Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(2): e2316104121, 2024 Jan 09.
Article de Anglais | MEDLINE | ID: mdl-38165941

RÉSUMÉ

The nuclear receptor corepressor (NCoR) forms a complex with histone deacetylase 3 (HDAC3) that mediates repressive functions of unliganded nuclear receptors and other transcriptional repressors by deacetylation of histone substrates. Recent studies provide evidence that NCoR/HDAC3 complexes can also exert coactivator functions in brown adipocytes by deacetylating and activating PPARγ coactivator 1α (PGC1α) and that signaling via receptor activator of nuclear factor kappa-B (RANK) promotes the formation of a stable NCoR/HDAC3/PGC1ß complex that coactivates nuclear factor kappa-B (NFκB)- and activator protein 1 (AP-1)-dependent genes required for osteoclast differentiation. Here, we demonstrate that activation of Toll-like receptor (TLR) 4, but not TLR3, the interleukin 4 (IL4) receptor nor the Type I interferon receptor, also promotes assembly of an NCoR/HDAC3/PGC1ß coactivator complex. Receptor-specific utilization of TNF receptor-associated factor 6 (TRAF6) and downstream activation of extracellular signal-regulated kinase 1 (ERK1) and TANK-binding kinase 1 (TBK1) accounts for the common ability of RANK and TLR4 to drive assembly of an NCoR/HDAC3/PGC1ß complex in macrophages. ERK1, the p65 component of NFκB, and the p300 histone acetyltransferase (HAT) are also components of the induced complex and are associated with local histone acetylation and transcriptional activation of TLR4-dependent enhancers and promoters. These observations identify a TLR4/TRAF6-dependent signaling pathway that converts NCoR from a corepressor of nuclear receptors to a coactivator of NFκB and AP-1 that may be relevant to functions of NCoR in other developmental and homeostatic processes.


Sujet(s)
Histone , Facteur-6 associé aux récepteurs de TNF , Activation de la transcription , Protéines corépressives/génétique , Histone/génétique , Histone/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur de transcription AP-1/métabolisme , Récepteur de type Toll-4/métabolisme , Transduction du signal , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Récepteurs cytoplasmiques et nucléaires/métabolisme
2.
J Invest Dermatol ; 144(1): 116-124.e4, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37478901

RÉSUMÉ

Cutibacterium acnes is a commensal bacterium on the skin that is generally well-tolerated, but different strain types have been hypothesized to contribute to the disease acne vulgaris. To understand how some strain types might contribute to skin inflammation, we generated a repository of C. acnes isolates from skin swabs of healthy subjects and subjects with acne and assessed their strain-level identity and capacity to stimulate cytokine release. Phylotype II K-type strains were more frequent on healthy and nonlesional skin of subjects with acne than those isolated from lesions. Phylotype IA-1 C-type strains were increased on lesional skin compared with those on healthy skin. The capacity to induce cytokines from cultured monocyte-derived dendritic cells was opposite to this action on sebocytes and keratinocytes and did not correlate with the strain types associated with the disease. Whole-genome sequencing revealed a linear plasmid in high-inflammatory isolates within similar strain types that had different proinflammatory responses. Single-cell RNA sequencing of mouse skin after intradermal injection showed that strains containing this plasmid induced a higher inflammatory response in dermal fibroblasts. These findings revealed that C. acnes strain type is insufficient to predict inflammation and that carriage of a plasmid could contribute to disease.


Sujet(s)
Acné juvénile , Dermatite , Animaux , Souris , Humains , Peau/microbiologie , Acné juvénile/microbiologie , Propionibacterium acnes/génétique , Plasmides/génétique , Inflammation , Cytokines/génétique
3.
Nat Immunol ; 24(11): 1825-1838, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37735593

RÉSUMÉ

Noncoding genetic variation drives phenotypic diversity, but underlying mechanisms and affected cell types are incompletely understood. Here, investigation of effects of natural genetic variation on the epigenomes and transcriptomes of Kupffer cells derived from inbred mouse strains identified strain-specific environmental factors influencing Kupffer cell phenotypes, including leptin signaling in Kupffer cells from a steatohepatitis-resistant strain. Cell-autonomous and non-cell-autonomous effects of genetic variation were resolved by analysis of F1 hybrid mice and cells engrafted into an immunodeficient host. During homeostasis, non-cell-autonomous trans effects of genetic variation dominated control of Kupffer cells, while strain-specific responses to acute lipopolysaccharide injection were dominated by actions of cis-acting effects modifying response elements for lineage-determining and signal-dependent transcription factors. These findings demonstrate that epigenetic landscapes report on trans effects of genetic variation and serve as a resource for deeper analyses into genetic control of transcription in Kupffer cells and macrophages in vitro.


Sujet(s)
Cellules de Küpffer , Transcriptome , Souris , Animaux , Épigénome , Souris de lignée C57BL , Variation génétique
4.
Mol Cell ; 83(19): 3421-3437.e11, 2023 10 05.
Article de Anglais | MEDLINE | ID: mdl-37751740

RÉSUMÉ

The nuclear receptor co-repressor (NCoR) complex mediates transcriptional repression dependent on histone deacetylation by histone deacetylase 3 (HDAC3) as a component of the complex. Unexpectedly, we found that signaling by the receptor activator of nuclear factor κB (RANK) converts the NCoR/HDAC3 co-repressor complex to a co-activator of AP-1 and NF-κB target genes that are required for mouse osteoclast differentiation. Accordingly, the dominant function of NCoR/HDAC3 complexes in response to RANK signaling is to activate, rather than repress, gene expression. Mechanistically, RANK signaling promotes RNA-dependent interaction of the transcriptional co-activator PGC1ß with the NCoR/HDAC3 complex, resulting in the activation of PGC1ß and inhibition of HDAC3 activity for acetylated histone H3. Non-coding RNAs Dancr and Rnu12, which are associated with altered human bone homeostasis, promote NCoR/HDAC3 complex assembly and are necessary for RANKL-induced osteoclast differentiation in vitro. These findings may be prototypic for signal-dependent functions of NCoR in other biological contexts.


Sujet(s)
Ostéoclastes , ARN , Humains , Souris , Animaux , Protéines corépressives/génétique , Ostéoclastes/métabolisme , Ligand de RANK/génétique , Corépresseur-1 de récepteur nucléaire/génétique , Corépresseur-1 de récepteur nucléaire/métabolisme , Expression des gènes
5.
Sci Transl Med ; 14(632): eabh1478, 2022 02 16.
Article de Anglais | MEDLINE | ID: mdl-35171653

RÉSUMÉ

Innate immune defense against deep tissue infection by Staphylococcus aureus is orchestrated by fibroblasts that become antimicrobial when triggered to differentiate into adipocytes. However, the role of this process in noninfectious human diseases is unknown. To investigate the potential role of adipogenesis by dermal fibroblasts in acne, a disorder triggered by Cutibacterium acnes, single-cell RNA sequencing was performed on human acne lesions and mouse skin challenged by C. acnes. A transcriptome consistent with adipogenesis was observed within specific fibroblast subsets from human acne and mouse skin lesions infected with C. acnes. Perifollicular dermal preadipocytes in human acne and mouse skin lesions showed colocalization of PREF1, an early marker of adipogenesis, and cathelicidin (Camp), an antimicrobial peptide. This capacity of C. acnes to specifically trigger production of cathelicidin in preadipocytes was dependent on TLR2. Treatment of wild-type mice with retinoic acid (RA) suppressed the capacity of C. acnes to form acne-like lesions, inhibited adipogenesis, and enhanced cathelicidin expression in preadipocytes, but lesions were unresponsive in Camp-/- mice, despite the anti-adipogenic action of RA. Analysis of inflamed skin of acne patients after retinoid treatment also showed enhanced induction of cathelicidin, a previously unknown beneficial effect of retinoids in difficult-to-treat acne. Overall, these data provide evidence that adipogenic fibroblasts are a critical component of the pathogenesis of acne and represent a potential target for therapy.


Sujet(s)
Acné juvénile , Anti-infectieux , Maladies de la peau , Animaux , Antibactériens/pharmacologie , Anti-infectieux/pharmacologie , Humains , Souris , Propionibacterium acnes/métabolisme , Staphylococcus aureus , Trétinoïne/pharmacologie
6.
J Clin Invest ; 131(21)2021 11 01.
Article de Anglais | MEDLINE | ID: mdl-34720087

RÉSUMÉ

Inflammatory disorders of the skin are frequently associated with inflammatory bowel diseases (IBDs). To explore mechanisms by which these organs communicate, we performed single-cell RNA-Seq analysis on fibroblasts from humans and mice with IBD. This analysis revealed that intestinal inflammation promoted differentiation of a subset of intestinal stromal fibroblasts into preadipocytes with innate antimicrobial host defense activity. Furthermore, this process of reactive adipogenesis was exacerbated if mouse skin was inflamed as a result of skin wounding or infection. Since hyaluronan (HA) catabolism is activated during skin injury and fibroblast-to-adipocyte differentiation is dependent on HA, we tested the hypothesis that HA fragments could alter colon fibroblast function by targeted expression of human hyaluronidase-1 in basal keratinocytes from mouse skin. Hyaluronidase expression in the skin activated intestinal stromal fibroblasts, altered the fecal microbiome, and promoted excessive reactive adipogenesis and increased inflammation in the colon after challenge with dextran sodium sulfate. The response to digested HA was dependent on expression of TLR4 by preadipocytes. Collectively, these results suggest that the association between skin inflammation and IBD may be due to recognition by mesenchymal fibroblasts in the colon of HA released during inflammation of the skin.


Sujet(s)
Colite/métabolisme , Fibroblastes/métabolisme , Maladies inflammatoires intestinales/métabolisme , Muqueuse intestinale/métabolisme , Peau/métabolisme , Animaux , Colite/génétique , Colite/anatomopathologie , Fibroblastes/anatomopathologie , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Muqueuse intestinale/anatomopathologie , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Souris , Souris knockout , Peau/anatomopathologie
7.
STAR Protoc ; 2(1): 100363, 2021 03 19.
Article de Anglais | MEDLINE | ID: mdl-33748781

RÉSUMÉ

Significant advancements in understanding disease mechanisms can occur through combined analysis of next-generation sequencing datasets generated using purified cell populations. Here, we detail our optimized protocol for purification of mouse hepatic macrophages (or other liver non-parenchymal populations) suitable for use in various next-generation sequencing protocols. An alternative framework is described for sorting pre-fixed hepatic nuclei populations. This strategy has the advantage of rapidly preserving the nuclei and can facilitate success with ChIP-seq for more challenging molecules. For complete details on the use and execution of these protocols, please refer to Muse et al. (2018), Sakai et al. (2019), and Seidman et al. (2020).


Sujet(s)
Séquençage après immunoprécipitation de la chromatine/méthodes , Immunoprécipitation de la chromatine/méthodes , Animaux , Noyau de la cellule , Hépatocytes/métabolisme , Séquençage nucléotidique à haut débit/méthodes , Souris , Analyse de séquence d'ADN , Facteurs de transcription/isolement et purification
8.
STAR Protoc ; 2(1): 100358, 2021 03 19.
Article de Anglais | MEDLINE | ID: mdl-33718886

RÉSUMÉ

Integrative analysis of next-generation sequencing data can help understand disease mechanisms. Specifically, ChIP-seq can illuminate where transcription regulators bind to regulate transcription. A major obstacle to performing this assay on primary cells is the low numbers obtained from tissues. The extensively validated ChIP-seq protocol presented here uses small volumes and single-pot on-bead library preparation to generate diverse high-quality ChIP-seq data. This protocol allows for medium-to-high-throughput ChIP-seq of low-abundance cells and can also be applied to other mammalian cells. For complete details on the use and execution of this protocol, please refer to Brigidi et al. (2019), Carlin et al. (2018), Heinz et al. (2018), Nott et al. (2019), Sakai et al. (2019), and Seidman et al. (2020).


Sujet(s)
Séquençage après immunoprécipitation de la chromatine , Animaux , Cellules cultivées , Humains , Souris
9.
Dermatol Online J ; 26(6)2020 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-32815691

RÉSUMÉ

Cutaneous side effects such as acneiform eruption, xerosis, and paronychia are frequently observed in patients undergoing treatment with epidermal growth factor receptor (EGFR) inhibitors for non-small cell lung cancer and other solid tumors. Interestingly, these side effects appear to positively correlate with length of remission, indicating that disruption of homeostatic EGFR signaling in the skin may serve as a marker of therapeutic EGFR inhibition in tumors. We report the case of a woman with metastatic lung cancer in remission being treated with the EGFR inhibitor, erlotinib, who experienced numerous commonly occurring adverse cutaneous reactions early in her treatment, and after two years of treatment developed eruptive nevi as well as a nevoid melanoma. Changes in pigmented lesions and the development of melanoma have been described during treatment with the BRAF inhibitor, vemurafenib, and are believed to relate to paradoxical activation of BRAF and the MAPK pathway. We speculate that a similar mechanism may occur during treatment with EGFR inhibitors. Therefore, thorough skin examinations are essential for patients undergoing long term treatment with erlotinib.


Sujet(s)
Récepteurs ErbB/antagonistes et inhibiteurs , Chlorhydrate d'erlotinib/effets indésirables , Naevus/induit chimiquement , Inhibiteurs de protéines kinases/effets indésirables , Tumeurs cutanées/induit chimiquement , Sujet âgé , Tumeurs du sein , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Femelle , Humains , Tumeurs du poumon/traitement médicamenteux , Mélanome/induit chimiquement
10.
Immunity ; 52(6): 1057-1074.e7, 2020 06 16.
Article de Anglais | MEDLINE | ID: mdl-32362324

RÉSUMÉ

Tissue-resident and recruited macrophages contribute to both host defense and pathology. Multiple macrophage phenotypes are represented in diseased tissues, but we lack deep understanding of mechanisms controlling diversification. Here, we investigate origins and epigenetic trajectories of hepatic macrophages during diet-induced non-alcoholic steatohepatitis (NASH). The NASH diet induced significant changes in Kupffer cell enhancers and gene expression, resulting in partial loss of Kupffer cell identity, induction of Trem2 and Cd9 expression, and cell death. Kupffer cell loss was compensated by gain of adjacent monocyte-derived macrophages that exhibited convergent epigenomes, transcriptomes, and functions. NASH-induced changes in Kupffer cell enhancers were driven by AP-1 and EGR that reprogrammed LXR functions required for Kupffer cell identity and survival to instead drive a scar-associated macrophage phenotype. These findings reveal mechanisms by which disease-associated environmental signals instruct resident and recruited macrophages to acquire distinct gene expression programs and corresponding functions.


Sujet(s)
Microenvironnement cellulaire/génétique , Reprogrammation cellulaire/génétique , Épigenèse génétique , Régulation de l'expression des gènes , Cellules myéloïdes/métabolisme , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/métabolisme , Animaux , Marqueurs biologiques , Séquençage après immunoprécipitation de la chromatine , Régime alimentaire , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Gene Ontology , Séquençage nucléotidique à haut débit , Cellules de Küpffer/immunologie , Cellules de Küpffer/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Souris , Stéatose hépatique non alcoolique/anatomopathologie , Spécificité d'organe/génétique , Spécificité d'organe/immunologie , Liaison aux protéines , Transduction du signal , Analyse sur cellule unique
11.
Cell Metab ; 31(1): 189-206.e8, 2020 01 07.
Article de Anglais | MEDLINE | ID: mdl-31761566

RÉSUMÉ

Oxidized phospholipids (OxPLs), which arise due to oxidative stress, are proinflammatory and proatherogenic, but their roles in non-alcoholic steatohepatitis (NASH) are unknown. Here, we show that OxPLs accumulate in human and mouse NASH. Using a transgenic mouse that expresses a functional single-chain variable fragment of E06, a natural antibody that neutralizes OxPLs, we demonstrate the causal role of OxPLs in NASH. Targeting OxPLs in hyperlipidemic Ldlr-/- mice improved multiple aspects of NASH, including steatosis, inflammation, fibrosis, hepatocyte death, and progression to hepatocellular carcinoma. Mechanistically, we found that OxPLs promote ROS accumulation to induce mitochondrial dysfunction in hepatocytes. Neutralizing OxPLs in AMLN-diet-fed Ldlr-/- mice reduced oxidative stress, improved hepatic and adipose-tissue mitochondrial function, and fatty-acid oxidation. These results suggest targeting OxPLs may be an effective therapeutic strategy for NASH.


Sujet(s)
Apoptose/génétique , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/métabolisme , Mitochondries/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stress oxydatif , Phospholipides/métabolisme , Anticorps à chaîne unique/usage thérapeutique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/traitement médicamenteux , Alimentation riche en graisse , Stéatose hépatique/complications , Stéatose hépatique/traitement médicamenteux , Gene Ontology , Hépatocytes/cytologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Humains , Inflammation/complications , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Cirrhose du foie/complications , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/métabolisme , Tumeurs du foie/traitement médicamenteux , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Microscopie électronique à transmission , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/anatomopathologie , Mitochondries/ultrastructure , Stéatose hépatique non alcoolique/complications , Stéatose hépatique non alcoolique/métabolisme , Obésité/traitement médicamenteux , Oxydoréduction , Phospholipides/sang , Phospholipides/immunologie , RNA-Seq , Espèces réactives de l'oxygène/métabolisme
12.
Immunity ; 51(4): 655-670.e8, 2019 10 15.
Article de Anglais | MEDLINE | ID: mdl-31587991

RÉSUMÉ

Tissue environment plays a powerful role in establishing and maintaining the distinct phenotypes of resident macrophages, but the underlying molecular mechanisms remain poorly understood. Here, we characterized transcriptomic and epigenetic changes in repopulating liver macrophages following acute Kupffer cell depletion as a means to infer signaling pathways and transcription factors that promote Kupffer cell differentiation. We obtained evidence that combinatorial interactions of the Notch ligand DLL4 and transforming growth factor-b (TGF-ß) family ligands produced by sinusoidal endothelial cells and endogenous LXR ligands were required for the induction and maintenance of Kupffer cell identity. DLL4 regulation of the Notch transcriptional effector RBPJ activated poised enhancers to rapidly induce LXRα and other Kupffer cell lineage-determining factors. These factors in turn reprogrammed the repopulating liver macrophage enhancer landscape to converge on that of the original resident Kupffer cells. Collectively, these findings provide a framework for understanding how macrophage progenitor cells acquire tissue-specific phenotypes.


Sujet(s)
Cellules de Küpffer/physiologie , Foie/métabolisme , Macrophages/physiologie , Cellules myéloïdes/physiologie , Animaux , Différenciation cellulaire , Cellules cultivées , Microenvironnement cellulaire , Reprogrammation cellulaire , Facteur de transcription CBF-1/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Foie/cytologie , Récepteurs hépatiques X/génétique , Protéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Souris transgéniques , Phénotype , Transduction du signal , Facteur de croissance transformant bêta/métabolisme
13.
Transl Psychiatry ; 9(1): 24, 2019 01 17.
Article de Anglais | MEDLINE | ID: mdl-30655503

RÉSUMÉ

SETD5, a gene linked to intellectual disability (ID) and autism spectrum disorder (ASD), is a member of the SET-domain family and encodes a putative histone methyltransferase (HMT). To date, the mechanism by which SETD5 haploinsufficiency causes ASD/ID remains an unanswered question. Setd5 is the highly conserved mouse homolog, and although the Setd5 null mouse is embryonic lethal, the heterozygote is viable. Morphological tracing and multielectrode array was used on cultured cortical neurons. MRI was conducted of adult mouse brains and immunohistochemistry of juvenile mouse brains. RNA-Seq was used to investigate gene expression in the developing cortex. Behavioral assays were conducted on adult mice. Setd5+/- cortical neurons displayed significantly reduced synaptic density and neuritic outgrowth in vitro, with corresponding decreases in network activity and synchrony by electrophysiology. A specific subpopulation of fetal Setd5+/- cortical neurons showed altered gene expression of neurodevelopment-related genes. Setd5+/- animals manifested several autism-like behaviors, including hyperactivity, cognitive deficit, and altered social interactions. Anatomical differences were observed in Setd5+/- adult brains, accompanied by a deficit of deep-layer cortical neurons in the developing brain. Our data converge on a picture of abnormal neurodevelopment driven by Setd5 haploinsufficiency, consistent with a highly penetrant risk factor.


Sujet(s)
Trouble du spectre autistique/génétique , Comportement animal , Haploinsuffisance/génétique , Methyltransferases/génétique , Neurones/métabolisme , Animaux , Trouble du spectre autistique/psychologie , Encéphale/anatomopathologie , Femelle , Prédisposition génétique à une maladie , Hétérozygote , Imagerie par résonance magnétique , Mâle , Souris , Souris knockout , Mutation
14.
Cell Metab ; 29(2): 457-474.e5, 2019 02 05.
Article de Anglais | MEDLINE | ID: mdl-30595478

RÉSUMÉ

The nature of obesity-associated islet inflammation and its impact on ß cell abnormalities remains poorly defined. Here, we explore immune cell components of islet inflammation and define their roles in regulating ß cell function and proliferation. Islet inflammation in obese mice is dominated by macrophages. We identify two islet-resident macrophage populations, characterized by their anatomical distributions, distinct phenotypes, and functional properties. Obesity induces the local expansion of resident intra-islet macrophages, independent of recruitment from circulating monocytes. Functionally, intra-islet macrophages impair ß cell function in a cell-cell contact-dependent manner. Increased engulfment of ß cell insulin secretory granules by intra-islet macrophages in obese mice may contribute to restricting insulin secretion. In contrast, both intra- and peri-islet macrophage populations from obese mice promote ß cell proliferation in a PDGFR signaling-dependent manner. Together, these data define distinct roles and mechanisms for islet macrophages in the regulation of islet ß cells.


Sujet(s)
Inflammation/immunologie , Cellules à insuline/métabolisme , Macrophages/immunologie , Obésité/métabolisme , Récepteurs aux facteurs de croissance dérivés des plaquettes/immunologie , Animaux , Lignée cellulaire , Prolifération cellulaire , Sécrétion d'insuline , Cellules à insuline/anatomopathologie , Macrophages/cytologie , Mâle , Souris , Souris de lignée C57BL , Souris obèse
15.
Dermatol Online J ; 25(11)2019 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-32045153

RÉSUMÉ

Bullous pemphigoid (BP) is an autoimmune blistering disorder that predominantly affects the elderly. Treatment regimens typically include topical and systemic immunosuppressive medications. Although effective, systemic corticosteroids are sometimes poorly tolerated in the elderly patient, contributing to the overall morbidity and mortality of BP. Dupilumab is a monoclonal antibody targeting interleukin 4 receptor alpha (IL4R?), approved for the treatment of atopic dermatitis, as well as moderate to severe asthma and chronic rhinosinusitis with nasal polyposis. In recent reports, dupilumab has been successfully used off-label to treat a variety of pruritic disorders, including chronic spontaneous urticaria [1], anal and genital itch [2], allergic contact dermatitis [3], and prurigo nodularis [4, 5]. We report here a case of an elderly patient with refractory BP whose symptoms of pruritus and blistering became well-controlled with the addition of dupilumab to the treatment regimen.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Pemphigoïde bulleuse/traitement médicamenteux , Prurit/traitement médicamenteux , Sujet âgé de 80 ans ou plus , Résistance aux substances , Granulocytes éosinophiles , Humains , Mâle , Pemphigoïde bulleuse/complications , Pemphigoïde bulleuse/immunologie , Prurit/étiologie , Lymphocytes auxiliaires Th2/métabolisme
16.
Proc Natl Acad Sci U S A ; 115(20): E4680-E4689, 2018 05 15.
Article de Anglais | MEDLINE | ID: mdl-29632203

RÉSUMÉ

Activation of liver X receptors (LXRs) with synthetic agonists promotes reverse cholesterol transport and protects against atherosclerosis in mouse models. Most synthetic LXR agonists also cause marked hypertriglyceridemia by inducing the expression of sterol regulatory element-binding protein (SREBP)1c and downstream genes that drive fatty acid biosynthesis. Recent studies demonstrated that desmosterol, an intermediate in the cholesterol biosynthetic pathway that suppresses SREBP processing by binding to SCAP, also binds and activates LXRs and is the most abundant LXR ligand in macrophage foam cells. Here we explore the potential of increasing endogenous desmosterol production or mimicking its activity as a means of inducing LXR activity while simultaneously suppressing SREBP1c-induced hypertriglyceridemia. Unexpectedly, while desmosterol strongly activated LXR target genes and suppressed SREBP pathways in mouse and human macrophages, it had almost no activity in mouse or human hepatocytes in vitro. We further demonstrate that sterol-based selective modulators of LXRs have biochemical and transcriptional properties predicted of desmosterol mimetics and selectively regulate LXR function in macrophages in vitro and in vivo. These studies thereby reveal cell-specific discrimination of endogenous and synthetic regulators of LXRs and SREBPs, providing a molecular basis for dissociation of LXR functions in macrophages from those in the liver that lead to hypertriglyceridemia.


Sujet(s)
Biomimétique , Desmostérol/pharmacologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Récepteurs hépatiques X/métabolisme , Macrophages/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Animaux , Cellules HepG2 , Hépatocytes/cytologie , Hépatocytes/effets des médicaments et des substances chimiques , Humains , Récepteurs hépatiques X/génétique , Macrophages/cytologie , Macrophages/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée C57BL , Régions promotrices (génétique) , Protéine-1 de liaison à l'élément de régulation des stérols/génétique
17.
Nat Med ; 23(11): 1309-1318, 2017 Nov.
Article de Anglais | MEDLINE | ID: mdl-29035364

RÉSUMÉ

The cellular mechanism(s) linking macrophages to norepinephrine (NE)-mediated regulation of thermogenesis have been a topic of debate. Here we identify sympathetic neuron-associated macrophages (SAMs) as a population of cells that mediate clearance of NE via expression of solute carrier family 6 member 2 (SLC6A2), an NE transporter, and monoamine oxidase A (MAOA), a degradation enzyme. Optogenetic activation of the sympathetic nervous system (SNS) upregulates NE uptake by SAMs and shifts the SAM profile to a more proinflammatory state. NE uptake by SAMs is prevented by genetic deletion of Slc6a2 or inhibition of the encoded transporter. We also observed an increased proportion of SAMs in the SNS of two mouse models of obesity. Genetic ablation of Slc6a2 in SAMs increases brown adipose tissue (BAT) content, causes browning of white fat, increases thermogenesis, and leads to substantial and sustained weight loss in obese mice. We further show that this pathway is conserved, as human sympathetic ganglia also contain SAMs expressing the analogous molecular machinery for NE clearance, which thus constitutes a potential target for obesity treatment.


Sujet(s)
Macrophages/métabolisme , Neurones/métabolisme , Norépinéphrine/métabolisme , Obésité/anatomopathologie , Système nerveux sympathique/anatomopathologie , Animaux , Récepteur-1 de la chimiokine CX3C/métabolisme , Analyse de profil d'expression de gènes , Homéostasie , Souris , Souris de lignée C57BL , Souris transgéniques , Transporteurs de la norépinéphrine/génétique , Obésité/génétique
18.
Microbiol Spectr ; 4(5)2016 10.
Article de Anglais | MEDLINE | ID: mdl-28087927

RÉSUMÉ

Macrophages play essential roles in the response to injury and infection and contribute to the development and/or homeostasis of the various tissues they reside in. Conversely, macrophages also influence the pathogenesis of metabolic, neurodegenerative, and neoplastic diseases. Mechanisms that contribute to the phenotypic diversity of macrophages in health and disease remain poorly understood. Here we review the recent application of genome-wide approaches to characterize the transcriptomes and epigenetic landscapes of tissue-resident macrophages. These studies are beginning to provide insights into how distinct tissue environments are interpreted by transcriptional regulatory elements to drive specialized programs of gene expression.


Sujet(s)
Macrophages/physiologie , Animaux , Étude d'association pangénomique , Humains , Activation des macrophages/génétique , Transcriptome
19.
J Biol Chem ; 286(36): 31105-12, 2011 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-21757758

RÉSUMÉ

The bacterial tmRNA·SmpB system facilitates recycling of stalled translational complexes in a process termed "ribosome rescue." During ribosome rescue, the nascent chain is tagged with the tmRNA-encoded ssrA peptide, which targets the tagged polypeptide for degradation. Translational pausing also induces a variety of recoding events such as frameshifts, ribosome hops, and stop codon readthrough. To examine the interplay between recoding and ribosome rescue, we determined the various fates of ribosomes that pause during translation termination. We expressed a model protein containing the C-terminal Asp-Pro nascent peptide motif (which interferes with translation termination) and quantified the protein chains produced by recoding and ssrA-peptide tagging. The nature and extent of translational recoding depended upon the codon for the C-terminal Pro residue, with CCU and CCC promoting efficient +1 frameshifting. In contrast, ssrA-peptide tagging was unaffected by C-terminal Pro coding. Moreover, +1 frameshifting was not suppressed by tmRNA·SmpB activity, suggesting that recoding and ribosome rescue are not competing events. However, cells lacking ribosomal protein L9 (ΔL9) exhibited a significant increase in recoding and a concomitant decrease in ssrA-peptide tagging. Pulse-chase analysis revealed that pre-termination ribosomes turn over more rapidly in ΔL9 cells, suggesting that increased recoding alleviates the translational arrest. Together, these results indicate that tmRNA·SmpB does not suppress transient ribosome pauses, but responds to prolonged translational arrest.


Sujet(s)
Biosynthèse des protéines , Ribosomes/métabolisme , Codon , Escherichia coli/génétique , ARN bactérien/physiologie , Protéines de liaison à l'ARN/physiologie , Protéines ribosomiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...