Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 21(9): 1034-1045, 2020 09.
Article in English | MEDLINE | ID: mdl-32661363

ABSTRACT

Skin wounds heal by coordinated induction of inflammation and tissue repair, but the initiating events are poorly defined. Here we uncover a fundamental role of commensal skin microbiota in this process and show that it is mediated by the recruitment and the activation of type I interferon (IFN)-producing plasmacytoid DC (pDC). Commensal bacteria colonizing skin wounds trigger activation of neutrophils to express the chemokine CXCL10, which recruits pDC and acts as an antimicrobial protein to kill exposed microbiota, leading to the formation of CXCL10-bacterial DNA complexes. These complexes and not complexes with host-derived DNA activate pDC to produce type I IFNs, which accelerate wound closure by triggering skin inflammation and early T cell-independent wound repair responses, mediated by macrophages and fibroblasts that produce major growth factors required for healing. These findings identify a key function of commensal microbiota in driving a central innate wound healing response of the skin.


Subject(s)
Dendritic Cells/immunology , Fibroblasts/immunology , Macrophages/immunology , Microbiota/immunology , Neutrophils/immunology , Skin/immunology , Animals , Cells, Cultured , Chemokine CXCL10/metabolism , Humans , Immunity, Innate , Inflammation , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/pathology , Symbiosis , Wound Healing
2.
Nat Immunol ; 19(1): 63-75, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29203862

ABSTRACT

Innate immune cells adjust to microbial and inflammatory stimuli through a process termed environmental plasticity, which links a given individual stimulus to a unique activated state. Here, we report that activation of human plasmacytoid predendritic cells (pDCs) with a single microbial or cytokine stimulus triggers cell diversification into three stable subpopulations (P1-P3). P1-pDCs (PD-L1+CD80-) displayed a plasmacytoid morphology and specialization for type I interferon production. P3-pDCs (PD-L1-CD80+) adopted a dendritic morphology and adaptive immune functions. P2-pDCs (PD-L1+CD80+) displayed both innate and adaptive functions. Each subpopulation expressed a specific coding- and long-noncoding-RNA signature and was stable after secondary stimulation. P1-pDCs were detected in samples from patients with lupus or psoriasis. pDC diversification was independent of cell divisions or preexisting heterogeneity within steady-state pDCs but was controlled by a TNF autocrine and/or paracrine communication loop. Our findings reveal a novel mechanism for diversity and division of labor in innate immune cells.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Gene Expression/immunology , Immunity, Innate/immunology , Adaptive Immunity/immunology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Gene Expression Profiling/methods , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Interferon Type I/metabolism , Lupus Erythematosus, Systemic/immunology , Microscopy, Electron, Transmission , Orthomyxoviridae/immunology , Psoriasis/immunology
3.
Nature ; 603(7899): 145-151, 2022 03.
Article in English | MEDLINE | ID: mdl-35045565

ABSTRACT

COVID-19, which is caused by infection with SARS-CoV-2, is characterized by lung pathology and extrapulmonary complications1,2. Type I interferons (IFNs) have an essential role in the pathogenesis of COVID-19 (refs 3-5). Although rapid induction of type I IFNs limits virus propagation, a sustained increase in the levels of type I IFNs in the late phase of the infection is associated with aberrant inflammation and poor clinical outcome5-17. Here we show that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, which controls immunity to cytosolic DNA, is a critical driver of aberrant type I IFN responses in COVID-19 (ref. 18). Profiling COVID-19 skin manifestations, we uncover a STING-dependent type I IFN signature that is primarily mediated by macrophages adjacent to areas of endothelial cell damage. Moreover, cGAS-STING activity was detected in lung samples from patients with COVID-19 with prominent tissue destruction, and was associated with type I IFN responses. A lung-on-chip model revealed that, in addition to macrophages, infection with SARS-CoV-2 activates cGAS-STING signalling in endothelial cells through mitochondrial DNA release, which leads to cell death and type I IFN production. In mice, pharmacological inhibition of STING reduces severe lung inflammation induced by SARS-CoV-2 and improves disease outcome. Collectively, our study establishes a mechanistic basis of pathological type I IFN responses in COVID-19 and reveals a principle for the development of host-directed therapeutics.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Interferon Type I/immunology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , SARS-CoV-2/immunology , Animals , COVID-19/metabolism , COVID-19/virology , Cells, Cultured , DNA, Mitochondrial/metabolism , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Female , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Macrophages/immunology , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/virology , SARS-CoV-2/pathogenicity , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/pathology
4.
Nat Immunol ; 16(9): 970-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26168081

ABSTRACT

Interleukin 17-producing helper T cells (T(H)17 cells) have a major role in protection against infections and in mediating autoimmune diseases, yet the mechanisms involved are incompletely understood. We found that interleukin 26 (IL-26), a human T(H)17 cell-derived cytokine, is a cationic amphipathic protein that kills extracellular bacteria via membrane-pore formation. Furthermore, T(H)17 cell-derived IL-26 formed complexes with bacterial DNA and self-DNA released by dying bacteria and host cells. The resulting IL-26-DNA complexes triggered the production of type I interferon by plasmacytoid dendritic cells via activation of Toll-like receptor 9, but independently of the IL-26 receptor. These findings provide insights into the potent antimicrobial and proinflammatory function of T(H)17 cells by showing that IL-26 is a natural human antimicrobial that promotes immune sensing of bacterial and host cell death.


Subject(s)
DNA, Bacterial/immunology , DNA/immunology , Immunity, Innate/immunology , Interleukins/immunology , Th17 Cells/immunology , Toll-Like Receptor 9/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Mice , Psoriasis/immunology , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism
5.
J Med Genet ; 61(5): 411-419, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38290824

ABSTRACT

BACKGROUND: Cutaneous epidermal nevi are genotypically diverse mosaic disorders. Pathogenic hotspot variants in HRAS, KRAS, and less frequently, NRAS and BRAF may cause isolated keratinocytic epidermal nevi and sebaceous nevi or several different syndromes when associated with extracutaneous anomalies. Therefore, some authors suggest the concept of mosaic RASopathies to group these different disorders. METHODS: In this paper, we describe three new cases of syndromic epidermal nevi caused by mosaic HRAS variants: one associating an extensive keratinocytic epidermal nevus with hypomastia, another with extensive mucosal involvement and a third combining a small sebaceous nevus with seizures and intellectual deficiency. Moreover, we performed extensive literature of all cases of syndromic epidermal nevi and related disorders with confirmed pathogenic postzygotic variants in HRAS, KRAS, NRAS or BRAF. RESULTS: Most patients presented with bone, ophthalmological or neurological anomalies. Rhabdomyosarcoma, urothelial cell carcinoma and pubertas praecox are also repeatedly reported. KRAS pathogenic variants are involved in 50% of the cases, especially in sebaceous nevi, oculoectodermal syndrome and encephalocraniocutaneous lipomatosis. They are frequently associated with eye and brain anomalies. Pathogenic variants in HRAS are rather present in syndromic keratinocytic epidermal nevi and phacomatosis pigmentokeratotica. CONCLUSION: This review delineates genotype/phenotype correlations of syndromic epidermal nevi with somatic RAS and BRAF pathogenic variants and may help improve their follow-up.


Subject(s)
Nevus , Skin Diseases , Skin Neoplasms , Humans , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins p21(ras)/genetics , Nevus/genetics , Nevus/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
6.
Contact Dermatitis ; 89(4): 284-289, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37286188

ABSTRACT

BACKGROUND: A popular antiseptic spray in Switzerland (Merfen spray), containing chlorhexidine digluconate, benzoxonium chloride and lauramine oxide, is frequently used to treat skin wounds. However, it is also increasingly reported as a major cause of adverse skin reactions, including allergic contact dermatitis (ACD). OBJECTIVES: To investigate the contact allergens responsible for ACD from this antiseptic. PATIENTS/METHODS: Patch tests were performed on seven patients with a clinical history compatible with contact dermatitis from this antiseptic mixture. RESULTS: All patients presented with acute eczematous reactions following contact with either Merfen spray alone, or with multiple products including this spray. Patients showed positive reactions to this product in both patch tests and repeated open application tests (ROATs). Four patients showed dose-dependent reactions to both benzoxonium chloride and lauramine oxide. One patient showed a dose-dependent reaction to the former and a non-dose-dependent reaction to the latter. Finally, two subjects showed responses only to lauramine oxide. One patient reacted to chlorhexidine digluconate 0.5% aq. in addition to both other allergens. CONCLUSIONS: Two commercially unavailable allergens, that is, benzoxonium chloride and/or lauramine oxide were identified as major causes of ACD from Merfen antiseptic spray, whereas chlorhexidine digluconate was a contributing culprit in only one patient.


Subject(s)
Anti-Infective Agents, Local , Dermatitis, Allergic Contact , Humans , Anti-Infective Agents, Local/adverse effects , Chlorhexidine/adverse effects , Allergens/adverse effects , Dermatitis, Allergic Contact/diagnosis , Dermatitis, Allergic Contact/etiology , Switzerland , Patch Tests/adverse effects , Oxides
7.
Rev Med Suisse ; 19(820): 632-636, 2023 Mar 29.
Article in French | MEDLINE | ID: mdl-36988171

ABSTRACT

Juvenile dermatomyositis is a rare multi-system auto-immune disease, particularly causing inflammation of skin and muscles of children. The diagnosis is based on the clinical picture with typical cutaneous lesions, which frequently are the first signs of the disease in contrast to muscle involvement. Muscular MRI is nowadays the first line investigation to diagnose myositis. Recently specific auto-antibodies have been detected allowing a better understanding of the disease and being important prognostic factors. An early diagnosis and aggressive treatment is crucial to induce remission of the disease, especially restore muscular function and to prevent severe complications such as calcinosis and lipodystrophy, which are difficult to treat as well as vital organ dysfunction.


La dermatomyosite juvénile est une maladie auto-immune multisystémique rare caractérisée par une faiblesse musculaire et/ou une éruption cutanée. Le diagnostic se pose par la reconnaissance des signes cutanés caractéristiques qui, contrairement à l'atteinte musculaire, se voit au stade initial de la maladie. L'IRM est l'examen de choix pour détecter la myosite. La mise en évidence récente d'auto-anticorps spécifiques de la maladie a permis de mieux comprendre et d'établir des pronostics sur l'évolution clinique des patients. Un diagnostic précoce et un traitement agressif sont cruciaux pour aider à obtenir une rémission. Ils permettent d'améliorer la fonction musculaire, de prévenir d'importantes séquelles cutanées difficilement traitables comme la calcinose et la lipodystrophie et d'éviter l'atteinte d'organes vitaux.


Subject(s)
Dermatomyositis , Muscular Diseases , Myositis , Child , Humans , Dermatomyositis/diagnosis , Dermatomyositis/therapy , Dermatomyositis/complications , Dermatologists , Myositis/therapy , Skin
8.
J Immunol ; 203(4): 911-921, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31235553

ABSTRACT

Th17 cells play a critical role in the adaptive immune response against extracellular bacteria, and the possible mechanisms by which they can protect against infection are of particular interest. In this study, we describe, to our knowledge, a novel IL-1ß dependent pathway for secretion of the antimicrobial peptide IL-26 from human Th17 cells that is independent of and more rapid than classical TCR activation. We find that IL-26 is secreted 3 hours after treating PBMCs with Mycobacterium leprae as compared with 48 hours for IFN-γ and IL-17A. IL-1ß was required for microbial ligand induction of IL-26 and was sufficient to stimulate IL-26 release from Th17 cells. Only IL-1RI+ Th17 cells responded to IL-1ß, inducing an NF-κB-regulated transcriptome. Finally, supernatants from IL-1ß-treated memory T cells killed Escherichia coli in an IL-26-dependent manner. These results identify a mechanism by which human IL-1RI+ "antimicrobial Th17 cells" can be rapidly activated by IL-1ß as part of the innate immune response to produce IL-26 to kill extracellular bacteria.


Subject(s)
Immunity, Innate/immunology , Interleukin-1beta/immunology , Interleukins/immunology , Lymphocyte Activation/immunology , Th17 Cells/immunology , Bacterial Infections/immunology , Humans , Interleukin-1beta/metabolism , Interleukins/metabolism , Th17 Cells/microbiology
9.
Rev Med Suisse ; 17(732): 646-652, 2021 Mar 31.
Article in French | MEDLINE | ID: mdl-33793103

ABSTRACT

Since the beginning of the COVID-19 pandemic, dermatologists around the world have reported patients with chilblain-like skin lesions on their toes, called COVID-toes. Surprisingly, the majority of these patients do not develop COVID-19 symptoms, and their nasal swabs and serological tests are unable to confirm SARS-CoV-2 infection, despite a clear exposure to the virus. Recent evidence suggests that these patients mount a robust type I interferon response to SARS-CoV-2, making them resistant to the infection. Because chilblains are hallmarks of excessive type I interferons, COVID-toes may represent the skin expression of interferon-mediated resistance to SARS-CoV-2. Uncovering the molecular patho-mechanisms of COVID-toes may provide new avenues to promote SARS-CoV-2 resistance and control the COVID-19 pandemic.


Depuis le début de la pandémie de Covid-19, les dermatologues du monde entier ont signalé des cas inexpliqués de pseudo-engelures, surnommées «â€…orteils Covid ¼ (COVID-toes). Cette recrudescence inhabituelle de pseudo-engelures est d'autant plus intrigante que la majorité des cas ne présentent pas de symptômes de Covid-19 ni de PCR ou de sérologie positive, malgré une exposition probable au SARS-CoV-2. Les données actuelles suggèrent que ces personnes sont prédisposées à induire une immunité innée robuste contre le SARS-CoV-2, ce qui les rend résistantes à l'infection. Une forte réponse interféron de type I, dont les pseudo-engelures sont l'expression clinique, pourrait favoriser une clairance rapide du virus, évitant à la fois la maladie et la séroconversion. Les mécanismes génétiques et moléculaires qui sous-tendent cette résistance naturelle au SARS-CoV-2 restent cependant à élucider.


Subject(s)
COVID-19 , Chilblains , Chilblains/epidemiology , Humans , Pandemics , SARS-CoV-2 , Toes
10.
J Immunol ; 200(10): 3364-3371, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29632142

ABSTRACT

Lupus erythematosus (LE) patients develop autoantibodies that form circulating immune complexes (ICs) with extracellular self-nucleic acids. These ICs are deposited into peripheral tissues, where they trigger detrimental organ inflammation. Recent evidence suggests that ICs contain LL37-DNA complexes derived from neutrophil extracellular traps (NETs) and that LE patients develop pathogenic autoantibodies against these structures, including Abs to LL37. However, the mechanism that leads to the generation of these Abs is unknown. In this study, we show that NETs directly trigger Ab production by human memory B cells. This occurs via LL37-DNA complexes present in NETs, which have the unique ability to gain access to endosomal compartments of B cells and to trigger TLR9 activation. In LE patients, NET-derived LL37-DNA complexes trigger polyclonal B cell activation via TLR9, but also specifically expand self-reactive memory B cells producing anti-LL37 Abs in an Ag-dependent manner. These findings suggest a unique link between neutrophils and B cells in which NETs trigger a concerted activation of TLR9 and BCR leading to anti-NET autoantibody production in lupus.


Subject(s)
B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Neutrophils/immunology , Antimicrobial Cationic Peptides/immunology , Autoantibodies/immunology , DNA/immunology , Extracellular Traps/immunology , Humans , Immunologic Memory/immunology , Toll-Like Receptor 9/immunology , Cathelicidins
11.
Rev Med Suisse ; 16(688): 618-621, 2020 Apr 01.
Article in French | MEDLINE | ID: mdl-32239834

ABSTRACT

Recent progress in molecular engineering, digital imaging and artificial intelligence improve human modern medicine to levels never seen before. Digital pathology becomes the new standard of patient care in dermatology and personalized medicine. It is increasingly used for digital exchange of histological slides, personalized consultations, tumor boards, quantitative image analysis for research purposes and in education. Digital pathology allows automatization and quantification with greater consistency and accuracy than light microscopy. Personalized dermatology is focusing on tailoring therapy to the individual characteristics of each patient and allow to use genetic information in order to develop a treatment plan, uniquely suited to each patient, which in turn leads to improved quality of care and management of each individual.


L'ingénierie moléculaire, l'imagerie digitale et l'intelligence artificielle (IA) améliorent la médecine moderne à des niveaux jamais vus auparavant. La pathologie digitale (PD) est progressivement utilisée pour l'échange digital de lames histologiques produites en routine, les consultations personnalisées, les tumor boards, l'analyse quantitative d'images à des buts de recherche et dans l'éducation, et enfin l'archivage. La PD permet l'automatisation et la quantification avec plus de cohérence et de précision que la microscopie optique. La dermatologie personnalisée se concentre sur l'adaptation de la thérapie aux caractéristiques individuelles de chaque patient et permet d'utiliser les données génétiques afin de développer un plan de traitement individuellement adapté, en améliorant la qualité des soins et la prise en charge.


Subject(s)
Dermatology/methods , Pathology/methods , Artificial Intelligence , Computers , Dermatologists , Humans
12.
Blood ; 129(17): 2420-2428, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28167662

ABSTRACT

The endoplasmic reticulum kinase inositol-requiring enzyme 1 (IRE1) and its downstream target X-box-binding protein 1 (XBP1) drive B-cell differentiation toward plasma cells and have been shown to contribute to multiple myeloma development; yet, little is known of the role of this pathway in diffuse large B-cell lymphoma (DLBCL). Here, we show that in the germinal center B-cell-like (GCB) DLBCL subtype, IRE1 expression is reduced to a level that prevents XBP1 activation. Gene expression profiles indicated that, in GCB DLBCL cancer samples, expression of IRE1 messenger RNA was inversely correlated with the levels and activity of the epigenetic repressor, histone methyltransferase enhancer of zeste homolog 2 (EZH2). Correspondingly, in GCB-derived cell lines, the IRE1 promoter carried increased levels of the repressive epigenetic mark histone 3 lysine 27 trimethylation. Pharmacological inhibition of EZH2 erased those marks and restored IRE1 expression and function in vitro and in vivo. Moreover, reconstitution of the IRE1-signaling pathway, by expression of the XBP1-active form, compromised GCB DLBCL tumor growth in a mouse xenograft cancer model. These findings indicate that IRE1-XBP1 downregulation distinguishes GCB DLBCL from other DLBCL subtypes and contributes to tumor growth.


Subject(s)
B-Lymphocytes/immunology , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Germinal Center/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , X-Box Binding Protein 1/genetics , Animals , B-Lymphocytes/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/immunology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Epigenesis, Genetic , Germinal Center/pathology , Histones/genetics , Histones/immunology , Humans , Indazoles/pharmacology , Leupeptins/pharmacology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Mice , Mice, 129 Strain , Plasma Cells/immunology , Plasma Cells/pathology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/immunology , Pyridones/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/immunology , Signal Transduction , X-Box Binding Protein 1/antagonists & inhibitors , X-Box Binding Protein 1/immunology , Xenograft Model Antitumor Assays
13.
Rev Med Suisse ; 20(867): 619-620, 2024 Mar 27.
Article in French | MEDLINE | ID: mdl-38563535
14.
Mol Pharm ; 15(1): 116-126, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29185769

ABSTRACT

By screening a drug library comprising FDA approved compounds, we discovered a potent interaction between the antifungal agent haloprogin and the experimental organometallic drug RAPTA-T, to synergistically induce cancer cell killing. The combination of these two small molecules, even at low doses, elicited an improved therapeutic response on tumor growth over either agent alone or the current treatment used in the clinic in the highly aggressive syngeneic B16F10 melanoma tumor model, where classical cytotoxic chemotherapeutic agents show little efficacy. The combination with the repurposed chemodrug haloprogin provides the basis for a new powerful treatment option for cutaneous melanoma. Importantly, because synergistic induction of tumor cell death is achieved with low individual drug doses, and cellular targets for RAPTA-T are different from those of classical chemotherapeutic drugs, a therapeutic strategy based on this approach could avoid toxicities and potentially resistance mechanisms, and could even inhibit metastatic progression.


Subject(s)
Antifungal Agents/therapeutic use , Drug Repositioning/methods , Melanoma/drug therapy , Organometallic Compounds/therapeutic use , Skin Neoplasms/drug therapy , Animals , Antifungal Agents/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Drug Synergism , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Organometallic Compounds/chemistry , Phenyl Ethers/therapeutic use , Melanoma, Cutaneous Malignant
15.
EMBO Rep ; 17(10): 1471-1484, 2016 10.
Article in English | MEDLINE | ID: mdl-27572820

ABSTRACT

Activation of the elongation factor 2 kinase (eEF2K) leads to the phosphorylation and inhibition of the elongation factor eEF2, reducing mRNA translation rates. Emerging evidence indicates that the regulation of factors involved in protein synthesis may be critical for controlling diverse biological processes including cancer progression. Here we show that inhibitors of the HIV aspartyl protease (HIV-PIs), nelfinavir in particular, trigger a robust activation of eEF2K leading to the phosphorylation of eEF2. Beyond its anti-viral effects, nelfinavir has antitumoral activity and promotes cell death. We show that nelfinavir-resistant cells specifically evade eEF2 inhibition. Decreased cell viability induced by nelfinavir is impaired in cells lacking eEF2K. Moreover, nelfinavir-mediated anti-tumoral activity is severely compromised in eEF2K-deficient engrafted tumors in vivo Our findings imply that exacerbated activation of eEF2K is detrimental for tumor survival and describe a mechanism explaining the anti-tumoral properties of HIV-PIs.


Subject(s)
Elongation Factor 2 Kinase/metabolism , Neoplasms/metabolism , Neoplasms/pathology , AMP-Activated Protein Kinases/metabolism , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Drug Resistance/genetics , Elongation Factor 2 Kinase/genetics , Female , Gene Expression , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Nelfinavir/chemistry , Nelfinavir/pharmacology , Neoplasms/genetics , Peptide Elongation Factor 2/metabolism , Phosphorylation , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , Tumor Burden
16.
J Immunol ; 196(4): 1900-9, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26773151

ABSTRACT

Type I IFNs (IFN-I) are key innate mediators that create a profound antiviral state and orchestrate the activation of almost all immune cells. Plasmacytoid dendritic cells (pDCs) are the most powerful IFN-I-producing cells and play important roles during viral infections, cancer, and autoimmune diseases. By comparing gene expression profiles of murine pDCs and conventional DCs, we found that CD28, a prototypic T cell stimulatory receptor, was highly expressed in pDCs. Strikingly, CD28 acted as a negative regulator of pDC IFN-I production upon TLR stimulation but did not affect pDC survival or maturation. Importantly, cell-intrinsic CD28 expression restrained pDC (and systemic) IFN-I production during in vivo RNA and DNA viral infections, limiting antiviral responses and enhancing viral growth early after exposure. Finally, CD28 also downregulated IFN-I response upon skin injury. Our study identified a new pDC regulatory mechanism by which the same CD28 molecule that promotes stimulation in most cells that express it is co-opted to negatively regulate pDC IFN-I production and limit innate responses.


Subject(s)
CD28 Antigens/immunology , Dendritic Cells/immunology , Interferon Type I/biosynthesis , Interferon Type I/immunology , Animals , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Transcriptome
19.
Proc Natl Acad Sci U S A ; 112(50): 15408-13, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26607445

ABSTRACT

Spontaneous CD8 T-cell responses occur in growing tumors but are usually poorly effective. Understanding the molecular and cellular mechanisms that drive these responses is of major interest as they could be exploited to generate a more efficacious antitumor immunity. As such, stimulator of IFN genes (STING), an adaptor molecule involved in cytosolic DNA sensing, is required for the induction of antitumor CD8 T responses in mouse models of cancer. Here, we find that enforced activation of STING by intratumoral injection of cyclic dinucleotide GMP-AMP (cGAMP), potently enhanced antitumor CD8 T responses leading to growth control of injected and contralateral tumors in mouse models of melanoma and colon cancer. The ability of cGAMP to trigger antitumor immunity was further enhanced by the blockade of both PD1 and CTLA4. The STING-dependent antitumor immunity, either induced spontaneously in growing tumors or induced by intratumoral cGAMP injection was dependent on type I IFNs produced in the tumor microenvironment. In response to cGAMP injection, both in the mouse melanoma model and an ex vivo model of cultured human melanoma explants, the principal source of type I IFN was not dendritic cells, but instead endothelial cells. Similarly, endothelial cells but not dendritic cells were found to be the principal source of spontaneously induced type I IFNs in growing tumors. These data identify an unexpected role of the tumor vasculature in the initiation of CD8 T-cell antitumor immunity and demonstrate that tumor endothelial cells can be targeted for immunotherapy of melanoma.


Subject(s)
Endothelial Cells/metabolism , Immunity , Membrane Proteins/metabolism , Neoplasms/immunology , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Dose-Response Relationship, Immunologic , Endothelial Cells/drug effects , Injections, Intralesional , Interferon Type I/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neoplasms/pathology , Nucleotides, Cyclic/administration & dosage , Nucleotides, Cyclic/pharmacology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/drug effects
20.
Rev Med Suisse ; 19(820): 603-604, 2023 03 29.
Article in French | MEDLINE | ID: mdl-36988166

Subject(s)
Pruritus , Humans
SELECTION OF CITATIONS
SEARCH DETAIL