infection.

13.
medRxiv ; 2021 May 25.
Article de Anglais | MEDLINE | ID: mdl-34075384

RÉSUMÉ

Plitidepsin is a marine-derived cyclic-peptide that inhibits SARS-CoV-2 replication at low nanomolar concentrations by the targeting of host protein eEF1A (eukaryotic translation-elongation-factor-1A). We evaluated a model of intervention with plitidepsin in hospitalized COVID-19 adult patients where three doses were assessed (1.5, 2 and 2.5 mg/day for 3 days, as a 90-minute intravenous infusion) in 45 patients (15 per dose-cohort). Treatment was well tolerated, with only two Grade 3 treatment-related adverse events observed (hypersensitivity and diarrhea). The discharge rates by Days 8 and 15 were 56.8% and 81.8%, respectively, with data sustaining dose-effect. A mean 4.2 log10 viral load reduction was attained by Day 15. Improvement in inflammation markers was also noted in a seemingly dose-dependent manner. These results suggest that plitidepsin impacts the outcome of patients with COVID-19. ONE-SENTENCE SUMMARY: Plitidepsin, an inhibitor of SARS-Cov-2 in vitro , is safe and positively influences the outcome of patients hospitalized with COVID-19.

14.
bioRxiv ; 2021 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-33791702

RÉSUMÉ

The recent emergence of SARS-CoV-2 variants with increased transmission, pathogenesis and immune resistance has jeopardised the global response to the COVID-19 pandemic. Determining the fundamental biology of viral variants and understanding their evolutionary trajectories will guide current mitigation measures, future genetic surveillance and vaccination strategies. Here we examine virus entry by the B.1.1.7 lineage, commonly referred to as the UK/Kent variant. Pseudovirus infection of model cell lines demonstrate that B.1.1.7 entry is enhanced relative to the Wuhan-Hu-1 reference strain, particularly under low expression of receptor ACE2. Moreover, the entry characteristics of B.1.1.7 were distinct from that of its predecessor strain containing the D614G mutation. These data suggest evolutionary tuning of spike protein function. Additionally, we found that amino acid deletions within the N-terminal domain (NTD) of spike were important for efficient entry by B.1.1.7. The NTD is a hotspot of diversity across sarbecoviruses, therefore, we further investigated this region by examining the entry of closely related CoVs. Surprisingly, Pangolin CoV spike entry was 50-100 fold enhanced relative to SARS-CoV-2; suggesting there may be evolutionary pathways by which SARSCoV-2 may further optimise entry. Swapping the NTD between Pangolin CoV and SARS-CoV-2 demonstrates that changes in this region alone have the capacity to enhance virus entry. Thus, the NTD plays a hitherto unrecognised role in modulating spike activity, warranting further investigation and surveillance of NTD mutations.

15.
bioRxiv ; 2021 Feb 04.
Article de Anglais | MEDLINE | ID: mdl-33501437

RÉSUMÉ

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in millions of deaths worldwide and massive societal and economic burden. Recently, a new variant of SARS-CoV-2, known as B.1.1.7, was first detected in the United Kingdom and is spreading in several other countries, heightening public health concern and raising questions as to the resulting effectiveness of vaccines and therapeutic interventions. We and others previously identified host-directed therapies with antiviral efficacy against SARS-CoV-2 infection. Less prone to the development of therapy resistance, host-directed drugs represent promising therapeutic options to combat emerging viral variants as host genes possess a lower propensity to mutate compared to viral genes. Here, in the first study of the full-length B.1.1.7 variant virus , we find two host-directed drugs, plitidepsin (aplidin; inhibits translation elongation factor eEF1A) and ralimetinib (inhibits p38 MAP kinase cascade), as well as remdesivir, to possess similar antiviral activity against both the early-lineage SARS-CoV-2 and the B.1.1.7 variant, evaluated in both human gastrointestinal and lung epithelial cell lines. We find that plitidepsin is over an order of magnitude more potent than remdesivir against both viruses. These results highlight the importance of continued development of host-directed therapeutics to combat current and future coronavirus variant outbreaks.

16.
PLoS Pathog ; 13(9): e1006577, 2017 Sep.
Article de Anglais | MEDLINE | ID: mdl-28863187

RÉSUMÉ

Early events in the human airways determining whether exposure to Mycobacterium tuberculosis (Mtb) results in acquisition of infection are poorly understood. Epithelial cells are the dominant cell type in the lungs, but little is known about their role in tuberculosis. We hypothesised that human primary airway epithelial cells are part of the first line of defense against Mtb-infection and contribute to the protective host response in the human respiratory tract. We modelled these early airway-interactions with human primary bronchial epithelial cells (PBECs) and alveolar macrophages. By combining in vitro infection and transwell co-culture models with a global transcriptomic approach, we identified PBECs to be inert to direct Mtb-infection, yet to be potent responders within an Mtb-activated immune network, mediated by IL1ß and type I interferon (IFN). Activation of PBECs by Mtb-infected alveolar macrophages and monocytes increased expression of known and novel antimycobacterial peptides, defensins and S100-family members and epithelial-myeloid interactions further shaped the immunological environment during Mtb-infection by promoting neutrophil influx. This is the first in depth analysis of the primary epithelial response to infection and offers new insights into their emerging role in tuberculosis through complementing and amplifying responses to Mtb.


Sujet(s)
Cellules épithéliales/microbiologie , Immunité innée , Poumon/microbiologie , Macrophages/immunologie , Mycobacterium tuberculosis/immunologie , Tuberculose/immunologie , Cellules cultivées , Humains , Interféron gamma/immunologie , Monocytes/immunologie
17.
PLoS Pathog ; 9(7): e1003442, 2013.
Article de Anglais | MEDLINE | ID: mdl-23853585

RÉSUMÉ

Suppressor of cytokine signalling 3 (SOCS3) negatively regulates STAT3 activation in response to several cytokines such as those in the gp130-containing IL-6 receptor family. Thus, SOCS3 may play a major role in immune responses to pathogens. In the present study, the role of SOCS3 in M. tuberculosis infection was examined. All Socs3(fl/fl) LysM cre, Socs3(fl/fl) lck cre (with SOCS3-deficient myeloid and lymphoid cells, respectively) and gp130(F/F) mice, with a mutation in gp130 that impedes binding to SOCS3, showed increased susceptibility to infection with M. tuberculosis. SOCS3 binding to gp130 in myeloid cells conveyed resistance to M. tuberculosis infection via the regulation of IL-6/STAT3 signalling. SOCS3 was redundant for mycobacterial control by macrophages in vitro. Instead, SOCS3 expression in infected macrophages and DCs prevented the IL-6-mediated inhibition of TNF and IL-12 secretion and contributed to a timely CD4+ cell-dependent IFN-γ expression in vivo. In T cells, SOCS3 expression was essential for a gp130-independent control of infection with M. tuberculosis, but was neither required for the control of infection with attenuated M. bovis BCG nor for M. tuberculosis in BCG-vaccinated mice. Socs3(fl/fl) lck cre mice showed an increased frequency of γδ+ T cells in different organs and an enhanced secretion of IL-17 by γδ+ T cells in response to infection. Socs3(fl/fl) lck cre γδ+ T cells impaired the control of infection with M. tuberculosis. Thus, SOCS3 expression in either lymphoid or myeloid cells is essential for resistance against M. tuberculosis via discrete mechanisms.


Sujet(s)
Immunité cellulaire , Poumon/immunologie , Mycobacterium tuberculosis/immunologie , Cellules myéloïdes/métabolisme , Protéines SOCS/métabolisme , Lymphocytes T/métabolisme , Tuberculose pulmonaire/immunologie , Animaux , Vaccin BCG/usage thérapeutique , Récepteur gp130 de cytokines/génétique , Récepteur gp130 de cytokines/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Cellules dendritiques/microbiologie , Poumon/microbiologie , Poumon/anatomopathologie , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/microbiologie , Souris , Souris knockout , Souris transgéniques , Mutation , Mycobacterium bovis/croissance et développement , Mycobacterium bovis/immunologie , Cellules myéloïdes/immunologie , Cellules myéloïdes/microbiologie , Facteur de transcription STAT-3/agonistes , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme , Transduction du signal , Organismes exempts d'organismes pathogènes spécifiques , Protéine-3 suppressive de la signalisation des cytokine , Protéines SOCS/génétique , Lymphocytes T/immunologie , Lymphocytes T/microbiologie , Tuberculose pulmonaire/microbiologie , Tuberculose pulmonaire/anatomopathologie , Tuberculose pulmonaire/prévention et contrôle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...