Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 12(1): 6664, 2021 11 18.
Article de Anglais | MEDLINE | ID: mdl-34795263

RÉSUMÉ

Crohn's disease is an inflammatory disease of the gastrointestinal tract characterized by an aberrant response to microbial and environmental triggers. This includes an altered microbiome dominated by Enterobacteriaceae and in particular adherent-invasive E. coli (AIEC). Clinical evidence implicates periods of psychological stress in Crohn's disease exacerbation, and disturbances in the gut microbiome might contribute to the pathogenic mechanism. Here we show that stress-exposed mice develop ileal dysbiosis, dominated by the expansion of Enterobacteriaceae. In an AIEC colonisation model, stress-induced glucocorticoids promote apoptosis of CD45+CD90+ cells that normally produce IL-22, a cytokine that is essential for the maintenance of ileal mucosal barrier integrity. Blockade of glucocorticoid signaling or administration of recombinant IL-22 restores mucosal immunity, prevents ileal dysbiosis, and blocks AIEC expansion. We conclude that psychological stress impairs IL-22-driven protective immunity in the gut, which creates a favorable niche for the expansion of pathobionts that have been implicated in Crohn's disease. Importantly, this work also shows that immunomodulation can counteract the negative effects of psychological stress on gut immunity and hence disease-associated dysbiosis.


Sujet(s)
Dysbiose/immunologie , Microbiome gastro-intestinal/immunologie , Immunité muqueuse/immunologie , Interleukines/immunologie , Muqueuse intestinale/immunologie , Stress psychologique/immunologie , Animaux , Adhérence bactérienne/immunologie , Maladie de Crohn/immunologie , Maladie de Crohn/microbiologie , Dysbiose/microbiologie , Enterobacteriaceae/classification , Enterobacteriaceae/génétique , Enterobacteriaceae/immunologie , Escherichia coli/immunologie , Escherichia coli/physiologie , Infections à Escherichia coli/immunologie , Infections à Escherichia coli/microbiologie , Microbiome gastro-intestinal/génétique , Humains , Iléum/immunologie , Iléum/microbiologie , Iléum/anatomopathologie , Interleukines/métabolisme , Mâle , Souris de lignée C57BL , Récepteurs aux glucocorticoïdes/immunologie , Récepteurs aux glucocorticoïdes/métabolisme , Antigènes Thy-1/immunologie , Antigènes Thy-1/métabolisme ,
2.
Am J Physiol Endocrinol Metab ; 321(3): E338-E350, 2021 09 01.
Article de Anglais | MEDLINE | ID: mdl-34280051

RÉSUMÉ

Obesity is associated with metabolic, immunological, and infectious disease comorbidities, including an increased risk of enteric infection and inflammatory bowel disease such as Crohn's disease (CD). Expansion of intestinal pathobionts such as adherent-invasive Escherichia coli (AIEC) is a common dysbiotic feature of CD, which is amplified by prior use of oral antibiotics. Although high-fat, high-sugar diets are associated with dysbiotic expansion of E. coli, it is unknown if the content of fat or another dietary component in obesogenic diets is sufficient to promote AIEC expansion. Here, we found that administration of an antibiotic combined with feeding mice an obesogenic low-fiber, high-sucrose, high-fat diet (HFD) that is typically used in rodent-obesity studies promoted AIEC intestinal expansion. Even a short-term (i.e., 1 day) pulse of HFD feeding before infection was sufficient to promote AIEC expansion, indicating that the magnitude of obesity was not the main driver of AIEC expansion. Controlled-diet experiments demonstrated that neither dietary fat nor sugar were the key determinants of AIEC colonization, but that lowering dietary fiber from approximately 13% to 5%-6% was sufficient to promote the intestinal expansion of AIEC when combined with antibiotics in mice. When combined with antibiotics, lowering fiber promoted AIEC intestinal expansion to a similar extent as widely used HFDs in mice. However, lowering dietary fiber was sufficient to promote AIEC intestinal expansion without affecting body mass. Our results show that low dietary fiber combined with oral antibiotics are environmental factors that promote the expansion of Crohn's disease-associated pathobionts in the gut.NEW & NOTEWORTHY It is commonly thought that obesity or a high-fat diet alters pathogenic bacteria and promotes inflammatory gut diseases. We found that lower dietary fiber is a key factor that expands a gut pathobiont linked to Crohn's disease, independent of obesity status in mice.


Sujet(s)
Maladie de Crohn/microbiologie , Fibre alimentaire/administration et posologie , Intestins/microbiologie , Obésité/microbiologie , Animaux , Escherichia coli/physiologie , Mâle , Souris , Souris de lignée C57BL
3.
J Mol Biol ; 431(16): 2970-2981, 2019 07 26.
Article de Anglais | MEDLINE | ID: mdl-31029703

RÉSUMÉ

Escherichia coli is one of the most genetically and phenotypically diverse species of bacteria. This remarkable diversity produces a plethora of clinical outcomes following infection and has informed much of what we currently know about host-pathogen interactions for a wide range of bacteria-host relationships. In studying the role of microbes in disease, adherent-invasive E. coli (AIEC) has emerged as having a strong association with Crohn's disease (CD). Thus, there has been an equally strong effort to uncover the root origins of AIEC, to appreciate how AIEC differs from other well-known pathogenic E. coli variants, and to understand its connection to disease. Emerging from a growing body of research on AIEC is the understanding that AIEC itself is remarkably diverse, both in phylogenetic origins, genetic makeup, and behavior in the host setting. Here, we describe the unique lifestyle of CD-associated AIEC and review recent research that is uncovering the inextricable link between AIEC and its host in the context of CD.


Sujet(s)
Maladie de Crohn/microbiologie , Infections à Escherichia coli/microbiologie , Escherichia coli/pathogénicité , Adhérence bactérienne , Escherichia coli/classification , Humains , Muqueuse intestinale/microbiologie , Phylogenèse
4.
PLoS Negl Trop Dis ; 12(8): e0006701, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-30133440

RÉSUMÉ

Leishmaniasis is a serious global health problem affecting many people worldwide. While patients with leishmaniasis can be treated with several agents, drug toxicicty and the emergence of resistant strains render available treatments ineffective in the long run. Inhibitors of the mammalian target of rapamycin (mTOR) have been demonstrated to exert anti-pathogen properties. In this study, we tested the therapeutic efficacy of several mTOR inhibitors in controlling infection with Leishmania major. Rapamycin, GSK-2126458 and KU-0063794 were administered to BALB/c mice, which had received an intrafootpad injection of the parasite. Footpad swelling and parasite burden were assessed, and cytokine production by mouse splenocytes and phenotypic changes in draining lymph node cells were evaluated. Treatment with a clinically relevant dose of rapamycin or with GSK-2126458, but not with KU-0063794, dramatically lowered both the footpad swelling and the parasite load in the draining lymph node. Importantly, the employed dose of rapamycin did not kill the promastigotes in vitro as judged by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and electron microscopy. Moreover, the IL-4 production capacity of splenocytes harvested from infected mice that were treated with rapamycin was significantly reduced. Consequently, the IFN-γ:IL-4 production ratio was elevated, suggesting a T helper-type 1 (Th1)-skewed cytokine profile. Finally, the expression level of CD69, an early activation marker, on splenic and lymph node CD4+ and CD8+ T cells was enhanced in rapamycin-treated mice. Taken together, our findings suggest that select mTOR inhibitors may be used in therapeutic settings for the management of leishmaniasis. We propose that the beneficial effects of such inhibitors stem from their immunomodulatory properties. Therefore, the adjuvanticity of mTOR inhibitors may also be considered in vaccination strategies against Leishmania species.


Sujet(s)
Leishmaniose/traitement médicamenteux , Morpholines/usage thérapeutique , Pyrimidines/usage thérapeutique , Quinoléines/usage thérapeutique , Sulfonamides/usage thérapeutique , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Animaux , Antiprotozoaires/usage thérapeutique , Antienzymes/usage thérapeutique , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Concentration inhibitrice 50 , Souris , Souris de lignée BALB C , Pyridazines , Sirolimus/pharmacologie
5.
Cancer Immunol Immunother ; 67(12): 1885-1896, 2018 Dec.
Article de Anglais | MEDLINE | ID: mdl-29470597

RÉSUMÉ

Mucosa-associated invariant T (MAIT) cells are a subset of innate-like T lymphocytes known for their ability to respond to MHC-related protein 1 (MR1)-restricted stimuli and select cytokine signals. They are abundant in humans and especially enriched in mucosal layers, common sites of neoplastic transformation. MAIT cells have been found within primary and metastatic tumors. However, whether they promote malignancy or contribute to anticancer immunity is unclear. On the one hand, MAIT cells produce IL-17A in certain locations and under certain circumstances, which could in turn facilitate neoangiogenesis, intratumoral accumulation of immunosuppressive cell populations, and cancer progression. On the other hand, they can express a potent arsenal of cytotoxic effector molecules, NKG2D and IFN-γ, all of which have established roles in cancer immune surveillance. In this review, we highlight MAIT cells' characteristics as they might pertain to cancer initiation, progression, or control. We discuss recent findings, including our own, that link MAIT cells to cancer, with a focus on colorectal carcinoma, as well as some of the outstanding questions in this active area of research. Finally, we provide a hypothetical picture in which MAIT cells constitute attractive targets in cancer immunotherapy.


Sujet(s)
Cellules T invariantes associées aux muqueuses/immunologie , Cellules T invariantes associées aux muqueuses/métabolisme , Tumeurs/immunologie , Animaux , Marqueurs biologiques , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/métabolisme , Cytokines/métabolisme , Humains , Surveillance immunologique , Immunophénotypage , Activation des lymphocytes , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Lymphocytes TIL/anatomopathologie , Cellules T invariantes associées aux muqueuses/anatomopathologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Sous-populations de lymphocytes T/anatomopathologie
6.
J Immunol ; 199(9): 3348-3359, 2017 11 01.
Article de Anglais | MEDLINE | ID: mdl-28939757

RÉSUMÉ

The interactions between programmed death-1 (PD-1) and its ligands hamper tumor-specific CD8+ T cell (TCD8) responses, and PD-1-based "checkpoint inhibitors" have shown promise in certain cancers, thus revitalizing interest in immunotherapy. PD-1-targeted therapies reverse TCD8 exhaustion/anergy. However, whether they alter the epitope breadth of TCD8 responses remains unclear. This is an important question because subdominant TCD8 are more likely than immunodominant clones to escape tolerance mechanisms and may contribute to protective anticancer immunity. We have addressed this question in an in vivo model of TCD8 responses to well-defined epitopes of a clinically relevant oncoprotein, large T Ag. We found that unlike other coinhibitory molecules (CTLA-4, LAG-3, TIM-3), PD-1 was highly expressed by subdominant TCD8, which correlated with their propensity to favorably respond to PD-1/PD-1 ligand-1 (PD-L1)-blocking Abs. PD-1 blockade increased the size of subdominant TCD8 clones at the peak of their primary response, and it also sustained their presence, thus giving rise to an enlarged memory pool. The expanded population was fully functional as judged by IFN-γ production and MHC class I-restricted cytotoxicity. The selective increase in subdominant TCD8 clonal size was due to their enhanced survival, not proliferation. Further mechanistic studies utilizing peptide-pulsed dendritic cells, recombinant vaccinia viruses encoding full-length T Ag or epitope mingenes, and tumor cells expressing T Ag variants revealed that anti-PD-1 invigorates subdominant TCD8 responses by relieving their lysis-dependent suppression by immunodominant TCD8 To our knowledge, our work constitutes the first report that interfering with PD-1 signaling potentiates epitope spreading in tumor-specific responses, a finding with clear implications for cancer immunotherapy and vaccination.


Sujet(s)
Lymphocytes T CD8+/immunologie , Épitopes/immunologie , Immunité cellulaire , Protéines tumorales/immunologie , Tumeurs expérimentales/immunologie , Récepteur-1 de mort cellulaire programmée/immunologie , Transduction du signal/immunologie , Animaux , Lymphocytes T CD8+/anatomopathologie , Mort cellulaire/génétique , Mort cellulaire/immunologie , Lignée cellulaire tumorale , Épitopes/génétique , Femelle , Interféron gamma/génétique , Interféron gamma/immunologie , Souris , Protéines tumorales/génétique , Tumeurs expérimentales/génétique , Récepteur-1 de mort cellulaire programmée/génétique , Transduction du signal/génétique
7.
Cancer Immunol Immunother ; 66(12): 1563-1575, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-28798979

RÉSUMÉ

Mucosa-associated invariant T (MAIT) cells are innate-like T lymphocytes that are unusually abundant in the human liver, a common site of colorectal carcinoma (CRC) metastasis. However, whether they contribute to immune surveillance against colorectal liver metastasis (CRLM) is essentially unexplored. In addition, whether MAIT cell functions can be impacted by chemotherapy is unclear. These are important questions given MAIT cells' potent immunomodulatory and inflammatory properties. Herein, we examined the frequencies and functions of peripheral blood, healthy liver tissue, tumor-margin and tumor-infiltrating MAIT cells in 21 CRLM patients who received no chemotherapy, FOLFOX, or a combination of FOLFOX and Avastin before they underwent liver resection. We found that MAIT cells, defined as CD3ε+Vα7.2+CD161++ or CD3ε+MR1 tetramer+ cells, were present within both healthy and tumor-afflicted hepatic tissues. Paired and grouped analyses of samples revealed the physical proximity of MAIT cells to metastatic lesions to drastically influence their functional competence. Accordingly, unlike those residing in the healthy liver compartment, tumor-infiltrating MAIT cells failed to produce IFN-γ in response to a panel of TCR and cytokine receptor ligands, and tumor-margin MAIT cells were only partially active. Furthermore, chemotherapy did not account for intratumoral MAIT cell insufficiencies. Our findings demonstrate for the first time that CRLM-penetrating MAIT cells exhibit wide-ranging functional impairments, which are dictated by their physical location but not by preoperative chemotherapy. Therefore, we propose that MAIT cells may provide an attractive therapeutic target in CRC and that their ligands may be combined with chemotherapeutic agents to treat CRLM.


Sujet(s)
Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Tumeurs du foie/immunologie , Tumeurs du foie/secondaire , Cellules T invariantes associées aux muqueuses/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Humains , Immunité muqueuse , Mâle , Adulte d'âge moyen , Cellules T invariantes associées aux muqueuses/anatomopathologie , Métastase tumorale , Microenvironnement tumoral
8.
PLoS Biol ; 15(6): e2001930, 2017 Jun.
Article de Anglais | MEDLINE | ID: mdl-28632753

RÉSUMÉ

Superantigens (SAgs) are potent exotoxins secreted by Staphylococcus aureus and Streptococcus pyogenes. They target a large fraction of T cell pools to set in motion a "cytokine storm" with severe and sometimes life-threatening consequences typically encountered in toxic shock syndrome (TSS). Given the rapidity with which TSS develops, designing timely and truly targeted therapies for this syndrome requires identification of key mediators of the cytokine storm's initial wave. Equally important, early host responses to SAgs can be accompanied or followed by a state of immunosuppression, which in turn jeopardizes the host's ability to combat and clear infections. Unlike in mouse models, the mechanisms underlying SAg-associated immunosuppression in humans are ill-defined. In this work, we have identified a population of innate-like T cells, called mucosa-associated invariant T (MAIT) cells, as the most powerful source of pro-inflammatory cytokines after exposure to SAgs. We have utilized primary human peripheral blood and hepatic mononuclear cells, mouse MAIT hybridoma lines, HLA-DR4-transgenic mice, MAIThighHLA-DR4+ bone marrow chimeras, and humanized NOD-scid IL-2Rγnull mice to demonstrate for the first time that: i) mouse and human MAIT cells are hyperresponsive to SAgs, typified by staphylococcal enterotoxin B (SEB); ii) the human MAIT cell response to SEB is rapid and far greater in magnitude than that launched by unfractionated conventional T, invariant natural killer T (iNKT) or γδ T cells, and is characterized by production of interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-2, but not IL-17A; iii) high-affinity MHC class II interaction with SAgs, but not MHC-related protein 1 (MR1) participation, is required for MAIT cell activation; iv) MAIT cell responses to SEB can occur in a T cell receptor (TCR) Vß-specific manner but are largely contributed by IL-12 and IL-18; v) as MAIT cells are primed by SAgs, they also begin to develop a molecular signature consistent with exhaustion and failure to participate in antimicrobial defense. Accordingly, they upregulate lymphocyte-activation gene 3 (LAG-3), T cell immunoglobulin and mucin-3 (TIM-3), and/or programmed cell death-1 (PD-1), and acquire an anergic phenotype that interferes with their cognate function against Klebsiella pneumoniae and Escherichia coli; vi) MAIT cell hyperactivation and anergy co-utilize a signaling pathway that is governed by p38 and MEK1/2. Collectively, our findings demonstrate a pathogenic, rather than protective, role for MAIT cells during infection. Furthermore, we propose a novel mechanism of SAg-associated immunosuppression in humans. MAIT cells may therefore provide an attractive therapeutic target for the management of both early and late phases of severe SAg-mediated illnesses.


Sujet(s)
Antigènes bactériens/toxicité , Anergie clonale , Modèles immunologiques , Cellules T invariantes associées aux muqueuses/immunologie , Staphylococcus aureus/immunologie , Streptococcus pyogenes/immunologie , Superantigènes/toxicité , Animaux , Antigènes bactériens/métabolisme , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/immunologie , Cellules de la moelle osseuse/métabolisme , Lignée cellulaire , Cellules cultivées , Anergie clonale/effets des médicaments et des substances chimiques , Croisements génétiques , Entérotoxines/métabolisme , Entérotoxines/toxicité , Femelle , Humains , Hybridomes , Immunité innée , Agranulocytes/cytologie , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/immunologie , Agranulocytes/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Souris , Souris de lignée NOD , Souris knockout , Souris SCID , Souris transgéniques , Cellules T invariantes associées aux muqueuses/cytologie , Cellules T invariantes associées aux muqueuses/effets des médicaments et des substances chimiques , Cellules T invariantes associées aux muqueuses/métabolisme , Organismes exempts d'organismes pathogènes spécifiques , Staphylococcus aureus/métabolisme , Streptococcus pyogenes/métabolisme , Superantigènes/métabolisme , Chimère obtenue par transplantation/sang , Chimère obtenue par transplantation/immunologie , Chimère obtenue par transplantation/métabolisme
10.
PLoS Pathog ; 12(5): e1005589, 2016 05.
Article de Anglais | MEDLINE | ID: mdl-27148737

RÉSUMÉ

Clinical and experimental studies have shown that estradiol (E2) confers protection against HIV and other sexually transmitted infections. Here, we investigated the underlying mechanism. Better protection in E2-treated mice, immunized against genital HSV-2, coincided with earlier recruitment and higher proportions of Th1 and Th17 effector cells in the vagina post-challenge, compared to placebo-treated controls. Vaginal APCs isolated from E2-treated mice induced 10-fold higher Th17 and Th1 responses, compared to APCs from progesterone-treated, placebo-treated, and estradiol-receptor knockout mice in APC-T cell co-cultures. CD11c+ DCs in the vagina were the predominant APC population responsible for priming these Th17 responses, and a potent source of IL-6 and IL-1ß, important factors for Th17 differentiation. Th17 responses were abrogated in APC-T cell co-cultures containing IL-1ß KO, but not IL-6 KO vaginal DCs, showing that IL-1ß is a critical factor for Th17 induction in the genital tract. E2 treatment in vivo directly induced high expression of IL-1ß in vaginal DCs, and addition of IL-1ß restored Th17 induction by IL-1ß KO APCs in co-cultures. Finally, we examined the role of IL-17 in anti-HSV-2 memory T cell responses. IL-17 KO mice were more susceptible to intravaginal HSV-2 challenge, compared to WT controls, and vaginal DCs from these mice were defective at priming efficient Th1 responses in vitro, indicating that IL-17 is important for the generation of efficient anti-viral memory responses. We conclude that the genital mucosa has a unique microenvironment whereby E2 enhances CD4+ T cell anti-viral immunity by priming vaginal DCs to induce Th17 responses through an IL-1-dependent pathway.


Sujet(s)
Lymphocytes T CD4+/immunologie , Cellules dendritiques/immunologie , Oestradiol/immunologie , Cellules Th17/immunologie , Vagin/immunologie , Animaux , Techniques de coculture , Cytokines/biosynthèse , Modèles animaux de maladie humaine , Test ELISA , Oestradiol/pharmacologie , Femelle , Cytométrie en flux , Herpès génital/immunologie , Herpèsvirus humain de type 2/immunologie , Interleukine-1/immunologie , Activation des lymphocytes/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Vagin/virologie
11.
Front Immunol ; 6: 401, 2015.
Article de Anglais | MEDLINE | ID: mdl-26322041

RÉSUMÉ

Dysregulated immune responses to infection, such as those encountered in sepsis, can be catastrophic. Sepsis is typically triggered by an overwhelming systemic response to an infectious agent(s) and is associated with high morbidity and mortality even under optimal critical care. Recent studies have implicated unconventional, innate-like T lymphocytes, including CD1d- and MR1-restricted T cells as effectors and/or regulators of inflammatory responses during sepsis. These cell types are typified by invariant natural killer T (iNKT) cells, variant NKT (vNKT) cells, and mucosa-associated invariant T (MAIT) cells. iNKT and vNKT cells are CD1d-restricted, lipid-reactive cells with remarkable immunoregulatory properties. MAIT cells participate in antimicrobial defense, and are restricted by major histocompatibility complex-related protein 1 (MR1), which displays microbe-derived vitamin B metabolites. Importantly, NKT and MAIT cells are rapid and potent producers of immunomodulatory cytokines. Therefore, they may be considered attractive targets during the early hyperinflammatory phase of sepsis when immediate interventions are urgently needed, and also in later phases when adjuvant immunotherapies could potentially reverse the dangerous state of immunosuppression. We will highlight recent findings that point to the significance or the therapeutic potentials of NKT and MAIT cells in sepsis and will also discuss what lies ahead in research in this area.

12.
Eur J Immunol ; 44(5): 1375-86, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24519467

RÉSUMÉ

The immune mechanisms underlying delayed induction of Th1-type immunity in the lungs following pulmonary mycobacterial infection remain poorly understood. We have herein investigated the underlying immune mechanisms for such delayed responses and whether a selected innate immune-modulating strategy can accelerate Th1-type responses. We have found that, in the early stage of pulmonary infection with attenuated Mycobacterium tuberculosis (M.tb H37Ra), the levels of infection in the lung continue to increase logarithmically until days 14 and 21 postinfection in C57BL/6 mice. The activation of innate immune responses, particularly DCs, in the lung is delayed. This results in a delay in the subsequent downstream immune responses including the migration of antigen-bearing DCs to the draining lymph node (dLN), the Th1-cell priming in dLN, and the recruitment of Th1 cells to the lung. However, single lung mucosal exposure to the TLR agonist FimH postinfection is able to accelerate protective Th1-type immunity via facilitating DC migration to the lung and draining lymph nodes, enhancing DC antigen presentation and Th1-cell priming. These findings hold implications for the development of immunotherapeutic and vaccination strategies and suggest that enhancement of early innate immune activation is a viable option for improving Th1-type immunity against pulmonary mycobacterial diseases.


Sujet(s)
Cellules dendritiques/immunologie , Immunité innée , Poumon/immunologie , Mycobacterium tuberculosis/immunologie , Lymphocytes auxiliaires Th1/immunologie , Tuberculose pulmonaire/immunologie , Animaux , Cellules dendritiques/microbiologie , Cellules dendritiques/anatomopathologie , Poumon/microbiologie , Poumon/anatomopathologie , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/microbiologie , Noeuds lymphatiques/anatomopathologie , Souris , Souris transgéniques , Lymphocytes auxiliaires Th1/anatomopathologie , Facteurs temps , Tuberculose pulmonaire/génétique , Tuberculose pulmonaire/anatomopathologie
13.
PLoS One ; 8(3): e59185, 2013.
Article de Anglais | MEDLINE | ID: mdl-23527127

RÉSUMÉ

Pulmonary tuberculosis (TB), caused by Mycobacterium tuberculosis, is the leading cause of death due to a bacterial pathogen. Emerging epidemiologic evidence suggests that the leading risk factor associated with TB mortality is cigarette smoke exposure. Despite this, it remains poorly understood what is the effect of cigarette smoke exposure on anti-TB immunity and whether its potential detrimental effect can be reversed by cigarette smoking cessation. In our current study, we have investigated the impact of both continuous and discontinuous cigarette smoke exposure on the development of anti-mycobacterial type 1 immunity in murine models. We find that while continuous cigarette smoke exposure severely impairs type 1 immunity in the lung, a short-term smoking cessation allows rapid restoration of anti-mycobacterial immunity. The ability of continuous cigarette smoke exposure to dampen type 1 protective immunity is attributed locally to its affects on innate immune cells in the lung. Continuous cigarette smoke exposure locally, by not systemically, impairs APC accumulation and their production of TNF, IL-12, and RANTES, blunts the recruitment of CD4+IFN-γ+ T cells to the lung, and weakens the formation of granuloma. On the other hand, smoking cessation was found to help restore type 1 immunity by rapidly improving the functionality of lung APCs, enhancing the recruitment of CD4+IFN-γ+ T cells to the lung, and promoting the formation of granuloma. Our study for the first time demonstrates that continuous, but not discontinuous, cigarette smoke exposure severely impedes the lung expression of anti-TB Th1 immunity via inhibiting innate immune activation and lung T cell recruitment. Our findings thus suggest cigarette smoking cessation to be beneficial to the control of pulmonary TB.


Sujet(s)
Cellules présentatrices d'antigène/immunologie , Immunité cellulaire/effets des médicaments et des substances chimiques , Arrêter de fumer , Fumer/effets indésirables , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Tuberculose pulmonaire/immunologie , Animaux , Lymphocytes T CD4+/immunologie , Test ELISA , Femelle , Poumon/microbiologie , Poumon/anatomopathologie , Souris , Souris de lignée C57BL , Monoxyde d'azote/métabolisme , Lymphocytes auxiliaires Th1/immunologie
14.
Front Immunol ; 4: 30, 2013.
Article de Anglais | MEDLINE | ID: mdl-23420646

RÉSUMÉ

Pulmonary tuberculosis, caused by Mycobacterium tuberculosis (M.tb) represents a leading global health concern, with 8.7 million newly emerging cases, and 1.4 million reported deaths annually. Despite an estimated one third of the world's population being infected, relatively few infected individuals ever develop active clinical disease. The ability of the host to remain latently infected while preventing disease is thought to be due to the generation of a robust type 1 immune response in the lung, capable of controlling, but not clearing, M.tb. A key feature of the type 1 immune response to M.tb is the formation of immune cellular aggregates termed granuloma. The granuloma structure has long been considered a hallmark of host's protective response toward M.tb. Historically, a correlative relationship between granuloma formation/maintenance and bacterial control has been seen in models where disrupted granuloma formation or structure was found to be fatal. Despite this established relationship much about the granuloma's role in M.tb immunity remains unknown. Recent publications suggest that the granuloma actually aids the persistence of M.tb and that the development of a necrotic granuloma is essential to person-to-person transmission. Our group and others have recently demonstrated that enclosed within the granuloma is a population of immunologically altered antigen-presenting cells and T lymphocyte populations. Of note, the ability of these populations to produce type 1 cytokines such as interferon-gamma, and bactericidal products including nitric oxide, are significantly reduced, while remaining competent to produce high levels immunosuppressive interleukin-10. These observations indicate that although the chronic granuloma represents a highly unique environment, it is more similar to that of a tumor than an active site of bacterial control. In this review we will explore what is known about this unique environment and its contribution to the persistence of M.tb.

15.
Clin Dev Immunol ; 2012: 628293, 2012.
Article de Anglais | MEDLINE | ID: mdl-22545059

RÉSUMÉ

Mycobacterium tuberculosis (M.tb), the causative bacterium of pulmonary tuberculosis (TB), is a serious global health concern. Central to M.tb effective immune avoidance is its ability to modulate the early innate inflammatory response and prevent the establishment of adaptive T-cell immunity for nearly three weeks. When compared with other intracellular bacterial lung pathogens, such as Legionella pneumophila, or even closely related mycobacterial species such as M. smegmatis, this delay is astonishing. Customarily, the alveolar macrophage (AM) acts as a sentinel, detecting and alerting surrounding cells to the presence of an invader. However, in the case of M.tb, this may be impaired, thus delaying the recruitment of antigen-presenting cells (APCs) to the lung. Upon uptake by APC populations, M.tb is able to subvert and delay the processing of antigen, MHC class II loading, and the priming of effector T cell populations. This delay ultimately results in the deferred recruitment of effector T cells to not only the lung interstitium but also the airway lumen. Therefore, it is of upmost importance to dissect the mechanisms that contribute to the delayed onset of immune responses following M.tb infection. Such knowledge will help design the most effective vaccination strategies against pulmonary TB.


Sujet(s)
Échappement immunitaire , Poumon/immunologie , Mycobacterium tuberculosis/immunologie , Lymphocytes T/immunologie , Tuberculose pulmonaire/immunologie , Immunité acquise , Cellules présentatrices d'antigène/immunologie , Cellules présentatrices d'antigène/microbiologie , Mouvement cellulaire/immunologie , Humains , Immunité innée , Poumon/microbiologie , Poumon/anatomopathologie , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/microbiologie , Mycobacterium smegmatis/immunologie , Mycobacterium tuberculosis/pathogénicité , Lymphocytes T/microbiologie , Facteurs temps , Vaccins antituberculeux/immunologie , Tuberculose pulmonaire/microbiologie , Tuberculose pulmonaire/anatomopathologie
16.
J Immunol ; 187(8): 4280-92, 2011 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-21908731

RÉSUMÉ

Immunopathology is a major cause of influenza-associated morbidity and mortality worldwide. However, the role and regulatory mechanisms of CD4 T cells in severe lung immunopathology following acute influenza infection are poorly understood. In this paper, we report that the emergence of immunopathogenic CD4 T cells is under the control of a transmembrane immunoadaptor DAP12 pathway during influenza infection. We find that the mice lacking DAP12 have unaltered viral clearance but easily succumb to influenza infection as a result of uncontrolled immunopathology. Such immunopathology is associated with markedly increased CD4 T cells displaying markedly increased cytotoxicity and Fas ligand expression. Furthermore, the immunopathogenic property of these CD4 T cells is transferrable. Thus, depletion of CD4 T cells or abrogation of Fas/Fas ligand signaling pathway improves survival and immunopathology. We further find that DAP12 expressed by dendritic cells plays an important role in controlling the immunopathogenic CD4 T cells during influenza infection. Our findings identify a novel pathway that controls the level of immune-pathogenic CD4 T cells during acute influenza infection.


Sujet(s)
Protéines adaptatrices de la transduction du signal/immunologie , Lymphocytes T CD4+/immunologie , Sous-type H1N1 du virus de la grippe A , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/anatomopathologie , Infections de l'appareil respiratoire/immunologie , Infections de l'appareil respiratoire/anatomopathologie , Transfert adoptif , Animaux , Liquide de lavage bronchoalvéolaire/composition chimique , Liquide de lavage bronchoalvéolaire/immunologie , Séparation cellulaire , Test ELISA , Cytométrie en flux , Immunohistochimie , Souris , Souris de lignée C57BL , Souris knockout , Pneumopathie infectieuse/immunologie , Pneumopathie infectieuse/anatomopathologie , Pneumopathie infectieuse/virologie , Infections de l'appareil respiratoire/virologie , Transduction du signal/immunologie
17.
Am J Pathol ; 178(4): 1622-34, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-21406169

RÉSUMÉ

The granuloma, a hallmark of host defense against pulmonary mycobacterial infection, has long been believed to be an active type 1 immune environment. However, the mechanisms regarding why granuloma fails to eliminate mycobacteria even in immune-competent hosts, have remained largely unclear. By using a model of pulmonary Mycobacterium bovis Bacillus Calmette-Guerin (BCG) infection, we have addressed this issue by comparing the immune responses within the airway luminal and granuloma compartments. We found that despite having a similar immune cellular profile to that in the airway lumen, the granuloma displayed severely suppressed type 1 immune cytokine but enhanced chemokine responses. Both antigen-presenting cells (APCs) and T cells in granuloma produced fewer type 1 immune molecules including tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), and nitric oxide. As a result, the granuloma APCs developed a reduced capacity to phagocytose mycobacteria and to induce T-cell proliferation. To examine the molecular mechanisms, we compared the levels of immune suppressive cytokine IL-10 in the airway lumen and granuloma and found that both granuloma APCs and T cells produced much more IL-10. Thus, IL-10 deficiency restored type 1 immune activation within the granuloma while having a minimal effect within the airway lumen. Hence, our study provides the first experimental evidence that, contrary to the conventional belief, the BCG-induced lung granuloma represents a symbiotic host-microbe microenvironment characterized by suppressed type 1 immune activation.


Sujet(s)
Granulome/microbiologie , Interleukine-10/métabolisme , Mycobacterium bovis/métabolisme , Animaux , Cellules présentatrices d'antigène/métabolisme , Vaccin BCG/métabolisme , Antigènes CD11b/biosynthèse , Antigènes CD11c/biosynthèse , Prolifération cellulaire , Femelle , Système immunitaire , Interféron gamma/métabolisme , Souris , Souris de lignée C57BL , Monoxyde d'azote/métabolisme , Symbiose , Lymphocytes T/cytologie , Lymphocytes T/microbiologie , Facteur de nécrose tumorale alpha/métabolisme
18.
Expert Rev Respir Med ; 5(1): 75-85, 2011 Feb.
Article de Anglais | MEDLINE | ID: mdl-21348588

RÉSUMÉ

Mycobacterium tuberculosis, the causative agent of pulmonary TB, causes chronic intracellular infection of lung-resident antigen-presenting cells, including macrophages and dendritic cells (DCs). Life-long bacterial control requires robust T-cell immune responses. Lung DCs are critical for initiating and co-ordinating adaptive immune responses against TB. The recent description of previously uncharacterized DC subsets has prompted the re-examination of lung DCs and their role in priming antimycobacterial T-cell responses. While there is some data on these new DCs in their naive state, very little is known about how these DCs respond to pulmonary mycobacterial infection. In this article, we attempt to identify the major antigen-presenting cell subsets that may be critical to lymph node homing, T-cell priming and controlling mycobacterial infection. We further examine the areas of DC heterogeneity that may relate to differential susceptibility between mouse strains. Furthermore, we discuss how DCs may be manipulated and exploited as a cell-based prophylactic TB vaccine and the prospect of using this strategy for post-M. tuberculosis exposure settings.


Sujet(s)
Cellules dendritiques/physiologie , Lymphocytes T/physiologie , Vaccins antituberculeux/usage thérapeutique , Tuberculose pulmonaire/prévention et contrôle , Animaux , Mouvement cellulaire/physiologie , Cellules dendritiques/immunologie , Cellules dendritiques/microbiologie , Modèles animaux de maladie humaine , Humains , Souris , Mycobacterium tuberculosis/immunologie , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/microbiologie , Muqueuse respiratoire/physiologie , Lymphocytes T/immunologie , Vaccins antituberculeux/immunologie , Tuberculose pulmonaire/immunologie , Tuberculose pulmonaire/physiopathologie
19.
J Gene Med ; 12(8): 693-704, 2010 Aug.
Article de Anglais | MEDLINE | ID: mdl-20694950

RÉSUMÉ

BACKGROUND: Virus-vectored vaccine is a powerful activator of CD8 T cell-mediated immunity and is especially amenable to respiratory mucosal immunization, offering hopes for use in humans with diminished helper CD4 T cell function. However, whether virus-mediated mucosal immunization can produce immune protective CD8 T cells without the CD4 T cell help remains to be investigated. METHODS: We used a replication-deficient adenovirus vector expressing an Mycobacterium tuberculosis antigen Ag85A for intranasal vaccination and evaluated its effect on CD8 T cell activation and protection in mice depleted of CD4 T cells. RESULTS: Intranasal vaccination of CD4 T cell-depleted mice led to suboptimal generation of Ag-specific tetramer(+) or interferon (IFN)-gamma-producing CD8 T cells in the lung and spleen but this was observed mainly at the early time after vaccination. Reduced CD8 T cell priming was also accompanied by decreased CD8 T cell responses (CTL). Nevertheless, the ratio of Ag-specific CD8 T cells to IFN-gamma-producing CD8 T cells in CD4 T cell-depleted hosts remained comparable to that in CD4 T cell-competent hosts. Furthermore, the 'unhelped' CD8 T cells also displayed a similar immune phenotype as the 'helped' counterparts. The animals with 'unhelped' CD8 T cells were as well-protected from pulmonary M. tuberculosis challenge as those with 'helped' CD8 T cells in the absence of CD4 T cells. CONCLUSIONS: The data obtained in the present study suggest that the fully immune protective CD8 T cells can still be generated by respiratory mucosal viral-mediated immunization without CD4 T cells and that CD8 T cells, 'helped' or 'unhelped', can confer significant protection against pulmonary tuberculosis independent of CD4 T cells.


Sujet(s)
Adenoviridae/génétique , Lymphocytes T CD8+/immunologie , Muqueuse respiratoire/immunologie , Acyltransferases/immunologie , Acyltransferases/métabolisme , Adenoviridae/métabolisme , Administration par voie nasale , Animaux , Antigènes bactériens/immunologie , Antigènes bactériens/métabolisme , Lymphocytes T CD8+/cytologie , Tests de cytotoxicité immunologique , Femelle , Vecteurs génétiques , Immunisation , Déplétion lymphocytaire , Souris , Souris de lignée BALB C , Lymphocytes T auxiliaires/cytologie , Lymphocytes T auxiliaires/immunologie , Vaccins antituberculeux/administration et posologie , Vaccins antituberculeux/immunologie
20.
J Immunol ; 184(4): 2048-56, 2010 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-20083661

RÉSUMÉ

Influenza viral infection is well-known to predispose to subsequent bacterial superinfection in the lung but the mechanisms have remained poorly defined. We have established a murine model of heterologous infections by an H1N1 influenza virus and Staphylococcus aureus. We found that indeed prior influenza infection markedly increased the susceptibility of mice to secondary S. aureus superinfection. Severe sickness and heightened bacterial infection in flu and S. aureus dual-infected animals were associated with severe immunopathology in the lung. We further found that flu-experienced lungs had an impaired NK cell response in the airway to subsequent S. aureus bacterial infection. Thus, adoptive transfer of naive NK cells to the airway of prior flu-infected mice restored flu-impaired antibacterial host defense. We identified that TNF-alpha production of NK cells played an important role in NK cell-mediated antibacterial host defense as NK cells in flu-experienced lungs had reduced TNF-alpha expression and adoptive transfer of TNF-alpha-deficient NK cells to the airway of flu-infected mice failed to restore flu-impaired antibacterial host defense. Defected NK cell function was found to be an upstream mechanism of depressed antibacterial activities by alveolar macrophages as contrast to naive wild-type NK cells, the NK cells from flu-infected or TNF-alpha-deficient mice failed to enhance S. aureus phagocytosis by alveolar macrophages. Together, our study identifies the weakened NK cell response in the lung to be a novel critical mechanism for flu-mediated susceptibility to bacterial superinfection.


Sujet(s)
Sous-type H1N1 du virus de la grippe A/immunologie , Cellules tueuses naturelles/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/microbiologie , Pneumopathie bactérienne/immunologie , Pneumopathie virale/immunologie , Infections à staphylocoques/immunologie , Surinfection/immunologie , Animaux , Prédisposition aux maladies/immunologie , Prédisposition aux maladies/anatomopathologie , Femelle , Cellules tueuses naturelles/microbiologie , Cellules tueuses naturelles/virologie , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/microbiologie , Macrophages alvéolaires/anatomopathologie , Souris , Souris de lignée C57BL , Infections à Orthomyxoviridae/anatomopathologie , Pneumopathie bactérienne/anatomopathologie , Pneumopathie bactérienne/virologie , Pneumopathie virale/microbiologie , Pneumopathie virale/anatomopathologie , Infections à staphylocoques/anatomopathologie , Infections à staphylocoques/virologie , Surinfection/microbiologie , Surinfection/anatomopathologie , Surinfection/virologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...