Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Int J Mol Sci ; 25(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38732169

ABSTRACT

Infections may affect the course of autoimmune inflammatory diseases of the central nervous system (CNS), such as multiple sclerosis (MS). Infections with lactate dehydrogenase-elevating virus (LDV) protected mice from developing experimental autoimmune encephalomyelitis (EAE), a mouse counterpart of MS. Uninfected C57BL/6 mice immunized with the myelin oligodendrocyte glycoprotein peptide (MOG35-55) experienced paralysis and lost weight at a greater rate than mice who had previously been infected with LDV. LDV infection decreased the presentation of the MOG peptide by CD11b+CD11c+ dendritic cells (DC) to pathogenic T lymphocytes. When comparing non-infected mice to infected mice, the histopathological examination of the CNS showed more areas of demyelination and CD45+ and CD3+, but not Iba1+ cell infiltration. These results suggest that the protective effect of LDV infection against EAE development is mediated by a suppression of myelin antigen presentation by a specific DC subset to autoreactive T lymphocytes. Such a mechanism might contribute to the general suppressive effect of infections on autoimmune diseases known as the hygiene hypothesis.


Subject(s)
Dendritic Cells , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Lactate dehydrogenase-elevating virus , Mice, Inbred C57BL , Multiple Sclerosis , Myelin-Oligodendrocyte Glycoprotein , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Myelin-Oligodendrocyte Glycoprotein/immunology , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Multiple Sclerosis/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/virology , Lactate dehydrogenase-elevating virus/immunology , CD11b Antigen/metabolism , CD11b Antigen/immunology , Antigen Presentation/immunology , Female , CD11c Antigen/metabolism , Cardiovirus Infections/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Front Immunol ; 14: 1224383, 2023.
Article in English | MEDLINE | ID: mdl-38146368

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a major health issue primarily caused by cigarette smoke (CS) and characterized by breathlessness and repeated airway inflammation. NLRP6 is a cytosolic innate receptor controlling intestinal inflammation and orchestrating the colonic host-microbial interface. However, its roles in the lungs remain largely unexplored. Using CS exposure models, our data show that airway inflammation is strongly impaired in Nlrp6-deficient mice with drastically fewer recruited neutrophils, a key cell subset in inflammation and COPD. We found that NLRP6 expression in lung epithelial cells is important to control airway and lung tissue inflammation in an inflammasome-dependent manner. Since gut-derived metabolites regulate NLRP6 inflammasome activation in intestinal epithelial cells, we investigated the link between NLRP6, CS-driven lung inflammation, and gut microbiota composition. We report that acute CS exposure alters gut microbiota in both wild-type (WT) and Nlrp6-deficient mice and that antibiotic treatment decreases CS-induced lung inflammation. In addition, gut microbiota transfer from dysbiotic Nlrp6-deficient mice to WT mice decreased airway lung inflammation in WT mice, highlighting an NLRP6-dependent gut-to-lung axis controlling pulmonary inflammation.


Subject(s)
Gastrointestinal Microbiome , Pneumonia , Receptors, Cell Surface , Tobacco Smoke Pollution , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/microbiology , Animals , Mice , Mice, Inbred C57BL , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/pathology , Feces/microbiology , Bacteria/classification , Bacteria/metabolism , Biodiversity , Gene Expression
3.
Int J Mol Sci ; 24(15)2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37569646

ABSTRACT

For effective treatments and preventive measures against severe COVID-19, it is essential to determine early markers of disease severity in different populations. We analysed the cytokine kinetics of 129 COVID-19 patients with mild symptoms, 68 severe cases, and 20 healthy controls for the first time in Rwanda. Pro-inflammatory (IFNγ, IL-6, TNFα), Treg (IL-10, TGFß1, TGFß3), Th9 (IL-9), Th17 (IL-17), and Th2 (IL-4, IL-13) cytokines, total IgM and IgG, as well as gene expressions of FoxP3, STAT5+, IFNγ-R1, and ROR alpha+, were measured at day 1, day 7, day 14, day 21, and day 28 post-infection. Severe cases showed a significantly stronger increase than mild patients in levels of all cytokines (except IL-9) and all gene expression on day 1 of infection. Some cytokine levels dropped to levels comparable to mild cases at later time points. Further analysis identified IFNγ as a marker of severity throughout the disease course, while TGFß1, IL-6, and IL-17 were markers of severity only at an early phase. Importantly, this study revealed a striking low IL-9 level and high IFNγ/IL-9 ratio in the plasma of patients who later died compared to mild and severe cases who recovered, suggesting that this could be an important biomarker for predicting the severity of COVID-19 and post-COVID-19 syndrome.


Subject(s)
COVID-19 , Cytokines , Humans , Cytokines/genetics , Interleukin-17/genetics , Interleukin-9/genetics , Interleukin-6 , Kinetics , Post-Acute COVID-19 Syndrome , Rwanda/epidemiology , Interferon-gamma , Patient Acuity
4.
Signal Transduct Target Ther ; 8(1): 155, 2023 04 17.
Article in English | MEDLINE | ID: mdl-37069149

ABSTRACT

Loss of function of the von Hippel-Lindau (VHL) tumor suppressor gene is a hallmark of clear cell renal cell carcinoma (ccRCC). The importance of heterogeneity in the loss of this tumor suppressor has been under reported. To study the impact of intratumoral VHL heterogeneity observed in human ccRCC, we engineered VHL gene deletion in four RCC models, including a new primary tumor cell line derived from an aggressive metastatic case. The VHL gene-deleted (VHL-KO) cells underwent epithelial-to-mesenchymal transition (EMT) and exhibited increased motility but diminished proliferation and tumorigenicity compared to the parental VHL-expressing (VHL+) cells. Renal tumors with either VHL+ or VHL-KO cells alone exhibit minimal metastatic potential. Combined tumors displayed rampant lung metastases, highlighting a novel cooperative metastatic mechanism. The poorly proliferative VHL-KO cells stimulated the proliferation, EMT, and motility of neighboring VHL+ cells. Periostin (POSTN), a soluble protein overexpressed and secreted by VHL non-expressing (VHL-) cells, promoted metastasis by enhancing the motility of VHL-WT cells and facilitating tumor cell vascular escape. Genetic deletion or antibody blockade of POSTN dramatically suppressed lung metastases in our preclinical models. This work supports a new strategy to halt the progression of ccRCC by disrupting the critical metastatic crosstalk between heterogeneous cell populations within a tumor.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Lung Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Genes, Tumor Suppressor , Lung Neoplasms/genetics
5.
Int J Mol Sci ; 23(20)2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36293524

ABSTRACT

The proper control of Plasmodium infection requires a finely balanced immune response. Here, we evaluated the implication of TGF-ß1 and TGF-ß3 in this process using novel monoclonal antibodies to measure their plasma concentrations in comparison with other cytokines and the expression of FOXP3 mRNA. Plasma cytokine levels were measured in 80 patients with severe anaemic malaria and 186 with a mild presentation using ELISA, and rtPCR was used to measure FOXP3 mRNA expression. While no mature TGF-ß isoforms were detected in the plasma, the latent TGF-ß1 and TGF-ß3 were strongly upregulated in patients with mild malaria and nearly undetected in patients with severe disease. Similar selective upregulation in mild patients was observed for IL-9 and FOXP3 mRNA, while IL-7, IL-10, IL-17, and IL-27, although higher in mild cases, were also detected in severe disease. In contrast, a clearly skewed trend of severe cases towards higher pro-inflammatory (IL-6, IL-13, TNF-α) and Th1 (IFN-γ) responses was observed, which was associated with a higher level of parasitaemia as well as lower IgG and higher IgM responses. Together, these results suggest that the stimulation of regulatory T cells through TGF-ß1/TGF-ß3 and IL-9 is paramount to an effective and balanced protective immunity in natural human malaria infection.


Subject(s)
Interleukin-27 , Malaria , Humans , Interleukin-10 , Transforming Growth Factor beta1/genetics , Interleukin-13 , Interleukin-17 , Interleukin-9/genetics , Tumor Necrosis Factor-alpha , Up-Regulation , Transforming Growth Factor beta3 , Interleukin-6 , Interleukin-7 , Cytokines , Transforming Growth Factor beta , RNA, Messenger , Immunoglobulin M , Immunoglobulin G , Forkhead Transcription Factors , Antibodies, Monoclonal
6.
Front Med (Lausanne) ; 9: 1057252, 2022.
Article in English | MEDLINE | ID: mdl-36714124

ABSTRACT

Introduction: Natural prevention of cancer development depends on an efficient immunosurveillance that may be modulated by environmental factors, including infections. Innate lymphoid cytotoxic cells have been shown to play a major role in this immunosurveillance. Interleukin-12 (IL-12) has been suggested to be a key factor in the activation of innate cytotoxic cells after infection, leading to the enhancement of cancer immunosurveillance. Methods: The aim of this work was to analyze in mouse experimental models by which mechanisms the interaction between infectious agent molecules and the early innate responses could enhance early inhibition of cancer growth and especially to assess the role of IL-12 by using novel antibodies specific for IL-12 heterodimers. Results: Ligation of toll-like receptor (TLR)9 by CpG-protected mice against plasmacytoma TEPC.1033.C2 cell early growth. This protection mediated by innate cytolytic cells was strictly dependent on IL-12 and partly on gamma-interferon. Moreover, the protective effect of CpG stimulation, and to a lesser extent of TLR3 and TLR7/8, and the role of IL-12 in this protection were confirmed in a model of early mesothelioma AB1 cell growth. Discussion: These results suggest that modulation of the mouse immune microenvironment by ligation of innate receptors deeply modifies the efficiency of cancer immunosurveillance through the secretion of IL-12, which may at least partly explain the inhibitory effect of previous infections on the prevalence of some cancers.

7.
Commun Biol ; 4(1): 1296, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34789823

ABSTRACT

TGFß is a potential target in cancer treatment due to its dual role in tumorigenesis and homeostasis. However, the expression of TGFß and its inhibition within the tumor microenvironment has mainly been investigated in stroma-heavy tumors. Using B16 mouse melanoma and CT26 colon carcinoma as models of stroma-poor tumors, we demonstrate that myeloid/dendritic cells are the main sources of TGFß1 and TGFß3. Depending on local expression of TGFß isoforms, isoform specific inhibition of either TGFß1 or TGFß3 may be effective. The TGFß signature of CT26 colon carcinoma is defined by TGFß1 and TGFß1 inhibition results in tumor delay; B16 melanoma has equal expression of both isoforms and inhibition of either TGFß1 or TGFß3 controls tumor growth. Using T cell functional assays, we show that the mechanism of tumor delay is through and dependent on enhanced CD8+ T cell function. To overcome the local immunosuppressive environment, we found that combining TGFß inhibition with immune checkpoint blockade results in improved tumor control. Our data suggest that TGFß inhibition in stroma poor tumors shifts the local immune environment to favor tumor suppression.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis , Transforming Growth Factor beta/pharmacology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
8.
Eur J Immunol ; 51(6): 1482-1493, 2021 06.
Article in English | MEDLINE | ID: mdl-33788263

ABSTRACT

The dimeric cytokine IL-12 is important in the control of various infections but also contributes to the pathology of certain diseases making it a potential target for therapy. However, its specific inhibition with antibodies is complicated by the fact that its two subunits are present in other cytokines: p40 in IL-23 and p35 in IL-35. This has led to erroneous conclusions like the alleged implication of IL-12 in experimental autoimmune encephalomyelitis (EAE). Here, we report the development of a mouse anti-mouse IL-12 vaccine and the production of monoclonal antibodies (mAbs) that do not react with p40 or p35 (in IL-35) but specifically recognize and functionally inhibit the IL-12 heterodimer. Using one of these mAbs, MM12A1.6, that strongly inhibited IFN-γ production and LPS-induced septic shock after viral infection, we demonstrate the critical role played by IL-12 in the rejection of male skin graft by female C57BL/6 syngeneic recipients and in the clearance of an immunogenic mastocytoma tumor variant by DBA/2 mice, but not in a parent to F1 immune aggression model nor in MOG-induced EAE, which was clearly prevented by anti-p40 mAb C17.8. Given this selective inhibition of IL-12, these mAbs provide new options for reassessing IL-12 function in vivo.


Subject(s)
Antibodies, Monoclonal/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Graft Rejection/immunology , Interleukin-12/metabolism , Mastocytoma/immunology , Multiple Sclerosis/immunology , Nidovirales Infections/immunology , Nidovirales/physiology , Protein Subunits/metabolism , Sepsis/immunology , Skin Transplantation , Animals , Antibodies, Monoclonal/isolation & purification , Disease Models, Animal , Epitopes , Humans , Hybridomas , Interleukin-12/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasms, Experimental , Protein Subunits/immunology
9.
J Immunother Cancer ; 9(2)2021 02.
Article in English | MEDLINE | ID: mdl-33637600

ABSTRACT

BACKGROUND: Transforming growth factor-ß (TGFß) is emerging as a promising target for cancer therapy, given its ability to promote progression of advanced tumors and to suppress anti-tumor immune responses. However, TGFß also plays multiple roles in normal tissues, particularly during organogenesis, raising toxicity concerns about TGFß blockade. Dose-limiting cardiovascular toxicity was observed, possibly due to the blockade of all three TGFß isoforms. The dominant isoform in tumors is TGFß1, while TGFß2 and TGFß3 seem to be more involved in cardiovascular development. Recent data indicated that selective targeting of TGFß1 promoted the efficacy of checkpoint inhibitor anti-PD1 in transplanted preclinical tumor models, without cardiovascular toxicity. METHODS: To further explore the therapeutic potential of isoform-specific TGFß blockade, we developed neutralizing mAbs targeting mature TGFß1 or TGFß3, and tested them, in parallel with anti-panTGFß mAb 1D11, in two preclinical models: the transplanted colon cancer model CT26, and the autochthonous melanoma model TiRP. RESULTS: We observed that the blockade of TGFß1, but not that of TGFß3, increased the efficacy of a prophylactic cellular vaccine against colon cancer CT26. This effect was similar to pan-TGFß blockade, and was associated with increased infiltration of activated CD8 T cells in the tumor, and reduced levels of regulatory T cells and myeloid-derived suppressor cells. In contrast, in the autochthonous TiRP melanoma model, we observed therapeutic efficacy of the TGFß1-specific mAb as a single agent, while the TGFß3 mAb was inactive. In this model, the anti-tumor effect of TGFß1 blockade was tumor intrinsic rather than immune mediated, as it was also observed in T-cell depleted mice. Mechanistically, TGFß1 blockade increased mouse survival by delaying the phenotype switch, akin to epithelial-to-mesenchymal transition (EMT), which transforms initially pigmented tumors into highly aggressive unpigmented tumors. CONCLUSIONS: Our results confirm TGFß1 as the relevant isoform to target for cancer therapy, not only in combination with checkpoint inhibitors, but also with other immunotherapies such as cancer vaccines. Moreover, TGFß1 blockade can also act as a monotherapy, through a tumor-intrinsic effect blocking the EMT-like transition. Because human melanomas that resist therapy often express a gene signature that links TGFß1 with EMT-related genes, these results support the clinical development of TGFß1-specific mAbs in melanoma.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/pharmacology , Colorectal Neoplasms/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Male , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice, Inbred BALB C , Mice, Transgenic , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/antagonists & inhibitors , Transforming Growth Factor beta3/immunology , Transforming Growth Factor beta3/metabolism , Tumor Microenvironment
10.
Front Immunol ; 11: 1085, 2020.
Article in English | MEDLINE | ID: mdl-32655552

ABSTRACT

In many infectious diseases, the immune response operates as a double-edged sword. While required for protective immunity, infection-induced inflammation can be detrimental if it is not properly controlled, causing collateral body damage and potentially leading to death. It is in this context that the potent anti-inflammatory cytokine interleukin-10 (IL-10) is required to dampen the pro-inflammatory immune response that hallmarks trypanosomosis. Effective control of this infection requires not just the action of antibodies specific for the parasite's variable surface glycoprotein (VSG) coat antigens, but also a pro-inflammatory immune response mediated mainly by IFNγ, TNF, and NO. However, strict control of inflammation is mandatory, as IL-10-deficient mice succumb from an unrestrained cytokine storm within 10 days of a Trypanosome brucei infection. The relevant cellular source of IL-10 and the associated molecular mechanisms implicated in its trypanosomosis associated production are poorly understood. Using an IL-10 reporter mouse strain (Vert-X), we demonstrate here that NK cells, CD8+ T cells and CD4+ T cells as well as B cells and plasma cells constitute potential cellular sources of IL-10 within the spleen and liver during acute infection. The IL-10 wave follows peak pro-inflammatory cytokine production, which accompanied the control of peak parasitemia. Similar results were observed following conventional experimental needle infection and physiological infections via T. brucei-infected tsetse flies. Our results show that conditional T cell-specific ablation of the IL-10 regulating Prdm1 gene (encoding for the Blimp-1 transcription factor), leads to an uncontrolled trypanosome-induced pro-inflammatory syndrome like the one observed in infected IL-10-deficient mice. This result indicates that the biological role of IL-10-derived from non-T cells, including NK cells, is of minor importance when considering host survival. The cytokine IL-27 that is also considered to be an IL-10 regulator, did not affect IL-10 production during infection. Together, these data suggest that T. brucei activates a Blimp-1-dependent IL-10 regulatory pathway in T cells that acts as a critical anti-inflammatory rheostat, mandatory for host survival during the acute phase of parasitemia.


Subject(s)
Cytokine Release Syndrome/prevention & control , Interleukin-10/biosynthesis , Positive Regulatory Domain I-Binding Factor 1/immunology , T-Lymphocytes/immunology , Trypanosoma brucei brucei , Trypanosomiasis, African/immunology , Animals , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Disease Models, Animal , Female , Inflammation/etiology , Inflammation/immunology , Inflammation/prevention & control , Insect Vectors/parasitology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukins/antagonists & inhibitors , Interleukins/deficiency , Interleukins/immunology , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/deficiency , Positive Regulatory Domain I-Binding Factor 1/genetics , Spleen/immunology , Trypanosomiasis, African/complications , Trypanosomiasis, African/parasitology , Tsetse Flies/parasitology
11.
Infect Agent Cancer ; 15: 30, 2020.
Article in English | MEDLINE | ID: mdl-32391074

ABSTRACT

BACKGROUND: Viral infections can reduce early cancer development through enhancement of cancer immunosurveillance. This study was performed to analyse this effect of viral infection in a mouse model of solid tumor. METHODS: The experimental model used was the effect of BALB/c mouse infection by lactate dehydrogenase-elevating virus on AB1 mesothelioma cancer development. RESULTS: Acute infection with lactate dehydrogenase-elevating virus strongly reduced in vivo early AB1 mesothelioma growth and death resulting from cancer development. This effect was not due to a direct cytolytic effect of the virus on AB1 cells, but to an in vivo activation of natural killer cells. Gamma-interferon production rather than cytotoxic activity against AB1 cells mediated this protective effect. This gamma-interferon production by natural killer cells was dependent on interleukin-12 production. CONCLUSIONS: Together with other reported effects of infectious agents on cancer development, this observation may support the hypothesis that enhancement of innate immunosurveillance against tumors may result from infection with common infectious agents through modulation of the host immune microenvironment.

12.
J Exp Med ; 217(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32453422

ABSTRACT

NKG2D is a danger sensor expressed on different subsets of innate and adaptive lymphocytes. Despite its established role as a potent activator of the immune system, NKG2D-driven regulation of CD4+ T helper (Th) cell-mediated immunity remains unclear. In this study, we demonstrate that NKG2D modulates Th1 and proinflammatory T-bet+ Th17 cell effector functions in vitro and in vivo. In particular, NKG2D promotes higher production of proinflammatory cytokines by Th1 and T-bet+ Th17 cells and reinforces their transcription of type 1 signature genes, including Tbx21. Conditional deletion of NKG2D in T cells impairs the ability of antigen-specific CD4+ T cells to promote inflammation in vivo during antigen-induced arthritis and experimental autoimmune encephalomyelitis, indicating that NKG2D is an important target for the amelioration of Th1- and Th17-mediated chronic inflammatory diseases.


Subject(s)
Arthritis, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cytokines/genetics , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th1 Cells/pathology , Th17 Cells/pathology
13.
Front Immunol ; 11: 144, 2020.
Article in English | MEDLINE | ID: mdl-32161582

ABSTRACT

Airborne ozone exposure causes severe lung injury and inflammation. The aryl hydrocarbon Receptor (AhR) (1), activated in pollutant-induced inflammation, is critical for cytokine production, especially IL-22 and IL-17A. The role of AhR in ozone-induced lung inflammation is unknown. We report here that chronic ozone exposure activates AhR with increased tryptophan and lipoxin A4 production in mice. AhR-/- mice show increased lung inflammation, airway hyperresponsiveness, and tissue remodeling with an increased recruitment of IL-17A and IL-22-expressing cells in comparison to control mice. IL-17A- and IL-22-neutralizing antibodies attenuate lung inflammation in AhR-/- and control mice. Enhanced lung inflammation and recruitment of ILC3, ILC2, and T cells were observed after T cell-specific AhR depletion using the AhRCD4cre-deficient mice. Together, the data demonstrate that ozone exposure activates AhR, which controls lung inflammation, airway hyperresponsiveness, and tissue remodeling via the reduction of IL-22 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Interleukins/metabolism , Lung Injury/chemically induced , Lung Injury/metabolism , Ozone/adverse effects , Pneumonia/chemically induced , Pneumonia/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , CD4-Positive T-Lymphocytes/immunology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/genetics , Interleukins/immunology , Lipoxins/metabolism , Lung Injury/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/drug therapy , Receptors, Aryl Hydrocarbon/genetics , Receptors, Interleukin-17/genetics , Respiratory Hypersensitivity/drug therapy , Tryptophan/metabolism , Interleukin-22
14.
Int J Pharm X ; 1: 100019, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31517284

ABSTRACT

PEGylation is a promising approach to increase the residence time of antibody fragments in the lungs and sustain their therapeutic effects. However, concerns arise as to the potential pulmonary toxicity of antibody fragments conjugated to high molecular weight (HMW) polyethylene glycol (PEG), notably after repeated administrations, and the possibility of PEG accumulation in the lungs. The purpose of this proof-of-concept study is to give insights about the safety of lung administration of a Fab' anti-IL17A antibody fragment conjugated to two-armed 40 kDa PEG (PEG40). The presence of the PEG40 moiety inside alveolar macrophages remained stable for at least 24 h after intratracheal administration of PEG40-Fab' to mice. PEG40 was then progressively cleared from alveolar macrophages. Incubation of PEG40 alone with macrophages in vitro did not significantly harm macrophages and did not affect phagocytosis or the production of inflammatory markers. After acute or chronic administration of PEG40-Fab' to mice, no signs of significant pulmonary toxicity or inflammatory cell accumulation were observed. A vacuolization of alveolar macrophages not associated with any inflammation was noticed when PEG40, PEG40-Fab', or unPEGylated Fab' were administered. To conclude this preliminary proof of concept study, acute or repeated pulmonary administrations of PEGylated Fab' appear safe in rodents.

15.
Haematologica ; 104(2): 392-402, 2019 02.
Article in English | MEDLINE | ID: mdl-30213828

ABSTRACT

In spite of considerable therapeutic progress, acute graft-versus-host disease still limits allogeneic hematopoietic cell transplantation. We recently reported that mouse infection with nidovirus lactate dehydrogenase elevating virus impairs disease in non-conditioned B6D2F1 recipients of parental B6 spleen cells. As this virus activates TLR7, we tested a pharmacological TLR7 ligand, R848, in this model and observed complete survival if donor and recipients were treated before transplantation. Mixed lymphocyte culture performed 48 h after R848-treatment of normal mice demonstrated that both T-cell allo-responsiveness and antigen presentation by CD11b+ and CD8α+ dendritic cells were inhibited. These inhibitions were dependent on IFNAR-1 signaling. In the B6 to B6D2F1 transplantation model, R848 decelerated, but did not abrogate, donor T-cell implantation and activation. However, it decreased interferon-gamma, tumor necrosis factor-alpha and interleukin-27 while upregulating active transforming growth factor-beta 1 plasma levels. In addition, donor and recipient Foxp3+ regulatory T-cell numbers were increased in recipient mice and their elimination compromised disease prevention. R848 also strongly improved survival of lethally irradiated BALB/c recipients of B6 hematopoietic cells and this also correlated with an upregulation of CD4 and CD8 Foxp3+ regulatory T cells that could be further increased by inhibition of interleukin-27. The combination of anti-interleukin-27p28 mono -clonal antibody and R848 showed strong synergy in preventing disease in the B6 to B6D2F1 transplantation model when recipients were sublethally irradiated and this also correlated with upregulation of regulatory T cells. We conclude that R848 modulates multiple aspects of graft-versus-host disease and offers potential for safe allogeneic bone marrow transplantation that can be further optimized by inhibition of interleukin-27.


Subject(s)
Antibodies, Monoclonal/pharmacology , Graft vs Host Disease/prevention & control , Imidazoles/pharmacology , Interleukin-27/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 7/metabolism , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Immunomodulation/drug effects , Ligands , Melanoma, Experimental , Mice , Neoplasm Transplantation , T-Lymphocytes, Regulatory/immunology
16.
Eur J Immunol ; 48(11): 1883-1891, 2018 11.
Article in English | MEDLINE | ID: mdl-30216414

ABSTRACT

The pathogenic role of IL-17 and GM-CSF has been unravelled in experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). However, in most models, EAE is characterised by a monophasic attack which is not representative of the relapsing nature nor the chronicity displayed in MS. Here, we used proteolipid protein peptide (PLP139-151 ) to trigger EAE-relapses (EAE-II) in SJL mice that had recovered from a primary-EAE episode (EAE-I). This procedure resulted in severe and irreversible disease that, unlike EAE-I, was not abolished by anti-IL-17-mAb. In contrast, prophylactic anti-GM-CSF-mAb treatment prevented EAE-I and -II. Strikingly, the expression of T-cell transcription factors and cytokines/chemokines in mice treated with anti-GM-CSF during both EAE episodes was silenced. Anti-GM-CSF-mAb treatment administered only during EAE-II did not completely prevent relapses but mice ultimately reached full recovery. Anti-GM-CSF treatment also strongly impaired and ultimately resolved monophasic MOG35-55 -induced EAE in C57Bl/6 mice. In such protected mice, anti-GM-CSF treatment also prevented a further relapse induced by MOG-revaccination. These results underscore the critical role of GM-CSF on pro-inflammatory mediator production. Furthermore, we observed a strong preventive and curative effect of anti-GM-CSF neutralisation in two EAE models, relapsing and chronic. Altogether these findings are relevant for further MS research.


Subject(s)
Antibodies, Monoclonal/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-17/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/metabolism , Recurrence , Transcription Factors/metabolism
17.
Sci Rep ; 8(1): 8652, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29872095

ABSTRACT

Host directed immunomodulation represents potential new adjuvant therapies in infectious diseases such as tuberculosis. Major cytokines like TNFα exert a multifold role in host control of mycobacterial infections. GM-CSF and its receptor are over-expressed during acute M. tuberculosis infection and we asked how GM-CSF neutralization might affect host response, both in immunocompetent and in immunocompromised TNFα-deficient mice. GM-CSF neutralizing antibodies, at a dose effectively preventing acute lung inflammation, did not affect M. tuberculosis bacterial burden, but increased the number of granuloma in wild-type mice. We next assessed whether GM-CSF neutralization might affect the control of M. tuberculosis by isoniazid/rifampicin chemotherapy. GM-CSF neutralization compromised the bacterial control under sub-optimal isoniazid/rifampicin treatment in TNFα-deficient mice, leading to exacerbated lung inflammation with necrotic granulomatous structures and high numbers of intracellular M. tuberculosis bacilli. In vitro, GM-CSF neutralization promoted M2 anti-inflammatory phenotype in M. bovis BCG infected macrophages, with reduced mycobactericidal NO production and higher intracellular M. bovis BCG burden. Thus, GM-CSF pathway overexpression during acute M. tuberculosis infection contributes to an efficient M1 response, and interfering with GM-CSF pathway in the course of infection may impair the host inflammatory response against M. tuberculosis.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunologic Factors/metabolism , Immunomodulation , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/pathology , Animals , Antibodies, Neutralizing/administration & dosage , Antitubercular Agents/administration & dosage , Cattle , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Immunologic Factors/antagonists & inhibitors , Isoniazid/administration & dosage , Macrophages/immunology , Mice , Mycobacterium bovis/immunology , Rifampin/administration & dosage , Treatment Outcome , Tuberculosis, Pulmonary/immunology
18.
Eur Cytokine Netw ; 28(3): 111-119, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-29187338

ABSTRACT

Mice infected with mouse hepatitis virus A59 (MHV-A59) develop hepatitis and autoantibodies (autoAb) to liver and kidney fumarylacetoacetate hydrolase (FAH), a fact closely related to the release of alarmins such as uric acid and/or high-mobility group box protein 1 (HMGB1). We studied the effect of neutralizing monoclonal antibodies (MAb) against IL-17A in our model of mouse MHV-A59-infection. MAb anti-IL-17F and anti-IFNγ were used to complement the study. Results showed that transaminase levels markedly decreased in MHV-A59-infected mice treated with MAb anti-IL-17A whereas plasmatic Ig concentration sharply increased. Conversely, MAb anti-IL-17F enhanced transaminase liberation and did not affect Ig levels. Serum IFNγ was detected in mice infected with MHV-A59 and its concentration increased after MAb anti-IL-17A administration. Besides, MAb anti-IFNγ greatly augmented transaminase plasmatic levels. IL-17A neutralization did not affect MHV-A59-induction of HMGB1 liberation and slightly augmented plasmatic uric acid concentration. However, mice treated with the MAb failed to produce autoAb to FAH. The above results suggest a reciprocal regulation of Th1 and Th17 cells acting on the different MHV-A59 effects. In addition, it is proposed that IL-17A is involved in alarmins adjuvant effects leading to autoAb expression.


Subject(s)
Interleukin-17/metabolism , Murine hepatitis virus/pathogenicity , Animals , Autoantibodies/immunology , Cell Differentiation/physiology , Female , Hydrolases/metabolism , Kidney/metabolism , Liver/metabolism , Mice , Murine hepatitis virus/immunology , Murine hepatitis virus/metabolism
19.
Clin Sci (Lond) ; 131(20): 2533-2548, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-29026003

ABSTRACT

T helper (Th)17 immune response participates in allergic lung inflammation and asthma is reduced in the absence of interleukin (IL)-17 in mice. Since IL-17A and IL-17F are induced and bind the shared receptor IL-17RA, we asked whether both IL-17A and IL-17F contribute to house dust mite (HDM) induced asthma. We report that allergic lung inflammation is attenuated in absence of either IL-17A or IL-17F with reduced airway hyperreactivity, eosinophilic inflammation, goblet cell hyperplasia, cytokine and chemokine production as found in absence of IL-17RA. Furthermore, specific antibody neutralization of either IL-17A or IL-17F given during the sensitization phase attenuated allergic lung inflammation and airway hyperreactivity. In vitro activation by HDM of primary dendritic cells revealed a comparable induction of CXCL1 and IL-6 expression and the response to IL-17A and IL-17F relied on IL-17RA signaling via the adaptor protein act1 in fibroblasts. Therefore, HDM-induced allergic respiratory response depends on IL-17RA via act1 signaling and inactivation of either IL-17A or IL-17F is sufficient to attenuate allergic asthma in mice.


Subject(s)
Asthma/drug therapy , Interleukin-17/antagonists & inhibitors , Pyroglyphidae/immunology , Allergens/immunology , Animals , Asthma/immunology , Dendritic Cells/immunology , Disease Models, Animal , Interleukin-17/immunology , Interleukin-6/immunology , Lung/immunology , Mice, Inbred C57BL , Th17 Cells/immunology , Th2 Cells/immunology
20.
Immun Inflamm Dis ; 5(2): 200-213, 2017 06.
Article in English | MEDLINE | ID: mdl-28474504

ABSTRACT

INTRODUCTION: Viruses have developed multiple mechanisms to alter immune reactions. In 1969, it was reported that lactate dehydrogenase-elevating virus (LDV), a single stranded positive sense mouse nidovirus, delays skin allograft rejection and inhibits spleen alterations in graft versus host disease (GVHD). As the underlying mechanisms have remained unresolved and given the need for new therapies of this disease, we reassessed the effects of the virus on GVHD and tried to uncover its mode of action. METHODS: GVHD was induced by transfer of parent (B6) spleen cells to non-infected or LDV-infected B6D2F1 recipients. In vitro mixed-lymhocyte culture (MLC) reactions were used to test the effects of the virus on antigen-presenting cells (APC) and responder T cells. RESULTS: LDV infection resulted in a threefold increase in survival rate with reduced weight loss and liver inflammation but with the establishment of permanent chimerism that correlated with decreased interleukine (IL)-27 and interferon (IFN)γ plasma levels. Infected mice showed a transient elimination of splenic CD11b+ and CD8α+ conventional dendritic cells (cDCs) required for allogeneic CD4 and CD8 T cell responses in vitro. This drop of APC numbers was not observed with APCs derived from toll-like receptor (TLR)7-deficient mice. A second effect of the virus was a decreased T cell proliferation and IFNγ production during MLC without detectable changes in Foxp3+ regulatory T cell (Tregs) numbers. Both cDC and responder T cell inhibition were type I IFN dependent. Although the suppressive effects were very transient, the GVHD inhibition was long-lasting. CONCLUSION: A type I IFN-dependent suppression of DC and T cells just after donor spleen cell transplantation induces permanent chimerism and donor cell implantation in a parent to F1 spleen cell transplantation model. If this procedure can be extended to full allogeneic bone marrow transplantation, it could open new therapeutic perspectives for hematopoietic stem cell transplantation (HSCT).


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Interferon Type I/immunology , Nidovirales Infections/immunology , Nidovirales/immunology , Allografts , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Interferon Type I/genetics , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...