Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Bioorg Med Chem Lett ; 100: 129630, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38307441

ABSTRACT

A series of macrocyclic PKCθ inhibitors based on a 1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one hinge binder has been studied. Different aromatic and heteroaromatic substituents have been explored in order to optimize potency, isoform selectivity as well as DMPK properties. The importance of the length of the macrocyclic linker has also been analyzed. In particular, it has been found that methyl substitutions on the linker can have a profound influence on both potency and metabolic stability. Several compounds showing very good profiles, suitable for in vivo testing, are disclosed.

2.
J Invest Dermatol ; 144(2): 252-262.e4, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37598867

ABSTRACT

Tissue transcriptomics is used to uncover molecular dysregulations underlying diseases. However, the majority of transcriptomics studies focus on single diseases with limited relevance for understanding the molecular relationship between diseases or for identifying disease-specific markers. In this study, we used a normalization approach to compare gene expression across nine inflammatory skin diseases. The normalized datasets were found to retain differential expression signals that allowed unsupervised disease clustering and identification of disease-specific gene signatures. Using the NS-Forest algorithm, we identified a minimal set of biomarkers and validated their use as diagnostic disease classifier. Among them, PTEN was identified as being a specific marker for cutaneous lupus erythematosus and found to be strongly expressed by lesional keratinocytes in association with pathogenic type I IFNs. In fact, PTEN facilitated the expression of IFN-ß and IFN-κ in keratinocytes by promoting activation and nuclear translocation of IRF3. Thus, cross-comparison of tissue transcriptomics is a valid strategy to establish a molecular disease classification and to identify pathogenic disease biomarkers.


Subject(s)
Dermatitis , Lupus Erythematosus, Cutaneous , Lupus Erythematosus, Systemic , Humans , Biomarkers/metabolism , Dermatitis/pathology , Gene Expression Profiling , Keratinocytes/metabolism , Lupus Erythematosus, Cutaneous/diagnosis , Lupus Erythematosus, Cutaneous/genetics , Lupus Erythematosus, Cutaneous/metabolism , Lupus Erythematosus, Systemic/genetics , PTEN Phosphohydrolase/genetics , Skin/pathology
3.
J Allergy Clin Immunol ; 153(1): 146-160, 2024 01.
Article in English | MEDLINE | ID: mdl-37506977

ABSTRACT

BACKGROUND: Prurigo nodularis (PN) is a chronic neuroimmune skin disease characterized by bilaterally distributed pruritic hyperkeratotic nodules on extremities and trunk. Neuroimmune dysregulation and chronic scratching are believed to both induce and maintain the characteristic lesions. OBJECTIVES: This study sought to provide a comprehensive view of the molecular pathogenesis of PN at the single-cell level to identify and outline key pathologic processes and the cell types involved. Features that distinguish PN skin from the skin of patients with atopic dermatitis were of particular interest. We further aimed to determine the impact of the IL31RA antagonist, nemolizumab, and its specificity at the single-cell level. METHODS: Single-cell RNA-sequencing of skin from 15 healthy donors and nonlesional and lesional skin from 6 patients each with PN and atopic dermatitis, combined with spatial-sequencing using the 10x Visium platform. Integration with bulk RNA-sequencing data from patients treated with nemolizumab. RESULTS: This study demonstrates that PN is an inflammatory skin disease characterized by both keratinocyte proliferation and activation of profibrotic responses. This study also demonstrates that the COL11A1+ fibroblast subset is a major contributor to fibrosis and is predominantly found in the papillary dermis of PN skin. Activation of fibrotic responses is the main distinguishing feature between PN and atopic dermatitis skin. This study further shows the broad effect of nemolizumab on PN cell types, with a prominent effect driving COL11A1+ fibroblast and keratinocyte responses toward normal. CONCLUSIONS: This study provides a high-resolution characterization of the cell types and cellular processes activated in PN skin, establishing PN as a chronic fibrotic inflammatory skin disease. It further demonstrates the broad effect of nemolizumab on pathological processes in PN skin.


Subject(s)
Dermatitis, Atopic , Prurigo , Humans , Prurigo/drug therapy , Dermatitis, Atopic/pathology , Skin/pathology , Chronic Disease , RNA , Pruritus/pathology
4.
JAMA Dermatol ; 159(9): 977-985, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37556125

ABSTRACT

Importance: Prurigo nodularis (PN) is a debilitating skin disease characterized by intense pruritus and hyperkeratotic skin nodules. Nemolizumab, a monoclonal antibody targeting interleukin 31 receptor α, is a promising novel therapy for the treatment of moderate to severe PN. The biological mechanisms by which nemolizumab promotes improvement of itch and skin lesions in PN are unknown. Objective: To characterize changes in plasma protein biomarkers associated with clinical response to nemolizumab in patients with PN. Design, Setting, and Participants: This multicenter cohort study included patients recruited from Austria, France, Germany, Poland, and the US from a phase 2 clinical trial. Adults diagnosed with moderate to severe PN with severe pruritus for at least 6 months were included in the original trial. Patients in the nemolizumab group were included in the present study if they achieved at least a 4-point decrease in the Peak Pruritus Numerical Rating Scale (PP-NRS) from baseline to week 12 during nemolizumab treatment. Placebo controls did not experience a 4-point decrease in PP-NRS. Mass spectrometry with tandem mass tags to enrich skin-specific protein detection was used to characterize changes in plasma protein expression in nemolizumab and placebo groups. Data were collected from November 2, 2017, to September 26, 2018, and analyzed from December 6, 2019, to April 8, 2022. Intervention: As part of the clinical trial, patients were treated with 3 doses of nemolizumab or placebo at 0, 4, and 8 weeks. Main Outcomes and Measures: Changes in plasma and epidermal protein expression in nemolizumab-treated patients compared with the placebo group at 0, 4, and 12 weeks. Results: Among the 38 patients included in the analysis (22 women and 16 men; mean [SD] age, 55.8 [15.8] years), enrichment analysis of canonical pathways, biological functions, and upstream regulators showed downregulation of terms involving inflammation (IL-6, acute-phase response, signal transducer and activator of transcription 3, and interferon γ), neural processes (synaptogenesis signaling and neuritogenesis), tissue remodeling and fibrosis (transforming growth factor ß1 and endothelin-1), and epidermal differentiation (epithelial mesenchymal transition) in the plasma of nemolizumab group. Conclusions and Relevance: In this cohort study, differences between nemolizumab and placebo groups included modulation of inflammatory signaling, neural development, and epithelial differentiation, suggesting a promising potential approach for clinical management of PN.


Subject(s)
Prurigo , Adult , Male , Humans , Female , Middle Aged , Prurigo/drug therapy , Prurigo/complications , Cohort Studies , Pruritus/etiology , Pruritus/complications , Biomarkers
5.
J Allergy Clin Immunol ; 149(4): 1329-1339, 2022 04.
Article in English | MEDLINE | ID: mdl-34857395

ABSTRACT

BACKGROUND: Prurigo nodularis (PN) is a debilitating, difficult-to-treat, intensely pruritic, chronic inflammatory skin disease characterized by hyperkeratotic skin nodules. The pathogenesis of PN is not well understood but is believed to involve cross talk between sensory nerve fibers, immune cells, and the epidermis. It is centered around the neuroimmune cytokine IL-31, driving an intractable itch-scratch cycle. OBJECTIVE: We sought to provide a comprehensive view of the transcriptomic changes in PN skin and characterize the mechanism of action of the anti-IL-31 receptor inhibitor nemolizumab. METHOD: RNA sequencing of biopsy samples obtained from a cohort of patients treated with the anti-IL-31 receptor inhibitor nemolizumab and taken at baseline and week 12. Generation and integration of patient data with RNA-Seq data generated from reconstructed human epidermis stimulated with IL-31 and other proinflammatory cytokines. RESULTS: Our results demonstrate that nemolizumab effectively decreases IL-31 responses in PN skin, leading to effective suppression of downstream inflammatory responses including TH2/IL-13 and TH17/IL-17 responses. This is accompanied by decreased keratinocyte proliferation and normalization of epidermal differentiation and function. Furthermore, our results demonstrate how transcriptomic changes associated with nemolizumab treatment correlate with improvement in lesions, pruritus, stabilization of extracellular matrix remodeling, and processes associated with cutaneous nerve function. CONCLUSION: These data demonstrate a broad response to IL-31 receptor inhibition with nemolizumab and confirm the critical upstream role of IL-31 in PN pathogenesis.


Subject(s)
Prurigo , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Chronic Disease , Cytokines/therapeutic use , Humans , Prurigo/drug therapy , Prurigo/genetics , Pruritus/drug therapy , Pruritus/genetics , Transcriptome
6.
Front Med (Lausanne) ; 8: 745822, 2021.
Article in English | MEDLINE | ID: mdl-34746181

ABSTRACT

Background and Objectives: Trifarotene is a topical retinoid selective for retinoic acid receptor gamma that was recently approved for treatment of acne vulgaris. We performed a gene expression analysis to identify the molecular and cellular impact of trifarotene treatment on acne papules. Methods: In this open-label prospective study, subjects with moderate inflammatory acne of the back were treated with trifarotene 0.005% or vehicle cream on dedicated areas for 27 days, and 4 biopsies were collected from each subject (1 from skin without a visible acne lesion and three at the site of an acne papule: one baseline, one after vehicle treatment, and one after trifarotene treatment). Large scale gene expression profiling of the biopsies was performed using Affymetrix technology, treatment-specific gene expression profiles were generated using statistical modeling, and pathway analysis was performed. Using single-cell RNAseq data, in silico deconvolution of transcriptomics data was performed to identify cellular signatures. Results: We discovered a unique set of 67 genes modulated by trifarotene that are primarily involved in cellular migration, inflammation, and extracellular matrix reorganization. Changes in cellular expression were similar in both trifarotene-treated and spontaneously-resolving lesions. However, only trifarotene treatment impacted SPP1+ macrophages, a subset of highly proliferative macrophages recently identified in fibrotic tissue. Conclusions: These results show that trifarotene has a novel action in acne treatment by affecting epidermal and immune components of acne pathogenesis.

7.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34279540

ABSTRACT

Inflammatory skin diseases including atopic dermatitis (AD) and psoriasis (PSO) are underpinned by dendritic cell (DC)-mediated T cell responses. Currently, the heterogeneous human cutaneous DC population is incompletely characterized, and its contribution to these diseases remains unclear. Here, we performed index-sorted single-cell flow cytometry and RNA sequencing of lesional and nonlesional AD and PSO skin to identify macrophages and all DC subsets, including the newly described mature LAMP3+BIRC3+ DCs enriched in immunoregulatory molecules (mregDC) and CD14+ DC3. By integrating our indexed data with published skin datasets, we generated a myeloid cell universe of DC and macrophage subsets in healthy and diseased skin. Importantly, we found that CD14+ DC3s increased in PSO lesional skin and co-produced IL1B and IL23A, which are pathological in PSO. Our study comprehensively describes the molecular characteristics of macrophages and DC subsets in AD and PSO at single-cell resolution, and identifies CD14+ DC3s as potential promoters of inflammation in PSO.


Subject(s)
Dermatitis, Atopic/pathology , Interleukin-1beta/metabolism , Interleukin-23 Subunit p19/metabolism , Langerhans Cells/pathology , Psoriasis/pathology , Dermatitis, Atopic/metabolism , Gene Expression , Gene Regulatory Networks , Humans , Interleukin-15/metabolism , Langerhans Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Macrophages/cytology , Psoriasis/metabolism , Single-Cell Analysis
8.
Front Med (Lausanne) ; 8: 639097, 2021.
Article in English | MEDLINE | ID: mdl-33644104

ABSTRACT

Pruritus represents one of the most common symptoms in dermatology and general medicine. Chronic pruritus severely impairs the quality of life of affected patients. During the last two decades a number of modulators and mediator of pruritus have been identified. Recently, Interleukin (IL)-31 and its receptor complex attracted significant interest, as clinical phase two studies demonstrated therapeutic efficacy of the neutralizing IL-31 receptor A (IL-31RA) antibody nemolizumab in patients suffering from atopic dermatitis or prurigo nodularis. IL-31 has also been shown to play relevant roles in allergic contact dermatitis, urticaria, mastocytosis, allergic rhinitis and asthma. Here, we summarize the current knowledge of the novel cytokine IL-31 and its receptor regarding cellular origin, regulation, signaling pathways and their involvement in biological processes such as pruritus, neuronal growth, inflammation, barrier dysfunction and tissue remodeling.

9.
Eur Respir J ; 52(4)2018 10.
Article in English | MEDLINE | ID: mdl-30190271

ABSTRACT

In line with the pathophysiological continuum described between nose and bronchus in allergic respiratory diseases, we assessed whether nasal epithelium could mirror the Type 2 T-helper cell (Th2) status of bronchial epithelium.Nasal and bronchial cells were collected by brushing from healthy controls (C, n=13), patients with allergic rhinitis and asthma (AR, n=12), and patients with isolated allergic rhinitis (R, n=14). Cellular composition was assessed by flow cytometry, gene expression was analysed by RNA sequencing and Th2, Type 17 T-helper cell (Th17) and interferon (IFN) signatures were derived from the literature.Infiltration by polymorphonuclear neutrophils (PMN) in the nose excluded 30% of the initial cohort. All bronchial samples from the AR group were Th2-high. The gene expression profile of nasal samples from the AR group correctly predicted the paired bronchial sample Th2 status in 71% of cases. Nevertheless, nasal cells did not appear to be a reliable surrogate for the Th2 response, in particular due to a more robust influence of the IFN response in 14 out of 26 nasal samples. The Th2 scores in the nose and bronchi correlated with mast cell count (both p<0.001) and number of sensitisations (p=0.006 and 0.002), while the Th17 scores correlated with PMN count (p=0.006 and 0.003).The large variability in nasal cell composition and type of inflammation restricts its use as a surrogate for assessing bronchial Th2 inflammation in AR patients.


Subject(s)
Asthma/immunology , Rhinitis, Allergic/immunology , Th17 Cells/cytology , Th2 Cells/cytology , Adult , Asthma/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Case-Control Studies , Female , Gene Expression , Humans , Inflammation/immunology , Interferons/metabolism , Male , Nasal Lavage Fluid/cytology , Respiratory Mucosa/metabolism , Rhinitis, Allergic/physiopathology , Sequence Analysis, RNA , Th17 Cells/immunology , Th2 Cells/immunology , Young Adult
10.
Med Sci Monit Basic Res ; 23: 368-372, 2017 Nov 27.
Article in English | MEDLINE | ID: mdl-29176546

ABSTRACT

BACKGROUND Migration of leukocytes into airways is the hallmark of allergic asthma. The aim of this study was to target the pathological process using pantethine, a pleiotropic natural compound which has been recently shown to down-regulate chemokine-driven T cell migration. MATERIAL AND METHODS Mice were sensitized to the Leishmania LACK antigen, then treated or not treated with pantethine and exposed to LACK or saline aerosol. After sacrifice of the animals, cells in the bronchoalveolar lavage were analyzed and inflammatory parameters were determined to evaluate inflammation seriousness. RESULTS As compared to untreated animals, pantethine-treated animals displayed a moderated response to the allergen, as documented by decreased infiltration of inflammatory cells (all types), in addition to reduced levels of lung Th2 cytokines and circulating LACK-specific IgE. CONCLUSIONS These data reveal the potential therapeutic importance of pantethine to moderate allergic asthma pathology. The compound has been previously shown to exert a broad range of protective activity in animals and in humans, with few or no adverse effects.


Subject(s)
Leukocytes/drug effects , Pantetheine/analogs & derivatives , Allergens/physiology , Animals , Antigens, Protozoan/immunology , Bronchoalveolar Lavage/methods , Cytokines/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Female , Inflammation/drug therapy , Inflammation/pathology , Leukocytes/physiology , Lung , Mice , Mice, Inbred BALB C , Pantetheine/metabolism , Pantetheine/pharmacology , Protozoan Proteins/immunology
11.
Sci Transl Med ; 8(362): 362ra143, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27797959

ABSTRACT

In the developed world, declining prevalence of some parasitic infections correlates with increased incidence of allergic and autoimmune disorders. Moreover, experimental human infection with some parasitic worms confers protection against inflammatory diseases in phase 2 clinical trials. Parasitic worms manipulate the immune system by secreting immunoregulatory molecules that offer promise as a novel therapeutic modality for inflammatory diseases. We identify a protein secreted by hookworms, anti-inflammatory protein-2 (AIP-2), that suppressed airway inflammation in a mouse model of asthma, reduced expression of costimulatory markers on human dendritic cells (DCs), and suppressed proliferation ex vivo of T cells from human subjects with house dust mite allergy. In mice, AIP-2 was primarily captured by mesenteric CD103+ DCs and suppression of airway inflammation was dependent on both DCs and Foxp3+ regulatory T cells (Tregs) that originated in the mesenteric lymph nodes (MLNs) and accumulated in distant mucosal sites. Transplantation of MLNs from AIP-2-treated mice into naïve hosts revealed a lymphoid tissue conditioning that promoted Treg induction and long-term maintenance. Our findings indicate that recombinant AIP-2 could serve as a novel curative therapeutic for allergic asthma and potentially other inflammatory diseases.


Subject(s)
Asthma/blood , Helminth Proteins/pharmacology , Hypersensitivity/therapy , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/immunology , Adult , Ancylostomatoidea , Animals , Antigens, CD/metabolism , Asthma/immunology , Cell Proliferation , Cytokines/immunology , Dendritic Cells/immunology , Disease Models, Animal , Female , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Inflammation , Integrin alpha Chains/metabolism , Male , Mice , Mucous Membrane/metabolism , Prevalence , Pyroglyphidae
12.
Med Sci (Paris) ; 32(3): 260-6, 2016 Mar.
Article in French | MEDLINE | ID: mdl-27011244

ABSTRACT

Allergic asthma and atopic dermatitis are diseases mainly resulting from the activation of Th2 cells, that produce cytokines favouring IgE production and eosinophilia but also of Th1 cells, that contribute to inflammation chronicity. Lymphocyte recruitment and retention of Th cells in target organs are 2 key events for asthma and atopic dermatitis pathogenesis. While lymphocyte migration is regulated by chemokines and lipid mediators such as leukotrienes and prostaglandins, factors involved in lymphocyte retention and survival within inflammatory tissues remain poorly understood. Recent works show that, in allergic diseases, there is an increased expression of fractalkine/CX3CL1 and its unique receptor CX3CR1 and that this chemokine does not act as chemoattractant. In allergic asthma, CX3CR1 expression regulates Th2 and Th1 cell survival in the inflammatory lung, while, in atopic dermatitis, it regulate Th2 and Th1 cell retention into the inflammatory site. Use of peptides blocking fractalkine binding to its receptor is currently tested in the treatment of asthma and atopic dermatitis.


Subject(s)
Chemokine CX3CL1/physiology , Hypersensitivity/genetics , Receptors, Chemokine/physiology , Animals , CX3C Chemokine Receptor 1 , Cell Movement/genetics , Cell Survival/genetics , Gene Expression Regulation , Humans , Hypersensitivity/immunology , Tissue Distribution
14.
Nat Rev Immunol ; 15(5): 308-22, 2015 May.
Article in English | MEDLINE | ID: mdl-25907459

ABSTRACT

The incidence of allergic diseases is increasing, both in developed and developing countries, concomitantly with the rise in living standards and the adoption of a 'western lifestyle'. For two decades, the hygiene hypothesis - which proposes that the lack of early childhood exposure to infectious agents increases susceptibility to allergic diseases in later life - provided the conceptual framework for unravelling the mechanisms that could account for the increased incidence of allergic diseases. In this Review, we discuss recent evidence that highlights the role of diet as a key factor influencing immune homeostasis and the development of allergic diseases through a complex interplay between nutrients, their metabolites and immune cell populations. Although further investigations are still required to understand these complex relationships, recent data have established a possible connection between metabolic homeostasis and allergic diseases.


Subject(s)
Asthma/immunology , Diet , Food , Homeostasis/immunology , Hypersensitivity/immunology , Immune System/immunology , Allergens/immunology , Breast Feeding , Humans , Hygiene Hypothesis , Obesity/immunology
15.
J Exp Med ; 211(6): 1185-96, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24821910

ABSTRACT

Atopic dermatitis (AD) is a chronic allergic dermatosis characterized by epidermal thickening and dermal inflammatory infiltrates with a dominant Th2 profile during the acute phase, whereas a Th1 profile is characteristic of the chronic stage. Among chemokines and chemokine receptors associated with inflammation, increased levels of CX3CL1 (fractalkine) and its unique receptor, CX3CR1, have been observed in human AD. We have thus investigated their role and mechanism of action in experimental models of AD and psoriasis. AD pathology and immune responses, but not psoriasis, were profoundly decreased in CX3CR1-deficient mice and upon blocking CX3CL1-CX3CR1 interactions in wild-type mice. CX3CR1 deficiency affected neither antigen presentation nor T cell proliferation in vivo upon skin sensitization, but CX3CR1 expression by both Th2 and Th1 cells was required to induce AD. Surprisingly, unlike in allergic asthma, where CX3CL1 and CX3CR1 regulate the pathology by controlling effector CD4(+) T cell survival within inflamed tissues, adoptive transfer experiments established CX3CR1 as a key regulator of CD4(+) T cell retention in inflamed skin, indicating a new function for this chemokine receptor. Therefore, although CX3CR1 and CX3CL1 act through distinct mechanisms in different pathologies, our results further indicate their interest as promising therapeutic targets in allergic diseases.


Subject(s)
Chemokine CX3CL1/immunology , Dermatitis, Atopic/immunology , Receptors, Chemokine/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CX3C Chemokine Receptor 1 , Cell Proliferation , Cells, Cultured , Chemokine CX3CL1/antagonists & inhibitors , Chemokine CX3CL1/genetics , Dermatitis, Atopic/genetics , Flow Cytometry , Humans , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Protein Binding/drug effects , Protein Binding/immunology , Receptors, Chemokine/genetics , Skin/metabolism , Skin/pathology , T-Lymphocytes/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
16.
J Surg Oncol ; 109(6): 597-605, 2014 May.
Article in English | MEDLINE | ID: mdl-24391050

ABSTRACT

BACKGROUND: Increased activity of Src has been found in several human cancers, and often is associated with poor clinical outcome. The present study aimed to determine whether Src activity is increased in gastrointestinal stromal tumors (GIST), and whether it correlates with established tumor or patient characteristics and prognosis. METHODS: Tumor/normal tissues of 29 patients were analyzed for Src activity/protein with kinase assays, and for VEGF/VEGFR with immunohistochemical staining. RESULTS: Src activity was higher in tumor than in normal tissues (P = 0.093). However, when imatinib responders were excluded from the analyses, it was significantly higher in the tumor tissue (P = 0.017). Additionally, it was higher in primary compared to recurrent tumors or metastasis (P = 0.04). Univariate survival analysis showed a longer overall survival for patients with high Src activity (P = 0.038). In multivariate analysis, the response to imatinib treatment was the only survival-influencing factor (P = 0.072). CONCLUSIONS: Src activity is increased in GIST. In contrast to most other tumor entities, it does not correlate with poor clinical outcome, but decreases during the progression from primary tumor to recurrence and metastasis, especially under therapy with imatinib. Additionally, our results show that higher Src activity is associated with longer overall survival.


Subject(s)
Gastrointestinal Neoplasms/metabolism , Gastrointestinal Stromal Tumors/metabolism , src-Family Kinases/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Exons , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/mortality , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/mortality , Humans , Imatinib Mesylate , Immunohistochemistry , Middle Aged , Multivariate Analysis , Mutation , Neoplasm Metastasis , Neoplasm Recurrence, Local/metabolism , Piperazines/therapeutic use , Prognosis , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/therapeutic use , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
17.
Eur J Immunol ; 41(8): 2341-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21557213

ABSTRACT

Interleukin-33 (IL-33) is thought to be released during cellular death as an alarming cytokine during the acute phase of disease, but its regulation in vivo is poorly understood. We investigated the expression of IL-33 in two mouse models of acute hepatitis by administering either carbon tetrachloride (CCl(4) ) or concanavalin A (ConA). IL-33 was overexpressed in both models but with a stronger induction in ConA-induced hepatitis. IL-33 was weakly expressed in vascular and sinusoidal endothelial cells from normal liver and was clearly induced in CCl(4) -treated mice. Surprisingly, we found that hepatocytes strongly expressed IL-33 exclusively in the ConA model. CD1d knock-out mice, which are deficient in NKT cells and resistant to ConA-induced hepatitis, no longer expressed IL-33 in hepatocytes following ConA administration. Interestingly, invariant NKT (iNKT) cells adoptively transferred into ConA-treated CD1d KO mouse restored IL-33 expression in hepatocytes. This strongly suggests that NKT cells are responsible for the induction of IL-33 in hepatocytes.


Subject(s)
Hepatitis, Animal/genetics , Hepatocytes/metabolism , Interleukins/genetics , Natural Killer T-Cells/metabolism , Acute Disease , Adoptive Transfer , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Carbon Tetrachloride , Concanavalin A , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Hepatitis, Animal/chemically induced , Hepatitis, Animal/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-33 , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukins/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Nat Med ; 16(11): 1305-12, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21037587

ABSTRACT

Allergic asthma is a T helper type 2 (T(H)2)-dominated disease of the lung. In people with asthma, a fraction of CD4(+) T cells express the CX3CL1 receptor, CX3CR1, and CX3CL1 expression is increased in airway smooth muscle, lung endothelium and epithelium upon allergen challenge. Here we found that untreated CX3CR1-deficient mice or wild-type (WT) mice treated with CX3CR1-blocking reagents show reduced lung disease upon allergen sensitization and challenge. Transfer of WT CD4(+) T cells into CX3CR1-deficient mice restored the cardinal features of asthma, and CX3CR1-blocking reagents prevented airway inflammation in CX3CR1-deficient recipients injected with WT T(H)2 cells. We found that CX3CR1 signaling promoted T(H)2 survival in the inflamed lungs, and injection of B cell leukemia/lymphoma-2 protein (BCl-2)-transduced CX3CR1-deficient T(H)2 cells into CX3CR1-deficient mice restored asthma. CX3CR1-induced survival was also observed for T(H)1 cells upon airway inflammation but not under homeostatic conditions or upon peripheral inflammation. Therefore, CX3CR1 and CX3CL1 may represent attractive therapeutic targets in asthma.


Subject(s)
Lung/immunology , Lung/pathology , Pneumonia/immunology , Receptors, Chemokine/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Animals , Antigens, Protozoan/immunology , Apoptosis , Bronchial Hyperreactivity/complications , Bronchial Hyperreactivity/immunology , CX3C Chemokine Receptor 1 , Cell Proliferation , Cell Survival , Hypersensitivity/complications , Hypersensitivity/immunology , Lymph Nodes/pathology , Mice , Mice, Transgenic , Phenotype , Pneumonia/complications , Protozoan Proteins/immunology , Receptors, Interleukin-8A/metabolism , Signal Transduction
19.
J Immunol ; 185(4): 2174-81, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20624944

ABSTRACT

The ability of NK cells to rapidly produce IFN-gamma is an important innate mechanism of resistance to many pathogens including Leishmania major. Molecular and cellular components involved in NK cell activation in vivo are still poorly defined, although a central role for dendritic cells has been described. In this study, we demonstrate that Ag-specific CD4(+) T cells are required to initiate NK cell activation early on in draining lymph nodes of L. major-infected mice. We show that early IFN-gamma secretion by NK cells is controlled by IL-2 and IL-12 and is dependent on CD40/CD40L interaction. These findings suggest that newly primed Ag-specific CD4(+) T cells could directly activate NK cells through the secretion of IL-2 but also indirectly through the regulation of IL-12 secretion by dendritic cells. Our results reveal an unappreciated role for Ag-specific CD4(+) T cells in the initiation of NK cell activation in vivo upon L. major infection and demonstrate bidirectional regulations between innate and adaptive immunity.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Leishmaniasis, Cutaneous/immunology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Leishmania major/immunology , Leishmaniasis, Cutaneous/parasitology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Molecular Sequence Data , Time Factors
20.
J Allergy Clin Immunol ; 124(3): 573-82, 582.e1-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19539982

ABSTRACT

BACKGROUND: Eosinophils are key players in T(H)2-driven pathologies, such as allergic lung inflammation. After IL-5- and eotaxin-mediated tissue recruitment, they release several cytotoxic and inflammatory mediators. However, their exact contribution to asthma remains controversial. Indeed, in human subjects anti-IL-5 treatment inhibits eosinophilia but not antigen-induced airway hyperresponsiveness (AHR). Likewise, lung fibrosis is abrogated in 2 strains of eosinophil-deficient mice, whereas AHR is inhibited in only one of them. Finally, eosinophils have been shown to attract T(H)2 lymphocytes at the inflammatory site. OBJECTIVE: The ability of eosinophils to promote AHR and lung inflammation independently of lymphocytes was investigated. METHODS: Adoptive transfers of resting or activated eosinophils from IL-5 transgenic mice were performed into naive BALB/c mice, mice with severe combined immunodeficiency, and IFN-gamma-deficient BALB/c recipients. RESULTS: Adoptively transferred eosinophils induced lung inflammation, fibrosis, collagen deposition, and AHR not only in BALB/c mice but also in recipient mice with severe combined immunodeficiency. Surprisingly, IFN-gamma expression was increased in lungs from eosinophil-transferred animals. Furthermore, IFN-gamma neutralization in recipients partially inhibited eosinophil-induced AHR. Moreover, IFN-gamma-deficient eosinophils or eosinophils treated with a blocking anti-IFN-gamma receptor antibody failed to induce AHR in IFN-gamma-deficient recipients. Finally, in vitro and at low concentrations, IFN-gamma increased eosinophil peroxidase release, potentiated chemotaxis, and prolonged survival, suggesting the existence of an autocrine mechanism. CONCLUSIONS: These results support the important and previously unsuspected contribution of eosinophils to lung inflammation independently of lymphocytes through production of IFN-gamma, the prototypical T(H)1 cytokine.


Subject(s)
Bronchial Hyperreactivity/immunology , Eosinophils/immunology , Interferon-gamma/metabolism , Interleukin-5/immunology , Lymphocytes/immunology , Pneumonia/immunology , Adoptive Transfer , Animals , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/metabolism , Chemotaxis/immunology , Eosinophil Peroxidase/analysis , Eosinophils/metabolism , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-5/metabolism , Lung/immunology , Lung/pathology , Lymphocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Mice, Transgenic , Pneumonia/etiology , Pneumonia/metabolism , Receptors, Interferon/immunology , Receptors, Interferon/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...