Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Virol ; 98(9): e0066924, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39194251

RÉSUMÉ

Respiratory infections are a major health burden worldwide. Respiratory syncytial virus (RSV) is among the leading causes of hospitalization in both young children and older adults. The onset of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic and the public health response had a profound impact on the normal seasonal outbreaks of other respiratory viruses. However, little is known about how a prior respiratory virus infection impacts SARS-CoV-2 disease outcomes. In this study, we examine the impact of a previous RSV infection on the disease severity of a subsequent SARS-CoV-2 challenge in BALB/c mice. Mice infected with RSV, followed by a SARS-CoV-2 challenge, 30 days later, exhibited decreased weight loss and increased survival as compared to control groups. Our results suggest a prior RSV infection can provide protection against a subsequent SARS-CoV-2 infection. IMPORTANCE: Severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus are respiratory viruses that are a major health burden worldwide. Severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus frequently have peak seasonal outbreaks during the winter months, and are capable of causing severe respiratory disease, often leading to hospitalization. The 2019 pandemic brought attention to the importance of understanding how co-circulating viruses can impact the disease severity of other respiratory viruses. It is known that many hospitalized patients are undergoing multiple viral infections at once, yet not much has been studied to understand the impact this has on other respiratory viruses or patients. How co-circulating viruses impact one another can provide critical knowledge for future interventions of hospitalized patients and potential vaccination strategies.


Sujet(s)
COVID-19 , Souris de lignée BALB C , Infections à virus respiratoire syncytial , SARS-CoV-2 , Animaux , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Infections à virus respiratoire syncytial/virologie , Souris , COVID-19/prévention et contrôle , COVID-19/virologie , SARS-CoV-2/immunologie , Femelle , Humains , Modèles animaux de maladie humaine , Virus respiratoires syncytiaux/physiologie , Virus respiratoires syncytiaux/immunologie
2.
Front Immunol ; 13: 989000, 2022.
Article de Anglais | MEDLINE | ID: mdl-36072595

RÉSUMÉ

Pharmacological ascorbate (i.e., intravenous infusions of vitamin C reaching ~ 20 mM in plasma) is under active investigation as an adjuvant to standard of care anti-cancer treatments due to its dual redox roles as an antioxidant in normal tissues and as a prooxidant in malignant tissues. Immune checkpoint inhibitors (ICIs) are highly promising therapies for many cancer patients but face several challenges including low response rates, primary or acquired resistance, and toxicity. Ascorbate modulates both innate and adaptive immune functions and plays a key role in maintaining the balance between pro and anti-inflammatory states. Furthermore, the success of pharmacological ascorbate as a radiosensitizer and a chemosensitizer in pre-clinical studies and early phase clinical trials suggests that it may also enhance the efficacy and expand the benefits of ICIs.


Sujet(s)
Antinéoplasiques , Tumeurs , Antinéoplasiques/usage thérapeutique , Acide ascorbique/pharmacologie , Acide ascorbique/usage thérapeutique , Humains , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Tumeurs/traitement médicamenteux
3.
Redox Biol ; 53: 102318, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35525024

RÉSUMÉ

PURPOSE: Platinum-based chemotherapy with or without immunotherapy is the mainstay of treatment for advanced stage non-small cell lung cancer (NSCLC) lacking a molecular driver alteration. Pre-clinical studies have reported that pharmacological ascorbate (P-AscH-) enhances NSCLC response to platinum-based therapy. We conducted a phase II clinical trial combining P-AscH- with carboplatin-paclitaxel chemotherapy. EXPERIMENTAL DESIGN: Chemotherapy naïve advanced stage NSCLC patients received 75 g ascorbate twice per week intravenously with carboplatin and paclitaxel every three weeks for four cycles. The primary endpoint was to improve tumor response per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 compared to the historical control of 20%. The trial was conducted as an optimal Simon's two-stage design. Blood samples were collected for exploratory analyses. RESULTS: The study enrolled 38 patients and met its primary endpoint with an objective response rate of 34.2% (p = 0.03). All were confirmed partial responses (cPR). The disease control rate was 84.2% (stable disease + cPR). Median progression-free and overall survival were 5.7 months and 12.8 months, respectively. Treatment-related adverse events (TRAE) included one grade 5 (neutropenic fever) and five grade 4 events (cytopenias). Cytokine and chemokine data suggest that the combination elicits an immune response. Immunophenotyping of peripheral blood mononuclear cells demonstrated an increase in effector CD8 T-cells in patients with a progression-free survival (PFS) ≥ 6 months. CONCLUSIONS: The addition of P-AscH- to platinum-based chemotherapy improved tumor response in advanced stage NSCLC. P-AscH- appears to alter the host immune response and needs further investigation as a potential adjuvant to immunotherapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Carboplatine/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Humains , Agranulocytes/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Paclitaxel/usage thérapeutique , Platine/usage thérapeutique
4.
J Immunol ; 208(3): 720-731, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-35022269

RÉSUMÉ

Respiratory infections are a leading cause of morbidity and mortality. The presence of multiple heterologous virus infections is routinely observed in a subset of individuals screened for the presence of respiratory viruses. However, the impact overlapping infections has on disease severity and the host immune response is not well understood. Respiratory syncytial virus (RSV) and influenza A virus (IAV) are two of the most common respiratory infections observed in hospitalized patients, particularly in the very young and aged populations. In this study, we examined how the order in which BALB/c mice were infected with both RSV and IAV impacts disease severity. RSV infection prior to an IAV infection was associated with decreased weight loss and increased survival as compared with IAV infection alone. In contrast, IAV infection prior to an RSV infection was associated with similar morbidity and mortality as compared with an IAV infection alone. Our results suggest that the order in which viral infections are acquired plays a critical role in the outcome of disease severity and the host immune response.


Sujet(s)
Virus de la grippe A/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à virus respiratoire syncytial/immunologie , Virus respiratoires syncytiaux/immunologie , Interférence virale/physiologie , Animaux , Anticorps antiviraux/immunologie , Lymphocytes T CD8+/immunologie , Co-infection/immunologie , Co-infection/virologie , Cytokines/immunologie , Femelle , Interféron de type I/immunologie , Souris , Souris de lignée BALB C , Infections à Orthomyxoviridae/prévention et contrôle
5.
Elife ; 102021 06 18.
Article de Anglais | MEDLINE | ID: mdl-34143731

RÉSUMÉ

Protective lung tissue-resident memory CD8+T cells (Trm) form after influenza A virus (IAV) infection. We show that IAV infection of mice generates CD69+CD103+and other memory CD8+T cell populations in lung-draining mediastinal lymph nodes (mLNs) from circulating naive or memory CD8+T cells. Repeated antigen exposure, mimicking seasonal IAV infections, generates quaternary memory (4M) CD8+T cells that protect mLN from viral infection better than 1M CD8+T cells. Better protection by 4M CD8+T cells associates with enhanced granzyme A/B expression and stable maintenance of mLN CD69+CD103+4M CD8+T cells, vs the steady decline of CD69+CD103+1M CD8+T cells, paralleling the durability of protective CD69+CD103+4M vs 1M in the lung after IAV infection. Coordinated upregulation in canonical Trm-associated genes occurs in circulating 4M vs 1M populations without the enrichment of canonical downregulated Trm genes. Thus, repeated antigen exposure arms circulating memory CD8+T cells with enhanced capacity to form long-lived populations of Trm that enhance control of viral infections of the mLN.


Sujet(s)
Lymphocytes T CD8+ , Noeuds lymphatiques , Animaux , Antigènes CD/génétique , Antigènes CD/immunologie , Antigènes CD/métabolisme , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Cellules cultivées , Femelle , Virus de la grippe A/immunologie , Poumon/cytologie , Poumon/immunologie , Noeuds lymphatiques/cytologie , Noeuds lymphatiques/immunologie , Souris , Souris de lignée C57BL , Infections à Orthomyxoviridae/immunologie , Transcriptome/génétique
6.
Immunohorizons ; 5(2): 59-69, 2021 02 03.
Article de Anglais | MEDLINE | ID: mdl-33536235

RÉSUMÉ

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection in young children. The T cell response plays a critical role in facilitating clearance of an acute RSV infection, and memory T cell responses are vital for protection against secondary RSV exposures. Tissue-resident memory (TRM) T cells have been identified as a subset of memory T cells that reside in nonlymphoid tissues and are critical for providing long-term immunity. There is currently limited information regarding the establishment and longevity of TRM T cell responses elicited following an acute RSV infection as well as their role in protection against repeated RSV infections. In this study, we examined the magnitude, phenotype, and protective capacity of TRM CD4 and CD8 T cells in the lungs of BALB/c mice following an acute RSV infection. TRM CD4 and CD8 T cells were established within the lungs and waned by 149 d following RSV infection. To determine the protective capacity of TRMs, FTY720 administration was used to prevent trafficking of peripheral memory T cells into the lungs prior to challenge of RSV-immune mice, with a recombinant influenza virus expressing either an RSV-derived CD4 or CD8 T cell epitope. We observed enhanced viral clearance in RSV-immune mice, suggesting that TRM CD8 T cells can contribute to protection against a secondary RSV infection. Given the protective capacity of TRMs, future RSV vaccine candidates should focus on the generation of these cell populations within the lung to induce effective immunity against RSV infection.


Sujet(s)
Déterminants antigéniques des lymphocytes T/immunologie , Mémoire immunologique , Vaccins antigrippaux/immunologie , Cellules T mémoire/immunologie , Infections à virus respiratoire syncytial/prévention et contrôle , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Déterminants antigéniques des lymphocytes T/administration et posologie , Déterminants antigéniques des lymphocytes T/génétique , Femelle , Chlorhydrate de fingolimod/pharmacologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/génétique , Poumon/immunologie , Poumon/virologie , Cellules T mémoire/effets des médicaments et des substances chimiques , Souris , Souris de lignée BALB C , Infections à virus respiratoire syncytial/immunologie , Virus respiratoires syncytiaux/immunologie , Vaccins synthétiques/administration et posologie , Vaccins synthétiques/immunologie
7.
PLoS Pathog ; 14(1): e1006810, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29293660

RÉSUMÉ

Memory CD8 T cells can provide protection from re-infection by respiratory viruses such as influenza and SARS. However, the relative contribution of memory CD8 T cells in providing protection against respiratory syncytial virus (RSV) infection is currently unclear. To address this knowledge gap, we utilized a prime-boost immunization approach to induce robust memory CD8 T cell responses in the absence of RSV-specific CD4 T cells and antibodies. Unexpectedly, RSV infection of mice with pre-existing CD8 T cell memory led to exacerbated weight loss, pulmonary disease, and lethal immunopathology. The exacerbated disease in immunized mice was not epitope-dependent and occurred despite a significant reduction in RSV viral titers. In addition, the lethal immunopathology was unique to the context of an RSV infection as mice were protected from a normally lethal challenge with a recombinant influenza virus expressing an RSV epitope. Memory CD8 T cells rapidly produced IFN-γ following RSV infection resulting in elevated protein levels in the lung and periphery. Neutralization of IFN-γ in the respiratory tract reduced morbidity and prevented mortality. These results demonstrate that in contrast to other respiratory viruses, RSV-specific memory CD8 T cells can induce lethal immunopathology despite mediating enhanced viral clearance.


Sujet(s)
Lymphocytes T CD8+/physiologie , Maladies du système immunitaire/immunologie , Maladies du système immunitaire/virologie , Mémoire immunologique , Infections à virus respiratoire syncytial/complications , Infections à virus respiratoire syncytial/immunologie , Animaux , Cellules cultivées , Femelle , Humains , Maladies du système immunitaire/anatomopathologie , Souris , Souris de lignée BALB C , Infections à virus respiratoire syncytial/anatomopathologie , Virus respiratoires syncytiaux/immunologie , Indice de gravité de la maladie
8.
Front Immunol ; 8: 1527, 2017.
Article de Anglais | MEDLINE | ID: mdl-29213267

RÉSUMÉ

Recent work has suggested that current mouse models may underrepresent the complexity of human immune responses. While most mouse immunology studies utilize inbred mouse strains, it is unclear if conclusions drawn from inbred mice can be extended to all mouse strains or generalized to humans. We recently described a "surrogate activation marker" approach that could be used to track polyclonal CD8 T cell responses in inbred and outbred mice and noted substantial discord in the magnitude and kinetics of CD8 T cell responses in individual outbred mice following infection. However, how the memory CD8 T cell response develops following infection and the correlates of memory CD8 T cell-mediated protection against re-infection in outbred mice remains unknown. In this study, we investigated development of pathogen-specific memory CD8 T cell responses in inbred C57B/6 and outbred National Institutes of Health Swiss mice following lymphocytic choriomeningitis virus or L. monocytogenes infection. Interestingly, the size of the memory CD8 T cell pool generated and rate of phenotypic progression was considerably more variable in individual outbred compared to inbred mice. Importantly, while prior infection provided both inbred and outbred cohorts of mice with protection against re-infection that was dependent on the dose of primary infection, levels of memory CD8 T cells generated and degree of protection against re-infection did not correlate with primary infection dose in all outbred mice. While variation in CD8 T cell responses to infection is not entirely surprising due to the genetic diversity present, analysis of infection-induced immunity in outbred hosts may reveal hidden complexity in CD8 T cell responses in genetically diverse populations and might help us further bridge the gap between mouse and human studies.

9.
PLoS Pathog ; 13(9): e1006569, 2017 Sep.
Article de Anglais | MEDLINE | ID: mdl-28910403

RÉSUMÉ

Sepsis is a systemic infection that enhances host vulnerability to secondary infections normally controlled by T cells. Using CLP sepsis model, we observed that sepsis induces apoptosis of circulating memory CD8 T-cells (TCIRCM) and diminishes their effector functions, leading to impaired CD8 T-cell mediated protection to systemic pathogen re-infection. In the context of localized re-infections, tissue resident memory CD8 T-cells (TRM) provide robust protection in a variety of infectious models. TRM rapidly 'sense' infection in non-lymphoid tissues and 'alarm' the host by enhancing immune cell recruitment to the site of the infection to accelerate pathogen clearance. Here, we show that compared to pathogen-specific TCIRCM, sepsis does not invoke significant numerical decline of Vaccinia virus induced skin-TRM keeping their effector functions (e.g., Ag-dependent IFN-γ production) intact. IFN-γ-mediated recruitment of immune cells to the site of localized infection was, however, reduced in CLP hosts despite TRM maintaining their 'sensing and alarming' functions. The capacity of memory CD8 T-cells in the septic environment to respond to inflammatory cues and arrive to the site of secondary infection/antigen exposure remained normal suggesting T-cell-extrinsic factors contributed to the observed lesion. Mechanistically, we showed that IFN-γ produced rapidly during sepsis-induced cytokine storm leads to reduced IFN-γR1 expression on vascular endothelium. As a consequence, decreased expression of adhesion molecules and/or chemokines (VCAM1 and CXCL9) on skin endothelial cells in response to TRM-derived IFN-γ was observed, leading to sub-optimal bystander-recruitment of effector cells and increased susceptibility to pathogen re-encounter. Importantly, as visualized by intravital 2-photon microscopy, exogenous administration of CXCL9/10 was sufficient to correct sepsis-induced impairments in recruitment of effector cells at the localized site of TRM antigen recognition. Thus, sepsis has the capacity to alter skin TRM anamnestic responses without directly impacting TRM number and/or function, an observation that helps to further define the immunoparalysis phase in sepsis survivors.


Sujet(s)
Lymphocytes T CD8+/immunologie , Mémoire immunologique/immunologie , Sepsie/immunologie , Virus de la vaccine/immunologie , Animaux , Antigènes/immunologie , Cytokines/métabolisme , Interféron gamma/biosynthèse , Peau/immunologie
10.
PLoS Pathog ; 13(7): e1006498, 2017 Jul.
Article de Anglais | MEDLINE | ID: mdl-28715493

RÉSUMÉ

Effective CD8 T cell responses are vital for the control of chronic viral infections. Many factors of the host immune response contribute to the maintenance of effector CD8 T cell responses versus CD8 T cell exhaustion during chronic infection. Specific MHC alleles and the degree of MHC heterogeneity are associated with enhanced CD8 T cell function and viral control during human chronic infection. However, it is currently unclear to what extent host genetics influences the establishment of chronic viral infection. In order to examine the impact of MHC heterogeneity versus non-MHC host genetics on the development of chronic viral infection, an F1 cross of B10.D2 x B6 (D2B6F1) and BALB.B x BALB/c (BCF1) mice were infected with the clone-13 (Cl-13) strain of lymphocytic choriomeningitis virus (LCMV). Following chronic Cl-13 infection both H-2bxd D2B6F1 and BCF1 mice demonstrated increased viral control compared to homozygous mice. Strikingly, H-2bxd D2B6F1 mice on a C57BL genetic background exhibited mortality following Cl-13 infection. CD8 T cell depletion prevented mortality in Cl-13-infected D2B6F1 mice indicating that mortality was CD8 T-cell-dependent. D2B6F1 mice maintained more CD8 T cell effector cytokine production and exhibited reduced expression of the T cell exhaustion marker PD-1. In addition, D2B6F1 mice also induced a larger Th1 response than BCF1 mice and Cl-13-induced mortality in D2B6F1 mice was also dependent on CD4 T-cell-mediated IFN-γ production. Thus, following a chronic viral infection, increased functionality of the CD8 T cell response allowed for more rapid viral clearance at the cost of enhanced immunopathology dependent on both MHC diversity and the genetic background of the host.


Sujet(s)
Lymphocytes T CD8+/immunologie , Chorioméningite lymphocytaire/génétique , Virus de la chorioméningite lymphocytaire/physiologie , Animaux , Lymphocytes T CD4+/immunologie , Femelle , Antigènes d'histocompatibilité de classe I/génétique , Antigènes d'histocompatibilité de classe I/immunologie , Humains , Chorioméningite lymphocytaire/immunologie , Chorioméningite lymphocytaire/anatomopathologie , Chorioméningite lymphocytaire/virologie , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL
11.
Respir Res ; 18(1): 136, 2017 07 11.
Article de Anglais | MEDLINE | ID: mdl-28693498

RÉSUMÉ

BACKGROUND: Inhibitors of mTOR, such as sirolimus, have been shown to induce thymus involution and inflammatory lung disease in mice. The latter effect supports the role of this serine/threonine kinase in ameliorating lung inflammation. Other studies have shown sirolimus reduces/delays lung disease associated with various strains of influenza A virus (IAV). Thus, the effects of mTOR inhibitors on influenza infection deserve further studies. METHODS: Here, we examined the changes in lung viral copies, pathology and pulmonary function associated with IAV (A/PR/8/34) infection in mice treated with sirolimus. RESULTS: Body weight loss peaked between days 6-11 post-infection and was more severe in IAV-infected mice that were administered sirolimus as compared to mice that received IAV alone (p = 0.030). Natural log viral gene copies, mean ± SD per mg lung tissue, in IAV-infected mice that were administered sirolimus were 17.31 ± 1.27 on day 4, 19.31 ± 7.46 on day 10, and 0 on day 25. The corresponding number of copies in mice that received IAV alone were 18.56 ± 0.95 on day 4 (p = 0.132), 1.52 ± 1.39 on day 10 (p = 0.008), and 0 on day 25. Lung pathology was evident on days 4, 10, and 25 post infection, with mean ± SD inflammatory score of 9.0 ± 4.5 in IAV-infected mice that were administered sirolimus, as compared to 11.5 ± 4.5 (p = 0.335) in mice received IAV alone (maximum score, 26.0). Impaired lung function was evident in IAV-infected mice on days 4 and 10, as demonstrated by increased airway resistance and decreased compliance. CONCLUSIONS: In this model, the effects of sirolimus on influenza infection included severe weight loss and modified viral replication, respiratory function and lung inflammation. The adverse events associated with sirolimus treatment are consistent with its potent immunosuppressive activity and, thus, preclude its use in IAV infection.


Sujet(s)
Virus de la grippe A/effets des médicaments et des substances chimiques , Grippe humaine/traitement médicamenteux , Poumon/effets des médicaments et des substances chimiques , Sirolimus/usage thérapeutique , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Charge virale/effets des médicaments et des substances chimiques , Animaux , Humains , Immunosuppresseurs/pharmacologie , Immunosuppresseurs/usage thérapeutique , Grippe humaine/anatomopathologie , Poumon/anatomopathologie , Souris , Souris de lignée BALB C , Sirolimus/pharmacologie , Charge virale/physiologie
12.
Nat Immunol ; 18(8): 931-939, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28604718

RÉSUMÉ

Activated CD8+ T cells differentiate into cytotoxic effector (TEFF) cells that eliminate target cells. How TEFF cell identity is established and maintained is not fully understood. We found that Runx3 deficiency limited clonal expansion and impaired upregulation of cytotoxic molecules in TEFF cells. Runx3-deficient CD8+ TEFF cells aberrantly upregulated genes characteristic of follicular helper T (TFH) cell lineage, including Bcl6, Tcf7 and Cxcr5. Mechanistically, the Runx3-CBFß transcription factor complex deployed H3K27me3 to Bcl6 and Tcf7 genes to suppress the TFH program. Ablating Tcf7 in Runx3-deficient CD8+ TEFF cells prevented the upregulation of TFH genes and ameliorated their defective induction of cytotoxic genes. As such, Runx3-mediated Tcf7 repression coordinately enforced acquisition of cytotoxic functions and protected the cytotoxic lineage integrity by preventing TFH-lineage deviation.


Sujet(s)
Sous-unité alpha 3 du facteur CBF/génétique , Lymphopoïèse/génétique , Lymphocytes T cytotoxiques/cytologie , Lymphocytes T auxiliaires/cytologie , Animaux , Lignage cellulaire , Test ELISA , Épigenèse génétique , Régulation de l'expression des gènes , Facteur nucléaire hépatocytaire HNF-1 alpha/génétique , Immunohistochimie , Souris , Protéines proto-oncogènes c-bcl-6/génétique , Récepteurs CXCR5/génétique , RT-PCR , Analyse de séquence d'ARN , Régulation positive
13.
Front Immunol ; 8: 40, 2017.
Article de Anglais | MEDLINE | ID: mdl-28191007

RÉSUMÉ

Unlike systemic infections, little is known about the role of repeated localized infections on (re)shaping pathogen-specific memory CD8 T cell responses. Here, we used primary (1°) and secondary (2°) intranasal influenza virus infections of mice as a model to study intrinsic memory CD8 T cell properties. We show that secondary antigen exposure, relative to a single infection, generates memory CD8 T cell responses of superior magnitude in multiple tissue compartments including blood, spleen, draining lymph nodes, and lung. Unexpectedly, regardless of the significantly higher number of 2° memory CD8 T cells, similar degree of protection against pulmonary challenge was observed in both groups of mice containing 1° or 2° memory CD8 T cells. Mechanistically, using pertussis toxin-induced migration block, we showed that superior antigen-driven proliferation and ability to relocate to the site of infection allowed 1° memory CD8 T cells to accumulate in the infected lung during the first few days after challenge, compensating for the initially lower cell numbers. Taken together, the history of antigen exposures to localized pulmonary infections, through altering basic cell biology, dictates dynamic properties of protective memory CD8 T cell responses. This knowledge has important implications for a design of novel and an improvement of existing vaccines and immunization strategies.

14.
Pediatr Res ; 80(5): 702-709, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27486703

RÉSUMÉ

BACKGROUND: Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in children under 1 y of age in the USA. The host immune response is believed to contribute to RSV-induced disease. We hypothesize that severe RSV infection in infants is mediated by insufficient regulation of the host immune response of regulatory T cells (Tregs) resulting in immunopathology. METHODS: Blood and nasal aspirates from 23 RSV-infected and 17 control infants under 1 y of age were collected. Treg frequencies were determined by flow cytometry from peripheral blood mononuclear cells. Analysis of 24 cytokines was measured by multiplex assay on nasal aspirates. RESULTS: We demonstrate that the frequency of activated Tregs is significantly reduced in the peripheral blood of RSV-infected infants compared with age-matched controls. Surprisingly, T helper (Th)17 related cytokines including interleukin (IL)-1ß, IL-17A, and IL-23 were associated with a reduction in clinical symptoms of respiratory distress. In addition, the amount of IL-33 protein in nasal washes, a cytokine important in maintaining Treg homeostasis in mucosal tissues, was decreased in RSV-infected children. CONCLUSION: These results suggest that decreased Treg numbers and an inability to properly control the host inflammatory response results in severe RSV infection.


Sujet(s)
Cytokines/sang , Infections à virus respiratoire syncytial/immunologie , Lymphocytes T régulateurs/immunologie , Bronchiolite/virologie , Études cas-témoins , Femelle , Humains , Nourrisson , Nouveau-né , Inflammation/sang , Interleukine-17/sang , Interleukine-1 bêta/sang , Sous-unité p19 de l'interleukine-23/sang , Interleukine-33/sang , Agranulocytes/cytologie , Mâle , Muqueuse nasale/immunologie , Pneumopathie infectieuse/virologie , Infections à virus respiratoire syncytial/sang , Virus respiratoire syncytial humain
15.
Virology ; 498: 128-135, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27573069

RÉSUMÉ

Respiratory syncytial virus (RSV) and the common commensal and opportunistic pathogen, non-typeable Haemophilus influenzae (NTHi) both serve as a frequent cause of respiratory infection in children. Although it is well established that some respiratory viruses can increase host susceptibility to secondary bacterial infections, few studies have examined how commensal bacteria could influence a secondary viral response. Here, we examined the impact of NTHi exposure on a subsequent RSV infection of human bronchial epithelial cells (16HBE14o-). Co-culture of 16HBE14o- cells with NTHi resulted in inhibition of viral gene expression following RSV infection. 16HBE14o- cells co-cultured with heat-killed NTHi failed to protect against an RSV infection, indicating that protection requires live bacteria. However, NTHi did not inhibit influenza A virus replication, indicating that NTHi-mediated protection was RSV-specific. Our data demonstrates that prior exposure to a commensal bacterium such as NTHi can elicit protection against a subsequent RSV infection.


Sujet(s)
Antibiose , Co-infection , Haemophilus influenzae/physiologie , Muqueuse respiratoire/microbiologie , Muqueuse respiratoire/virologie , Infections à virus respiratoire syncytial/virologie , Virus respiratoire syncytial humain/physiologie , Lignée cellulaire , Régulation de l'expression des gènes viraux , Humains , Réplication virale
16.
J Clin Invest ; 125(9): 3477-90, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-26241055

RÉSUMÉ

Due to their ability to rapidly proliferate and produce effector cytokines, memory CD8+ T cells increase protection following reexposure to a pathogen. However, low inflammatory immunizations do not provide memory CD8+ T cells with a proliferation advantage over naive CD8+ T cells, suggesting that cell-extrinsic factors enhance memory CD8+ T cell proliferation in vivo. Herein, we demonstrate that inflammatory signals are critical for the rapid proliferation of memory CD8+ T cells following infection. Using murine models of viral infection and antigen exposure, we found that type I IFN-driven expression of IL-15 in response to viral infection prepares memory CD8+ T cells for rapid division independently of antigen reexposure by transiently inducing cell-cycle progression via a pathway dependent on mTOR complex-1 (mTORC1). Moreover, exposure to IL-15 allowed more rapid division of memory CD8+ T cells following antigen encounter and enhanced their protective capacity against viral infection. Together, these data reveal that inflammatory IL-15 promotes optimal responses by memory CD8+ T cells.


Sujet(s)
Lymphocytes T CD8+/immunologie , Division cellulaire/immunologie , Mémoire immunologique , Interleukine-15/immunologie , Animaux , Lymphocytes T CD8+/cytologie , Division cellulaire/génétique , Humains , Interféron de type I/génétique , Interféron de type I/immunologie , Interleukine-15/génétique , Complexe-1 cible mécanistique de la rapamycine , Souris , Souris knockout , Complexes multiprotéiques/génétique , Complexes multiprotéiques/immunologie , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/immunologie
17.
PLoS Pathog ; 11(3): e1004757, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25769044

RÉSUMÉ

There is no currently licensed vaccine for respiratory syncytial virus (RSV) despite being the leading cause of lower respiratory tract infections in children. Children previously immunized with a formalin-inactivated RSV (FI-RSV) vaccine exhibited enhanced respiratory disease following natural RSV infection. Subsequent studies in animal models have implicated roles for CD4 T cells, eosinophils and non-neutralizing antibodies in mediating enhanced respiratory disease. However, the underlying immunological mechanisms responsible for the enhanced respiratory disease and other disease manifestations associated with FI-RSV vaccine-enhanced disease remain unclear. We demonstrate for the first time that while CD4 T cells mediate all aspects of vaccine-enhanced disease, distinct CD4 T cell subsets orchestrate discrete and specific disease parameters. A Th2-biased immune response, but not eosinophils specifically, was required for airway hyperreactivity and mucus hypersecretion. In contrast, the Th1-associated cytokine TNF-α was necessary to mediate airway obstruction and weight loss. Our data demonstrate that individual disease manifestations associated with FI-RSV vaccine-enhanced disease are mediated by distinct subsets of CD4 T cells.


Sujet(s)
Vaccins contre les virus respiratoires syncytiaux/immunologie , Virus respiratoires syncytiaux/immunologie , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th2/immunologie , Facteur de nécrose tumorale alpha/immunologie , Animaux , Humains , Souris , Souris de lignée BALB C , Souris knockout , Facteur de nécrose tumorale alpha/génétique
18.
J Virol ; 88(16): 9010-6, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24899187

RÉSUMÉ

UNLABELLED: The migration of pathogen-specific T cells into nonlymphoid tissues, such as the lung, is critical to control peripheral infections. Use of in vivo intravascular labeling of leukocytes has allowed for improved discrimination between cells located in the blood from cells present within peripheral tissues, such as the lung. This is particularly important in the lung, which is comprised of an intricate network of blood vessels that harbors a large proportion of the total blood volume at any given time. Recent work has demonstrated that >80% of antigen-specific effector CD8 T cells remain in the pulmonary vasculature following an intratracheal infection with a systemic viral pathogen. However, it remains unclear what proportion of effector CD8 T cells are located within lung tissue following a localized respiratory viral infection. We confirm that most effector and memory CD8 T cells are found in the vasculature after an intranasal infection with the systemic pathogens lymphocytic choriomeningitis virus (LCMV) or vaccinia virus (VACV). In contrast, following pulmonary viral infections with either respiratory syncytial virus (RSV) or influenza A virus (IAV), 80 to 90% of the antigen-specific effector CD8 T cells were located within lung tissue. Similarly, the majority of antigen-specific CD4 T cells were present within lung tissue during a pulmonary viral infection. Furthermore, a greater proportion of gamma interferon-positive (IFN-γ(+)) effector CD8 and CD4 T cells were located within lung tissue following a localized respiratory viral infection. Our results indicate that T cells exhibit significantly altered distribution patterns dependent upon the tissue tropism of the infection. IMPORTANCE: The migration of T cells to nonlymphoid sites, such as the lung, is critical to mediate clearance of viral infections. The highly vascularized lung holds up to 40% of blood, and thus, the T cell response may be a reflection of lymphocytes localized to the pulmonary vasculature instead of lung tissue. We examined the localization of T cell responses within the lung following either a localized or systemic viral infection. We demonstrate that following intranasal infection with a systemic pathogen, most T cells are localized to the pulmonary vasculature. In contrast, T cells are primarily localized to lung tissue following a respiratory viral infection. Our results demonstrate vast differences in the localization of T cell responses within the lung parenchyma between pathogens that can replicate locally versus systemically and that intravascular antibody labeling can be utilized to assess the localization patterns of T cell responses in nonlymphoid organs.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/virologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/virologie , Poumon/immunologie , Tropisme/immunologie , Animaux , Virus de la grippe A/immunologie , Interféron gamma/immunologie , Poumon/virologie , Virus de la chorioméningite lymphocytaire/immunologie , Souris , Souris de lignée C57BL , Infections à virus respiratoire syncytial/immunologie , Infections à virus respiratoire syncytial/virologie , Virus respiratoires syncytiaux/immunologie , Infections de l'appareil respiratoire/immunologie , Infections de l'appareil respiratoire/virologie
19.
Virology ; 454-455: 263-9, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24725953

RÉSUMÉ

We have previously reported that lung cellular bioenergetics (cellular respiration and ATP) increased in 4-10 week-old BALB/c mice infected with respiratory syncytial virus (RSV). This study examined the kinetics and changes in cellular bioenergetics in ≤ 2-week-old C57BL/6 mice following RSV infection. Mice (5-14 days old) were inoculated intranasally with RSV and the lungs were examined on days 1-10 post-infection. Histopathology and electron microscopy revealed preserved pneumocyte architectures and organelles. Increased lung cellular bioenergetics was noted from days 1-10 post-infection. Cellular GSH remained unchanged. These results indicate that the increased lung cellular respiration (measured by mitochondrial O2 consumption) and ATP following RSV infection is independent of either age or genetic background of the host.


Sujet(s)
Métabolisme énergétique , Poumon/physiopathologie , Poumon/virologie , Infections à virus respiratoire syncytial/physiopathologie , Infections à virus respiratoire syncytial/virologie , Virus respiratoires syncytiaux/physiologie , Adénosine triphosphate/biosynthèse , Animaux , Animaux nouveau-nés , Histocytochimie , Souris , Souris de lignée C57BL , Microscopie électronique , Consommation d'oxygène
20.
PLoS One ; 9(3): e90720, 2014.
Article de Anglais | MEDLINE | ID: mdl-24608125

RÉSUMÉ

Coronaviruses cause respiratory disease in humans that can range from mild to severe. However, the pathogenesis of pulmonary coronavirus infections is poorly understood. Mouse hepatitis virus type 1 (MHV-1) is a group 2 coronavirus capable of causing severe morbidity and mortality in highly susceptible C3H/HeJ mice. We have previously shown that both CD4 and CD8 T cells play a critical role in mediating MHV-1-induced disease. Here we evaluated the role of alveolar macrophages (AM) in modulating the adaptive immune response and subsequent disease. Depletion of AM using clodronate liposomes administered prior to MHV-1 infection was associated with a significant amelioration of MHV-1-induced morbidity and mortality. AM depletion resulted in a decreased number of virus-specific CD4 T cells in the lung airways. In addition, a significant increase in the frequency and total number of Tregs in the lung tissue and lung airways was observed following MHV-1 infection in mice depleted of AM. Our results indicate that AM play a critical role in modulating MHV-1-induced morbidity and mortality.


Sujet(s)
Infections à coronavirus/immunologie , Macrophages alvéolaires/immunologie , Animaux , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/métabolisme , Cellules cultivées , Coronavirus , Femelle , Poumon/immunologie , Poumon/virologie , Souris , Souris de lignée C3H , Virus de l'hépatite murine/pathogénicité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE