Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 12: 839216, 2022.
Article in English | MEDLINE | ID: mdl-35967864

ABSTRACT

In human cutaneous leishmaniasis (HCL) caused by Leishmania (L.) major, the cutaneous lesions heal spontaneously and induce a Th1-type immunity that confers solid protection against reinfection. The same holds true for the experimental leishmaniasis induced by L. major in C57BL/6 mice where residual parasites persist after spontaneous clinical cure and induce sustainable memory immune responses and resistance to reinfection. Whether residual parasites also persist in scars of cured HCL caused by L. major is still unknown. Cutaneous scars from 53 volunteers with healed HCL caused by L. major were biopsied and the tissue sample homogenates were analyzed for residual parasites by four methods: i) microscope detection of amastigotes, ii) parasite culture by inoculation on biphasic medium, iii) inoculation of tissue exctracts to the footpad of BALB/c mice, an inbred strain highly susceptible to L. major, and iv) amplification of parasite kDNA by a highly sensitive real-time PCR (RT-PCR). Our results show that the scars of healed lesions of HCL caused by L. major do not contain detectable residual parasites, suggesting that this form likely induces a sterile cure at least within the scars. This feature contrasts with other Leishmania species causing chronic, diffuse, or recidivating forms of leishmaniasis where parasites do persist in healed lesions. The possibility that alternative mechanisms to parasite persistence are needed to boost and maintain long-term immunity to L. major, should be taken into consideration in vaccine development against L. major infection.


Subject(s)
Leishmania major , Leishmaniasis, Cutaneous , Parasites , Animals , Cicatrix , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Reinfection
2.
PLoS Genet ; 18(1): e1010010, 2022 01.
Article in English | MEDLINE | ID: mdl-35041643

ABSTRACT

Calcific aortic valve disease (CAVD) is characterized by a fibrocalcific process. The regulatory mechanisms that drive the fibrotic response in the aortic valve (AV) are poorly understood. Long noncoding RNAs derived from super-enhancers (lncRNA-SE) control gene expression and cell fate. Herein, multidimensional profiling including chromatin immunoprecipitation and sequencing, transposase-accessible chromatin sequencing, genome-wide 3D chromatin contacts of enhancer-promoter identified LINC01013 as an overexpressed lncRNA-SE during CAVD. LINC01013 is within a loop anchor, which has contact with the promoter of CCN2 (CTGF) located at ~180 kb upstream. Investigation showed that LINC01013 acts as a decoy factor for the negative transcription elongation factor E (NELF-E), whereby it controls the expression of CCN2. LINC01013-CCN2 is part of a transforming growth factor beta 1 (TGFB1) network and exerts a control over fibrogenesis. These findings illustrate a novel mechanism whereby a dysregulated lncRNA-SE controls, through a looping process, the expression of CCN2 and fibrogenesis of the AV.


Subject(s)
Aortic Valve Stenosis/genetics , Aortic Valve/pathology , Calcinosis/genetics , Chromatin/metabolism , Connective Tissue Growth Factor/genetics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Aged , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Calcinosis/metabolism , Chromatin Immunoprecipitation Sequencing , Enhancer Elements, Genetic , Female , Humans , Male , Middle Aged , Promoter Regions, Genetic , Signal Transduction , Transforming Growth Factor beta1/metabolism , Up-Regulation
3.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33803922

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by a sustained elevation of pulmonary artery (PA) pressure, right ventricular failure, and premature death. Enhanced proliferation and resistance to apoptosis (as seen in cancer cells) of PA smooth muscle cells (PASMCs) is a major pathological hallmark contributing to pulmonary vascular remodeling in PAH, for which current therapies have only limited effects. Emerging evidence points toward a critical role for Enhancer of Zeste Homolog 2 (EZH2) in cancer cell proliferation and survival. However, its role in PAH remains largely unknown. The aim of this study was to determine whether EZH2 represents a new factor critically involved in the abnormal phenotype of PAH-PASMCs. We found that EZH2 is overexpressed in human lung tissues and isolated PASMCs from PAH patients compared to controls as well as in two animal models mimicking the disease. Through loss- and gain-of-function approaches, we showed that EZH2 promotes PAH-PASMC proliferation and survival. By combining quantitative transcriptomic and proteomic approaches in PAH-PASMCs subjected or not to EZH2 knockdown, we found that inhibition of EZH2 downregulates many factors involved in cell-cycle progression, including E2F targets, and contributes to maintain energy production. Notably, we found that EZH2 promotes expression of several nuclear-encoded components of the mitochondrial translation machinery and tricarboxylic acid cycle genes. Overall, this study provides evidence that, by overexpressing EZH2, PAH-PASMCs remove the physiological breaks that normally restrain their proliferation and susceptibility to apoptosis and suggests that EZH2 or downstream factors may serve as therapeutic targets to combat pulmonary vascular remodeling.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Proteome/genetics , Pulmonary Arterial Hypertension/genetics , Transcriptome/genetics , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Citric Acid Cycle/genetics , Epigenesis, Genetic/genetics , Female , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Lung/metabolism , Lung/pathology , Male , Middle Aged , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/growth & development , Pulmonary Artery/pathology , Rats
4.
iScience ; 24(3): 102241, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33748722

ABSTRACT

Genome-wide association studies for calcific aortic valve stenosis (CAVS) previously reported strong signal for noncoding variants at 1p21.2. Previous study using Mendelian randomization suggested that the locus controls the expression of PALMD encoding Palmdelphin (PALMD). However, the molecular regulation at the locus and the impact of PALMD on the biology of the aortic valve is presently unknown. 3D genetic mapping and CRISPR activation identified rs6702619 as being located in a distant-acting enhancer, which controls the expression of PALMD. DNA-binding assay showed that the risk variant modified the DNA shape, which prevented the recruitment of NFATC2 and lowered the expression of PALMD. In co-expression network analysis, a module encompassing PALMD was enriched in actin-based process. Mass spectrometry and functional assessment showed that PALMD is a regulator of actin polymerization. In turn, lower level of PALMD promoted the activation of myocardin-related transcription factor and fibrosis, a key pathobiological process underpinning CAVS.

5.
Nucleic Acids Res ; 47(16): 8424-8438, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31251802

ABSTRACT

ENPP2, which encodes for the enzyme autotaxin (ATX), is overexpressed during chronic inflammatory diseases and various cancers. However, the molecular mechanism involved in the ENPP2 transcription remains elusive. Here, in HEK 293T cells, we demonstrated that lipopolysaccharide (LPS) increased the transcription process at ENPP2 locus through a NF-кB pathway and a reduction of H3K27me3 level, a histone repressive mark, by the demethylase UTX. Simultaneously, the H3K27me3 demethylase JMJD3/KDM6B was recruited to the transcription start site (TSS), within the gene body and controlled the expression of ENPP2 in a non-enzymatic manner. Mass spectrometry data revealed a novel interaction for JMJD3 with DDX21, a RNA helicase that unwinds R-loops created by nascent transcript and DNA template. Upon LPS treatment, JMJD3 is necessary for DDX21 recruitment at ENPP2 locus allowing the resolution of aberrant R-loops. CRISPR-Cas9-mediated deletion of a distant-acting enhancer decreased the expression of ENPP2 and lowered the recruitment of JMJD3-DDX21 complex at TSS and its progression through the gene body. Taken together, these findings revealed that enhancer-mediated enrichment of novel JMJD3-DDX21 interaction at ENPP2 locus is necessary for nascent transcript synthesis via the resolution of aberrant R-loops formation in response to inflammatory stimulus.


Subject(s)
DEAD-box RNA Helicases/genetics , DNA/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Phosphoric Diester Hydrolases/genetics , RNA, Messenger/genetics , Transcription, Genetic/drug effects , CRISPR-Cas Systems , DEAD-box RNA Helicases/metabolism , DNA/chemistry , DNA/metabolism , Enhancer Elements, Genetic , Gene Editing/methods , Gene Expression Regulation , HEK293 Cells , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Inflammation , Jumonji Domain-Containing Histone Demethylases/metabolism , Lipopolysaccharides/pharmacology , Models, Biological , NF-kappa B/genetics , NF-kappa B/metabolism , Nucleic Acid Conformation , Phosphoric Diester Hydrolases/metabolism , Protein Binding , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , Signal Transduction , Transcription Initiation Site
6.
Eur Heart J ; 40(17): 1362-1373, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30395215

ABSTRACT

AIMS: Calcific aortic valve stenosis (CAVS) is characterized by a fibrocalcific process. Studies have shown an association between CAVS and the activation of platelets. It is believed that shear stress associated with CAVS promotes the activation of platelets. However, whether platelets actively participate to the mineralization of the aortic valve (AV) and the progression of CAVS is presently unknown. To identify the role of platelets into the pathobiology of CAVS. METHODS AND RESULTS: Explanted control non-mineralized and mineralized AVs were examined by scanning electron microscope (SEM) for the presence of activated platelets. In-depth functional assays were carried out with isolated human valve interstitial cells (VICs) and platelets as well as in LDLR-/- apoB100/100 IGFII (IGFII) mice. Scanning electron microscope and immunogold markings for glycoprotein IIb/IIIa (GPIIb/IIIa) revealed the presence of platelet aggregates with fibrin in endothelium-denuded areas of CAVS. In isolated VICs, collagen-activated platelets induced an osteogenic programme. Platelet-derived adenosine diphosphate induced the release of autotaxin (ATX) by VICs. The binding of ATX to GPIIb/IIIa of platelets generated lysophosphatidic acid (LysoPA) with pro-osteogenic properties. In IGFII mice with CAVS, platelet aggregates were found at the surface of AVs. Administration of activated platelets to IGFII mice accelerated the development of CAVS by 2.1-fold, whereas a treatment with Ki16425, an antagonist of LysoPA receptors, prevented platelet-induced mineralization of the AV and the progression of CAVS. CONCLUSIONS: These findings suggest a novel role for platelets in the progression of CAVS.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Blood Platelets/metabolism , Calcinosis/metabolism , Osteogenesis , Animals , Aortic Valve/metabolism , Aortic Valve/ultrastructure , Apolipoprotein B-100/metabolism , Disease Progression , Humans , Integrin beta3/metabolism , Lysophospholipids/metabolism , Mice , Microscopy, Electron, Scanning/methods , Phosphoric Diester Hydrolases/metabolism , Platelet Membrane Glycoprotein IIb/metabolism
7.
Clin Chim Acta ; 485: 173-177, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29966619

ABSTRACT

INTRODUCTION: Aortic valve bioprostheses, which do not mandate chronic anticoagulation, are prone to structural valve degeneration (SVD). The processes involved in SVD are likely multifactorial. We hypothesized that inflammation and macrophage activation could be involved in SVD. METHODS: In 203 patients with an aortic valve bioprosthesis, we evaluated the association between the macrophage activation marker soluble CD14 (sCD14) and SVD. RESULTS: After a mean follow-up of 8 ±â€¯3 years, 42 (21%) patients developed SVD. Patients with SVD had higher peak (44 ±â€¯13 mmHg vs. 25 ±â€¯12 mmHg, p < .0001) and mean (24 ±â€¯7 mmHg vs. 12 ±â€¯5 mmHg, p < .0001) transprosthetic gradients. On univariable analysis, low-density lipoprotein cholesterol (LDL) and sCD14 were associated with SVD. After correction for covariates, sCD14 (OR: 1.12, 95%CI: 1.02-1.23, p = .01) remained independently associated with SVD. In turn, sCD14 was associated with the HOMA index and high-density lipoprotein (HDL) level. Patients with a metabolic syndrome (MetS) had higher level of sCD14. In a model corrected for age, sex, HOMA and HDL, the MetS remained independently associated with sCD14 levels (ß = 0.65, SE = 0.30, p = .03). CONCLUSION: Circulating level of sCD14 is an independent predictor of SVD. In turn, patients with MetS have higher sCD14 levels.


Subject(s)
Bioprosthesis , CD4 Antigens/analysis , Heart Valve Prosthesis , Metabolic Syndrome/diagnosis , Aged , Biomarkers/analysis , Cholesterol, LDL/analysis , Female , Humans , Male , Recombinant Proteins/analysis
8.
Cardiovasc Res ; 114(11): 1525-1535, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29726894

ABSTRACT

Aims: Calcific aortic valve disease (CAVD) is characterized by the osteogenic transition of valve interstitial cells (VICs). In CAVD, lysophosphatidic acid (LysoPA), a lipid mediator with potent osteogenic activity, is produced in the aortic valve (AV) and is degraded by membrane-associated phospholipid phosphatases (PLPPs). We thus hypothesized that a dysregulation of PLPPs could participate to the osteogenic reprograming of VICs during CAVD. Methods and results: The expression of PLPPs was examined in human control and mineralized AVs and comprehensive analyses were performed to document the gene regulation and impact of PLPPs on the osteogenic transition of VICs. We found that PLPP3 gene and enzymatic activity were downregulated in mineralized AVs. Multidimensional gene profiling in 21 human AVs showed that expression of PLPP3 was inversely correlated with the level of 5-methylcytosine (5meC) located in an intronic mammalian interspersed repeat (MIR) element. Bisulphite pyrosequencing in a larger series of 67 AVs confirmed that 5meC in intron 1 was increased by 2.2-fold in CAVD compared with control AVs. In isolated cells, epigenome editing with clustered regularly interspersed short palindromic repeats-Cas9 system containing a deficient Cas9 fused with DNA methyltransferase (dCas9-DNMT) was used to increase 5meC in the intronic enhancer and showed that it reduced significantly the expression of PLPP3. Knockdown experiments showed that lower expression of PLPP3 in VICs promotes an osteogenic programme. Conclusions: DNA methylation of a MIR-based enhancer downregulates the expression of PLPP3 and promotes the mineralization of the AV.


Subject(s)
Aortic Valve Stenosis/genetics , Aortic Valve/enzymology , Aortic Valve/pathology , Calcinosis/genetics , DNA Methylation , DNA Transposable Elements , Osteogenesis/genetics , Phosphatidate Phosphatase/genetics , Promoter Regions, Genetic , 5-Methylcytosine/metabolism , Aged , Aortic Valve Stenosis/enzymology , Aortic Valve Stenosis/pathology , CRISPR-Cas Systems , Calcinosis/enzymology , Calcinosis/pathology , Calcium/metabolism , Case-Control Studies , Down-Regulation , Gene Editing/methods , Gene Expression Profiling/methods , HEK293 Cells , Humans , Lysophospholipids/metabolism , Male , Middle Aged , Phosphatidate Phosphatase/metabolism
9.
Cardiovasc Res ; 113(11): 1351-1363, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28472283

ABSTRACT

AIMS: Oxidatively modified lipoproteins may promote the development/progression of calcific aortic valve stenosis (CAVS). Oxidative transformation of low-density lipoprotein (OxLDL) generates lysophosphatidic acid (LPA), a lipid mediator that accumulates in mineralized aortic valves. LPA activates at least six different G protein-coupled receptors, which may play a role in the pathophysiology of CAVS. We hypothesized that LPA derived from OxLDL may promote a NF-κB signature that drives osteogenesis in the aortic valve. METHODS AND RESULTS: The role of OxLDL-LPA was examined in isolated valve interstitial cells (VICs) and the molecular pathway was validated in human explanted aortic valves and in a mouse model of CAVS. We found that OxLDL-LPA promoted the mineralization and osteogenic transition of VICs through LPAR1 and the activation of a RhoA-NF-κB pathway. Specifically, we identified that RhoA/ROCK activated IκB kinase alpha, which promoted the phosphorylation of p65 on serine 536 (p65 pS536). p65 pS536 was recruited to the BMP2 promoter and directed an osteogenic program not responsive to the control exerted by the inhibitor of kappa B. In LDLR-/-/ApoB100/100/IGFII transgenic mice (IGFII), which develop CAVS under a high-fat and high-sucrose diet the administration of Ki16425, a Lpar1 blocker, reduced by three-fold the progression rate of CAVS and also decreased the osteogenic activity as measured with a near-infrared fluorescent probe that recognizes hydroxyapatite of calcium. CONCLUSIONS: OxLDL-LPA promotes an osteogenic program in the aortic valve through a LPAR1-RhoA/ROCK-p65 pS536 pathway. LPAR1 may represent a suitable target to prevent the progression of CAVS.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Lipoproteins, LDL/metabolism , NF-kappa B/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Aortic Valve/metabolism , Humans , Lysophospholipids/pharmacology , Mice , Phosphorylation , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism
10.
Infect Genet Evol ; 50: 110-120, 2017 06.
Article in English | MEDLINE | ID: mdl-27818279

ABSTRACT

Zoonotic cutaneous leishmaniasis caused by Leishmania (L.) major parasites affects urban and suburban areas in the center and south of Tunisia where the disease is endemo-epidemic. Several cases were reported in human patients for which infection due to L. major induced lesions with a broad range of severity. However, very little is known about the mechanisms underlying this diversity. Our hypothesis is that parasite genomic variability could, in addition to the host immunological background, contribute to the intra-species clinical variability observed in patients and explain the lesion size differences observed in the experimental model. Based on several epidemiological, in vivo and in vitro experiments, we focused on two clinical isolates showing contrasted severity in patients and BALB/c experimental mice model. We used DNA-seq as a high-throughput technology to facilitate the identification of genetic variants with discriminating potential between both isolates. Our results demonstrate that various levels of heterogeneity could be found between both L. major isolates in terms of chromosome or gene copy number variation (CNV), and that the intra-species divergence could surprisingly be related to single nucleotide polymorphisms (SNPs) and Insertion/Deletion (InDels) events. Interestingly, we particularly focused here on genes affected by both types of variants and correlated them with the observed gene CNV. Whether these differences are sufficient to explain the severity in patients is obviously still open to debate, but we do believe that additional layers of -omic information is needed to complement the genomic screen in order to draw a more complete map of severity determinants.


Subject(s)
Chromosomes/chemistry , Endemic Diseases , Gene Dosage , Leishmania major/genetics , Leishmaniasis, Cutaneous/epidemiology , Phylogeny , Animals , DNA, Protozoan/genetics , Female , Follow-Up Studies , Genomics , Humans , INDEL Mutation , Leishmania major/classification , Leishmania major/isolation & purification , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/transmission , Mice , Mice, Inbred BALB C , Phylogeography , Polymorphism, Single Nucleotide , Severity of Illness Index , Tunisia/epidemiology
12.
Parasitology ; 143(12): 1615-21, 2016 10.
Article in English | MEDLINE | ID: mdl-27312247

ABSTRACT

Synthesized lipophilic tyrosyl ester derivatives with increasing lipophilicity were effective against Leishmania (L.) major and Leishmania infantum species in vitro. These findings prompted us to test in vivo leishmanicidal properties of these molecules and their potential effect on the modulation of immune responses. The experimental BALB/c model of cutaneous leishmaniasis was used in this study. Mice were infected with L. major parasites and treated with three in vitro active tyrosyl esters derivatives. Among these tested tyrosylcaprate (TyC) compounds, only TyC10 exhibited an in vivo anti-leishmanial activity, when injected sub-cutaneously (s.c.). TyC10 treatment of L. major-infected BALB/c mice resulted in a decrease of lesion development and parasite load. TyC10 s.c. treatment of non-infected mice induced an imbalance in interferon γ/interleukin 4 (IFN-γ/IL-4) ratio cytokines towards a Th1 response. Our results indicate that TyC10 s.c. treatment improves lesions' healing and parasite clearance and may act on the cytokine balance towards a Th1 protective response by decreasing IL-4 and increasing IFN-γ transcripts. TyC10 is worthy of further investigation to uncover its mechanism of action that could lead to consider this molecule as a potential drug candidate.


Subject(s)
Antiprotozoal Agents/administration & dosage , Immunologic Factors/administration & dosage , Leishmania major/drug effects , Leishmaniasis, Cutaneous/drug therapy , Animals , Cytokines/metabolism , Disease Models, Animal , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Parasite Load , Th1 Cells/immunology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
13.
Infect Genet Evol ; 43: 179-85, 2016 09.
Article in English | MEDLINE | ID: mdl-27137082

ABSTRACT

Tunisia is endemic for zoonotic cutaneous leishmaniasis (ZCL), a parasitic disease caused by Leishmania (L.) major. ZCL displays a wide clinical polymorphism, with severe forms present more frequently in emerging foci where naive populations are dominant. In this study, we applied the multi-locus microsatellite typing (MLMT) using ten highly informative and discriminative markers to investigate the genetic structure of 35 Tunisian Leishmania (L.) major isolates collected from patients living in five different foci of Central Tunisia (two old and three emerging foci). Phylogenetic reconstructions based on genetic distances showed that nine of the ten tested loci were homogeneous in all isolates with homozygous alleles, whereas one locus (71AT) had a 58/64-bp bi-allelic profile with an allele linked to emerging foci. Promastigote-stage parasites with the 58-bp allele tend to be more resistant to in vitro complement lysis. These results, which stress the geographical dependence of the genetic micro-heterogeneity, may improve our understanding of the ZCL epidemiology and clinical outcome.


Subject(s)
DNA, Protozoan/genetics , Endemic Diseases , Genome, Protozoan , Leishmania major/genetics , Leishmaniasis, Cutaneous/epidemiology , Life Cycle Stages/genetics , Phylogeny , Alleles , Animals , Genetic Heterogeneity , Genetic Loci , Humans , Leishmania major/classification , Leishmania major/growth & development , Leishmania major/isolation & purification , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/transmission , Microsatellite Repeats , Multilocus Sequence Typing , Psychodidae/parasitology , Tunisia/epidemiology , Zoonoses
14.
PLoS Negl Trop Dis ; 7(10): e2478, 2013.
Article in English | MEDLINE | ID: mdl-24098824

ABSTRACT

BACKGROUND: Leishmania (L.) are intracellular protozoan parasites able to survive and replicate in the hostile phagolysosomal environment of infected macrophages. They cause leishmaniasis, a heterogeneous group of worldwide-distributed affections, representing a paradigm of neglected diseases that are mainly embedded in impoverished populations. To establish successful infection and ensure their own survival, Leishmania have developed sophisticated strategies to subvert the host macrophage responses. Despite a wealth of gained crucial information, these strategies still remain poorly understood. MicroRNAs (miRNAs), an evolutionarily conserved class of endogenous 22-nucleotide non-coding RNAs, are described to participate in the regulation of almost every cellular process investigated so far. They regulate the expression of target genes both at the levels of mRNA stability and translation; changes in their expression have a profound effect on their target transcripts. METHODOLOGY/PRINCIPAL FINDINGS: We report in this study a comprehensive analysis of miRNA expression profiles in L. major-infected human primary macrophages of three healthy donors assessed at different time-points post-infection (three to 24 h). We show that expression of 64 out of 365 analyzed miRNAs was consistently deregulated upon infection with the same trends in all donors. Among these, several are known to be induced by TLR-dependent responses. GO enrichment analysis of experimentally validated miRNA-targeted genes revealed that several pathways and molecular functions were disturbed upon parasite infection. Finally, following parasite infection, miR-210 abundance was enhanced in HIF-1α-dependent manner, though it did not contribute to inhibiting anti-apoptotic pathways through pro-apoptotic caspase-3 regulation. CONCLUSIONS/SIGNIFICANCE: Our data suggest that alteration in miRNA levels likely plays an important role in regulating macrophage functions following L. major infection. These results could contribute to better understanding of the dynamics of gene expression in host cells during leishmaniasis.


Subject(s)
Gene Expression Profiling , Host-Pathogen Interactions , Leishmania major/immunology , Macrophages/immunology , Macrophages/parasitology , MicroRNAs/biosynthesis , Blood Donors , Cells, Cultured , Healthy Volunteers , Humans
15.
Neurochem Res ; 36(8): 1546-57, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21533646

ABSTRACT

Manganese (Mn) is an essential trace element required for ubiquitous enzymatic reactions. Chronic overexposure to this metal may promote potent neurotoxic effects. The mechanism of Mn toxicity is not well established, but several studies indicate that oxidative stress play major roles in the Mn-induced neurodegenerative processes. Silymarin (SIL) has antioxidant properties and stabilizes intracellular antioxidant defense systems. The aim of this study was to evaluate the toxic effects of MnCl(2) on the mouse neuroblastoma cell lines (Neuro-2A), to characterize the toxic mechanism associated with Mn exposure and to investigate whether SIL could efficiently protect against neurotoxicity induced by Mn. A significant increase in LDH release activity was observed in Neuro-2A cells associated with a significant decrease in cellular viability upon 24 h exposure to MnCl(2) at concentrations of 200 and 800 µM (P < 0.05) when compared with control unexposed cells. In addition, exposure cells to MnCl(2) (200 and 800 µM), increases oxidant biomarkers and alters enzymatic and non enzymatic antioxidant systems. SIL treatment significantly reduced the levels of LDH, nitric oxide, reactive oxygen species and the oxidants/antioxidants balance in Neuro-2A cells as compared to Mn-exposed cells. These results suggested that silymarin is a powerful antioxidant through a mechanism related to its antioxidant activity, able to interfere with radical-mediated cell death. SIL may be useful in diseases known to be aggravated by reactive oxygen species and in the development of novel treatments for neurodegenerative disorders such as Alzheimer or Parkinson diseases.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Line, Tumor/drug effects , Manganese/toxicity , Neuroblastoma/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Silymarin/pharmacology , Animals , Antioxidants/metabolism , Cell Membrane/drug effects , Cell Membrane/enzymology , Humans , Lipid Peroxidation , Mice , Oxidation-Reduction , Reactive Oxygen Species/metabolism
16.
J Mol Histol ; 36(6-7): 381-90, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16482425

ABSTRACT

The expression pattern of VEGF, p53 and ICAM-1 was studied in conjunctiva of diabetic patients with and without retinopathy. All patients underwent a complete ophthalmic examination, including retinal fluorescein angiography. Indirect immunoperoxidase method was performed on 20 eyes of 20 patients with type II diabetes without DR and on 5 eyes of 5 patients with PDR. A control study was performed on 6 normal conjunctiva undertaken during cataract surgery. Immunoreactivity of VEGF, p53 and ICAM-1 was found in epithelial, fibroblast and vascular endothelial cells. For the same duration of diabetes, a strong to moderate or weak immunoreactivity was observed in the conjunctiva of patients without retinopathy. In patients with PDR, the expression was strong for all these proteins. The immunoreactivity was correlated between VEGF, p53 and ICAM-1. In the normal conjunctiva, a weak to negative immunostaining was observed. The presence of these proteins in the conjunctiva of diabetic patients without retinopathy may add new data in the pathogenesis of diabetic retinopathy. Further studies are needed to confirm this hypothesis.


Subject(s)
Conjunctiva/pathology , Diabetes Mellitus, Type 2/metabolism , Intercellular Adhesion Molecule-1/metabolism , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Diabetic Retinopathy/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...