Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nat Immunol ; 19(9): 954-962, 2018 09.
Article in English | MEDLINE | ID: mdl-30127438

ABSTRACT

Controlling the balance between immunity and immunopathology is crucial for host resistance to pathogens. After infection, activation of the hypothalamic-pituitary-adrenal (HPA) axis leads to the production of glucocorticoids. However, the pleiotropic effects of these steroid hormones make it difficult to delineate their precise role(s) in vivo. Here we found that the regulation of natural killer (NK) cell function by the glucocorticoid receptor (GR) was required for host survival after infection with mouse cytomegalovirus (MCMV). Mechanistically, endogenous glucocorticoids produced shortly after infection induced selective and tissue-specific expression of the checkpoint receptor PD-1 on NK cells. This glucocorticoid-PD-1 pathway limited production of the cytokine IFN-γ by spleen NK cells, which prevented immunopathology. Notably, this regulation did not compromise viral clearance. Thus, the fine tuning of NK cell functions by the HPA axis preserved tissue integrity without impairing pathogen elimination, which reveals a novel aspect of neuroimmune regulation.


Subject(s)
Glucocorticoids/metabolism , Herpesviridae Infections/immunology , Killer Cells, Natural/physiology , Muromegalovirus/physiology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cells, Cultured , Female , Hypothalamo-Hypophyseal System , Immunity, Innate , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , Organ Specificity , Pituitary-Adrenal System , Receptors, Glucocorticoid/genetics , Signal Transduction , Viral Load
2.
Nature ; 594(7861): 94-99, 2021 06.
Article in English | MEDLINE | ID: mdl-34012116

ABSTRACT

Inflammation is a defence response to tissue damage that requires tight regulation in order to prevent impaired healing. Tissue-resident macrophages have a key role in tissue repair1, but the precise molecular mechanisms that regulate the balance between inflammatory and pro-repair macrophage responses during healing remain poorly understood. Here we demonstrate a major role for sensory neurons in promoting the tissue-repair function of macrophages. In a sunburn-like model of skin damage in mice, the conditional ablation of sensory neurons expressing the Gαi-interacting protein (GINIP) results in defective tissue regeneration and in dermal fibrosis. Elucidation of the underlying molecular mechanisms revealed a crucial role for the neuropeptide TAFA4, which is produced in the skin by C-low threshold mechanoreceptors-a subset of GINIP+ neurons. TAFA4 modulates the inflammatory profile of macrophages directly in vitro. In vivo studies in Tafa4-deficient mice revealed that TAFA4 promotes the production of IL-10 by dermal macrophages after UV-induced skin damage. This TAFA4-IL-10 axis also ensures the survival and maintenance of IL-10+TIM4+ dermal macrophages, reducing skin inflammation and promoting tissue regeneration. These results reveal a neuroimmune regulatory pathway driven by the neuropeptide TAFA4 that promotes the anti-inflammatory functions of macrophages and prevents fibrosis after tissue damage, and could lead to new therapeutic perspectives for inflammatory diseases.


Subject(s)
Cytokines/metabolism , Macrophages/metabolism , Regeneration , Sensory Receptor Cells/metabolism , Wound Healing , Animals , Cell Survival , Cytokines/deficiency , Disease Models, Animal , Female , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Macrophages/radiation effects , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/radiation effects , Skin/pathology , Skin/radiation effects , Sunburn/complications , Sunburn/etiology , Sunburn/metabolism , Sunburn/pathology , Ultraviolet Rays/adverse effects
3.
Gut ; 72(6): 1115-1128, 2023 06.
Article in English | MEDLINE | ID: mdl-36175116

ABSTRACT

OBJECTIVE: In the management of patients with IBD, there is a need to identify prognostic markers and druggable biological pathways to improve mucosal repair and probe the efficacy of tumour necrosis factor alpha biologics. Vnn1 is a pantetheinase that degrades pantetheine to pantothenate (vitamin B5, a precursor of coenzyme A (CoA) biosynthesis) and cysteamine. Vnn1 is overexpressed by inflamed colonocytes. We investigated its contribution to the tolerance of the intestinal mucosa to colitis-induced injury. DESIGN: We performed an RNA sequencing study on colon biopsy samples from patients with IBD stratified according to clinical severity and modalities of treatment. We generated the VIVA mouse transgenic model, which specifically overexpresses Vnn1 on intestinal epithelial cells and explored its susceptibility to colitis. We developed a pharmacological mimicry of Vnn1 overexpression by administration of Vnn1 derivatives. RESULTS: VNN1 overexpression on colonocytes correlates with IBD severity. VIVA mice are resistant to experimentally induced colitis. The pantetheinase activity of Vnn1 is cytoprotective in colon: it enhances CoA regeneration and metabolic adaptation of colonocytes; it favours microbiota-dependent production of short chain fatty acids and mostly butyrate, shown to regulate mucosal energetics and to be reduced in patients with IBD. This prohealing phenotype is recapitulated by treating control mice with the substrate (pantethine) or the products of pantetheinase activity prior to induction of colitis. In severe IBD, the protection conferred by the high induction of VNN1 might be compromised because its enzymatic activity may be limited by lack of available substrates. In addition, we identify the elevation of indoxyl sulfate in urine as a biomarker of Vnn1 overexpression, also detected in patients with IBD. CONCLUSION: The induction of Vnn1/VNN1 during colitis in mouse and human is a compensatory mechanism to reinforce the mucosal barrier. Therefore, enhancement of vitamin B5-driven metabolism should improve mucosal healing and might increase the efficacy of anti-inflammatory therapy.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Mice , Animals , Colitis/metabolism , Colon/pathology , Intestinal Mucosa/metabolism , Inflammatory Bowel Diseases/genetics , Fatty Acids, Volatile/metabolism , Vitamins , Dextran Sulfate , Disease Models, Animal
4.
Cell Mol Life Sci ; 79(7): 355, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35678896

ABSTRACT

Thymically-derived Foxp3+ regulatory T cells (Treg) critically control immunological tolerance. These cells are generated in the medulla through high affinity interactions with medullary thymic epithelial cells (mTEC) expressing the Autoimmune regulator (Aire). Recent advances have revealed that thymic Treg contain not only developing but also recirculating cells from the periphery. Although Aire is implicated in the generation of Foxp3+ Treg, its role in the biology of recirculating Treg remains elusive. Here, we show that Aire regulates the suppressive signature of recirculating Treg independently of the remodeling of the medullary 3D organization throughout life where Treg reside. Accordingly, the adoptive transfer of peripheral Foxp3+ Treg in AireKO recipients led to an impaired suppressive signature upon their entry into the thymus. Furthermore, recirculating Treg from AireKO mice failed to attenuate the severity of multiorgan autoimmunity, demonstrating that their suppressive function is altered. Using bone marrow chimeras, we reveal that mTEC-specific expression of Aire controls the suppressive signature of recirculating Treg. Finally, mature mTEC lacking Aire were inefficient in stimulating peripheral Treg both in polyclonal and antigen-specific co-culture assays. Overall, this study demonstrates that Aire confers to mTEC the ability to restimulate recirculating Treg, unravelling a novel function for this master regulator in Treg biology.


Subject(s)
Immune Tolerance , T-Lymphocytes, Regulatory , Animals , Autoimmunity , Epithelial Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Thymus Gland
5.
Nat Immunol ; 10(1): 75-82, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029904

ABSTRACT

NKp46+CD3- natural killer lymphocytes isolated from blood, lymphoid organs, lung, liver and uterus can produce granule-dependent cytotoxicity and interferon-gamma. Here we identify in dermis, gut lamina propria and cryptopatches distinct populations of NKp46+CD3- cells with a diminished capacity to degranulate and produce interferon-gamma. In the gut, expression of the transcription factor RORgammat, which is involved in the development of lymphoid tissue-inducer cells, defined a previously unknown subset of NKp46+CD3- lymphocytes. Unlike RORgammat- lamina propria and dermis natural killer cells, gut RORgammat+NKp46+ cells produced interleukin 22. Our data show that lymphoid tissue-inducer cells and natural killer cells shared unanticipated similarities and emphasize the heterogeneity of NKp46+CD3- cells in innate immunity, lymphoid organization and local tissue repair.


Subject(s)
Dermis/immunology , Intestinal Mucosa/immunology , Natural Killer T-Cells/immunology , Receptors, Retinoic Acid/physiology , Receptors, Thyroid Hormone/physiology , Transcription Factors/physiology , Animals , CD3 Complex/metabolism , Cell Division , Humans , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Natural Cytotoxicity Triggering Receptor 1/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3 , Peyer's Patches/immunology , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Transcription Factors/genetics , Interleukin-22
6.
Clin Infect Dis ; 70(11): 2405-2412, 2020 05 23.
Article in English | MEDLINE | ID: mdl-31309973

ABSTRACT

BACKGROUND: Etiological diagnosis is a key to therapeutic adaptation and improved prognosis, particularly for infections such as endocarditis. In blood culture-negative endocarditis (BCNE), 22% of cases remain undiagnosed despite an updated comprehensive syndromic approach. This prompted us to develop a new diagnostic approach. METHODS: Eleven valves from 10 BCNE patients were analyzed using a method that combines human RNA bait-depletion with phi29 DNA polymerase-based multiple displacement amplification and shotgun DNA sequencing. An additional case in which a microbe was serendipitously visualized by immunofluorescence was analyzed using the same method, but after laser capture microdissection. RESULTS: Background DNA prevented any diagnosis in cases analyzed without microdissection because the majority of sequences were contaminants. Moraxella sequences were dramatically enriched in the stained microdissected region of the additional case. A consensus genome sequence of 2.4 Mbp covering more than 94% of the Moraxella osloensis KSH reference genome was reconstructed with 234X average coverage. Several antibiotic-resistance genes were observed. Etiological diagnosis was confirmed using Western blot and specific polymerase chain reaction with sequencing on a different valve sample. CONCLUSIONS: Microdissection could be a key to the metagenomic diagnosis of infectious diseases when a microbe is visualized but remains unidentified despite an updated optimal approach. Moraxella osloensis should be tested in blood culture-negative endocarditis.


Subject(s)
Endocarditis, Bacterial , Endocarditis , Blood Culture , Endocarditis/diagnosis , Endocarditis, Bacterial/diagnosis , Humans , Metagenomics , Moraxella
7.
J Neuroinflammation ; 16(1): 191, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31660979

ABSTRACT

BACKGROUND: Targeting angiogenesis has been and continues to be an attractive therapeutic modality in glioblastoma (GBM) patients. However, GBM rapidly becomes refractory to anti-VEGF therapies. Myeloid cell infiltration is an important determinant of tumor progression. Given that VEGF is a modulator of the innate immune response we sought to analyze the dynamics of this response in a mouse model of GBM undergoing anti-VEGF therapy. METHODS: We grafted GL261-DsRed cells in transgenic Thy1-CFP//LysM-EGFP//CD11c-EYFP reporter mice. We combined recurrent spectral two-photon imaging with multiparametric cytometry, immunostaining, and brain clearing to characterize at two critical stages of tumor development (day 21 and day 28 after tumor grafting) the nature and spatial distribution of the innate response in control and bevacizumab-treated mice. RESULTS: We report that at an early stage (21 day), VEGF blockade has a detectable effect on the number of microglial cells but only a mild effect on the number of infiltrating myeloid cells. At a later stage (day 28), the treatment resulted in a specific adjustment of dendritic cell subsets. In treated mice, the number of monocytes and their monocyte-derived dendritic cells (moDC) progeny was increased by approximately twofold compared to untreated mice. In agreement, by in vivo quantitative imaging, we observed that treatment increased the number of LysM-EGFP cells traveling in tumor blood vessels and doubled the densities of both infiltrated LysM-EGFP monocytes and double-labeled EGFP/EYFP moDC. The treatment also led to an increased density of conventional cDCs2 subset together with a decrease of cDCs1 subset, necessary for the development of anti-tumor immunity. Finally, we describe differential spatial cell distributions and two immune cell-traveling routes into the brain. LysM-EGFP cells distributed as a gradient from the meninges towards the tumor whereas CD11c-EYFP/MHCII+ cells were located in the basal area of the tumor. Brain clearing also revealed a flow of CD11c-EYFP cells following the corpus callosum. CONCLUSION: We uncovered new features in the dynamics of innate immune cells in GBM-bearing mice and deciphered precisely the key populations, i.e., DC subsets controlling immune responses, that are affected by VEGF blockade. Since despite differences, human pathogenesis presents similarities with our mouse model, the data provide new insights into the effect of bevacizumab at the cellular level.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Burden/drug effects , Tumor Burden/physiology , Xenograft Model Antitumor Assays/methods
8.
Biochim Biophys Acta ; 1862(4): 662-669, 2016 04.
Article in English | MEDLINE | ID: mdl-26850476

ABSTRACT

BACKGROUND AND AIMS: Pretreatment with clofibrate, a peroxisome proliferator-activated receptor alpha (PPARa) agonist, protects mice from acetaminophen (APAP) injury. Protection is not due to alterations in APAP metabolism and is dependent on PPARa expression. Gene array analysis revealed that mice receiving clofibrate have enhanced hepatic Vanin-1 (Vnn1) gene expression, a response that is also PPARa dependent. METHODS: We examined the role of Vnn1 by comparing the responses of Vnn1 knockout and wild-type mice following APAP hepatotoxicity. APAP metabolism, hepatotoxicity, and compensatory hepatocyte proliferation and immune responses were assessed. RESULTS: Vnn1 knockout mice are more susceptible to APAP hepatotoxicity despite no differences in hepatic glutathione content, gene expression of APAP metabolizing enzymes, or hepatic capacity to bioactivate or detoxify APAP ex vivo. Together, these data strongly suggest that the susceptibility of Vnn1 knockout mice is not due to differences in APAP metabolism. Immunochemistry revealed a lack of proliferating cell nuclear antigen-positive hepatocytes and F4/80-positive macrophages in and around areas of centrilobular necrosis in APAP-treated Vnn1 knockouts. Hepatic gene induction of pro-inflammatory cytokines was either significantly reduced or completely blunted in these mice. This was correlated with a reduction in early recruitment of cells positive for granulocyte differentiation antigen 1 or integrin alpha M. Heightened toxicity was also observed in CCl4 and ConA hepatitis models in the absence of Vnn1. CONCLUSIONS: These results indicate that mice lacking Vnn1 have deficiencies in compensatory repair and immune responses following toxic APAP exposure and that these mechanisms may contribute to the enhanced hepatotoxicity seen.


Subject(s)
Acetaminophen/adverse effects , Amidohydrolases/deficiency , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/immunology , Liver/immunology , Acetaminophen/pharmacology , Amidohydrolases/immunology , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Clofibrate/pharmacology , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hepatocytes/immunology , Hepatocytes/pathology , Liver/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , PPAR alpha/genetics , PPAR alpha/immunology
9.
PLoS Pathog ; 11(5): e1004897, 2015 May.
Article in English | MEDLINE | ID: mdl-25954804

ABSTRACT

In mice, plasmacytoid dendritic cells (pDC) and natural killer (NK) cells both contribute to resistance to systemic infections with herpes viruses including mouse Cytomegalovirus (MCMV). pDCs are the major source of type I IFN (IFN-I) during MCMV infection. This response requires pDC-intrinsic MyD88-dependent signaling by Toll-Like Receptors 7 and 9. Provided that they express appropriate recognition receptors such as Ly49H, NK cells can directly sense and kill MCMV-infected cells. The loss of any one of these responses increases susceptibility to infection. However, the relative importance of these antiviral immune responses and how they are related remain unclear. In humans, while IFN-I responses are essential, MyD88 is dispensable for antiviral immunity. Hence, a higher redundancy has been proposed in the mechanisms promoting protective immune responses against systemic infections by herpes viruses during natural infections in humans. It has been assumed, but not proven, that mice fail to mount protective MyD88-independent IFN-I responses. In humans, the mechanism that compensates MyD88 deficiency has not been elucidated. To address these issues, we compared resistance to MCMV infection and immune responses between mouse strains deficient for MyD88, the IFN-I receptor and/or Ly49H. We show that selective depletion of pDC or genetic deficiencies for MyD88 or TLR9 drastically decreased production of IFN-I, but not the protective antiviral responses. Moreover, MyD88, but not IFN-I receptor, deficiency could largely be compensated by Ly49H-mediated antiviral NK cell responses. Thus, contrary to the current dogma but consistent with the situation in humans, we conclude that, in mice, in our experimental settings, MyD88 is redundant for IFN-I responses and overall defense against a systemic herpes virus infection. Moreover, we identified direct NK cell sensing of infected cells as one mechanism able to compensate for MyD88 deficiency in mice. Similar mechanisms likely contribute to protect MyD88- or IRAK4-deficient patients from viral infections.


Subject(s)
Herpesviridae Infections/immunology , Host-Pathogen Interactions , Interferon Type I/metabolism , Killer Cells, Natural/immunology , Muromegalovirus/immunology , Myeloid Differentiation Factor 88/metabolism , Receptor, Interferon alpha-beta/agonists , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Gene Expression Profiling , Gene Expression Regulation , Herpesviridae Infections/blood , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Immunity, Innate , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/virology , Interferon Type I/blood , Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Mice, Inbred BALB C , Mice, Knockout , Mice, Mutant Strains , Muromegalovirus/physiology , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , NK Cell Lectin-Like Receptor Subfamily A/deficiency , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Primary Immunodeficiency Diseases , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/metabolism , Spleen/virology , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
10.
Proc Natl Acad Sci U S A ; 111(30): 11115-20, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024217

ABSTRACT

The transcription factor NF-κB is central to inflammatory signaling and activation of innate and adaptive immune responses. Activation of the NF-κB pathway is tightly controlled by several negative feedback mechanisms, including A20, an ubiquitin-modifying enzyme encoded by the tnfaip3 gene. Mice with selective deletion of A20 in myeloid, dendritic, or B cells recapitulate some human inflammatory pathology. As we observed high expression of A20 transcripts in dysfunctional CD8 T cells in an autochthonous melanoma, we analyzed the role of A20 in regulation of CD8 T-cell functions, using mice in which A20 was selectively deleted in mature conventional T cells. These mice developed lymphadenopathy and some organ infiltration by T cells but no splenomegaly and no detectable pathology. A20-deleted CD8 T cells had increased sensitivity to antigen stimulation with production of large amounts of IL-2 and IFNγ, correlated with sustained nuclear expression of NF-κB components reticuloendotheliosis oncogene c-Rel and p65. Overexpression of A20 by retroviral transduction of CD8 T cells dampened their intratumor accumulation and antitumor activity. In contrast, relief from the A20 brake in NF-κB activation in adoptively transferred antitumor CD8 T cells led to improved control of melanoma growth. Tumor-infiltrating A20-deleted CD8 T cells had enhanced production of IFNγ and TNFα and reduced expression of the inhibitory receptor programmed cell death 1. As manipulation of A20 expression in CD8 T cells did not result in pathologic manifestations in the mice, we propose it as a candidate to be targeted to increase antitumor efficiency of adoptive T-cell immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cysteine Endopeptidases/immunology , Immunity, Cellular , Intracellular Signaling Peptides and Proteins/immunology , Melanoma/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/pathology , Cysteine Endopeptidases/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Intracellular Signaling Peptides and Proteins/genetics , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
Am J Pathol ; 185(11): 3039-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26343328

ABSTRACT

Tissue pantetheinase, encoded by the VNN1 gene, regulates response to stress, and previous studies have shown that VNN genes contribute to the susceptibility to malaria. Herein, we evaluated the role of pantetheinase on erythrocyte homeostasis and on the development of malaria in patients and in a new mouse model of pantetheinase insufficiency. Patients with cerebral malaria have significantly reduced levels of serum pantetheinase activity (PA). In mouse, we show that a reduction in serum PA predisposes to severe malaria, including cerebral malaria and severe anemia. Therefore, scoring pantetheinase in serum may serve as a severity marker in malaria infection. This disease triggers an acute stress in erythrocytes, which enhances cytoadherence and hemolysis. We speculated that serum pantetheinase might contribute to erythrocyte resistance to stress under homeostatic conditions. We show that mutant mice with a reduced serum PA are anemic and prone to phenylhydrazine-induced anemia. A cytofluorometric and spectroscopic analysis documented an increased frequency of erythrocytes with an autofluorescent aging phenotype. This is associated with an enhanced oxidative stress and shear stress-induced hemolysis. Red blood cell transfer and bone marrow chimera experiments show that the aging phenotype is not cell intrinsic but conferred by the environment, leading to a shortening of red blood cell half-life. Therefore, serum pantetheinase level regulates erythrocyte life span and modulates the risk of developing complicated malaria.


Subject(s)
Amidohydrolases/blood , Erythrocytes/physiology , Malaria/physiopathology , Adolescent , Adult , Amidohydrolases/metabolism , Anemia , Animals , Child , Child, Preschool , Disease Models, Animal , Disease Susceptibility , Female , GPI-Linked Proteins/blood , GPI-Linked Proteins/metabolism , Homeostasis , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Young Adult
12.
Blood ; 123(22): 3462-5, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24677543

ABSTRACT

Follicular lymphoma (FL) is a B-cell neoplasm resulting from the transformation of germinal center (GC) B cells. Although t(14;18) and ectopic B-cell lymphoma 2 (BCL2) expression constitute the genetic hallmark of FL, t(14;18)(pos) B cells bearing genotypic and phenotypic features of FL cells can be found in the blood of most healthy individuals. Nevertheless, the localization of these FL-like cells (FLLCs) in nonmalignant GC-rich tissues and the functional consequences of BCL2 overexpression have not been evaluated thus far. Among 85 reactive lymph node (RLN) samples, 14% were found to contain high levels of t(14;18) by quantitative polymerase chain reaction. In t(14;18)(hi) RLNs, CD20(pos)BCL2(pos)CD10(pos) FLLCs consistently accumulated within the GC, essentially as nonproliferative CXCR4(neg) centrocytes. Moreover, they displayed a reduced response to proliferative stimuli in vitro. Altogether, our findings provide new insights into in situ FLLC functional properties and suggest that these cells have not acquired the ultimate genetic events leading to FL transformation.


Subject(s)
Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 18 , Lymphoma, B-Cell/genetics , Lymphoma, Follicular/genetics , Translocation, Genetic , Adult , Aged , Antigens, CD20/genetics , Antigens, CD20/metabolism , Cell Transformation, Neoplastic/genetics , Female , Germinal Center/metabolism , Germinal Center/pathology , Humans , Male , Middle Aged , Neprilysin/genetics , Neprilysin/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
13.
Immunology ; 145(1): 24-33, 2015 May.
Article in English | MEDLINE | ID: mdl-25367158

ABSTRACT

CD8 T cells contribute to long-term protection against Listeria monocytogenes infection by differentiating into memory T cells. These rapidly respond to antigen or inflammation upon secondary infection. In this study we used CD8 T cells from OT1 mice and CD4 T cells from OT2 mice expressing a fluorescent chimeric granzyme (GZMB-Tom) protein to monitor the primary response to infection with ovalbumin-expressing L. monocytogenes (Lm-OVA). We show that, unlike poorly responding CD4 T cells, CD8 T cells readily proliferated and expressed high levels of GZMB-Tom as early as 2 days after infection. FACS analysis showed GZMB-Tom expression in undivided CD8 T cells, with its level increasing over one to four divisions. OT1 T cells were visualized in the T-cell zone by confocal microscopy. This showed GZMB-Tom-containing granules oriented towards MHCII-positive cells. Twenty hours later, most OT1 T cells had divided but their level of GZMB-Tom expression was reduced. Recently divided OT1 cells failed to express GZMB-Tom. Fourteen hours after secondary infection, GZMB-Tom was re-expressed in memory OT1 T cells responding either to Lm-OVA or L. monocytogenes. Differences in the activation phenotype and in the splenic distribution of OT1 T cells were observed, depending on the challenge. Notably, OTI T cells with polarized granules were only observed after challenge with cognate antigen. This work showed that the GZMB-Tom knock-in mice in which GZMB-Tom faithfully reproduced GZMB expression, provide useful tools to dissect mechanisms leading to the development of anti-bacterial effector and memory CD8 T cells and reactivation of the memory response to cognate antigen or inflammatory signals.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Gene Expression Regulation, Enzymologic/immunology , Granzymes/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Gene Expression Regulation, Enzymologic/genetics , Granzymes/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Listeriosis/genetics , Listeriosis/pathology , Mice , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , Time Factors
14.
J Immunol ; 190(11): 5526-33, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23616571

ABSTRACT

Galectin-1 (GAL1) is an S-type lectin with multiple functions, including the control of B cell homeostasis. GAL1 expression was reported to be under the control of the plasma cell master regulator BLIMP-1. GAL1 was detected at the protein level in LPS-stimulated B cells and was shown to promote Ig secretion in vitro. However, the pattern of GAL1 expression and function of GAL1 in B cells in vivo are still unclear. In this study, we show that, among B cells, GAL1 is only expressed by differentiating plasma cells following T-dependent or T-independent immunization. Using GAL1-deficient mice we demonstrate that GAL1 expression is required for the maintenance of Ag-specific Ig titers and Ab-secreting cell numbers. Using an in vitro differentiation assay we find that GAL1-deficient plasmablasts can develop normally but die rapidly, through caspase 8 activation, under serum starvation-induced death conditions. TUNEL assays show that in vivo-generated GAL1-deficient plasma cells exhibit an increased sensitivity to apoptosis. Taken together, our data indicate that endogenous GAL1 supports plasma cell survival and participates in the regulation of the humoral immune response.


Subject(s)
Galectin 1/metabolism , Homeostasis/immunology , Immunity, Humoral , Plasma Cells/immunology , Plasma Cells/metabolism , Animals , Antigens/immunology , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Galectin 1/genetics , Homeostasis/genetics , Immunity, Humoral/genetics , Immunoglobulin G/blood , Immunophenotyping , Lymphocyte Count , Mice , Mice, Knockout , Plasma Cells/cytology , Spleen/immunology , Spleen/metabolism
15.
Proc Natl Acad Sci U S A ; 109(8): 3006-11, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22315398

ABSTRACT

In response to inflammatory stimulation, dendritic cells (DCs) have a remarkable pattern of differentiation that exhibits specific mechanisms to control the immune response. Here we show that in response to polyriboinosinic:polyribocytidylic acid (pI:C), DCs mount a specific integrated stress response during which the transcription factor ATF4 and the growth arrest and DNA damage-inducible protein 34 (GADD34/Ppp1r15a), a phosphatase 1 (PP1) cofactor, are expressed. In agreement with increased GADD34 levels, an extensive dephosphorylation of the translation initiation factor eIF2α was observed during DC activation. Unexpectedly, although DCs display an unusual resistance to protein synthesis inhibition induced in response to cytosolic dsRNA, GADD34 expression did not have a major impact on protein synthesis. GADD34, however, was shown to be required for normal cytokine production both in vitro and in vivo. These observations have important implications in linking further pathogen detection with the integrated stress response pathways.


Subject(s)
Cytokines/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/enzymology , Poly I-C/pharmacology , Protein Phosphatase 1/metabolism , Protein Subunits/metabolism , Activating Transcription Factor 4/metabolism , Animals , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Eukaryotic Initiation Factor-2/metabolism , Interferon-beta/metabolism , Mice , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Up-Regulation/drug effects , eIF-2 Kinase/metabolism
16.
Genes Chromosomes Cancer ; 53(1): 52-66, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24249258

ABSTRACT

MYC is a potent oncogene involved in ∼70% of human cancers, inducing tumorigenesis with high penetrance and short latency in experimental transgenic models. Accordingly, MYC is recognized as a major driver of T-cell acute lymphoblastic leukemia (T-ALL) in human and zebrafish/mouse models, and uncovering the context by which MYC-mediated malignant transformation initiates and develops remains a considerable challenge. Because MYC is a very complex oncogene, highly dependent on the microenvironment and cell-intrinsic context, we generated transgenic mice (tgMyc(spo)) in which ectopic Myc activation occurs sporadically (<10(-6) thymocytes) within otherwise normal thymic environment, thereby mimicking the unicellular context in which oncogenic alterations initiate human tumors. We show that while Myc(+) clones in tgMyc(spo) mice develop and initially proliferate in thymus and the periphery, no tumor or clonal expansion progress in aging mice (n = 130), suggesting an unexpectedly low ability of Myc to initiate efficient tumorigenesis. Furthermore, to determine the relevance of this observation in human pathogenesis we analyzed a human T-ALL case at diagnosis and relapse using the molecular stigmata of V(D)J recombination as markers of malignant progression; we similarly demonstrate that despite the occurrence of TAL1 and MYC translocations in early thymocyte ontogeny, subsequent oncogenic alterations were required to drive oncogenesis. Altogether, our data suggest that although central to T-ALL, MYC overexpression per se is inefficient in triggering the cascade of events leading to malignant transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Genes, myc/physiology , Intracellular Signaling Peptides and Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Blast Crisis/genetics , Blast Crisis/pathology , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Recurrence , Translocation, Genetic , V(D)J Recombination
17.
Blood ; 117(24): 6552-61, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21511956

ABSTRACT

In the bone marrow (BM), stromal cells constitute a supportive tissue indispensable for the generation of pro-B/pre-BI, pre-BII, and immature B lymphocytes. IL-7-producing stromal cells constitute a cellular niche for pro-B/pre-BI cells, but no specific stromal cell microenvironment was identified for pre-BII cells expressing a functional pre-B cell receptor (pre-BCR). However expression of the pre-BCR represents a crucial checkpoint during B-cell development. We recently demonstrated that the stromal cell derived-galectin1 (GAL1) is a ligand for the pre-BCR, involved in the proliferation and differentiation of normal mouse pre-BII cells. Here we show that nonhematopoietic osteoblasts and reticular cells in the BM express GAL1. We observed that pre-BII cells, unlike the other B-cell subsets, were specifically localized in close contact with GAL1(+) reticular cells. We also determined that IL-7(+) and GAL1(+) cells represent 2 distinct mesenchymal populations with different BM localization. These results demonstrate the existence of a pre-BII specific stromal cell niche and indicate that early B cells move from IL-7(+) to GAL1(+) supportive BM niches during their development.


Subject(s)
Bone Marrow , Galectin 1/metabolism , Precursor Cells, B-Lymphoid/physiology , Stem Cell Niche/physiology , Stromal Cells/physiology , Animals , Bone Marrow/metabolism , Bone Marrow/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Differentiation/immunology , Cells, Cultured , Green Fluorescent Proteins/genetics , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pre-B Cell Receptors/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Stem Cell Niche/cytology , Stem Cell Niche/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
18.
iScience ; 26(6): 106910, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37378323

ABSTRACT

Severe acute malnutrition (SAM) is a multifactorial disease affecting millions of children worldwide. It is associated with changes in intestinal physiology, microbiota, and mucosal immunity, emphasizing the need for multidisciplinary studies to unravel its full pathogenesis. We established an experimental model in which weanling mice fed a high-deficiency diet mimic key anthropometric and physiological features of SAM in children. This diet alters the intestinal microbiota (less segmented filamentous bacteria, spatial proximity to epithelium), metabolism (decreased butyrate), and immune cell populations (depletion of LysoDC in Peyer's patches and intestinal Th17 cells). A nutritional intervention leads to a fast zoometric and intestinal physiology recovery but to an incomplete restoration of the intestinal microbiota, metabolism, and immune system. Altogether, we provide a preclinical model of SAM and have identified key markers to target with future interventions during the education of the immune system to improve SAM whole defects.

19.
Nat Commun ; 14(1): 4290, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37463962

ABSTRACT

Endo-lysosomes transport along microtubules and clustering in the perinuclear area are two necessary steps for microbes to activate specialized phagocyte functions. We report that RUN and FYVE domain-containing protein 3 (RUFY3) exists as two alternative isoforms distinguishable by the presence of a C-terminal FYVE domain and by their affinity for phosphatidylinositol 3-phosphate on endosomal membranes. The FYVE domain-bearing isoform (iRUFY3) is preferentially expressed in primary immune cells and up-regulated upon activation by microbes and Interferons. iRUFY3 is necessary for ARL8b + /LAMP1+ endo-lysosomes positioning in the pericentriolar organelles cloud of LPS-activated macrophages. We show that iRUFY3 controls macrophages migration, MHC II presentation and responses to Interferon-γ, while being important for intracellular Salmonella replication. Specific inactivation of rufy3 in phagocytes leads to aggravated pathologies in mouse upon LPS injection or bacterial pneumonia. This study highlights the role of iRUFY3 in controlling endo-lysosomal dynamics, which contributes to phagocyte activation and immune response regulation.


Subject(s)
Antigen Presentation , Lipopolysaccharides , Animals , Mice , Endosomes/metabolism , Lipopolysaccharides/metabolism , Lysosomes/metabolism , Phagocytes
20.
Mol Microbiol ; 80(3): 628-40, 2011 May.
Article in English | MEDLINE | ID: mdl-21362067

ABSTRACT

The oxidative burst produced by the NADPH oxidase (Phox) is an essential weapon used by host cells to eradicate engulfed pathogens. In Salmonella typhimurium, oxidative stress resistance has been previously proposed to be mediated by the pathogenicity island 2 type III secretion system (T3SS-2), periplasmic superoxide dismutases and cytoplasmic catalases/peroxidases. Here, we fused an OxyR-dependent promoter to the gfp to build the ahpC-gfp transcriptional fusion. This reporter was used to monitor hydrogen peroxide levels as sensed by Salmonella during the course of an infection. We showed that the expression of this fusion was under the exclusive control of reactive oxygen species produced by the host. The ahpC-gfp expression was noticeably modified in the absence of bacterial periplasmic superoxide dismutases or cytoplasmic catalases/peroxidases. Surprisingly, inactivation of the T3SS-2 had no effect on the ahpC-gfp expression. All together, these results led to a model in which Salmonella resistance relies on its arsenal of detoxifying enzymes to cope with Phox-mediated oxidative stress.


Subject(s)
Hydrogen Peroxide/metabolism , Macrophages/microbiology , Reactive Oxygen Species/metabolism , Respiratory Burst , Salmonella typhimurium/drug effects , Animals , Artificial Gene Fusion , Cells, Cultured , Disease Models, Animal , Gene Expression Profiling , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inactivation, Metabolic , Mice , Mice, Inbred C57BL , Microbial Viability/drug effects , Phagosomes/metabolism , Phagosomes/microbiology , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/physiology , Spleen/microbiology , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL