Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
JHEP Rep ; 4(7): 100508, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35712694

ABSTRACT

Background & Aims: High-dose irradiation is an essential tool to help control the growth of hepatic tumors, but it can cause radiation-induced liver disease (RILD). This life-threatening complication manifests itself months following radiation therapy and is characterized by fibrosis of the pericentral sinusoids. In this study, we aimed to establish a mouse model of RILD to investigate the underlying mechanism of radiation-induced liver fibrosis. Methods: Using a small animal image-guided radiation therapy platform, an irradiation scheme delivering 50 Gy as a single dose to a focal point in mouse livers was designed. Tissues were analyzed 1 and 6 days, and 6 and 20 weeks post-irradiation. Irradiated livers were assessed by histology, immunohistochemistry, imaging mass cytometry and RNA sequencing. Mitochondrial function was assessed using high-resolution respirometry. Results: At 6 and 20 weeks post-irradiation, pericentral fibrosis was visible in highly irradiated areas together with immune cell infiltration and extravasation of red blood cells. RNA sequencing analysis showed gene signatures associated with acute DNA damage, p53 activation, senescence and its associated secretory phenotype and fibrosis. Moreover, gene profiles of mitochondrial damage and an increase in mitochondrial DNA heteroplasmy were detected. Respirometry measurements of hepatocytes in vitro confirmed irradiation-induced mitochondrial dysfunction. Finally, the highly irradiated fibrotic areas showed markers of reactive oxygen species such as decreased glutathione and increased lipid peroxides and a senescence-like phenotype. Conclusions: Based on our mouse model of RILD, we propose that irradiation-induced mitochondrial DNA instability contributes to the development of fibrosis via the generation of excessive reactive oxygen species, p53 pathway activation and a senescence-like phenotype. Lay summary: Irradiation is an efficient cancer therapy, however, its applicability to the liver is limited by life-threatening radiation-induced hepatic fibrosis. We have developed a new mouse model of radiation-induced liver fibrosis, that recapitulates the human disease. Our model highlights the role of mitochondrial DNA instability in the development of irradiation-induced liver fibrosis. This new model and subsequent findings will help increase our understanding of the hepatic reaction to irradiation and to find strategies that protect the liver, enabling the expanded use of radiotherapy to treat hepatic tumors.

2.
Mol Cancer Ther ; 19(2): 614-626, 2020 02.
Article in English | MEDLINE | ID: mdl-31744898

ABSTRACT

Radiotherapy (RT) along with surgery is the mainstay of treatment in head and neck squamous cell carcinoma (HNSCC). Radioresistance represents a major source of treatment failure, underlining the urgent necessity to explore and implement effective radiosensitization strategies. The MET receptor widely participates in the acquisition and maintenance of an aggressive phenotype in HNSCC and modulates the DNA damage response following ionizing radiation (IR). Here, we assessed MET expression and mutation status in primary and metastatic lesions within a cohort of patients with advanced HNSCC. Moreover, we investigated the radiosensitization potential of the MET inhibitor tepotinib in a panel of cell lines, in vitro and in vivo, as well as in ex vivo patient-derived organotypic tissue cultures (OTC). MET was highly expressed in 62.4% of primary tumors and in 53.6% of lymph node metastases (LNM), and in 6 of 9 evaluated cell lines. MET expression in primaries and LNMs was significantly associated with decreased disease control in univariate survival analyses. Tepotinib abrogated MET phosphorylation and to distinct extent MET downstream signaling. Pretreatment with tepotinib resulted in variable radiosensitization, enhanced DNA damage, cell death, and G2-M-phase arrest. Combination of tepotinib with IR led to significant radiosensitization in one of two tested in vivo models. OTCs revealed differential patterns of response toward tepotinib, irradiation, and combination of both modalities. The molecular basis of tepotinib-mediated radiosensitization was studied by a CyTOF-based single-cell mass cytometry approach, which uncovered that MET inhibition modulated PI3K activity in cells radiosensitized by tepotinib but not in the resistant ones.


Subject(s)
Protein Kinase Inhibitors/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Disease Models, Animal , Humans , Mice , Xenograft Model Antitumor Assays
4.
Sci Rep ; 7(1): 11657, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912492

ABSTRACT

The effects of epicutaneous allergen administration on systemic immune responses in allergic and non-allergic individuals has not been investigated with defined allergen molecules. We studied the effects of epicutaneous administration of rBet v 1 and rBet v 1 fragments on systemic immune responses in allergic and non-allergic subjects. We conducted a clinical trial in which rBet v 1 and two hypoallergenic rBet v 1 fragments were applied epicutaneously by atopy patch testing (APT) to 15 birch pollen (bp) allergic patients suffering from atopic dermatitis, 5 bp-allergic patients suffering from rhinoconjunctivitis only, 5 patients with respiratory allergy without bp allergy and 5 non-allergic individuals. Epicutaneous administration of rBet v 1 and rBet v 1 fragments led to strong and significant increases of allergen-specific T cell proliferation (CLA+ and CCR4+T cell responses) only in bp-allergic patients with a positive APT reaction. There were no relevant changes of Bet v 1-specific IgE and IgG responses. No changes were noted in allergic subjects without bp allergy and in non-allergic subjects. Epicutaneous allergen application boosts specific T cell but not antibody responses mainly in allergic, APT-positive patients suggesting IgE-facilitated allergen presentation as mechanism for its effects on systemic allergen-specific immune responses.


Subject(s)
Allergens/immunology , Immunity, Cellular , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Administration, Cutaneous , Allergens/administration & dosage , Cytokines/metabolism , Desensitization, Immunologic , Humans , Hypersensitivity/immunology , Hypersensitivity/therapy , Immunization , Immunization, Secondary , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism
5.
Proc Natl Acad Sci U S A ; 114(14): E2891-E2900, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28320933

ABSTRACT

Type 1 regulatory T (Tr1) cells play a pivotal role in restraining human T-cell responses toward environmental allergens and protecting against allergic diseases. Still, the precise molecular cues that underlie their transcriptional and functional specification remain elusive. Here, we show that the cytokine activin-A instructs the generation of CD4+ T cells that express the Tr1-cell-associated molecules IL-10, inducible T-Cell costimulator (ICOS), lymphocyte activation gene 3 protein (LAG-3), and CD49b, and exert strongly suppressive functions toward allergic responses induced by naive and in vivo-primed human T helper 2 cells. Moreover, mechanistic studies reveal that activin-A signaling induces the activation of the transcription factor interferon regulatory factor (IRF4), which, along with the environmental sensor aryl hydrocarbon receptor, forms a multipartite transcriptional complex that binds in IL-10 and ICOS promoter elements and controls gene expression in human CD4+ T cells. In fact, IRF4 silencing abrogates activin-A-driven IL10 and ICOS up-regulation and impairs the suppressive functions of human activin-A-induced Tr1-like (act-A-iTr1) cells. Importantly, using a humanized mouse model of allergic asthma, we demonstrate that adoptive transfer of human act-A-iTr1 cells, both in preventive and therapeutic protocols, confers significant protection against cardinal asthma manifestations, including pulmonary inflammation. Overall, our findings uncover an activin-A-induced IRF4-aryl hydrocarbon receptor (AhR)-dependent transcriptional network, which generates suppressive human Tr1 cells that may be harnessed for the control of allergic diseases.


Subject(s)
Activins/metabolism , Asthma/prevention & control , Basic Helix-Loop-Helix Transcription Factors/metabolism , Interferon Regulatory Factors/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Regulatory/immunology , Activins/pharmacology , Animals , Asthma/immunology , Asthma/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Disease Models, Animal , Gene Expression Regulation , Humans , Hypersensitivity/metabolism , Hypersensitivity/prevention & control , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice, SCID , Promoter Regions, Genetic , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
6.
J Allergy Clin Immunol ; 137(2): 601-609.e8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26518092

ABSTRACT

BACKGROUND: Late allergic reactions are common in the course of allergen-specific immunotherapy and even occur with allergy vaccines with reduced IgE reactivity. OBJECTIVE: We sought to study atopy patch test (APT) reactions and T-cell responses to the recombinant birch pollen allergen Bet v 1 and recombinant hypoallergenic T-cell epitope-containing Bet v 1 fragments in patients with birch pollen allergy with and without atopic dermatitis (AD). METHODS: A clinical study was conducted in 15 patients with birch pollen allergy with AD (group 1), 5 patients with birch pollen allergy without AD (group 2), 5 allergic patients without birch pollen allergy (group 3), and 5 nonallergic subjects (group 4) by performing skin prick tests and APTs with rBet v 1 and hypoallergenic rBet v 1 fragments. T-cell, cutaneous lymphocyte antigen (CLA)(+) and CCR4(+) T-cell and cytokine responses were studied by thymidine uptake, carboxyfluorescein diacetate succinimidyl ester staining, and Luminex technology, respectively. RESULTS: rBet v 1 and hypoallergenic rBet v 1 fragments induced APT reactions in not only most of the patients with birch pollen allergy with AD (11/15) but also in most of those without AD (4/5). Patients with birch pollen allergy with AD had higher Bet v 1-specific proliferation of CLA(+) and CCR4(+) T cells compared with patients with birch pollen allergy without AD. There were no differences in Bet v 1-specific CLA(+) and CCR4(+) proliferation and cytokine secretion in patients with and without APT reactions. CONCLUSION: Hypoallergenic rBet v 1 fragments induce T cell-dependent late reactions not only in patients with birch pollen allergy with AD but also in those without AD, which can be determined based on APT results but not based on in vitro parameters.


Subject(s)
Allergens/immunology , Antigens, Plant/immunology , Hypersensitivity, Delayed/diagnosis , Hypersensitivity, Delayed/immunology , Patch Tests , T-Lymphocytes/immunology , Adult , Betula/adverse effects , Cytokines/biosynthesis , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Female , Histamine Release , Humans , Hypersensitivity, Delayed/metabolism , Hypersensitivity, Immediate/diagnosis , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/metabolism , Lymphocyte Activation/immunology , Male , Pollen/immunology , Rhinitis, Allergic, Seasonal/diagnosis , Rhinitis, Allergic, Seasonal/immunology , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes/metabolism , Young Adult
7.
J Immunol ; 193(5): 2196-206, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25063866

ABSTRACT

We identified two mast cell subsets characterized by the differential expression of surface CD25 (IL-2Rα) and by different abilities to produce cytokines and to proliferate, both in vitro and in vivo. CD25 can be expressed on the surface of immune cells in the absence of the other chains of the IL-2R, which are indispensable for IL-2 signaling. We show that functional differences between the two mast cell populations were dependent on CD25 itself, which directly modulated proliferation and cytokine responses. These effects were completely independent from IL-2 or the expression of the other chains of the high-affinity IL-2R, indicating an autonomous and previously unappreciated role for CD25 in regulating cell functions. Cells genetically ablated for CD25 completely recapitulated the CD25-negative phenotype and never acquired the properties characteristic of CD25-positive mast cells. Finally, adoptive transfer experiments in the mouse demonstrated a different impact of these populations in models of anaphylaxis and contact sensitivity. Our findings indicate a general role for CD25 in contexts where IL-2 signaling is not involved, and may have important implications for all mast cell-related diseases, as well as in all cell types expressing CD25 independently of its IL-2-related functions.


Subject(s)
Gene Expression Regulation/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2/immunology , Mast Cells/immunology , Signal Transduction/immunology , 3T3 Cells , Animals , Gene Expression Regulation/genetics , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Mast Cells/cytology , Mice , Mice, Knockout , Signal Transduction/genetics
8.
J Exp Med ; 211(7): 1273-80, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24958850

ABSTRACT

The preexisting HIV-1-specific T cell repertoire must influence both the immunodominance of T cells after infection and immunogenicity of vaccines. We directly compared two methods for measuring the preexisting CD4(+) T cell repertoire in healthy HIV-1-negative volunteers, the HLA-peptide tetramer enrichment and T cell library technique, and show high concordance (r = 0.989). Using the library technique, we examined whether naive, central memory, and/or effector memory CD4(+) T cells specific for overlapping peptides spanning the entire HIV-1 proteome were detectable in 10 HLA diverse, HIV-1-unexposed, seronegative donors. HIV-1-specific cells were detected in all donors at a mean of 55 cells/million naive cells and 38.9 and 34.1 cells/million in central and effector memory subsets. Remarkably, peptide mapping showed most epitopes recognized by naive (88%) and memory (56%) CD4(+) T cells had been previously reported in natural HIV-1 infection. Furthermore, 83% of epitopes identified in preexisting memory subsets shared epitope length matches (8-12 amino acids) with human microbiome proteins, suggestive of a possible cross-reactive mechanism. These results underline the power of a proteome-wide analysis of peptide recognition by human T cells for the identification of dominant antigens and provide a baseline for optimizing HIV-1-specific helper cell responses by vaccination.


Subject(s)
Antigens, Viral/immunology , Epitopes, T-Lymphocyte/immunology , HIV-1/immunology , Immunologic Memory , Microbiota/immunology , Peptides/immunology , Proteome/immunology , Viral Proteins/immunology , AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes , Cross Reactions , Female , Humans , Male
9.
Eur J Immunol ; 43(11): 2797-809, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24258910

ABSTRACT

Following antigen encounter and subsequent resolution of the immune response, a single naïve T cell is able to generate multiple subsets of memory T cells with different phenotypic and functional properties and gene expression profiles. Single-cell technologies, first and foremost flow cytometry, have revealed the complex heterogeneity of the memory T-cell compartment and its organization into subsets. However, a consensus has still to be reached, both at the semantic (nomenclature) and phenotypic level, regarding the identification of these subsets. Here, we review recent developments in the characterization of the heterogeneity of the memory T-cell compartment, and propose a unified classification of both human and nonhuman primate T cells on the basis of phenotypic traits and in vivo properties. Given that vaccine studies and adoptive cell transfer immunotherapy protocols are influenced by these recent findings, it is important to use uniform methods for identifying and discussing functionally distinct subsets of T cells.


Subject(s)
Cell Differentiation/immunology , Immunologic Memory/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Gene Expression , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
10.
J Virol ; 87(3): 1779-88, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23175378

ABSTRACT

Coinfection with Plasmodium falciparum malaria and Epstein-Barr virus (EBV) is a major risk factor for endemic Burkitt lymphoma (eBL), still one of the most prevalent pediatric cancers in equatorial Africa. Although malaria infection has been associated with immunosuppression, the precise mechanisms that contribute to EBV-associated lymphomagenesis remain unclear. In this study, we used polychromatic flow cytometry to characterize CD8(+) T-cell subsets specific for EBV-derived lytic (BMFL1 and BRLF1) and latent (LMP1, LMP2, and EBNA3C) antigens in individuals with divergent malaria exposure. No malaria-associated differences in EBV-specific CD8(+) T-cell frequencies were observed. However, based on a multidimensional analysis of CD45RO, CD27, CCR7, CD127, CD57, and PD-1 expression, we found that individuals living in regions with intense and perennial (holoendemic) malaria transmission harbored more differentiated EBV-specific CD8(+) T-cell populations that contained fewer central memory cells than individuals living in regions with little or no (hypoendemic) malaria. This profile shift was most marked for EBV-specific CD8(+) T-cell populations that targeted latent antigens. Importantly, malaria exposure did not skew the phenotypic properties of either cytomegalovirus (CMV)-specific CD8(+) T cells or the global CD8(+) memory T-cell pool. These observations define a malaria-associated aberration localized to the EBV-specific CD8(+) T-cell compartment that illuminates the etiology of eBL.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Coinfection/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/pathogenicity , Malaria, Falciparum/epidemiology , Malaria, Falciparum/immunology , Plasmodium falciparum/pathogenicity , Africa/epidemiology , Child , Child, Preschool , Epstein-Barr Virus Infections/complications , Flow Cytometry , Humans , Infant , Malaria, Falciparum/complications , T-Lymphocyte Subsets/immunology
11.
J Immunol ; 189(4): 1800-11, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22786768

ABSTRACT

A panel of 133 allergens derived from 28 different sources, including fungi, trees, grasses, weeds, and indoor allergens, was surveyed utilizing prediction of HLA class II-binding peptides and ELISPOT assays with PBMC from allergic donors, resulting in the identification of 257 T cell epitopes. More than 90% of the epitopes were novel, and for 14 allergen sources were the first ever identified to our knowledge. The epitopes identified in the different allergen sources summed up to a variable fraction of the total extract response. In cases of allergens in which the identified T cell epitopes accounted for a minor fraction of the extract response, fewer known protein sequences were available, suggesting that for low epitope coverage allergen sources, additional allergen proteins remain to be identified. IL-5 and IFN-γ responses were measured as prototype Th2 and Th1 responses, respectively. Whereas in some cases (e.g., orchard grass, Alternaria, cypress, and Russian thistle) IL-5 production greatly exceeded IFN-γ, in others (e.g., Aspergillus, Penicillum, and alder) the production of IFN-γ exceeded IL-5. Thus, different allergen sources are associated with variable polarization of the responding T cells. The present study represents the most comprehensive survey to date of human allergen-derived T cell epitopes. These epitopes might be used to characterize T cell phenotype/T cell plasticity as a function of seasonality, or as a result of specific immunotherapy treatment or varying disease severity (asthma or rhinitis).


Subject(s)
Allergens/immunology , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes/immunology , Cytokines/biosynthesis , Humans , Hypersensitivity/immunology
12.
J Infect Dis ; 201(2): 272-84, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20001854

ABSTRACT

BACKGROUND: Variability in human immunodeficiency virus (HIV) disease progression cannot be fully predicted by CD4(+) T cell counts or viral load (VL). Because central memory T (T(CM)) cells play a critical role in the pathogenesis of simian immunodeficiency virus disease, we hypothesized that quantifying these cells in early HIV infection could provide prognostic information. METHODS: We measured expression of CD45RO, chemokine (C-C motif) receptor (CCR) 5, CCR7, CD27, and CD28 to enumerate naive and memory subsets in samples from recently infected individuals. We also quantified proliferation, CD127 expression, and cell-associated VL. Disease progression was compared across subgroups defined by these measurements, using Kaplan-Meier survival curves and multivariate Cox proportional hazards regression. RESULTS: Four hundred sixty-six subjects contributed 101 events. The proportion or absolute count of T(CM) cells did not correlate with disease progression, defined as the time to AIDS or death. However, significant associations were observed for proliferation within CD4(+) or CD8(+) T cells, loss of naive or CD127(+) memory CD8(+) T cells, and CD4(+) T cell-associated VL. CONCLUSIONS: Our results demonstrate that the extent of the immunopathogenesis established early in HIV infection predicts the course of future disease. Because antiretroviral drug treatment reverses such defects in part, our study provides mechanistic clues to why early use of antiretrovirals may prove beneficial.


Subject(s)
HIV Infections/immunology , Immunologic Memory , T-Lymphocytes/virology , Acquired Immunodeficiency Syndrome/immunology , Adolescent , Adult , Anti-Retroviral Agents/administration & dosage , CD28 Antigens/immunology , Disease Progression , Drug Administration Schedule , Female , HIV Infections/drug therapy , Humans , Kaplan-Meier Estimate , Leukocyte Common Antigens/immunology , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Receptors, CCR5/immunology , Receptors, CCR7/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Young Adult
13.
Infect Immun ; 75(5): 2359-65, 2007 May.
Article in English | MEDLINE | ID: mdl-17339357

ABSTRACT

Infection with Leishmania major is enhanced when the sand fly Lutzomyia longipalpis salivary peptide maxadilan (MAX) is injected along with the parasite. Here we determined the effect that MAX has on the secretion of cytokines and nitric oxide (NO) and on parasite survival in macrophages (MPhis). The cytokines produced by MPhis can enhance a type 1 response, which will increase NO and the killing of intracellular pathogens such as L. major, or a type 2 response, leading to antibody production that is ineffective against intracellular pathogens such as L. major. A mouse macrophage cell line (RAW 264.7) was stimulated with various concentrations of MAX and lipopolysaccharide (LPS), and the supernatants were collected after 1, 2, and 3 days. Supernatants were assayed for interleukin-12p70 (IL-12p70), IL-10, IL-6, transforming growth factor beta (TGF-beta), NO, and tumor necrosis factor alpha (TNF-alpha). Our results indicate that the addition of MAX upregulates the cytokines associated with a type 2 response (IL-10, IL-6, and TGF-beta) but downregulates type 1 cytokines (IL-12p70 and TNF-alpha) and NO. MAX was also added to L. major-infected mouse peritoneal exudate cells (PECs), and the parasite load increased significantly. The enhanced parasite load correlated with decreased NO production by PECs that were stimulated with LPS and gamma interferon in the presence of MAX. The ability of MAX to foster a type 2 response, to enhance parasite survival, and to decrease NO argues that MAX may be crucial for the early survival of Leishmania in the vertebrate host, and therefore, MAX holds considerable promise as an antigenic component for a vaccine against Leishmania.


Subject(s)
Cytokines/biosynthesis , Insect Proteins/immunology , Leishmania major/growth & development , Macrophages/immunology , Psychodidae/metabolism , Th2 Cells/immunology , Animals , Cell Line , Cells, Cultured , Female , Insect Proteins/metabolism , Insect Proteins/pharmacology , Leishmania major/pathogenicity , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/parasitology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred C3H , Nitric Oxide/biosynthesis , Th2 Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...