Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Am J Pathol ; 194(3): 447-458, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38159722

ABSTRACT

Corneal scarring is the third leading cause of global blindness. Neovascularization of ocular tissues is a major predisposing factor in scar development. Although corneal transplantation is effective in restoring vision, some patients are at high risk for graft rejection due to the presence of blood vessels in the injured cornea. Current treatment options for controlling corneal scarring are limited, and outcomes are typically poor. In this study, topical application of a small-molecule inhibitor of galectin-3, GB1265, in mouse models of corneal wound healing, led to the reduction of the following in injured corneas: i) corneal angiogenesis; ii) corneal fibrosis; iii) infiltration of immune cells; and iv) expression of the proinflammatory cytokine IL-1ß. Four independent techniques (RNA sequencing, NanoString, real-time quantitative RT-PCR, and Western blot analysis) determined that decreased corneal opacity in the galectin-3 inhibitor-treated corneas was associated with decreases in the numbers of genes and signaling pathways known to promote fibrosis. These findings allowed for a high level of confidence in the conclusion that galectin-3 inhibition by the small-molecule inhibitor GB1265 has dual anti-angiogenic and anti-scarring effects. Targeting galectin-3 by GB1265 is, thus, attractive for the development of innovative therapies for a myriad of ocular and nonocular diseases characterized by pathologic angiogenesis and fibrosis.


Subject(s)
Cicatrix , Corneal Injuries , Animals , Mice , Humans , Cicatrix/metabolism , Galectin 3/metabolism , Cornea/metabolism , Corneal Injuries/metabolism , Wound Healing/physiology , Disease Models, Animal , Neovascularization, Pathologic/pathology , Fibrosis
2.
Invest Ophthalmol Vis Sci ; 64(2): 11, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36749596

ABSTRACT

Purpose: The current study was designed to examine the role of the NLRP3 inflammasome pathway in the clearance of Pseudomonas aeruginosa (PA) infection in mouse corneas. Methods: Corneas of wild type and NLRP3-/- mice were infected with PA. The severity of bacterial keratitis was graded on days 1 and 3 post-infection by slit lamp, and then corneas were harvested for: (i) bacterial enumeration, (ii) immune cell analysis by flow cytometry, (iii) immunoblotting analysis of cleaved caspase-1 and IL-1ß, and (iv) IL-1ß quantification by ELISA. In parallel experiments, severity of keratitis was examined in the wild-type mice receiving a subconjunctival injection of a highly selective NLRP3 inhibitor immediately prior to infection. Results: Compared to wild type mice, NLRP3-/- mice exhibited more severe infection, as indicated by an increase in opacity score and an increase in bacterial load. The hallmark of inflammasome assembly is the activation of proinflammatory caspase-1 and IL-1ß by cleavage of their precursors, pro-caspase-1 and pro-IL-1ß, respectively. Accordingly, increased severity of infection in the NLRP3-/- mice was associated with reduced levels of cleaved forms of caspase-1 and IL-1ß and reduced IL-1ß+ neutrophil infiltration in infected corneas. Likewise, corneas of mice receiving subconjunctival injections of NLRP3 inhibitor exhibited increased bacterial load, and reduced IL-1ß expression. Conclusions: Activation of NLRP3 pathway is required for the clearance of PA infection in mouse corneas.


Subject(s)
Keratitis , Pseudomonas Infections , Animals , Mice , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pseudomonas , Keratitis/microbiology , Caspase 1/metabolism , Pseudomonas Infections/microbiology , Interleukin-1beta/metabolism , Mice, Inbred C57BL
3.
J Immunol ; 210(4): 398-407, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36603009

ABSTRACT

Pseudomonas aeruginosa provokes a painful, sight-threatening corneal infection. It progresses rapidly and is difficult to treat. In this study, using a mouse model of P. aeruginosa keratitis, we demonstrate the importance of a carbohydrate-binding protein, galectin-8 (Gal-8), in regulation of the innate immune response. First, using two distinct strains of P. aeruginosa, we established that Gal-8-/- mice are resistant to P. aeruginosa keratitis. In contrast, mice deficient in Gal-1, Gal-3, and Gal-9 were fully susceptible. Second, the addition of exogenous rGal-8 to LPS (TLR4 ligand)-stimulated bone marrow-derived macrophages suppressed 1) the activation of the NF-κB pathway, and 2) formation of the MD-2/TLR4 complex. Additionally, the expression level of reactive oxygen species was substantially higher in infected Gal-8-/- bone marrow neutrophils (BMNs) compared with the Gal-8+/+ BMNs, and the P. aeruginosa killing capacity of Gal-8-/- BMNs was considerably higher compared with that of Gal-8+/+ BMNs. In the bacterial killing assays, the addition of exogenous rGal-8 almost completely rescued the phenotype of Gal-8-/- BMNs. Finally, we demonstrate that a subconjunctival injection of a Gal-8 inhibitor markedly reduces the severity of infection in the mouse model of P. aeruginosa keratitis. These data lead us to conclude that Gal-8 downmodulates the innate immune response by suppressing activation of the TLR4 pathway and clearance of P. aeruginosa by neutrophils. These findings have broad implications for developing novel therapeutic strategies for treatment of conditions resulting from the hyperactive immune response both in ocular as well as nonocular tissues.


Subject(s)
Keratitis , Pseudomonas Infections , Animals , Mice , Pseudomonas aeruginosa , Toll-Like Receptor 4 , Immunity, Innate , Galectins , Mice, Inbred C57BL
4.
J Clin Pharmacol ; 62 Suppl 2: S110-S118, 2022 12.
Article in English | MEDLINE | ID: mdl-36461746

ABSTRACT

New therapeutic modalities carry with them great promise for the treatment of rare diseases. They also present unique development challenges including immunogenicity, which can impact the safety and efficacy of those new modalities. In this review, an overview of the basic function of the immune system and its possible interaction with new therapeutic modalities is presented. A juxtaposition of immunogenicity in the rare disease space versus traditional clinical programs is hereby being proposed. A clinical pharmacology viewpoint of immunogenicity, proposed approaches to account for immunogenicity in clinical data, bioanalytical considerations, and effects of route of administration and production changes on immunogenicity are discussed.


Subject(s)
Pharmacology, Clinical , Rare Diseases , Humans , Rare Diseases/drug therapy
5.
Sci Transl Med ; 12(564)2020 10 07.
Article in English | MEDLINE | ID: mdl-33028709

ABSTRACT

Acute graft-versus-host disease (aGVHD) remains a major complication of allogeneic hematopoietic cell transplantation (HCT). CD146 and CCR5 are proteins that mark activated T helper 17 (Th17) cells. The Th17 cell phenotype is promoted by the interaction of the receptor ICOS on T cells with ICOS ligand (ICOSL) on dendritic cells (DCs). We performed multiparametric flow cytometry in a cohort of 156 HCT recipients and conducted experiments with aGVHD murine models to understand the role of ICOSL+ DCs. We observed an increased frequency of ICOSL+ plasmacytoid DCs, correlating with CD146+CCR5+ T cell frequencies, in the 64 HCT recipients with gastrointestinal aGVHD. In murine models, donor bone marrow cells from ICOSL-deficient mice compared to those from wild-type mice reduced aGVHD-related mortality. Reduced aGVHD resulted from lower intestinal infiltration of pDCs and pathogenic Th17 cells. We transplanted activated human ICOSL+ pDCs along with human peripheral blood mononuclear cells into immunocompromised mice and observed infiltration of intestinal CD146+CCR5+ T cells. We found that prophylactic administration of a dual human ICOS/CD28 antagonist (ALPN-101) prevented aGVHD in this model better than did the clinically approved belatacept (CTLA-4-Fc), which binds CD80 (B7-1) and CD86 (B7-2) and interferes with the CD28 T cell costimulatory pathway. When started at onset of aGVHD signs, ALPN-101 treatment alleviated symptoms of ongoing aGVHD and improved survival while preserving antitumoral cytotoxicity. Our data identified ICOSL+-pDCs as an aGVHD biomarker and suggest that coinhibition of the ICOSL/ICOS and B7/CD28 axes with one biologic drug may represent a therapeutic opportunity to prevent or treat aGVHD.


Subject(s)
CD28 Antigens , Graft vs Host Disease , Abatacept , Animals , Dendritic Cells , Graft vs Host Disease/drug therapy , Inducible T-Cell Co-Stimulator Protein , Leukocytes, Mononuclear , Mice
6.
J Immunol ; 202(10): 3053-3064, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30979817

ABSTRACT

Acute graft-versus-host disease (aGVHD) hinders the efficacy of allogeneic hematopoietic cell transplantation (HCT). Plasma levels of soluble membrane-bound ST2 (ST2) are elevated in human and murine aGVHD and correlated to type 1 T cells response. ST2 signals through the adapter protein MyD88. The role of MyD88 in T cells during aGVHD has yet to be elucidated. We found that knocking out MyD88 in the donor T cells protected against aGVHD independent of IL-1R and TLR4 signaling in two murine HCT models. This protection was entirely driven by MyD88-/- CD4 T cells. Transplanting donor MyD88-/- conventional T cells (Tcons) with wild-type (WT) or MyD88-/- regulatory T cells (Tregs) lowered aGVHD severity and mortality. Transcriptome analysis of sorted MyD88-/- CD4 T cells from the intestine 10 d post-HCT showed lower levels of Il1rl1 (gene of ST2), Ifng, Csf2, Stat5, Batf, and Jak2 Transplanting donor ST2-/- Tcons with WT or ST2-/- Tregs showed a similar phenotype with what we observed when using donor MyD88-/- Tcons. Decreased ST2 was confirmed at the protein level with less secretion of soluble ST2 and more expression of ST2 compared with WT T cells. Our data suggest that Treg suppression from lack of MyD88 signaling in donor Tcons during alloreactivity uses the ST2 but not the IL-1R or TLR4 pathways, and ST2 represents a potential aGVHD therapeutic target sparing Tregs.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Interleukin-1 Receptor-Like 1 Protein/deficiency , Myeloid Differentiation Factor 88/deficiency , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Interleukin-1 Receptor-Like 1 Protein/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , T-Lymphocytes, Regulatory/pathology , Transplantation, Homologous
7.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30694220

ABSTRACT

Soluble stimulation-2 (ST2) is increased during graft-versus-host disease (GVHD), while Tregs that express ST2 prevent GVHD through unknown mechanisms. Transplantation of Foxp3- T cells and Tregs that were collected and sorted from different Foxp3 reporter mice indicated that in mice that developed GVHD, ST2+ Tregs were thymus derived and predominantly localized to the intestine. ST2-/- Treg transplantation was associated with reduced total intestinal Treg frequency and activation. ST2-/- versus WT intestinal Treg transcriptomes showed decreased Treg functional markers and, reciprocally, increased Rorc expression. Rorc-/- T cells transplantation enhanced the frequency and function of intestinal ST2+ Tregs and reduced GVHD through decreased gut-infiltrating soluble ST2-producing type 1 and increased IL-4/IL-10-producing type 2 T cells. Cotransfer of ST2+ Tregs sorted from Rorc-/- mice with WT CD25-depleted T cells decreased GVHD severity and mortality, increased intestinal ST2+KLRG1+ Tregs, and decreased type 1 T cells after transplantation, indicating an intrinsic mechanism. Ex vivo IL-33-stimulated Tregs (TregIL-33) expressed higher amphiregulin and displayed better immunosuppression, and adoptive transfer prevented GVHD better than control Tregs or TregIL-33 cultured with IL-23/IL-17. Amphiregulin blockade by neutralizing antibody in vivo abolished the protective effect of TregIL-33. Our data show that inverse expression of ST2 and RORγt in intestinal Tregs determines GVHD and that TregIL-33 has potential as a cellular therapy avenue for preventing GVHD.


Subject(s)
Graft vs Host Disease/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Intestines/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Bone Marrow Transplantation , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Immune Tolerance , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-10/metabolism , Interleukin-17 , Interleukin-23 , Interleukin-33/metabolism , Interleukin-4/metabolism , Lectins, C-Type/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Immunologic/genetics , T-Lymphocytes/transplantation , T-Lymphocytes, Regulatory/metabolism , Transcriptome
8.
JCI Insight ; 3(14)2018 07 26.
Article in English | MEDLINE | ID: mdl-30046004

ABSTRACT

Soluble cytokine receptors function as decoy receptors to attenuate cytokine-mediated signaling and modulate downstream cellular responses. Dysregulated overproduction of soluble receptors can be pathological, such as soluble ST2 (sST2), a prognostic biomarker in cardiovascular diseases, ulcerative colitis, and graft-versus-host disease (GVHD). Although intervention using an ST2 antibody improves survival in murine GVHD models, sST2 is a challenging target for drug development because it binds to IL-33 via an extensive interaction interface. Here, we report the discovery of small-molecule ST2 inhibitors through a combination of high-throughput screening and computational analysis. After in vitro and in vivo toxicity assessment, 3 compounds were selected for evaluation in 2 experimental GVHD models. We show that the most effective compound, iST2-1, reduces plasma sST2 levels, alleviates disease symptoms, improves survival, and maintains graft-versus-leukemia activity. Our data suggest that iST2-1 warrants further optimization to develop treatment for inflammatory diseases mediated by sST2.


Subject(s)
Drug Discovery , Interleukin-1 Receptor-Like 1 Protein/drug effects , Interleukin-1 Receptor-Like 1 Protein/metabolism , Proteomics , Receptors, Cytokine/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biomarkers , Cell Line, Tumor , Computational Biology , Drug Evaluation, Preclinical , Graft vs Host Disease , High-Throughput Screening Assays , Interleukin-33/metabolism , Leukemia/drug therapy , Mice , Models, Animal , Stem Cell Transplantation
9.
J Exp Med ; 214(12): 3577-3596, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-29038366

ABSTRACT

Allogeneic immune cells, particularly T cells in donor grafts, recognize and eliminate leukemic cells via graft-versus-leukemia (GVL) reactivity, and transfer of these cells is often used for high-risk hematological malignancies, including acute myeloid leukemia. Unfortunately, these cells also attack host normal tissues through the often fatal graft-versus-host disease (GVHD). Full separation of GVL activity from GVHD has yet to be achieved. Here, we show that, in mice and humans, a population of interleukin-9 (IL-9)-producing T cells activated via the ST2-IL-33 pathway (T9IL-33 cells) increases GVL while decreasing GVHD through two opposing mechanisms: protection from fatal immunity by amphiregulin expression and augmentation of antileukemic activity compared with T9, T1, and unmanipulated T cells through CD8α expression. Thus, adoptive transfer of allogeneic T9IL-33 cells offers an attractive approach for separating GVL activity from GVHD.


Subject(s)
Cell Differentiation/immunology , Immunity , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Amphiregulin/metabolism , Animals , Cell Communication , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic , Epithelial Cells/metabolism , Graft vs Host Disease/pathology , Humans , Immunologic Memory , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Interleukin-9/metabolism , Intestines/pathology , Leukemia/immunology , Leukemia/pathology , Mice, Inbred BALB C , Phenotype , Proto-Oncogene Proteins/metabolism , Signal Transduction , Survival Analysis , Trans-Activators/metabolism
11.
Cell Immunol ; 310: 116-122, 2016 12.
Article in English | MEDLINE | ID: mdl-27614844

ABSTRACT

The contribution of dysregulated innate immune responses to the pathogenesis of allergic disease remains largely unknown. Herein, we addressed the role of Toll-like receptor signaling in airway inflammation by studying mice rendered deficient for the myeloid differentiation factor 88 (MyD88-/-) which results in concurrent deficiencies in TLR and IL-1R1 signaling pathways. We show that the lack of MyD88 offers a partial protection from allergic disease evidenced by reduced airway eosinophilia and production of the Th17-associated effector cytokine IL-17A. By contrast, airway hyperreactivity and Th2 cytokine production, the cardinal features of allergic disease, remained unchanged. We found that the impaired IL-17A production in MyD88-/- mice was associated with defective CD4+ T cells, which failed to respond to IL-23 stimulation. The total number of Th17-associated effectors in lymph nodes was likewise decreased. Taken together, our results demonstrate that MyD88-dependent mechanisms are critical for orchestrating lung inflammatory responses, in terms of IL-17A production, as well as eosinophil and neutrophil recruitment.


Subject(s)
Eosinophils/immunology , Hypersensitivity/immunology , Myeloid Differentiation Factor 88/metabolism , Neutrophils/immunology , Pneumonia/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Cell Movement/genetics , Cells, Cultured , Interleukin-17/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics
12.
Eur J Immunol ; 46(9): 2247-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27334749

ABSTRACT

T-cell polyspecificity, predicting that individual T cells recognize a continuum of related ligands, implies that multiple antigens can tolerize T cells specific for a given self-antigen. We previously showed in C57BL/6 mice that part of the CD4(+) T-cell repertoire specific for myelin oligodendrocyte glycoprotein (MOG) 35-55 also recognizes the neuronal antigen neurofilament medium (NF-M) 15-35. Such bi-specific CD4(+) T cells are frequent and produce inflammatory cytokines after stimulation. Since T cells recognizing two self-antigens would be expected to be tolerized more efficiently, this finding prompted us to study how polyspecificity impacts tolerance. We found that similar to MOG, NF-M is expressed in the thymus by medullary thymic epithelial cells, a tolerogenic population. Nevertheless, the frequency, phenotype, and capacity to transfer experimental autoimmune encephalomyelitis (EAE) of MOG35-55 -reactive CD4(+) T cells were increased in MOG-deficient but not in NF-M-deficient mice. We found that presentation of NF-M15-35 by I-A(b) on dendritic cells is of short duration, suggesting unstable MHC class II binding. Consistently, introducing an MHC-anchoring residue into NF-M15-35 (NF-M15-35 T20Y) increased its immunogenicity, activating a repertoire able to induce EAE. Our results show that in C57BL/6 mice bi-specific encephalitogenic T cells manage to escape tolerization due to inefficient exposure to two self-antigens.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immune Tolerance , Myelin Proteins/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurons/immunology , Animals , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/genetics , Neurofilament Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Cell Antigen Receptor Specificity/genetics , T-Cell Antigen Receptor Specificity/immunology , Thymus Gland/immunology , Thymus Gland/metabolism
13.
JCI Insight ; 1(6)2016 May 05.
Article in English | MEDLINE | ID: mdl-27195312

ABSTRACT

Gastrointestinal graft-versus-host-disease (GI-GVHD) is a life-threatening complication occurring after allogeneic hematopoietic cell transplantation (HCT), and a blood biomarker that permits stratification of HCT patients according to their risk of developing GI-GVHD would greatly aid treatment planning. Through in-depth, large-scale proteomic profiling of presymptomatic samples, we identified a T cell population expressing both CD146, a cell adhesion molecule, and CCR5, a chemokine receptor that is upregulated as early as 14 days after transplantation in patients who develop GI-GVHD. The CD4+CD146+CCR5+ T cell population is Th17 prone and increased by ICOS stimulation. shRNA knockdown of CD146 in T cells reduced their transmigration through endothelial cells, and maraviroc, a CCR5 inhibitor, reduced chemotaxis of the CD4+CD146+CCR5+ T cell population toward CCL14. Mice that received CD146 shRNA-transduced human T cells did not lose weight, showed better survival, and had fewer CD4+CD146+CCR5+ T cells and less pathogenic Th17 infiltration in the intestine, even compared with mice receiving maraviroc with control shRNA- transduced human T cells. Furthermore, the frequency of CD4+CD146+CCR5+ Tregs was increased in GI-GVHD patients, and these cells showed increased plasticity toward Th17 upon ICOS stimulation. Our findings can be applied to early risk stratification, as well as specific preventative therapeutic strategies following HCT.

14.
Brain ; 139(Pt 5): 1433-46, 2016 05.
Article in English | MEDLINE | ID: mdl-27000832

ABSTRACT

Polyspecific T cells recognizing multiple distinct self-antigens have been identified in multiple sclerosis and other organ-specific autoimmune diseases, but their pathophysiological relevance remains undetermined. Using a mouse model of multiple sclerosis, we show that autoimmune encephalomyelitis induction is strictly dependent on reactivation of pathogenic T cells by a peptide (35-55) derived from myelin oligodendrocyte glycoprotein (MOG). This disease-inducing response wanes after onset. Strikingly, the progression of disease is driven by the in situ activation and expansion of a minority of MOG35-55-specific T cells that also recognize neurofilament-medium (NF-M)15-35, an intermediate filament protein expressed in neurons. This mobilization of bispecific T cells is critical for disease progression as adoptive transfer of NF-M15-35/MOG35-55 bispecific T cell lines caused full-blown disease in wild-type but not NF-M-deficient recipients. Moreover, specific tolerance through injection of NF-M15-35 peptide at the peak of disease halted experimental autoimmune encephalomyelitis progression. Our findings highlight the importance of polyspecific autoreactive T cells in the aggravation and perpetuation of central nervous system autoimmunity.


Subject(s)
Autoantigens/immunology , Autoimmunity , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Peptide Fragments/immunology
15.
Sci Transl Med ; 7(308): 308ra160, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26446957

ABSTRACT

Graft-versus-host disease (GVHD) remains a devastating complication after allogeneic hematopoietic cell transplantation (HCT). We previously identified high plasma soluble suppression of tumorigenicity 2 (sST2) as a biomarker of the development of GVHD and death. sST2 sequesters interleukin-33 (IL-33), limiting its availability to T cells expressing membrane-bound ST2 (mST2) [T helper 2 (TH2) cells and ST2(+)FoxP3(+) regulatory T cells]. We report that blockade of sST2 in the peritransplant period with a neutralizing monoclonal antibody (anti-ST2 mAb) reduced GVHD severity and mortality. We identified intestinal stromal cells and T cells as major sources of sST2 during GVHD. ST2 blockade decreased systemic interferon-γ, IL-17, and IL-23 but increased IL-10 and IL-33 plasma levels. ST2 blockade also reduced sST2 production by IL-17-producing T cells while maintaining protective mST2-expressing T cells, increasing the frequency of intestinal myeloid-derived suppressor cells, and decreasing the frequency of intestinal CD103 dendritic cells. Finally, ST2 blockade preserved graft-versus-leukemia activity in a model of green fluorescent protein (GFP)-positive MLL-AF9 acute myeloid leukemia. Our findings suggest that ST2 is a therapeutic target for severe GVHD and that the ST2/IL-33 pathway could be investigated in other T cell-mediated immune disorders with loss of tolerance.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Graft vs Host Disease/drug therapy , Receptors, Cell Surface/antagonists & inhibitors , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/therapeutic use , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft vs Host Disease/metabolism , Mice , Mice, Inbred BALB C
16.
Front Immunol ; 6: 14, 2015.
Article in English | MEDLINE | ID: mdl-25674088

ABSTRACT

Hematopoietic stem-cell transplantation (HSCT) is the most potent curative therapy for many malignant and non-malignant disorders. Unfortunately, a major complication of HSCT is graft-versus-host disease (GVHD), which is mediated by tissue damage resulting from the conditioning regimens before the transplantation and the alloreaction of dual immune components (activated donor T-cells and recipient's antigen-presenting cells). This tissue damage leads to the release of alarmins and the triggering of pathogen-recognition receptors that activate the innate immune system and subsequently the adaptive immune system. Alarmins, which are of endogenous origin, together with the exogenous pathogen-associated molecular patterns (PAMPs) elicit similar responses of danger signals and represent the group of damage-associated molecular patterns (DAMPs). Effector cells of innate and adaptive immunity that are activated by PAMPs or alarmins can secrete other alarmins and amplify the immune responses. These complex interactions and loops between alarmins and PAMPs are particularly potent at inducing and then aggravating the GVHD reaction. In this review, we highlight the role of these tissue damaging molecules and their signaling pathways. Interestingly, some DAMPs and PAMPs are organ specific and GVHD-induced and have been shown to be interesting biomarkers. Some of these molecules may represent potential targets for novel therapeutic approaches.

17.
J Immunol ; 193(7): 3267-77, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25135834

ABSTRACT

The recognition of multiple ligands by a single TCR is an intrinsic feature of T cell biology, with important consequences for physiological and pathological processes. Polyspecific T cells targeting distinct self-antigens have been identified in healthy individuals as well as in the context of autoimmunity. We have previously shown that the 2D2 TCR recognizes the myelin oligodendrocyte glycoprotein epitope (MOG)35-55 as well as an epitope within the axonal protein neurofilament medium (NF-M15-35) in H-2(b) mice. In this study, we assess whether this cross-reactivity is a common feature of the MOG35-55-specific T cell response. To this end, we analyzed the CD4 T cell response of MOG35-55-immunized C57BL/6 mice for cross-reactivity with NF-M15-35. Using Ag recall responses, we established that an important proportion of MOG35-55-specific CD4 T cells also responded to NF-M15-35 in all mice tested. To study the clonality of this response, we analyzed 22 MOG35-55-specific T cell hybridomas expressing distinct TCR. Seven hybridomas were found to cross-react with NF-M15-35. Using an alanine scan of NF-M18-30 and an in silico predictive model, we dissected the molecular basis of cross-reactivity between MOG35-55 and NF-M15-35. We established that NF-M F24, R26, and V27 proved important TCR contacts. Strikingly, the identified TCR contacts are conserved within MOG38-50. Our data indicate that due to linear sequence homology, part of the MOG35-55-specific T cell repertoire of all C57BL/6 mice also recognizes NF-M15-35, with potential implications for CNS autoimmunity.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Myelin Sheath/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurofilament Proteins/immunology , Receptors, Antigen/immunology , Animals , Autoantigens/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , CD4-Positive T-Lymphocytes/pathology , Cross Reactions/genetics , Cross Reactions/immunology , Mice , Mice, Knockout , Myelin Sheath/genetics , Myelin-Oligodendrocyte Glycoprotein/genetics , Neurofilament Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Antigen/genetics
18.
PLoS One ; 5(7): e11484, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20628601

ABSTRACT

BACKGROUND: Infections may be associated with exacerbation of allergic and autoimmune diseases. Paradoxically, epidemiological and experimental data have shown that some microorganisms can also prevent these pathologies. This observation is at the origin of the hygiene hypothesis according to which the decline of infections in western countries is at the origin of the increased incidence of both Th1-mediated autoimmune diseases and Th2-mediated allergic diseases over the last decades. We have tested whether Toll-like receptor (TLR) stimulation can recapitulate the protective effect of infectious agents on allergy and autoimmunity. METHODS AND FINDINGS: Here, we performed a systematic study of the disease-modifying effects of a set of natural or synthetic TLR agonists using two experimental models, ovalbumin (OVA)-induced asthma and spontaneous autoimmune diabetes, presenting the same genetic background of the non obese diabetic mouse (NOD) that is highly susceptible to both pathologies. In the same models, we also investigated the effect of probiotics. Additionally, we examined the effect of the genetic invalidation of MyD88 on the development of allergic asthma and spontaneous diabetes. We demonstrate that multiple TLR agonists prevent from both allergy and autoimmunity when administered parenterally. Probiotics which stimulate TLRs also protect from these two diseases. The physiological relevance of these findings is further suggested by the major acceleration of OVA-induced asthma in MyD88 invalidated mice. Our results strongly indicate that the TLR-mediated effects involve immunoregulatory cytokines such as interleukin (IL)-10 and transforming growth factor (TGF)-beta and different subsets of regulatory T cells, notably CD4+CD25+FoxP3+ T cells for TLR4 agonists and NKT cells for TLR3 agonists. CONCLUSIONS/SIGNIFICANCE: These observations demonstrate that systemic administration of TLR ligands can suppress both allergic and autoimmune responses. They provide a plausible explanation for the hygiene hypothesis. They also open new therapeutic perspectives for the prevention of these pathologies.


Subject(s)
Asthma/drug therapy , Asthma/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Animals , Bacterial Outer Membrane Proteins/therapeutic use , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Imidazoles/therapeutic use , Lipopolysaccharides/therapeutic use , Mice , Mice, Inbred NOD , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Probiotics/therapeutic use , RNA, Double-Stranded/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism
20.
Int Immunopharmacol ; 8(12): 1626-32, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-18692598

ABSTRACT

It is well documented that compounds from rhizomes of Zingiber officinale, commonly called ginger, have anti-inflammatory properties. Here, we show that ginger can exert such functions in vivo, namely in a mouse model of Th2-mediated pulmonary inflammation. The preparation of ginger aqueous extract (Zo.Aq) was characterized by mass spectrometry as an enriched fraction of n-gingerols. Intraperitoneal injections of this extract before airway challenge of ovalbumin (OVA)-sensitized mice resulted in a marked decrease in the recruitment of eosinophils to the lungs as attested by cell counts in bronchoalveolar lavage (BAL) fluids and histological examination. Resolution of airway inflammation induced by Zo.Aq was accompanied by a suppression of the Th2 cell-driven response to allergen in vivo. Thus, IL-4, IL-5 and eotaxin levels in the lungs as well as specific IgE titres in serum were clearly diminished in ginger-treated mice relative to their controls after allergen sensitization and challenge. Finally, we found that [6]-gingerol, a major constituent of ginger, was sufficient to suppress eosinophilia in our model of inflammation. This is the first evidence that ginger can suppress Th2-mediated immune responses and might thus provide a possible therapeutic application in allergic asthma.


Subject(s)
Asthma/drug therapy , Eosinophilia/prevention & control , Phytotherapy , Plant Extracts/therapeutic use , Th2 Cells/immunology , Zingiber officinale , Animals , Asthma/immunology , Zingiber officinale/chemistry , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Ovalbumin/immunology , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...