Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 59
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Metab ; 6(6): 1053-1075, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38684889

RÉSUMÉ

Promoting brown adipose tissue (BAT) activity innovatively targets obesity and metabolic disease. While thermogenic activation of BAT is well understood, the rheostatic regulation of BAT to avoid excessive energy dissipation remains ill-defined. Here, we demonstrate that adenylyl cyclase 3 (AC3) is key for BAT function. We identified a cold-inducible promoter that generates a 5' truncated AC3 mRNA isoform (Adcy3-at), whose expression is driven by a cold-induced, truncated isoform of PPARGC1A (PPARGC1A-AT). Male mice lacking Adcy3-at display increased energy expenditure and are resistant to obesity and ensuing metabolic imbalances. Mouse and human AC3-AT are retained in the endoplasmic reticulum, unable to translocate to the plasma membrane and lack enzymatic activity. AC3-AT interacts with AC3 and sequesters it in the endoplasmic reticulum, reducing the pool of adenylyl cyclases available for G-protein-mediated cAMP synthesis. Thus, AC3-AT acts as a cold-induced rheostat in BAT, limiting adverse consequences of cAMP activity during chronic BAT activation.


Sujet(s)
Adenylate Cyclase , Tissu adipeux brun , Basse température , Adenylate Cyclase/métabolisme , Adenylate Cyclase/génétique , Tissu adipeux brun/métabolisme , Animaux , Souris , Mâle , Humains , Thermogenèse/génétique , Métabolisme énergétique , AMP cyclique/métabolisme , Souris knockout
2.
Sci Signal ; 17(824): eadc9662, 2024 02 20.
Article de Anglais | MEDLINE | ID: mdl-38377177

RÉSUMÉ

The IL-6-gp130-STAT3 signaling axis is a major regulator of inflammation. Activating mutations in the gene encoding gp130 and germline gain-of-function mutations in STAT3 (STAT3GOF) are associated with multi-organ autoimmunity, severe morbidity, and adverse prognosis. To dissect crucial cellular subsets and disease biology involved in activated gp130 signaling, the gp130-JAK-STAT3 axis was constitutively activated using a transgene, L-gp130, specifically targeted to T cells. Activating gp130 signaling in T cells in vivo resulted in fatal, early onset, multi-organ autoimmunity in mice that resembled human STAT3GOF disease. Female mice had more rapid disease progression than male mice. On a cellular level, gp130 signaling induced the activation and effector cell differentiation of T cells, promoted the expansion of T helper type 17 (TH17) cells, and impaired the activity of regulatory T cells. Transcriptomic profiling of CD4+ and CD8+ T cells from these mice revealed commonly dysregulated genes and a gene signature that, when applied to human transcriptomic data, improved the segregation of patients with transcriptionally diverse STAT3GOF mutations from healthy controls. The findings demonstrate that increased gp130-STAT3 signaling leads to TH17-driven autoimmunity that phenotypically resembles human STAT3GOF disease.


Sujet(s)
Auto-immunité , Lymphocytes T CD8+ , Humains , Mâle , Femelle , Souris , Animaux , Récepteur gp130 de cytokines/génétique , Récepteur gp130 de cytokines/métabolisme , Auto-immunité/génétique , Lymphocytes T CD8+/métabolisme , Transduction du signal , Inflammation , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme
3.
Vaccines (Basel) ; 10(2)2022 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-35214745

RÉSUMÉ

Erythropoiesis and megakaryo-/thrombopoiesis occur in the bone marrow proceeding from common, even bipotent, progenitor cells. Recently, we have shown that protective vaccination accelerates extramedullary hepatic erythroblastosis in response to blood-stage malaria of Plasmodium chabaudi. Here, we investigated whether protective vaccination also accelerates extramedullary hepatic megakaryo-/thrombopoiesis. Female Balb/c mice were twice vaccinated with a non-infectious vaccine before infecting with 106 P. chabaudi-parasitized erythrocytes. Using gene expression microarrays and quantitative real-time PCR, transcripts of genes known to be expressed in the bone marrow by cells of the megakaryo-/thrombocytic lineage were compared in livers of vaccination-protected and unprotected mice on days 0, 1, 4, 8, and 11 p.i. Livers of vaccination-protected mice responded with expression of megakaryo-/thrombocytic genes faster to P. chabaudi than those of unvaccinated mice, evidenced at early patency on day 4 p.i., when livers exhibited significantly higher levels of malaria-induced transcripts of the genes Selp and Pdgfb (p-values < 0.0001), Gp5 (p-value < 0.001), and Fli1, Runx1, Myb, Mpl, Gp1ba, Gp1bb, Gp6, Gp9, Pf4, and Clec1b (p-values < 0.01). Together with additionally analyzed genes known to be related to megakaryopoiesis, our data suggest that protective vaccination accelerates liver-intrinsic megakaryo-/thrombopoiesis in response to blood-stage malaria that presumably contributes to vaccination-induced survival of otherwise lethal blood-stage malaria.

4.
Nat Neurosci ; 24(7): 913-929, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-34002087

RÉSUMÉ

Pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus represent key regulators of metabolic homeostasis. Electrophysiological and single-cell sequencing experiments have revealed a remarkable degree of heterogeneity of these neurons. However, the exact molecular basis and functional consequences of this heterogeneity have not yet been addressed. Here, we have developed new mouse models in which intersectional Cre/Dre-dependent recombination allowed for successful labeling, translational profiling and functional characterization of distinct POMC neurons expressing the leptin receptor (Lepr) and glucagon like peptide 1 receptor (Glp1r). Our experiments reveal that POMCLepr+ and POMCGlp1r+ neurons represent largely nonoverlapping subpopulations with distinct basic electrophysiological properties. They exhibit a specific anatomical distribution within the arcuate nucleus and differentially express receptors for energy-state communicating hormones and neurotransmitters. Finally, we identify a differential ability of these subpopulations to suppress feeding. Collectively, we reveal a notably distinct functional microarchitecture of critical metabolism-regulatory neurons.


Sujet(s)
Comportement alimentaire/physiologie , Hypothalamus/physiologie , Neurones/physiologie , Pro-opiomélanocortine/métabolisme , Animaux , Métabolisme énergétique/physiologie , Homéostasie/physiologie , Hypothalamus/cytologie , Souris , Souris transgéniques , Neurones/cytologie
5.
Vaccines (Basel) ; 8(4)2020 Nov 13.
Article de Anglais | MEDLINE | ID: mdl-33202767

RÉSUMÉ

The role of natural killer (NK) cells in the liver as first-line post infectionem (p.i.) effectors against blood-stage malaria and their responsiveness to protective vaccination is poorly understood. Here, we investigate the effect of vaccination on NK cell-associated genes induced in the liver by blood-stage malaria of Plasmodium chabaudi. Female Balb/c mice were vaccinated at weeks 3 and 1 before being infected with 106P. chabaudi-parasitized erythrocytes. Genes preferentially expressed by NK cells were investigated in livers of vaccination-protected and non-protected mice on days 0, 1, 4, 8, and 11 p.i. using microarrays, qRT-PCR, and chromosome landscape analysis. Blood-stage malaria induces expression of specific genes in the liver at different phases of infection, i.e., Itga1 in expanding liver-resident NK (lrNK) cells, Itga2 in immigrating conventional NK (cNK) cells; Eomes and Tbx21 encoding transcription factors; Ncr1, Tnfsf10, Prf1, Gzma, Gzmb, Gzmc, Gzmm, and Gzmk encoding cytolytic effectors; natural killer gene complex (NKC)-localized genes encoding the NK cell receptors KLRG1, KLRK1, KLRAs1, 2, 5, 7, KLRD1, KLRC1, KLRC3, as well as the three receptors KLRB1A, KLRB1C, KLRB1F and their potential ligands CLEC2D and CLEC2I. Vaccination enhances this malaria-induced expression of genes, but impairs Gzmm expression, accelerates decline of Tnfsf10 and Clec2d expression, whereas it accelerates increased expression of Clec2i, taking a very similar time course as that of genes encoding plasma membrane proteins of erythroblasts, whose malaria-induced extramedullary generation in the liver is known to be accelerated by vaccination. Collectively, vaccination reshapes the response of the liver NK cell compartment to blood-stage malaria. Particularly, the malaria-induced expansion of lrNK cells peaking on day 4 p.i. is highly significantly (p < 0.0001) reduced by enhanced immigration of peripheral cNK cells, and KLRB1F:CLEC2I interactions between NK cells and erythroid cells facilitate extramedullary erythroblastosis in the liver, thus critically contributing to vaccination-induced survival of otherwise lethal blood-stage malaria of P. chabaudi.

6.
Malar J ; 19(1): 49, 2020 Jan 29.
Article de Anglais | MEDLINE | ID: mdl-31996238

RÉSUMÉ

BACKGROUND: Vaccination induces survival of otherwise lethal blood-stage infections of the experimental malaria Plasmodium chabaudi. Blood-stage malaria induces extramedullary erythropoiesis in the liver. This study investigates how vaccination affects the course of malaria-induced expression of erythrocytic genes in the liver. METHODS: Female Balb/c mice were vaccinated at week 3 and week 1 before challenging with 106 P. chabaudi-parasitized erythrocytes. The non-infectious vaccine consisted of erythrocyte ghosts isolated from P. chabaudi-infected erythrocytes. Gene expression microarrays and quantitative real-time PCR were used to compare mRNA expression of different erythrocytic genes in the liver of vaccination-protected and non-protected mice during infections on days 0, 1, 4, 8, and 11 p.i. RESULTS: Global transcriptomics analyses reveal vaccination-induced modifications of malaria-induced increases in hepatic gene expression on days 4 and 11 p.i. On these days, vaccination also alters hepatic expression of the erythropoiesis-involved genes Ermap, Kel, Rhd, Rhag, Slc4a1, Gypa, Add2, Ank1, Epb4.1, Epb4.2, Epb4.9, Spta1, Sptb, Tmod1, Ahsp, Acyp1, Gata1, Gfi1b, Tal1, Klf1, Epor, and Cldn13. In vaccination-protected mice, expression of these genes, except Epb4.1, is significantly higher on day 4 p.i. than in un-protected non-vaccinated mice, reaches maximal expression at peak parasitaemia on day 8 p.i., and is slowed down or even decreased towards the end of crisis phase on day 11 p.i.. After day 1 p.i., Epor expression takes about the same course as that of the other erythroid genes. Hepatic expression of Epo, however, is delayed in both vaccinated and non-vaccinated mice for the first 4 days p.i. and is maximal at significantly higher levels in vaccinated mice on day 8 p.i., before declining towards the end of crisis phase on day 11 p.i. CONCLUSION: The present data indicate that vaccination accelerates malaria-induced erythroblastosis in the liver for 1-2 days. This may contribute to earlier replenishment of peripheral red blood cells by liver-derived reticulocytes, which may favour final survival of otherwise lethal blood-stage malaria, since reticulocytes are not preferred as host cells by P. chabaudi.


Sujet(s)
Érythropoïèse/immunologie , Foie/anatomopathologie , Paludisme/sang , Plasmodium chabaudi/immunologie , Vaccination/effets indésirables , Animaux , Membrane érythrocytaire/immunologie , Érythropoïèse/génétique , Femelle , Foie/parasitologie , Paludisme/anatomopathologie , Vaccins contre le paludisme/effets indésirables , Souris , Souris de lignée BALB C , Analyse en composantes principales , Réaction de polymérisation en chaine en temps réel , Organismes exempts d'organismes pathogènes spécifiques , Transcriptome
7.
Leukemia ; 34(3): 771-786, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31690822

RÉSUMÉ

The proximal DNA damage response kinase ATM is frequently inactivated in human malignancies. Germline mutations in the ATM gene cause Ataxia-telangiectasia (A-T), characterized by cerebellar ataxia and cancer predisposition. Whether ATM deficiency impacts on tumor initiation or also on the maintenance of the malignant state is unclear. Here, we show that Atm reactivation in initially Atm-deficient B- and T cell lymphomas induces tumor regression. We further find a reduced T cell abundance in B cell lymphomas from Atm-defective mice and A-T patients. Using T cell-specific Atm-knockout models, as well as allogeneic transplantation experiments, we pinpoint impaired immune surveillance as a contributor to cancer predisposition and development. Moreover, we demonstrate that Atm-deficient T cells display impaired proliferation capacity upon stimulation, due to replication stress. Altogether, our data indicate that T cell-specific restoration of ATM activity or allogeneic hematopoietic stem cell transplantation may prevent lymphomagenesis in A-T patients.


Sujet(s)
Lymphomes/immunologie , Lymphocytes T/immunologie , Allèles , Animaux , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Prolifération cellulaire , Étoposide/pharmacologie , Transplantation de cellules souches hématopoïétiques , Lymphomes/métabolisme , Souris , Souris knockout , Lymphocytes T/métabolisme , Transplantation homologue , Résultat thérapeutique
8.
JCI Insight ; 4(15)2019 08 08.
Article de Anglais | MEDLINE | ID: mdl-31391340

RÉSUMÉ

Aberrant activity of the glycoprotein 130 130/JAK/STAT3 (gp130/JAK/STAT3) signaling axis is a recurrent event in inflammation and cancer. In particular, it is associated with a wide range of hematological malignancies, including multiple myeloma and leukemia. Novel targeted therapies have only been successful for some subtypes of these malignancies, underlining the need for developing robust mouse models to better dissect the role of this pathway in specific tumorigenic processes. Here, we investigated the role of selective gp130/JAK/STAT3 activation by generating a conditional mouse model. This model targeted constitutively active, cell-autonomous gp130 activity to B cells, as well as to the entire hematopoietic system. We found that regardless of the timing of activation in B cells, constitutively active gp130 signaling resulted in the formation specifically of mature B cell lymphomas and plasma cell disorders with full penetrance, only with different latencies, where infiltrating CD138+ cells were a dominant feature in every tumor. Furthermore, constitutively active gp130 signaling in all adult hematopoietic cells also led to the development specifically of largely mature, aggressive B cell cancers, again with a high penetrance of CD138+ tumors. Importantly, gp130 activity abrogated the differentiation block induced by a B cell-targeted Myc transgene and resulted in a complete penetrance of the gp130-associated, CD138+, mature B cell lymphoma phenotype. Thus, gp130 signaling selectively provides a strong growth and differentiation advantage for mature B cells and directs lymphomagenesis specifically toward terminally differentiated B cell cancers.


Sujet(s)
Lymphocytes B/immunologie , Différenciation cellulaire/immunologie , Récepteur gp130 de cytokines/métabolisme , Lymphomes/immunologie , Plasmocytome/immunologie , Transduction du signal/immunologie , Animaux , Carcinogenèse/génétique , Carcinogenèse/immunologie , Différenciation cellulaire/génétique , Récepteur gp130 de cytokines/génétique , Modèles animaux de maladie humaine , Femelle , Humains , Janus kinases/métabolisme , Activation des lymphocytes/génétique , Lymphomes/génétique , Lymphomes/anatomopathologie , Mâle , Souris , Plasmocytome/génétique , Plasmocytome/anatomopathologie , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Facteur de transcription STAT-3/métabolisme
9.
Malar J ; 17(1): 215, 2018 May 29.
Article de Anglais | MEDLINE | ID: mdl-29843710

RÉSUMÉ

BACKGROUND: The role of the liver for survival of blood-stage malaria is only poorly understood. In experimental blood-stage malaria with Plasmodium chabaudi, protective vaccination induces healing and, thus, survival of otherwise lethal infections. This model is appropriate to study the role of the liver in vaccination-induced survival of blood-stage malaria. METHODS: Female Balb/c mice were vaccinated with a non-infectious vaccine consisting of plasma membranes isolated in the form of erythrocyte ghosts from P. chabaudi-infected erythrocytes at week 3 and week 1 before infection with P. chabaudi blood-stage malaria. Gene expression microarrays and quantitative real-time PCR were used to investigate the response of the liver, in terms of expression of mRNA and long intergenic non-coding (linc)RNA, to vaccination-induced healing infections and lethal P. chabaudi malaria at early patency on day 4 post infection, when parasitized erythrocytes begin to appear in peripheral blood. RESULTS: In vaccination-induced healing infections, 23 genes were identified to be induced in the liver by > tenfold at p < 0.01. More than one-third were genes known to be involved in erythropoiesis, such as Kel, Rhag, Ahsp, Ermap, Slc4a1, Cldn13 Gata1, and Gfi1b. Another group of > tenfold expressed genes include genes involved in natural cytotoxicity, such as those encoding killer cell lectin-like receptors Klrb1a, Klrc3, Klrd1, the natural cytotoxicity-triggering receptor 1 Ncr1, as well as the granzyme B encoding Gzmb. Additionally, a series of genes involved in the control of cell cycle and mitosis were identified: Ccnb1, Cdc25c, Ckap2l were expressed > tenfold only in vaccination-protected mice, and the expression of 22 genes was at least 100% higher in vaccination-protected mice than in non-vaccinated mice. Furthermore, distinct lincRNA species were changed by > threefold in livers of vaccination-protected mice, whereas lethal malaria induced different lincRNAs. CONCLUSION: The present data suggest that protective vaccination accelerates the malaria-induced occurrence of extramedullary erythropoiesis, generation of liver-resident cytotoxic cells, and regeneration from malaria-induced injury in the liver at early patency, which may be critical for final survival of otherwise lethal blood-stage malaria of P. chabaudi.


Sujet(s)
Expression des gènes , Vaccins contre le paludisme/immunologie , Paludisme/génétique , Plasmodium chabaudi/physiologie , Animaux , Femelle , Foie/métabolisme , Foie/parasitologie , Paludisme/immunologie , Souris , Souris de lignée BALB C , Séquençage par oligonucléotides en batterie , Réaction de polymérisation en chaine en temps réel , Organismes exempts d'organismes pathogènes spécifiques
10.
Parasitol Res ; 117(4): 1115-1129, 2018 Apr.
Article de Anglais | MEDLINE | ID: mdl-29404749

RÉSUMÉ

Current knowledge about liver responses to blood-stage malaria and their modulation by vaccination is still unclear. This study investigated effects of protective vaccination on liver gene and lincRNA expression of Balb/c mice at early prepatency of Plasmodium chabaudi blood-stage malaria. When a blood-stage vaccine was used to induce > 80% survival of otherwise lethal malaria, significant differences (p < 0.01) were detectable in global liver gene expression between vaccination-protected (potentially surviving) and non-protected non-vaccinated mice on day 1 p.i.. In the livers of protected mice, gene expression microarrays identified 224 and 419 genes, whose expression was up- and downregulated by > 3-fold, respectively. There were 24 genes upregulated by > 10-fold, including 10 IFN-inducible genes encompassing GTPases Irgm1, 2, and 3, and guanylate-binding protein Gbp11, the IL-1 decoy receptors Il1f9 and Il1ra1, the Il6 gene, and the gene for facilitated glucose transportation. Moreover, the IL-18 decoy receptor gene Il18bp, Gzmb, the genes Lif and Osmr encoding proteins of the IL-6 family, and the taurine transporter gene Slc6a6 were expressed > 3-fold in vaccinated mice. The genes Gbp10, 6, 4 were expressed by > 50% in vaccination-protected than in non-vaccinated mice. In addition, 43 lincRNA species were up- and 36 downregulated. Our data suggested novel regulatory elements of potential anti-malaria activity activated by protective vaccination in the liver, evidenced in response to early prepatent infections in vaccination-protected mice of otherwise lethal blood-stage malaria of P. chabaudi.


Sujet(s)
Régulation de l'expression des gènes/génétique , Foie/métabolisme , Vaccins contre le paludisme/immunologie , Paludisme/prévention et contrôle , Plasmodium chabaudi/immunologie , ARN long non codant/biosynthèse , Animaux , Régulation négative/génétique , Femelle , Expression des gènes , Foie/parasitologie , Paludisme/parasitologie , Souris , Souris de lignée BALB C , Analyse sur microréseau , ARN long non codant/génétique , Régulation positive/génétique , Vaccination
11.
Parasitol Res ; 116(5): 1463-1477, 2017 May.
Article de Anglais | MEDLINE | ID: mdl-28315013

RÉSUMÉ

Epigenetic mechanisms such as DNA methylation are increasingly recognized to be critical for vaccination efficacy and outcome of different infectious diseases, but corresponding information is scarcely available for host defense against malaria. In the experimental blood-stage malaria Plasmodium chabaudi, we investigate the possible effects of a blood-stage vaccine on DNA methylation of gene promoters in the liver, known as effector against blood-stage malaria, using DNA methylation microarrays. Naturally susceptible Balb/c mice acquire, by protective vaccination, the potency to survive P. chabaudi malaria and, concomitantly, modifications of constitutive DNA methylation of promoters of numerous genes in the liver; specifically, promoters of 256 genes are hyper(=up)- and 345 genes are hypo(=down)-methylated (p < 0.05). Protective vaccination also leads to changes in promoter DNA methylation upon challenge with P. chabaudi at peak parasitemia on day 8 post infection (p.i.), when 571 and 1013 gene promoters are up- and down-methylated, respectively, in relation to constitutive DNA methylation (p < 0.05). Gene set enrichment analyses reveal that both vaccination and P. chabaudi infections mainly modify promoters of those genes which are most statistically enriched with functions relating to regulation of transcription. Genes with down-methylated promoters encompass those encoding CX3CL1, GP130, and GATA2, known to be involved in monocyte recruitment, IL-6 trans-signaling, and onset of erythropoiesis, respectively. Our data suggest that vaccination may epigenetically improve parts of several effector functions of the liver against blood-stage malaria, as, e.g., recruitment of monocyte/macrophage to the liver accelerated liver regeneration and extramedullary hepatic erythropoiesis, thus leading to self-healing of otherwise lethal P. chabaudi blood-stage malaria.


Sujet(s)
Méthylation de l'ADN/génétique , Foie/métabolisme , Macrophages/immunologie , Vaccins contre le paludisme/immunologie , Paludisme/immunologie , Monocytes/immunologie , Plasmodium chabaudi/immunologie , Animaux , Chimiokine CX3CL1/biosynthèse , Chimiokine CX3CL1/génétique , Récepteur gp130 de cytokines/biosynthèse , Récepteur gp130 de cytokines/génétique , Épigenèse génétique , Femelle , Facteur de transcription GATA-2/biosynthèse , Facteur de transcription GATA-2/génétique , Interleukine-6/génétique , Paludisme/parasitologie , Paludisme/prévention et contrôle , Souris , Souris de lignée BALB C , Séquençage par oligonucléotides en batterie , Parasitémie/immunologie , Régions promotrices (génétique)/génétique , Vaccination
12.
Front Microbiol ; 7: 1087, 2016.
Article de Anglais | MEDLINE | ID: mdl-27471498

RÉSUMÉ

Protective vaccination induces self-healing of otherwise fatal blood-stage malaria of Plasmodium chabaudi in female Balb/c mice. To trace processes critically involved in self-healing, the liver, an effector against blood-stage malaria, is analyzed for possible changes of its transcriptome in vaccination-protected in comparison to non-protected mice toward the end of the crisis phase. Gene expression microarray analyses reveal that vaccination does not affect constitutive expression of mRNA and lincRNA. However, malaria induces significant (p < 0.01) differences in hepatic gene and lincRNA expression in vaccination-protected vs. non-vaccinated mice toward the end of crisis phase. In vaccination-protected mice, infections induce up-regulations of 276 genes and 40 lincRNAs and down-regulations of 200 genes and 43 lincRNAs, respectively, by >3-fold as compared to the corresponding constitutive expressions. Massive up-regulations, partly by >100-fold, are found for genes as RhD, Add2, Ank1, Ermap, and Slc4a, which encode proteins of erythrocytic surface membranes, and as Gata1 and Gfi1b, which encode transcription factors involved in erythrocytic development. Also, Cldn13 previously predicted to be expressed on erythroblast surfaces is up-regulated by >200-fold, though claudins are known as main constituents of tight junctions acting as paracellular barriers between epithelial cells. Other genes are up-regulated by <100- and >10-fold, which can be subgrouped in genes encoding proteins known to be involved in mitosis, in cell cycle regulation, and in DNA repair. Our data suggest that protective vaccination enables the liver to respond to P. chabaudi infections with accelerated regeneration and extramedullary erythropoiesis during crisis, which contributes to survival of otherwise lethal blood-stage malaria.

13.
Parasitol Res ; 115(5): 1835-43, 2016 May.
Article de Anglais | MEDLINE | ID: mdl-26809341

RÉSUMÉ

Protective vaccination induces self-healing of otherwise lethal blood-stage infections of Plasmodium chabaudi malaria. Here, we investigate mRNA expression patterns of all 12 members of the Toll-like receptor (Tlr) gene family in the liver, a major effector organ against blood-stage malaria, during lethal and vaccination-induced self-healing infections of P. chabaudi in female Balb/c mice. Gene expression microarrays reveal that all 12 Tlr genes are constitutively expressed, though at varying levels, and specifically respond to infection. Protective vaccination does not affect constitutive expression of any of the 12 Tlr genes but leads to differential expression (p < 0.05) of seven Tlrs (1, 2, 4, 7, 8, 12, and 13) in response to malaria. Quantitative PCR substantiates differential expression at p < 0.01. There is an increased expression of Tlr2 by approximately five-fold on day 1 post-infection (p.i.) and Tlr1 by approximately threefold on day 4 p.i.. At peak parasitemia on day 8 p.i., none of the 12 Tlrs display any differential expression. After peak parasitemia, towards the end of the crisis phase on day 11 p.i., expression of Tlrs 1, 4, and 12 is increased by approximately four-, two-, and three-fold, respectively, and that of Tlr7 is decreased by approximately two-fold. Collectively, our data suggest that though all 12 members of the Tlr gene family are specifically responsive to malaria in the liver, not only Tlr2 at the early stage of infection but also the Tlrs 1, 4, 7, and 12 towards the end of crisis phase are critical for vaccination-induced resolution and survival of otherwise lethal blood-stage malaria.


Sujet(s)
Foie/métabolisme , Vaccins contre le paludisme/immunologie , Paludisme/prévention et contrôle , Plasmodium chabaudi , Récepteurs de type Toll/métabolisme , Animaux , Femelle , Régulation de l'expression des gènes , Foie/immunologie , Paludisme/immunologie , Paludisme/métabolisme , Paludisme/parasitologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Analyse sur microréseau , Parasitémie , Récepteurs de type Toll/génétique , Vaccination
14.
Front Microbiol ; 7: 2155, 2016.
Article de Anglais | MEDLINE | ID: mdl-28123381

RÉSUMÉ

MicroRNAs are increasingly recognized as epigenetic regulators for outcome of diverse infectious diseases and vaccination efficacy, but little information referring to this exists for malaria. This study investigates possible effects of both protective vaccination and P. chabaudi malaria on the miRNome of the liver as an effector against blood-stage malaria using miRNA microarrays and quantitative PCR. Plasmodium chabaudi blood-stage malaria takes a lethal outcome in female Balb/c mice, but a self-healing course after immunization with a non-infectious blood-stage vaccine. The liver robustly expresses 71 miRNA species at varying levels, among which 65 miRNA species respond to malaria evidenced as steadily increasing or decreasing expressions reaching highest or lowest levels toward the end of the crisis phase on day 11 p.i. in lethal malaria. Protective vaccination does not affect constitutive miRNA expression, but leads to significant (p < 0.05) changes in the expression of 41 miRNA species, however evidenced only during crisis. In vaccination-induced self-healing infections, 18 miRNA-species are up- and 14 miRNA-species are down-regulated by more than 50% during crisis in relation to non-vaccinated mice. Vaccination-induced self-healing and survival of otherwise lethal infections of P. chabaudi activate epigenetic miRNA-regulated remodeling processes in the liver manifesting themselves during crisis. Especially, liver regeneration is accelerated as suggested by upregulation of let-7a-5p, let-7b-5p, let-7c-5p, let-7d-5p, let-7f-5p, let-7g-5p, let-7i-5p, miR-26a, miR-122-5p, miR30a, miR27a, and mir-29a, whereas the up-regulated expression of miR-142-3p by more than 100% is compatible with the view of enhanced hepatic erythropoiesis, possibly at expense of megakaryopoiesis, during crisis of P. chabaudi blood-stage malaria.

15.
Parasitol Res ; 114(4): 1581-93, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25663104

RÉSUMÉ

Plant-based natural products are promising sources for identifying novel agents with potential anti-Eimeria activity. This study explores possible effects of berberine on Eimeria papillata infections in the jejunum of male Swiss albino mice. Berberine chloride, when daily administered to mice during infection, impairs intracellular development and multiplication of E. papillata, evidenced as 60% reduction of maximal fecal output of oocysts on day 5 p.i. Concomitantly, berberine attenuates the inflammatory response, evidenced as decreased messenger RNA (mRNA) expression of IL-1ß, IL-6, TNFα, IFNγ, and iNOS, as well as the oxidative stress response, evidenced as impaired increase in malondialdehyde, nitrate, and H2O2 and as prevented decrease in glutathione and catalase activity. Berberine also alters gene expression in the infected jejunum. On day 5 p.i., mRNA expression of 29 genes with annotated functions is more than 10-fold upregulated and that of 14 genes downregulated. Berberine downregulates the genes Xaf1, Itgb3bp, and Faim3 involved in apoptotic processes and upregulates genes involved in innate immune responses, as e.g., Colec11, Saa2, Klra8, Clec1b, and Crtam, especially the genes Cpa3, Fcer1a, and Mcpt1, Mcpt2, and Mcpt4 involved in mast cell activity. Additionally, 18 noncoding lincRNA species are differentially expressed more than 10-fold under berberine. Our data suggest that berberine induces hosts to exert anti-Eimeria activity by attenuating the inflammatory and oxidative stress response, by impairing apoptotic processes, and by activating local innate immune responses and epigenetic mechanisms in the host jejunum. Berberine has the potential as an anti-Eimeria food additive in animal farming.


Sujet(s)
Antiprotozoaires/pharmacologie , Berbérine/pharmacologie , Coccidiose/traitement médicamenteux , Coccidiose/génétique , Eimeria/effets des médicaments et des substances chimiques , Jéjunum/parasitologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Coccidiose/métabolisme , Coccidiose/parasitologie , Eimeria/physiologie , Expression des gènes/effets des médicaments et des substances chimiques , Glutathion/métabolisme , Humains , Interleukine-6/génétique , Interleukine-6/métabolisme , Jéjunum/métabolisme , Mâle , Malonaldéhyde/métabolisme , Souris , Stress oxydatif/effets des médicaments et des substances chimiques
16.
Adv Nutr ; 6(1): 73-82, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25593145

RÉSUMÉ

Viral and bacterial infections are often associated with deficiencies in macronutrients and micronutrients, including the essential trace element selenium. In selenium deficiency, benign strains of Coxsackie and influenza viruses can mutate to highly pathogenic strains. Dietary supplementation to provide adequate or supranutritional selenium supply has been proposed to confer health benefits for patients suffering from some viral diseases, most notably with respect to HIV and influenza A virus (IAV) infections. In addition, selenium-containing multimicronutrient supplements improved several clinical and lifestyle variables in patients coinfected with HIV and Mycobacterium tuberculosis. Selenium status may affect the function of cells of both adaptive and innate immunity. Supranutritional selenium promotes proliferation and favors differentiation of naive CD4-positive T lymphocytes toward T helper 1 cells, thus supporting the acute cellular immune response, whereas excessive activation of the immune system and ensuing host tissue damage are counteracted through directing macrophages toward the M2 phenotype. This review provides an up-to-date overview on selenium in infectious diseases caused by viruses (e.g., HIV, IAV, hepatitis C virus, poliovirus, West Nile virus) and bacteria (e.g., M. tuberculosis, Helicobacter pylori). Data from epidemiologic studies and intervention trials, with selenium alone or in combination with other micronutrients, and animal experiments are discussed against the background of dietary selenium requirements to alter immune functions.


Sujet(s)
Adjuvants immunologiques/usage thérapeutique , Infections bactériennes/traitement médicamenteux , Compléments alimentaires , Système immunitaire/effets des médicaments et des substances chimiques , Sélénium/usage thérapeutique , Oligoéléments/usage thérapeutique , Maladies virales/traitement médicamenteux , Adjuvants immunologiques/pharmacologie , Animaux , Infections bactériennes/microbiologie , Humains , Sélénium/déficit , Sélénium/pharmacologie , Oligoéléments/déficit , Oligoéléments/pharmacologie , Maladies virales/virologie
17.
J Steroid Biochem Mol Biol ; 145: 121-30, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25448745

RÉSUMÉ

Testosterone (T) is known to masculinize the female phenotype of the liver, evidenced as up- and down-regulated expressions of male- and female-predominant genes, respectively, involved in hepatic metabolism. This study is aimed at identifying epigenetic modifications of promoters of these differently expressed genes in the liver after masculinization by T of adult female C57BL/6 mice using methylated DNA immunoprecipitation and NimbleGen microarrays. Among the 17,354 promoters examined, 82 promoters in the liver have been identified to be significantly changed by T (p<0.05), with 47 and 35 promoters exhibiting increased and decreased DNA methylation, respectively. Most of these promoters display the changes of DNA methylation in their Ups-regions, which are between +500 and +2000 bp upstream from the transcription start site (TSS) of the genes. Less T-induced modifications have been detected in the Cor-regions of the promoters, i.e., +500 to -500 bp around the TSS. Only 13 and 7 Cor-promoters are hyper- and hypo-methylated, respectively, among which are 10 hyper- and 5 hypo-methylated promoters of genes with annotated functions. Surprisingly, the promoters are largely unmethylated in those genes whose expression has been previously found to be permanently deregulated by T in the liver, as e.g. the T-upregulated male-predominant genes Cyp7b1, Cyp2d9, Cyp4a10, Ugt2b1, Ugt2b38, Hsd3b5, Slco1a1 as well as the T-downregulated female-predominant genes Cyp2b9, Cyp2b13, Cyp3a41, Cyp3a44, Fmo3, Sult2a2, respectively. Though methylatable, the promoter DNA of Ar, Esr1, and Esr2 remained unaffected by T. However, T decreases DNA-methylation of the Cor-promoter region of Ddc encoding the AR-coactivator dopa decarboxylase. Among the identified 15 Cor-promoters of genes with annotated functions are also those of Defb43, Cst11, and Sele involved in innate immunity. Our data support the view that T may exert long-lasting epigenetic effects on functions of the liver-inherent immune system.


Sujet(s)
Méthylation de l'ADN/effets des médicaments et des substances chimiques , Épigenèse génétique/effets des médicaments et des substances chimiques , Régions promotrices (génétique) , Testostérone/pharmacologie , Animaux , Ilots CpG , Femelle , Immunoprécipitation , Foie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Séquençage par oligonucléotides en batterie , Phénotype
18.
Front Microbiol ; 5: 559, 2014.
Article de Anglais | MEDLINE | ID: mdl-25408684

RÉSUMÉ

The liver is well known as that organ which is obligately required for the intrahepatocyte development of the pre-erythrocytic stages of the malaria-causative agent Plasmodium. However, largely neglected is the fact that the liver is also a central player of the host defense against the morbidity- and mortality-causing blood stages of the malaria parasites. Indeed, the liver is equipped with a unique immune system that acts locally, however, with systemic impact. Its main "antipodal" functions are to recognize and to generate effective immunoreactivity against pathogens on the one hand, and to generate tolerance to avoid immunoreactivity with "self" and harmless substances as dietary compounds on the other hand. This review provides an introductory survey of the liver-inherent immune system: its pathogen recognition receptors including Toll-like receptors (TLRs) and its major cell constituents with their different facilities to fight and eliminate pathogens. Then, evidence is presented that the liver is also an essential organ to overcome blood-stage malaria. Finally, we discuss effector responses of the liver-inherent immune system directed against blood-stage malaria: activation of TLRs, acute phase response, phagocytic activity, cytokine-mediated pro- and anti-inflammatory responses, generation of "protective" autoimmunity by extrathymic T cells and B-1 cells, and T cell-mediated repair of liver injuries mainly produced by malaria-induced overreactions of the liver-inherent immune system.

19.
Parasitol Res ; 113(10): 3547-56, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25185667

RÉSUMÉ

Eimeriosis, a widespread infectious disease of livestock, is caused by coccidian protozoans of the genus Eimeria. These obligate intracellular parasites strike the digestive tract of their hosts and give rise to enormous economic losses, particularly in poultry, ruminants including cattle, and rabbit farming. Vaccination, though a rational prophylactic measure, has not yet been as successful as initially thought. Numerous broad-spectrum anti-coccidial drugs are currently in use for treatment and prophylactic control of eimeriosis. However, increasing concerns about parasite resistance, consumer health, and environmental safety of the commercial drugs warrant efforts to search for novel agents with anti-Eimeria activity. This review summarizes current approaches to prevent and treat eimeriosis such as vaccination and commercial drugs, as well as recent attempts to use dietary antioxidants as novel anti-Eimeria agents. In particular, the trace elements selenium and zinc, the vitamins A and E, and natural products extracted from garlic, barberry, pomegranate, sweet wormwood, and other plants are discussed. Several of these novel anti-Eimeria agents exhibit a protective role against oxidative stress that occurs not only in the intestine of Eimeria-infected animals, but also in their non-parasitized tissues, in particular, in the first-pass organ liver. Currently, it appears to be promising to identify safe combinations of low-cost natural products with high anti-Eimeria efficacy for a potential use as feed supplementation in animal farming.


Sujet(s)
Produits biologiques/pharmacologie , Coccidiose/médecine vétérinaire , Eimeria/effets des médicaments et des substances chimiques , Maladies de la volaille/traitement médicamenteux , Oligoéléments/pharmacologie , Vitamines/pharmacologie , Animaux , Antioxydants/usage thérapeutique , Produits biologiques/composition chimique , Bovins , Coccidiose/traitement médicamenteux , Coccidiose/parasitologie , Coccidiose/prévention et contrôle , Mâle , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Volaille , Maladies de la volaille/parasitologie , Maladies de la volaille/prévention et contrôle , Lapins , Vaccination/médecine vétérinaire
20.
Parasitol Res ; 113(10): 3609-20, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25056943

RÉSUMÉ

Testosterone (T) is known to induce persistent susceptibility to Plasmodium chabaudi malaria. Pathogens recognizing Toll-like receptors (TLRs), though potentially important against malaria, have not yet been examined for their T-sensitivity. Here, we investigate effects of T and P. chabaudi on mRNA expression and promoter DNA methylation of Tlr1-9 genes in the liver of female C57BL/6 mice. These are treated with T or vehicle for 3 weeks, and then treatment is discontinued for 12 weeks, before challenging with P. chabaudi for 8 days. Our data reveal that T induces a 9.1-fold downregulation of Tlr6 mRNA and 6.3-fold upregulation of Tlr8 mRNA. Blood-stage infections induce significant increases in mRNA expression of Tlr1, 2, 4, 6, 7, and 8 varying between 2.5-fold and 21-fold in control mice. In T-pretreated mice, these Tlr genes are also significantly responsive to infections. However, the malaria-induced upregulations of the relative mRNA expressions of Tlr6 and Tlr8 are 5.6-fold higher and 6.5-fold lower in T-pretreated mice than in control mice. Infections induce a massive DNA down-methylation of the Tlr6 gene promoter in control mice, which is still more pronounced in T-pretreated mice, while significant changes are not detectable for the DNA methylation status of the Tlr8 promoter. Our data support the view that hepatic expression of Tlr6, but not that of Tlr8 is epigenetically controlled, and that the dysregulations of Tlr6 and Tlr8 critically contribute to T-induced persistent susceptibility to P. chabaudi malaria, possibly by dys-balancing responses of TLR6-mediated pathogen recognition and TLR8-mediated generation of anti-malaria "protective" autoimmunity.


Sujet(s)
Paludisme/métabolisme , Plasmodium chabaudi/immunologie , Régions promotrices (génétique)/génétique , Testostérone/pharmacologie , Récepteur de type Toll-6/génétique , Récepteur de type Toll-8/génétique , Animaux , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Paludisme/immunologie , Souris , Souris de lignée C57BL , Séquençage par oligonucléotides en batterie , ARN messager/génétique , ARN messager/métabolisme , Organismes exempts d'organismes pathogènes spécifiques , Régulation positive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE