Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stephane Lajoie is active.

Publication


Featured researches published by Stephane Lajoie.


Nature Immunology | 2010

Complement-mediated regulation of the IL-17A axis is a central genetic determinant of the severity of experimental allergic asthma

Stephane Lajoie; Ian P. Lewkowich; Yusuke Suzuki; Jennifer R. Clark; Alyssa Sproles; Krista Dienger; Alison L. Budelsky; Marsha Wills-Karp

Severe asthma is associated with the production of interleukin 17A (IL-17A). The exact role of IL-17A in severe asthma and the factors that drive its production are unknown. Here we demonstrate that IL-17A mediated severe airway hyperresponsiveness (AHR) in susceptible strains of mice by enhancing IL-13-driven responses. Mechanistically, we demonstrate that IL-17A and AHR were regulated by allergen-driven production of anaphylatoxins, as mouse strains deficient in complement factor 5 (C5) or the complement receptor C5aR mounted robust IL-17A responses, whereas mice deficient in C3aR had fewer IL-17-producing helper T cells (TH17 cells) and less AHR after allergen challenge. The opposing effects of C3a and C5a were mediated through their reciprocal regulation of IL-23 production. These data demonstrate a critical role for complement-mediated regulation of the IL-23–TH17 axis in severe asthma.


PLOS ONE | 2008

Allergen Uptake, Activation, and IL-23 Production by Pulmonary Myeloid DCs Drives Airway Hyperresponsiveness in Asthma-Susceptible Mice

Ian P. Lewkowich; Stephane Lajoie; Jennifer R. Clark; Nancy S. Herman; Alyssa Sproles; Marsha Wills-Karp

Maladaptive, Th2-polarized inflammatory responses are integral to the pathogenesis of allergic asthma. As regulators of T cell activation, dendritic cells (DCs) are important mediators of allergic asthma, yet the precise signals which render endogenous DCs “pro-asthmatic”, and the extent to which these signals are regulated by the pulmonary environment and host genetics, remains unclear. Comparative phenotypic and functional analysis of pulmonary DC populations in mice susceptible (A/J), or resistant (C3H) to experimental asthma, revealed that susceptibility to airway hyperresponsiveness is associated with preferential myeloid DC (mDC) allergen uptake, and production of Th17-skewing cytokines (IL-6, IL-23), whereas resistance is associated with increased allergen uptake by plasmacytoid DCs. Surprisingly, adoptive transfer of syngeneic HDM-pulsed bone marrow derived mDCs (BMDCs) to the lungs of C3H mice markedly enhanced lung IL-17A production, and rendered them susceptible to allergen-driven airway hyperresponsiveness. Characterization of these BMDCs revealed levels of antigen uptake, and Th17 promoting cytokine production similar to that observed in pulmonary mDCs from susceptible A/J mice. Collectively these data demonstrate that the lung environment present in asthma-resistant mice promotes robust pDC allergen uptake, activation, and limits Th17-skewing cytokine production responsible for driving pathologic T cell responses central to the development of allergen-induced airway hyperresponsiveness.


European Journal of Immunology | 2015

Differential control of CD4+ T-cell subsets by the PD-1/PD-L1 axis in a mouse model of allergic asthma

Jaclyn W. McAlees; Stephane Lajoie; Krista Dienger; Alyssa Sproles; Phoebe K. Richgels; Yanfen Yang; Marat Khodoun; Miyuki Azuma; Hideo Yagita; Patricia C. Fulkerson; Marsha Wills-Karp; Ian P. Lewkowich

Studies examining the role of PD‐1 family members in allergic asthma have yielded conflicting results. Using a mouse model of allergic asthma, we demonstrate that blockade of PD‐1/PD‐L1 has distinct influences on different CD4+ T‐cell subsets. PD‐1/PD‐L1 blockade enhances airway hyperreactivity (AHR), not by altering the magnitude of the underlying Th2‐type immune response, but by allowing the development of a concomitant Th17‐type immune response. Supporting differential CD4+ T‐cell responsiveness to PD‐1‐mediated inhibition, naïve PD‐1−/− mice displayed elevated Th1 and Th17 levels, but diminished Th2 cytokine levels, and ligation of PD‐1 in WT cells limited cytokine production by in vitro polarized Th1 and Th17 cells, but slightly enhanced cytokine production by in vitro polarized Th2 cells. Furthermore, PD‐1 ligation enhanced Th2 cytokine production by naïve T cells cultured under nonpolarizing conditions. These data demonstrate that different CD4+ T‐cell subsets respond differentially to PD‐1 ligation and may explain some of the variable results observed in control of allergic asthma by the PD‐1 family members. As the PD‐1/PD‐L1 axis limits asthma severity by constraining Th17 cell activity, this suggests that severe allergic asthma may be associated with a defective PD‐1/PD‐L1 regulatory axis in some individuals.


Mucosal Immunology | 2013

PD-L2 modulates asthma severity by directly decreasing dendritic cell IL-12 production

Ian P. Lewkowich; Stephane Lajoie; Sl Stoffers; Y Suzuki; Phoebe K. Richgels; Krista Dienger; Alyssa Sproles; Hideo Yagita; Qutayba Hamid; Marsha Wills-Karp

Studies examining the role of programmed death 1 (PD-1) ligand 2 (PD-L2)/PD-1 in asthma have yielded conflicting results. To clarify its role, we examined the PD-L2 expression in biopsies from human asthmatics and the lungs of aeroallergen-treated mice. PD-L2 expression in bronchial biopsies correlated with the severity of asthma. In mice, allergen exposure increased PD-L2 expression on pulmonary myeloid dendritic cells (DCs), and PD-L2 blockade diminished allergen-induced airway hyperresponsiveness (AHR). By contrast, PD-1 blockade had no impact, suggesting that PD-L2 promotes AHR in a PD-1-independent manner. Decreased AHR was associated with enhanced serum interleukin (IL)-12 p40, and in vitro stimulation of DCs with allergen and PD-L2-Fc reduced IL-12 p70 production, suggesting that PD-L2 inhibits allergen-driven IL-12 production. In our model, IL-12 did not diminish T helper type 2 responses but rather directly antagonized IL-13-inducible gene expression, highlighting a novel role for IL-12 in regulation of IL-13 signaling. Thus, allergen-driven enhancement of PD-L2 signaling through a PD-1-independent mechanism limits IL-12 secretion, exacerbating AHR.


Clinical & Experimental Allergy | 2014

IL‐21 receptor signalling partially mediates Th2‐mediated allergic airway responses

Stephane Lajoie; Ian P. Lewkowich; Nancy S. Herman; Alyssa Sproles; John T. Pesce; Thomas A. Wynn; Michael J. Grusby; Qutayba Hamid; Marsha Wills-Karp

Interleukin‐21 (IL‐21) has been implicated in the development of Th2‐mediated immune responses; however, the exact role it plays in allergic diseases is not well understood.


Infection and Immunity | 2016

Role of Serum Amyloid A, Granulocyte-Macrophage Colony-Stimulating Factor, and Bone Marrow Granulocyte-Monocyte Precursor Expansion in Segmented Filamentous Bacterium-Mediated Protection from Entamoeba histolytica

Stacey L. Burgess; Mahmoud M. Saleh; Carrie A. Cowardin; Erica L. Buonomo; Zannatun Noor; Koji Watanabe; Mayuresh M. Abhyankar; Stephane Lajoie; Marsha Wills-Karp; William A. Petri

ABSTRACT Intestinal segmented filamentous bacteria (SFB) protect from ameba infection, and protection is transferable with bone marrow dendritic cells (BMDCs). SFB cause an increase in serum amyloid A (SAA), suggesting that SAA might mediate SFBs effects on BMDCs. Here we further explored the role of bone marrow in SFB-mediated protection. Transient gut colonization with SFB or SAA administration alone transiently increased the H3K27 histone demethylase Jmjd3, persistently increased bone marrow Csf2ra expression and granulocyte monocyte precursors (GMPs), and protected from ameba infection. Pharmacologic inhibition of Jmjd3 H3K27 demethylase activity during SAA treatment or blockade of granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling in SFB-colonized mice prevented GMP expansion, decreased gut neutrophils, and blocked protection from ameba infection. These results indicate that alteration of the microbiota and systemic exposure to SAA can influence myelopoiesis and susceptibility to amebiasis via epigenetic mechanisms. Gut microbiota-marrow communication is a previously unrecognized mechanism of innate protection from infection.


Current Allergy and Asthma Reports | 2016

Epithelial Cell Regulation of Allergic Diseases

Naina Gour; Stephane Lajoie

Allergic diseases, which have escalated in prevalence in recent years, arise as a result of maladaptive immune responses to ubiquitous environmental stimuli. Why only certain individuals mount inappropriate type 2 immune responses to these otherwise harmless allergens has remained an unanswered question. Mounting evidence suggests that the epithelium, by sensing its environment, is the central regulator of allergic diseases. Once considered to be a passive barrier to allergens, epithelial cells at mucosal surfaces are now considered to be the cornerstone of the allergic diathesis. Beyond their function as maintaining barrier at mucosal surfaces, mucosal epithelial cells through the secretion of mediators like IL-25, IL-33, and TSLP control the fate of downstream allergic immune responses. In this review, we will discuss the advances in recent years regarding the process of allergen recognition and secretion of soluble mediators by epithelial cells that shape the development of the allergic response.


Immunity | 2013

New Twist on an Ancient Innate Immune Pathway

Stephane Lajoie; Marsha Wills-Karp

Activation of the complement system has long been known to be regulated by a series of steps involving fluid-phase convertases. In this issue of Immunity, Liszewski et al. (2013) report the discovery of an intracellular cathepsin-L-dependent C3 activation pathway.


The Journal of Allergy and Clinical Immunology | 2017

IL-17A enhances IL-13 activity by enhancing IL-13–induced signal transducer and activator of transcription 6 activation

Sara L. Hall; Theresa Baker; Stephane Lajoie; Phoebe K. Richgels; Yanfen Yang; Jaclyn W. McAlees; Adelaide van Lier; Marsha Wills-Karp; Umasundari Sivaprasad; Thomas H. Acciani; Timothy D. LeCras; Jocelyn M. Biagini Myers; Melinda Butsch Kovacic; Ian P. Lewkowich

Background: Increased IL‐17A production has been associated with more severe asthma; however, the mechanisms whereby IL‐17A can contribute to IL‐13–driven pathology in asthmatic patients remain unclear. Objective: We sought to gain mechanistic insight into how IL‐17A can influence IL‐13–driven responses. Methods: The effect of IL‐17A on IL‐13–induced airway hyperresponsiveness, gene expression, mucus hypersecretion, and airway inflammation was assessed by using in vivo models of IL‐13–induced lung pathology and in vitro culture of murine fibroblast cell lines and primary fibroblasts and human epithelial cell lines or primary human epithelial cells exposed to IL‐13, IL‐17A, or both. Results: Compared with mice given intratracheal IL‐13 alone, those exposed to IL‐13 and IL‐17A had augmented airway hyperresponsiveness, mucus production, airway inflammation, and IL‐13–induced gene expression. In vitro, IL‐17A enhanced IL‐13–induced gene expression in asthma‐relevant murine and human cells. In contrast to the exacerbating influence of IL‐17A on IL‐13–induced responses, coexposure to IL‐13 inhibited IL‐17A–driven antimicrobial gene expression in vivo and in vitro. Mechanistically, in both primary human and murine cells, the IL‐17A–driven increase in IL‐13–induced gene expression was associated with enhanced IL‐13–driven signal transducer and activator of transcription 6 activation. Conclusions: Our data suggest that IL‐17A contributes to asthma pathophysiology by increasing the capacity of IL‐13 to activate intracellular signaling pathways, such as signal transducer and activator of transcription 6. These data represent the first mechanistic explanation of how IL‐17A can directly contribute to the pathogenesis of IL‐13–driven pathology. GRAPHICAL ABSTRACT Figure. No caption available.


Science immunology | 2018

Dysregulated invertebrate tropomyosin–dectin-1 interaction confers susceptibility to allergic diseases

Naina Gour; Stephane Lajoie; Ursula Smole; Marquitta J. White; Donglei Hu; Pagé Goddard; Scott Huntsman; Celeste Eng; Angel C. Y. Mak; Sam S. Oh; Jung Hyun Kim; Annu Sharma; Sophie Plante; Ikhlass Haj Salem; Yvonne Resch; Xiao Xiao; Nu Yao; Anju Singh; Susanne Vrtala; Jamila Chakir; Esteban G. Burchard; Andrew P. Lane; Marsha Wills-Karp

Invertebrate tropomyosin homologs are ligands for dectin-1. Dectin-1 limits allergic responses Aberrant activation of pattern recognition receptors (PRRs) drives inflammation in autoimmune and allergic diseases. Here, Gour et al. have identified invertebrate tropomyosin from house dust mite and shrimp as a ligand for dectin-1. Dectin-1 is a PRR that has been demonstrated to recognize fungal β-glucans to antifungal immune responses. Here, the authors report that engagement of dectin-1 by invertebrate tropomyosins limits type 2 inflammation and that dectin-1–deficient mice are more prone to allergic airway inflammation. They have also established that expression of dectin-1 is repressed in allergic individuals. By identifying invertebrate tropomyosin orthologs as dectin-1 ligands, the study establishes the importance of dectin-1 in limiting allergic responses. The key factors underlying the development of allergic diseases—the propensity for a minority of individuals to develop dysfunctional responses to harmless environmental molecules—remain undefined. We report a pathway of immune counter-regulation that suppresses the development of aeroallergy and shrimp-induced anaphylaxis. In mice, signaling through epithelially expressed dectin-1 suppresses the development of type 2 immune responses through inhibition of interleukin-33 (IL-33) secretion and the subsequent recruitment of IL-13–producing innate lymphoid cells. Although this homeostatic pathway is functional in respiratory epithelial cells from healthy humans, it is dramatically impaired in epithelial cells from asthmatic and chronic rhinosinusitis patients, resulting in elevated IL-33 production. Moreover, we identify an association between a single-nucleotide polymorphism (SNP) in the dectin-1 gene loci and reduced pulmonary function in two cohorts of asthmatics. This intronic SNP is a predicted eQTL (expression quantitative trait locus) that is associated with reduced dectin-1 expression in human tissue. We identify invertebrate tropomyosin, a ubiquitous arthropod-derived molecule, as an immunobiologically relevant dectin-1 ligand that normally serves to restrain IL-33 release and dampen type 2 immunity in healthy individuals. However, invertebrate tropomyosin presented in the context of impaired dectin-1 function, as observed in allergic individuals, leads to unrestrained IL-33 secretion and skewing of immune responses toward type 2 immunity. Collectively, we uncover a previously unrecognized mechanism of protection against allergy to a conserved recognition element omnipresent in our environment.

Collaboration


Dive into the Stephane Lajoie's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian P. Lewkowich

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alyssa Sproles

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Krista Dienger

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer R. Clark

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Phoebe K. Richgels

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Naina Gour

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Jaclyn W. McAlees

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yanfen Yang

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge