Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 43
Filtrer
1.
Basic Res Cardiol ; 117(1): 48, 2022 10 07.
Article de Anglais | MEDLINE | ID: mdl-36205817

RÉSUMÉ

Although p38 MAP Kinase α (p38 MAPKα) is generally accepted to play a central role in the cardiac stress response, to date its function in maladaptive cardiac hypertrophy is still not unambiguously defined. To induce a pathological type of cardiac hypertrophy we infused angiotensin II (AngII) for 2 days via osmotic mini pumps in control and tamoxifen-inducible, cardiomyocyte (CM)-specific p38 MAPKα KO mice (iCMp38αKO) and assessed cardiac function by echocardiography, complemented by transcriptomic, histological, and immune cell analysis. AngII treatment after inactivation of p38 MAPKα in CM results in left ventricular (LV) dilatation within 48 h (EDV: BL: 83.8 ± 22.5 µl, 48 h AngII: 109.7 ± 14.6 µl) and an ectopic lipid deposition in cardiomyocytes, reflecting a metabolic dysfunction in pressure overload (PO). This was accompanied by a concerted downregulation of transcripts for oxidative phosphorylation, TCA cycle, and fatty acid metabolism. Cardiac inflammation involving neutrophils, macrophages, B- and T-cells was significantly enhanced. Inhibition of adipose tissue lipolysis by the small molecule inhibitor of adipocytetriglyceride lipase (ATGL) Atglistatin reduced cardiac lipid accumulation by 70% and neutrophil infiltration by 30% and went along with an improved cardiac function. Direct targeting of neutrophils by means of anti Ly6G-antibody administration in vivo led to a reduced LV dilation in iCMp38αKO mice and an improved systolic function (EF: 39.27 ± 14%). Thus, adipose tissue lipolysis and CM lipid accumulation augmented cardiac inflammation in iCMp38αKO mice. Neutrophils, in particular, triggered the rapid left ventricular dilatation. We provide the first evidence that p38 MAPKα acts as an essential switch in cardiac adaptation to PO by mitigating metabolic dysfunction and inflammation. Moreover, we identified a heart-adipose tissue-immune cell crosstalk, which might serve as new therapeutic target in cardiac pathologies.


Sujet(s)
Défaillance cardiaque , Myocytes cardiaques , Tissu adipeux/métabolisme , Angiotensine-II/métabolisme , Animaux , Cardiomégalie/métabolisme , Acides gras/métabolisme , Inflammation/métabolisme , Triacylglycerol lipase/métabolisme , Triacylglycerol lipase/usage thérapeutique , Lipides/usage thérapeutique , Souris , Souris de lignée C57BL , Myocytes cardiaques/métabolisme , Granulocytes neutrophiles/métabolisme , Tamoxifène/métabolisme , Tamoxifène/usage thérapeutique , p38 Mitogen-Activated Protein Kinases/métabolisme , p38 Mitogen-Activated Protein Kinases/usage thérapeutique
3.
Mol Nutr Food Res ; 66(21): e2100991, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35094491

RÉSUMÉ

SCOPE: While cocoa flavanol (CF) consumption improves cardiovascular risk biomarkers, molecular mechanisms underlying their protective effects are not understood. OBJECTIVE: To investigate nutri(epi)genomic effects of CF and identify regulatory networks potential mediating vascular health benefits. METHODS AND RESULTS: Twenty healthy middle-aged men consume CF (bi-daily 450 mg) or control drinks for 1 month. Microarray analysis identifies 2235 differentially expressed genes (DEG) involved in processes regulating immune response, cell adhesion, or cytoskeleton organization. Distinct patterns of DEG correlate with CF-related changes in endothelial function, arterial stiffness, and blood pressure. DEG profile negatively correlates with expression profiles of cardiovascular disease patients. CF modulated DNA methylation profile of genes implicates in cell adhesion, actin cytoskeleton organization, or cell signaling. In silico docking analyses indicate that CF metabolites have the potential of binding to cell signaling proteins and transcription factors. Incubation of plasma obtained after CF consumption decrease monocyte to endothelial adhesion and dose-dependently increase nitric oxide-dependent chemotaxis of circulating angiogenic cells further validating the biological functions of CF metabolites. CONCLUSION: In healthy humans, CF consumption may mediate vascular protective effects by modulating gene expression and DNA methylation towards a cardiovascular protective effect, in agreement with clinical results, by preserving integrity of immunological-endothelial barrier functions.


Sujet(s)
Cacaoyer , Flavonols , Adulte d'âge moyen , Mâle , Humains , Flavonols/pharmacologie , Cacaoyer/composition chimique , Polyphénols/pharmacologie , Pression sanguine , Génomique , Méthode en double aveugle
4.
mBio ; 12(2)2021 03 30.
Article de Anglais | MEDLINE | ID: mdl-33785629

RÉSUMÉ

Infection with the obligate intracellular bacterium Chlamydia trachomatis is the most common bacterial sexually transmitted disease worldwide. Since no vaccine is available to date, antimicrobial therapy is the only alternative in C. trachomatis infection. However, changes in chlamydial replicative activity and the occurrence of chlamydial persistence caused by diverse stimuli have been proven to impair treatment effectiveness. Here, we report the mechanism for C. trachomatis regulating host signaling processes and mitochondrial function, which can be used for chlamydial metabolic reprogramming during treatment with ß-lactam antimicrobials. Activation of signal transducer and activator of transcription 3 (STAT3) is a well-known host response in various bacterial and viral infections. In C. trachomatis infection, inactivation of STAT3 by host protein tyrosine phosphatases increased mitochondrial respiration in both the absence and presence of ß-lactam antimicrobials. However, during treatment with ß-lactam antimicrobials, C. trachomatis increased the production of citrate as well as the activity of host ATP-citrate lyase involved in fatty acid synthesis. Concomitantly, chlamydial metabolism switched from the tricarboxylic acid cycle to fatty acid synthesis. This metabolic switch was a unique response in treatment with ß-lactam antimicrobials and was not observed in gamma interferon (IFN-γ)-induced persistent infection. Inhibition of fatty acid synthesis was able to attenuate ß-lactam-induced chlamydial persistence. Our findings highlight the importance of the mitochondrion-fatty acid interplay for the metabolic reprogramming of C. trachomatis during treatment with ß-lactam antimicrobials.IMPORTANCE The mitochondrion generates most of the ATP in eukaryotic cells, and its activity is used for controlling the intracellular growth of Chlamydia trachomatis Furthermore, mitochondrial activity is tightly connected to host fatty acid synthesis that is indispensable for chlamydial membrane biogenesis. Phospholipids, which are composed of fatty acids, are the central components of the bacterial membrane and play a crucial role in the protection against antimicrobials. Chlamydial persistence that is induced by various stimuli is clinically relevant. While one of the well-recognized inducers, ß-lactam antimicrobials, has been used to characterize chlamydial persistence, little is known about the role of mitochondria in persistent infection. Here, we demonstrate how C. trachomatis undergoes metabolic reprogramming to switch from the tricarboxylic acid cycle to fatty acid synthesis with promoted host mitochondrial activity in response to treatment with ß-lactam antimicrobials.


Sujet(s)
Antibactériens/pharmacologie , Infections à Chlamydia/microbiologie , Chlamydia trachomatis/effets des médicaments et des substances chimiques , Chlamydia trachomatis/métabolisme , Acides gras/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , bêta-Lactames/pharmacologie , Infections à Chlamydia/traitement médicamenteux , Chlamydia trachomatis/génétique , Cellules HeLa , Humains , Mitochondries/métabolisme , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme
5.
Sci Rep ; 9(1): 11021, 2019 07 30.
Article de Anglais | MEDLINE | ID: mdl-31363109

RÉSUMÉ

The p38MAPK downstream targets MAPKAP kinases (MK) 2 and 3 are critical for the regulation of the macrophage response to LPS. The extents to which these two kinases act cooperatively and distinctly in regulating LPS-induced inflammatory cytokine expression are still unclear. To address this uncertainty, whole transcriptome analyses were performed using bone marrow-derived macrophages (BMDM) generated from MK2-/- or MK2/3-/- animals and their wild-type littermates. The results suggest that in BMDM, MK2 and MK3 not only cooperatively regulate the transcript expression of signaling intermediates, including IL-10, IL-19, CXCL2 and the IL-4 receptor (IL-4R)α subunit, they also exert distinct regulatory effects on the expression of specific transcripts. Based on the differential regulation of gene expression by MK2 and MK3, at least six regulatory patterns were identified. Importantly, we confirmed our previous finding, which showed that in the absence of MK2, MK3 negatively regulates IFN-ß. Moreover, this genome-wide analysis identified the regulation of Cr1A, NOD1 and Serpina3f as similar to that of IFN-ß. In the absence of MK2, MK3 also delayed the nuclear translocation of NFκB by delaying the ubiquitination and subsequent degradation of IκBß, reflecting the substantial plasticity of the response of BMDM to LPS.


Sujet(s)
Protéines et peptides de signalisation intracellulaire/métabolisme , Système de signalisation des MAP kinases , Macrophages/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Transcriptome , Animaux , Cellules cultivées , Chimiokine CXCL2 , Interféron gamma/génétique , Interféron gamma/métabolisme , Interleukines/génétique , Interleukines/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Lipopolysaccharides/toxicité , Macrophages/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Protein-Serine-Threonine Kinases/génétique , Récepteurs de surface cellulaire/génétique , Récepteurs de surface cellulaire/métabolisme , p38 Mitogen-Activated Protein Kinases/génétique , p38 Mitogen-Activated Protein Kinases/métabolisme
6.
Transl Psychiatry ; 9(1): 156, 2019 05 31.
Article de Anglais | MEDLINE | ID: mdl-31150013

RÉSUMÉ

Currently, the clinical diagnosis of schizophrenia relies solely on self-reporting and clinical interview, and likely comprises heterogeneous biological subsets. Such subsets may be defined by an underlying biology leading to solid biomarkers. A transgenic rat model modestly overexpressing the full-length, non-mutant Disrupted-in-Schizophrenia 1 (DISC1) protein (tgDISC1 rat) was generated that defines such a subset, inspired by our previous identification of insoluble DISC1 protein in post mortem brains from patients with chronic mental illness. Besides specific phenotypes such as DISC1 protein pathology, abnormal dopamine homeostasis, and changes in neuroanatomy and behavior, this animal model also shows subtle disturbances in overarching signaling pathways relevant for schizophrenia. In a reverse-translational approach, assuming that both the animal model and a patient subset share common disturbed signaling pathways, we identified differentially expressed transcripts from peripheral blood mononuclear cells of tgDISC1 rats that revealed an interconnected set of dysregulated genes, led by decreased expression of regulator of G-protein signaling 1 (RGS1), chemokine (C-C) ligand 4 (CCL4), and other immune-related transcripts enriched in T-cell and macrophage signaling and converging in one module after weighted gene correlation network analysis. Testing expression of this gene network in two independent cohorts of patients with schizophrenia versus healthy controls (n = 16/50 and n = 54/45) demonstrated similar expression changes. The two top markers RGS1 and CCL4 defined a subset of 27% of patients with 97% specificity. Thus, analogous aberrant signaling pathways can be identified by a blood test in an animal model and a corresponding schizophrenia patient subset, suggesting that in this animal model tailored pharmacotherapies for this patient subset could be achieved.


Sujet(s)
Marqueurs biologiques/sang , Réseaux de régulation génique , Schizophrénie , Transduction du signal/génétique , Animaux , Chimiokine CCL4/sang , Études de cohortes , Modèles animaux de maladie humaine , Mâle , Protéines de tissu nerveux/génétique , Protéines RGS/sang , Rats , Rat Sprague-Dawley , Rats transgéniques , Schizophrénie/sang , Schizophrénie/classification , Schizophrénie/génétique , Schizophrénie/immunologie , Sensibilité et spécificité
7.
Oncotarget ; 9(32): 22703-22716, 2018 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-29854309

RÉSUMÉ

Glioblastoma is the most aggressive type of glioma. The Wingless (Wnt) signaling pathway has been shown to promote stem cell properties and resistance to radio- and chemotherapy in glioblastoma. Here, we demonstrate that pharmacological Wnt pathway inhibition using the porcupine inhibitor LGK974 acts synergistically with temozolomide (TMZ), the chemotherapeutic drug currently used as standard treatment for glioblastoma, to suppress in vitro growth of glioma cells. Synergistic growth inhibition was independent of the O6-alkylguanine DNA alkyltransferase (MGMT) promoter methylation status. Transcriptomic analysis revealed that expression of aldehyde dehydrogenase 3A1 (ALDH3A1) was significantly down-regulated when cells were treated with LGK974 and TMZ. Suppressing ALDH3A1 expression increased the efficacy of TMZ and reduced clonogenic potential accompanied by decreased expression of stem cell markers CD133, Nestin and Sox2. Taken together, our study suggests that previous observations concerning Wnt signaling blockade to reduce chemoresistance in glioblastoma is at least in part mediated by inhibition of ALDH3A1.

8.
PLoS Biol ; 16(6): e2004408, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29927970

RÉSUMÉ

We show that the cyclin-dependent kinase inhibitor 1B (CDKN1B)/p27, previously known as a cell cycle inhibitor, is also localized within mitochondria. The migratory capacity of endothelial cells, which need intact mitochondria, is completely dependent on mitochondrial p27. Mitochondrial p27 improves mitochondrial membrane potential, increases adenosine triphosphate (ATP) content, and is required for the promigratory effect of caffeine. Domain mapping of p27 revealed that the N-terminus and C-terminus are required for those improvements. Further analysis of those regions revealed that the translocation of p27 into the mitochondria and its promigratory activity depend on serine 10 and threonine 187. In addition, mitochondrial p27 protects cardiomyocytes against apoptosis. Moreover, mitochondrial p27 is necessary and sufficient for cardiac myofibroblast differentiation. In addition, p27 deficiency and aging decrease respiration in heart mitochondria. Caffeine does not increase respiration in p27-deficient animals, whereas aged mice display improvement after 10 days of caffeine in drinking water. Moreover, caffeine induces transcriptome changes in a p27-dependent manner, affecting mostly genes relevant for mitochondrial processes. Caffeine also reduces infarct size after myocardial infarction in prediabetic mice and increases mitochondrial p27. Our data characterize mitochondrial p27 as a common denominator that improves mitochondria-dependent processes and define an increase in mitochondrial p27 as a new mode of action of caffeine.


Sujet(s)
Caféine/pharmacologie , Cardiotoniques/pharmacologie , Inhibiteur p27 de kinase cycline-dépendante/métabolisme , Mitochondries/métabolisme , Infarctus du myocarde/anatomopathologie , Myocytes cardiaques/physiologie , Adénosine triphosphate/métabolisme , Animaux , Apoptose/physiologie , Différenciation cellulaire/physiologie , Lignée cellulaire , Mouvement cellulaire/physiologie , Inhibiteur p27 de kinase cycline-dépendante/génétique , Cellules endothéliales/physiologie , Cellules HEK293 , Humains , Potentiel de membrane mitochondriale/physiologie , Souris , Souris de lignée C57BL , Souris knockout , Myocytes cardiaques/cytologie , Transport des protéines/physiologie
9.
Toxicol Appl Pharmacol ; 354: 40-55, 2018 09 01.
Article de Anglais | MEDLINE | ID: mdl-29753005

RÉSUMÉ

It is widely accepted that human brain development has unique features that cannot be represented by rodents. Obvious reasons are the evolutionary distance and divergent physiology. This might lead to false predictions when rodents are used for safety or pharmacological efficacy studies. For a better translation of animal-based research to the human situation, human in vitro systems might be useful. In this study, we characterize developing neural progenitor cells from prenatal human and time-matched rat and mouse brains by analyzing the changes in their transcriptome profile during neural differentiation. Moreover, we identify hub molecules that regulate neurodevelopmental processes like migration and differentiation. Consequences of modulation of three of those hubs on these processes were studied in a species-specific context. We found that although the gene expression profiles of the three species largely differ qualitatively and quantitatively, they cluster in similar GO terms like cell migration, gliogenesis, neurogenesis or development of multicellular organism. Pharmacological modulation of the identified hub molecules triggered species-specific cellular responses. This study underlines the importance of understanding species differences on the molecular level and advocates the use of human based in vitro models for pharmacological and toxicological research.


Sujet(s)
Encéphale/physiologie , Analyse de profil d'expression de gènes/méthodes , Cellules souches neurales/physiologie , Transcriptome , Animaux , Encéphale/embryologie , Encéphale/métabolisme , Régulation de l'expression des gènes au cours du développement , Réseaux de régulation génique , Humains , Souris , Cellules souches neurales/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Rats , Appréciation des risques , Spécificité d'espèce , Sphéroïdes de cellules , Facteurs temps
10.
Microb Drug Resist ; 24(9): 1305-1315, 2018 Nov.
Article de Anglais | MEDLINE | ID: mdl-29750595

RÉSUMÉ

Enterobacter cloacae complex is a common cause of hospital outbreaks. A retrospective and prospective molecular analysis of carbapenem-resistant clinical isolates in a tertiary care center demonstrated an outbreak of a German-imipenemase-1 (GIM-1) metallo-beta-lactamase-producing Enterobacter hormaechei ssp. steigerwaltii affecting 23 patients between 2009 and 2016. Thirty-three isolates were sequence type 89 by conventional multilocus sequence typing (MLST) and displayed a maximum difference of 49 out of 3,643 targets in the ad-hoc core-genome MLST (cgMLST) scheme (SeqSphere+ software; Ridom, Münster, Germany). The relatedness of all isolates was confirmed by further maximum-likelihood phylogeny. One clonal complex of highly related isolates (≤15 allele difference in cgMLST) contained 17 patients, but epidemiological data only suggested five transmission events. The blaGIM-1-gene was embedded in a class-1-integron (In770) and the Tn21-subgroup transposon Tn6216 (KC511628) on a 25-kb plasmid. Environmental screening detected one colonized sink trap in a service room. The outbreak was self-limited as no further blaGIM-1-positive E. hormaechei has been isolated since 2016. Routine molecular screening of carbapenem-nonsusceptible gram-negative isolates detected a long-term, low-frequency outbreak of a GIM-1-producing E. hormaechei ssp. steigerwaltii clone. This highlights the necessity of molecular surveillance.


Sujet(s)
Enterobacter cloacae/génétique , Enterobacter cloacae/isolement et purification , Infections à Enterobacteriaceae/microbiologie , bêta-Lactamases/génétique , Antibactériens/pharmacologie , Carbapénèmes/pharmacologie , Infection croisée/traitement médicamenteux , Infection croisée/microbiologie , Épidémies de maladies , Enterobacter cloacae/effets des médicaments et des substances chimiques , Infections à Enterobacteriaceae/traitement médicamenteux , Allemagne , Humains , Intégrons/génétique , Tests de sensibilité microbienne/méthodes , Typage par séquençage multilocus/méthodes , Plasmides/génétique , Études prospectives , Études rétrospectives , Centres de soins tertiaires
11.
Sci Rep ; 8(1): 255, 2018 01 10.
Article de Anglais | MEDLINE | ID: mdl-29321478

RÉSUMÉ

Cholestasis is caused by autoimmune reactions, drug-induced hepatotoxicity, viral infections of the liver and the obstruction of bile ducts by tumours or gallstones. Cholestatic conditions are associated with impaired innate and adaptive immunity, including alterations of the cellular functions of monocytes, macrophages, NK cells and T-cells. Bile acids act as signalling molecules, affecting lipopolysaccharide (LPS)-induced cytokine expression in primary human macrophages. The present manuscript investigates the impact of bile acids, such as taurolithocholic acid (TLC), on the transcriptome of human macrophages in the presence or absence of LPS. While TLC itself has almost no effect on gene expression under control conditions, this compound modulates the expression of 202 out of 865 transcripts in the presence of LPS. Interestingly, pathway analysis revealed that TLC specifically supressed the expression of genes involved in mediating pro-inflammatory effects, phagocytosis, interactions with pathogens and autophagy as well as the recruitment of immune cells, such as NK cells, neutrophils and T cells. These data indicate a broad influence of bile acids on inflammatory responses and immune functions in macrophages. These findings may contribute to the clinical observation that patients with cholestasis present a lack of response to bacterial or viral infections.


Sujet(s)
Acides et sels biliaires/métabolisme , Reprogrammation cellulaire , Macrophages/métabolisme , Phénotype , Acides et sels biliaires/pharmacologie , Marqueurs biologiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/génétique , Chimiotaxie , Cytokines/génétique , Cytokines/métabolisme , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Humains , Médiateurs de l'inflammation/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Lipopolysaccharides/immunologie , Activation des macrophages/génétique , Activation des macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Transcriptome
12.
J Tissue Eng Regen Med ; 12(1): 114-128, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-27966263

RÉSUMÉ

Mesenchymal stromal cells (MSCs) play a pivotal role in modern therapeutic approaches in bone-healing disorders. Although bone marrow-derived MSCs are most frequently used, the knowledge that many other adult tissues represent promising sources for potent MSCs has gained acceptance. In the present study, the osteogenic differentiation potential of porcine skin fibroblasts (FBs), as well as bone marrow- (BMSCs), adipose tissue- (ASCs) and dental pulp-derived stromal cells (DSCs) were evaluated. However, additional application of BMP-2 significantly elevated the delayed osteogenic differentiation capacity of ASC and FB cultures, and in DSC cultures the supplementation of platelet-rich plasma increased osteogenic differentiation potential to a comparable level of the good differentiable BMSCs. Furthermore, microarray gene expression performed in an exemplary manner for ASCs and BMSCs revealed that ASCs and BMSCs use different gene expression patterns for osteogenic differentiation under standard media conditions, as diverse MSCs are imprinted dependent from their tissue niche. However, after increasing the differentiation potential of ASCs to a comparable level as shown in BMSCs, a small subset of identical key molecules was used to differentiate in the osteogenic lineage. Until now, the importance of identified genes seems to be underestimated for osteogenic differentiation. Apparently, the regulation of transmembrane protein 229A, interleukin-33 and the fibroblast growth factor receptor-2 in the early phase of osteogenic differentiation is needed for optimum results. Based on these results, bone regeneration strategies of MSCs have to be adjusted, and in vivo studies on the osteogenic capacities of the different types of MCSs are warranted. Copyright © 2016 The Authors Tissue Engineering and Regenerative Medicine published by John Wiley & Sons, Ltd.


Sujet(s)
Cellules souches mésenchymateuses/cytologie , Spécificité d'organe , Ostéogenèse , Tissu adipeux/cytologie , Phosphatase alcaline/métabolisme , Animaux , Cellules de la moelle osseuse/cytologie , Protéine morphogénétique osseuse de type 2/métabolisme , Différenciation cellulaire , Pulpe dentaire/cytologie , Femelle , Fibroblastes/cytologie , Régulation de l'expression des gènes , Cellules souches mésenchymateuses/métabolisme , Ostéogenèse/génétique , Phénotype , Plasma riche en plaquettes/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Suidae , Porc miniature
13.
PLoS One ; 12(7): e0181383, 2017.
Article de Anglais | MEDLINE | ID: mdl-28753664

RÉSUMÉ

Mycoplasma hominis is the second smallest facultative pathogen of the human urogenital tract. With less than 600 protein-encoding genes, it represents an ideal model organism for the study of host-pathogen interactions. For a comprehensive characterisation of the M. hominis action in infection a customized Mho microarray, which was based on two genome sequences (PG21 and LBD-4), was designed to analyze the dynamics of the mycoplasma transcriptome during infection and validated for M. hominis strain FBG. RNA preparation was evaluated and adapted to ensure the highest recovery of mycoplasmal mRNAs from in vitro HeLa cell infection assays. Following cRNA hybridization, the read-out strategy of the hybridization results was optimized and confirmed by RT-PCR. A statistically robust infection assay with M. hominis strain FBG enabled the identification of differentially regulated key effector molecules such as critical cytoadhesins (4 h post infection (pI)), invasins (48 h pI) and proteins associated with establishing chronic infection of the host (336 h pI). Of the 294 differentially regulated genes (>2-fold) 128 (43.5%) encoded hypothetical proteins, including lipoproteins that seem to play a central role as virulence factors at each stage of infection: P75 as a novel cytoadhesin candidate, which is also differentially upregulated in chronic infection; the MHO_2100 protein, a postulated invasin and the MHO_730-protein, a novel ecto-nuclease and domain of an ABC transporter, the function of which in chronic infection has still to be elucidated. Implementation of the M. hominis microarray strategy led to a comprehensive identification of to date unknown candidates for virulence factors at relevant stages of host cell infection.


Sujet(s)
Mycoplasma hominis/génétique , Mycoplasma hominis/pathogénicité , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Génome bactérien/génétique , Cellules HeLa , Humains , ARN messager/génétique , RT-PCR
14.
Immunol Cell Biol ; 95(7): 630-639, 2017 08.
Article de Anglais | MEDLINE | ID: mdl-28377612

RÉSUMÉ

Aberrantly activated CD4+ T memory cells play a central role in the development of type-1-diabetes. Interleukin-7 promotes generation of autoimmune memory T cells and increased Interleukin-7 availability is associated with type-1-diabetes susceptibility. T-cell-mediated immune pathology at onset of type-1-diabetes is well defined, but characteristics of long-term symptomatic disease stages remain largely elusive. In the present study, memory CD4+ T-cell activation and cytokine expression as well as sensitivity to Interleukin-7 in vitro were compared between patients with type-1-diabetes at clinical onset (n=25), long-term symptomatic disease (median duration 4.5 years, n=19) and matched healthy controls (n=21). T-cell responses of type-1-diabetes patients were characterized by higher frequencies of cytokine and activation marker expressing CD4+ memory T cells as compared to healthy controls. Notably, correction for individual cytokine expression levels revealed qualitative differences of cytokine profiles characterized by significantly increased TNFα and decreased IL-2-expressing T-cell proportions in long-term type-1-diabetes patients. IL-7-mediated T-cell co-stimulation induced quantitative and qualitative cytokine expression differences highly similar to type-1-diabetes-specific profiles. In addition, CD4+ memory T cells from children with long-term type-1-diabetes were more sensitive to in vitro IL-7 co-stimulation. Global transcriptome analysis revealed IL-7 induced expression differences of CD4+ T cells, including increased IL-2R expression and effects on subsequent T-cell receptor activation. We conclude that long-term symptomatic type-1-diabetes patients differed in memory T-cell cytokine profiles and Interleukin-7 co-stimulation. Regulation of IL-2 expression and sensitivity are affected with possible consequences for disease course and severity at long-term type-1-diabetes stages.


Sujet(s)
Lymphocytes T CD4+/immunologie , Diabète de type 1/immunologie , Interleukine-2/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Adolescent , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Études cas-témoins , Enfant , Enfant d'âge préscolaire , Diabète de type 1/anatomopathologie , Femelle , Analyse de profil d'expression de gènes , Humains , Mémoire immunologique/effets des médicaments et des substances chimiques , Interféron gamma/métabolisme , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Interleukine-7/pharmacologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Mâle , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs CCR7/métabolisme
15.
Free Radic Res ; 51(1): 91-102, 2017 Jan.
Article de Anglais | MEDLINE | ID: mdl-28090786

RÉSUMÉ

Acute myocardial infarction is the leading cause of mortality in the industrialized world. While it is essential to attempt an early reperfusion of ischemic myocardial territories, reperfusion itself adds damage to the heart, the ischemia-reperfusion (I/R) injury. Particularly the injury resulting from the very first minutes of reperfusion remains incompletely understood. MicroRNAs (miRNAs) are dynamic regulators in I/R injury. Nitric oxide (•NO) signaling, in turn, interacts with miRNA signaling. Our previous investigations showed that •NO signaling in I/R could be modulated by nitrite. We therefore sought to investigate the role of miRNAs in nitrite cardioprotection with focus on the first few minutes of reperfusion. The study was conducted in mice in vivo with 30 min of ischemia and 5 min of reperfusion. Mice received a single-dose of nitrite or saline intracardially 5 min prior to reperfusion. We identified nine miRNAs to be up-regulated after 5 min of reperfusion. The up-regulation of almost half of those miRNAs (miR-125a-5p, miR-146b, miR-339-3p, miR-433) was inhibited by nitrite treatment, perpetuating baseline values. In silico analysis revealed the Irak-M gene to be a target of miR-146b and miR-339-3p. Correspondingly, a rise in Irak-M transcript and protein levels occurred by nitrite treatment within the early phase of reperfusion. The results demonstrate that already a very short phase of reperfusion is sufficient for significant dysregulation in cardiac miRNAs expression and that nitrite preserves baseline values of miRNAs in the scale of only a few minutes. These findings hint at a potential novel cardioprotective mechanism of nitrite signaling.


Sujet(s)
microARN/métabolisme , Infarctus du myocarde/métabolisme , Lésion de reperfusion myocardique/métabolisme , Myocarde/métabolisme , Nitrite de sodium/pharmacologie , Animaux , Apoptose , Expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Gene Ontology , Mâle , Souris , microARN/génétique , Myocarde/anatomopathologie , Phosphorylation oxydative , Transduction du signal
16.
PLoS Pathog ; 12(12): e1006043, 2016 Dec.
Article de Anglais | MEDLINE | ID: mdl-27936238

RÉSUMÉ

Trehalose biosynthesis is considered an attractive target for the development of antimicrobials against fungal, helminthic and bacterial pathogens including Mycobacterium tuberculosis. The most common biosynthetic route involves trehalose-6-phosphate (T6P) synthase OtsA and T6P phosphatase OtsB that generate trehalose from ADP/UDP-glucose and glucose-6-phosphate. In order to assess the drug target potential of T6P phosphatase, we generated a conditional mutant of M. tuberculosis allowing the regulated gene silencing of the T6P phosphatase gene otsB2. We found that otsB2 is essential for growth of M. tuberculosis in vitro as well as for the acute infection phase in mice following aerosol infection. By contrast, otsB2 is not essential for the chronic infection phase in mice, highlighting the substantial remodelling of trehalose metabolism during infection by M. tuberculosis. Blocking OtsB2 resulted in the accumulation of its substrate T6P, which appears to be toxic, leading to the self-poisoning of cells. Accordingly, blocking T6P production in a ΔotsA mutant abrogated otsB2 essentiality. T6P accumulation elicited a global upregulation of more than 800 genes, which might result from an increase in RNA stability implied by the enhanced neutralization of toxins exhibiting ribonuclease activity. Surprisingly, overlap with the stress response caused by the accumulation of another toxic sugar phosphate molecule, maltose-1-phosphate, was minimal. A genome-wide screen for synthetic lethal interactions with otsA identified numerous genes, revealing additional potential drug targets synergistic with OtsB2 suitable for combination therapies that would minimize the emergence of resistance to OtsB2 inhibitors.


Sujet(s)
Protéines bactériennes/métabolisme , Mycobacterium tuberculosis/enzymologie , Phosphoric monoester hydrolases/métabolisme , Oses phosphates/métabolisme , Tréhalose/analogues et dérivés , Tuberculose/enzymologie , Animaux , Chromatographie sur couche mince , Modèles animaux de maladie humaine , Femelle , Analyse de profil d'expression de gènes , Techniques de knock-down de gènes , Glucosyltransferases/métabolisme , Techniques in vitro , Souris , Souris de lignée C57BL , Résonance magnétique nucléaire biomoléculaire , Réaction de polymérisation en chaine en temps réel , Tréhalose/métabolisme
17.
Sci Transl Med ; 8(362): 362ra146, 2016 10 26.
Article de Anglais | MEDLINE | ID: mdl-27797962

RÉSUMÉ

In inflammation-associated progressive neuroinflammatory disorders, such as multiple sclerosis (MS), inflammatory infiltrates containing T helper 1 (TH1) and TH17 cells cause demyelination and neuronal degeneration. Regulatory T cells (Treg) control the activation and infiltration of autoreactive T cells into the central nervous system (CNS). In MS and experimental autoimmune encephalomyelitis (EAE) in mice, Treg function is impaired. We show that a recently approved drug, Nle4-d-Phe7-α-melanocyte-stimulating hormone (NDP-MSH), induced functional Treg, resulting in amelioration of EAE progression in mice. NDP-MSH also prevented immune cell infiltration into the CNS by restoring the integrity of the blood-brain barrier. NDP-MSH exerted long-lasting neuroprotective effects in mice with EAE and prevented excitotoxic death and reestablished action potential firing in mouse and human neurons in vitro. Neuroprotection by NDP-MSH was mediated via signaling through the melanocortin-1 and orphan nuclear 4 receptors in mouse and human neurons. NDP-MSH may be of benefit in treating neuroinflammatory diseases such as relapsing-remitting MS and related disorders.


Sujet(s)
Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Récepteur de la mélanocortine de type 1/métabolisme , Hormone mélanotrope alpha/analogues et dérivés , Potentiels d'action , Animaux , Barrière hémato-encéphalique , Cellules de la moelle osseuse/métabolisme , Prolifération cellulaire , Système nerveux central/immunologie , Évolution de la maladie , Cytométrie en flux , Analyse de profil d'expression de gènes , Acide glutamique/composition chimique , Hippocampe/métabolisme , Imagerie par résonance magnétique , Souris , Souris de lignée C57BL , Neurones/métabolisme , Liaison aux protéines , Récepteur de la mélanocortine de type 1/génétique , Lymphocytes T régulateurs/cytologie , Hormone mélanotrope alpha/pharmacologie
18.
J Invest Dermatol ; 136(11): 2260-2269, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27430407

RÉSUMÉ

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor involved in adaptive cell functions, and it is highly active in the epidermis. AhR ligands can accelerate keratinocyte differentiation, but the precise role of AhR in the skin barrier is unknown. Our study showed that transepidermal water loss, a parameter of skin barrier integrity, is high in AhR-deficient mice. Experiments with conditionally AhR-deficient mouse lines identified keratinocytes as the primary cell population responsible for high transepidermal water loss. Electron microscopy showed weaker intercellular connectivity in the epidermis of keratinocytes in AhR-knockout mice, and gene expression analysis identified many barrier-associated genes as AhR targets. Moreover, AhR-deficient mice had higher interindividual differences in their microbiome. Interestingly, removing AhR ligands from the diet of wild-type mice mimicked AhR deficiency with respect to the impaired barrier; conversely, re-addition of the plant-derived ligand indole-3-carbinol rescued the barrier deficiency even in aged mice. Our results suggest that functional AhR expression is critical for skin barrier integrity and that AhR represents a molecular target for the development of therapeutic approaches for skin barrier diseases, including by dietary intervention.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , ADN/génétique , Régulation de l'expression des gènes , Kératinocytes/métabolisme , Récepteurs à hydrocarbure aromatique/génétique , Maladies de la peau/génétique , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/biosynthèse , Différenciation cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Kératinocytes/ultrastructure , Souris , Souris de lignée C57BL , Microscopie électronique , Récepteurs à hydrocarbure aromatique/biosynthèse , Maladies de la peau/métabolisme , Maladies de la peau/anatomopathologie
19.
Int J Cancer ; 139(10): 2343-52, 2016 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-27389073

RÉSUMÉ

New drugs for the treatment of multiple myeloma (MM) comprise immunomodulatory substances such as lenalidomide and related compounds. While lenalidomide has found its way into first-line treatment as well as into relapse therapy, little is known about lenalidomide effects on normal hematopoietic stem and progenitor cells (HSPCs). In this study, we investigated whether HSPCs are influenced by lenalidomide on a phenotypic, functional and gene expression level. For that purpose, samples from patients with MM were obtained who underwent equivalent first-line treatment including induction therapy, cytotoxic stem cell mobilization and high-dose melphalan therapy followed by autologous blood stem cell transplantation and a subsequent uniform lenalidomide consolidation treatment within a prospective clinical trial. We found that after six months of lenalidomide therapy, the number of CD34(+) HSPCs decreased. Additionally, lenalidomide affects the numerical composition of hematopoietic cells in the bone marrow while it does not affect long-term HSPC proliferation in vitro. We found a significant amplification of fetal hemoglobin (HbF) expression on a transcriptional level and can confirm a stimulated erythropoiesis on a phenotypic level. These effects were accompanied by silencing of the TGF-ß signaling pathway on the gene expression and protein level that is known to be amplified in active MM. However, these pleiotropic effects gave no evidence for mutagenic potential. In conclusion, lenalidomide does not exert long-term effects on proliferation of HSPCs but instead promotes erythropoiesis by shifting hemoglobin expression toward HbF and by silencing the TGF-ß signaling pathway.


Sujet(s)
Érythropoïèse/effets des médicaments et des substances chimiques , Myélome multiple/sang , Myélome multiple/traitement médicamenteux , Myélopoïèse/effets des médicaments et des substances chimiques , Thalidomide/analogues et dérivés , Inhibiteurs de l'angiogenèse/usage thérapeutique , Moelle osseuse/effets des médicaments et des substances chimiques , Chimiothérapie de consolidation , Hémoglobine foetale/métabolisme , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Humains , Lénalidomide , Adulte d'âge moyen , Thalidomide/usage thérapeutique
20.
Mol Cell Neurosci ; 75: 36-43, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-27346077

RÉSUMÉ

BACKGROUND: MicroRNAs (miRNAs) regulate gene expression in physiological as well as in pathological processes, including chronic pain. Whether deletion of a gene can affect expression of the miRNAs that associate with the deleted gene mRNA remains elusive. We investigated the effects of brain-derived neurotrophic factor (Bdnf) gene deletion on the expression of miR-1 in dorsal root ganglion (DRG) neurons and its pain-associated downstream targets heat shock protein 60 (Hsp60) and connexin 43 (Cx43) in tamoxifen-inducible conditional knockout mice, Bdnf(fl/fl); Advillin-CreER(T2) (Bdnf cKO). RESULTS: Efficient Bdnf gene deletion was confirmed in DRG of Bdnf cKO mice by Real-Time qRT-PCR and ELISA 10days after completed tamoxifen treatment. In DRG, miR-1 expression was reduced 0.44-fold (p<0.05; Real-time qRT-PCR) in Bdnf cKO compared to floxed wildtype littermate control Bdnf(fl/fl) mice (WT). While Hsp60 protein expression was increased 1.85-fold (p<0.05; Western blot analysis), expression levels of Cx43 and the miR-1-associated transcription factors MEF2a and SRF remained unchanged. When analyzing Bdnf cKO mice 32days after complete tamoxifen treatment to investigate whether observed expression alterations remain permanently, we found no significant differences between Bdnf cKO and WT mice. However, miRNA microarray analysis revealed that 167 miRNAs altered (p<0.05) in DRG of these mice following Bdnf gene deletion. CONCLUSIONS: Our results indicate that deletion of Bdnf in DRG neurons leads to a temporary dysregulation of miR-1, suggesting an impairment of a presumable feedback loop between BDNF protein and its targeting miR-1. This appears to affect its downstream protein Hsp60 and as a consequence might influence the phenotype after inducible Bdnf gene deletion. While this appears to be a MEF2a-/SRF-independent and transient effect, expression levels of various other miRNAs may remain permanently altered.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/génétique , Ganglions sensitifs des nerfs spinaux/métabolisme , microARN/génétique , Animaux , Facteur neurotrophique dérivé du cerveau/métabolisme , Chaperonine-60/génétique , Chaperonine-60/métabolisme , Connexine 43/génétique , Connexine 43/métabolisme , Femelle , Délétion de gène , Mâle , Souris , Souris de lignée C57BL , microARN/métabolisme , Protéines mitochondriales/génétique , Protéines mitochondriales/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...