47 results on '"Inflammation/genetics"'
Search Results
2. Splenic monocytes drive pathogenic subretinal inflammation in age-related macular degeneration
- Author
-
Roubeix, Christophe, Nous, Caroline, Augustin, Sébastien, Ronning, Kaitryn E, Mathis, Thibaud, Blond, Frédéric, Lagouge-Roussey, Pauline, Crespo-Garcia, Sergio, Sullivan, Patrick M, Gautier, Emmanuel L, Reichhart, Nadine, Sahel, José-Alain, Burns, Marie E, Paques, Michel, Sørensen, Torben Lykke, Strauss, Olaf, Guillonneau, Xavier, Delarasse, Cécile, Sennlaub, Florian, Roubeix, Christophe, Nous, Caroline, Augustin, Sébastien, Ronning, Kaitryn E, Mathis, Thibaud, Blond, Frédéric, Lagouge-Roussey, Pauline, Crespo-Garcia, Sergio, Sullivan, Patrick M, Gautier, Emmanuel L, Reichhart, Nadine, Sahel, José-Alain, Burns, Marie E, Paques, Michel, Sørensen, Torben Lykke, Strauss, Olaf, Guillonneau, Xavier, Delarasse, Cécile, and Sennlaub, Florian
- Abstract
Age-related macular degeneration (AMD) is invariably associated with the chronic accumulation of activated mononuclear phagocytes in the subretinal space. The mononuclear phagocytes are composed of microglial cells but also of monocyte-derived cells, which promote photoreceptor degeneration and choroidal neovascularization. Infiltrating blood monocytes can originate directly from bone marrow, but also from a splenic reservoir, where bone marrow monocytes develop into angiotensin II receptor (ATR1)+ splenic monocytes. The involvement of splenic monocytes in neurodegenerative diseases such as AMD is not well understood. Using acute inflammatory and well-phenotyped AMD models, we demonstrate that angiotensin II mobilizes ATR1+ splenic monocytes, which we show are defined by a transcriptional signature using single-cell RNA sequencing and differ functionally from bone marrow monocytes. Splenic monocytes participate in the chorio-retinal infiltration and their inhibition by ATR1 antagonist and splenectomy reduces the subretinal mononuclear phagocyte accumulation and pathological choroidal neovascularization formation. In aged AMD-risk ApoE2-expressing mice, a chronic AMD model, ATR1 antagonist and splenectomy also inhibit the chronic retinal inflammation and associated cone degeneration that characterizes these mice. Our observation of elevated levels of plasma angiotensin II in AMD patients, suggests that similar events take place in clinical disease and argue for the therapeutic potential of ATR1 antagonists to inhibit splenic monocytes for the treatment of blinding AMD.
- Published
- 2024
3. Loss of Shp1 impairs myeloid cell function and causes lethal inflammation in zebrafish larvae
- Author
-
Allers, Maaike, Bakker, Petra A, Hoeksma, Jelmer, Spaink, Herman P, den Hertog, Jeroen, Allers, Maaike, Bakker, Petra A, Hoeksma, Jelmer, Spaink, Herman P, and den Hertog, Jeroen
- Abstract
PTPN6 encodes SHP1, a protein tyrosine phosphatase with an essential role in immune cell function. SHP1 mutations are associated with neutrophilic dermatoses and emphysema in humans, which resembles the phenotype seen in motheaten mice that lack functional SHP1. To investigate the function of Shp1 in developing zebrafish embryos, we generated a ptpn6 knockout zebrafish line lacking functional Shp1. Shp1 knockout caused severe inflammation and lethality around 17 days post fertilization (dpf). During early development, the myeloid lineage was affected, resulting in a decrease in the number of neutrophils and a concomitant increase in the number of macrophages. The number of emerging hematopoietic stem and progenitor cells (HSPCs) was decreased, but due to hyperproliferation, the number of HSPCs was higher in ptpn6 mutants than in siblings at 5 dpf. Finally, the directional migration of neutrophils and macrophages was decreased in response to wounding, and fewer macrophages were recruited to the wound site. Yet, regeneration of the caudal fin fold was normal. We conclude that loss of Shp1 impaired neutrophil and macrophage function, and caused severe inflammation and lethality at the larval stage.
- Published
- 2023
4. The lncRNA LUCAT1 is elevated in inflammatory disease and restrains inflammation by regulating the splicing and stability of NR4A2
- Author
-
Tim Vierbuchen, Shiuli Agarwal, John L. Johnson, Liraz Galia, Xuqiu Lei, Karina Stein, David Olagnier, Karoline I. Gaede, Christian Herzmann, Christian K. Holm, Holger Heine, Athma Pai, Aisling O’Hara Hall, Kasper Hoebe, and Katherine A. Fitzgerald
- Subjects
Inflammation ,Cell Proliferation/genetics ,Multidisciplinary ,Nuclear Receptor Subfamily 4, Group A, Member 2/genetics ,Cell Movement ,Inflammation/genetics ,Nuclear Receptor Subfamily 4, Group A, Member 2 ,Cell Movement/genetics ,Humans ,Receptors, Cytoplasmic and Nuclear ,RNA, Long Noncoding ,RNA, Long Noncoding/genetics ,Cell Proliferation - Abstract
The nuclear long non-coding RNA LUCAT1 has previously been identified as a negative feedback regulator of type I interferon and inflammatory cytokine expression in human myeloid cells. Here, we define the mechanistic basis for the suppression of inflammatory gene expression by LUCAT1. Using comprehensive identification of RNA-binding proteins by mass spectrometry as well as RNA immunoprecipitation, we identified proteins important in processing and alternative splicing of mRNAs as LUCAT1-binding proteins. These included heterogeneous nuclear ribonucleoprotein C, M, and A2B1. Consistent with this finding, cells lacking LUCAT1 have altered splicing of selected immune genes. In particular, upon lipopolysaccharide stimulation, the splicing of the nuclear receptor 4A2 (NR4A2) gene was particularly affected. As a consequence, expression of NR4A2 was reduced and delayed in cells lacking LUCAT1. NR4A2-deficient cells had elevated expression of immune genes. These observations suggest that LUCAT1 is induced to control the splicing and stability of NR4A2, which is in part responsible for the anti-inflammatory effect of LUCAT1. Furthermore, we analyzed a large cohort of patients with inflammatory bowel disease as well as asthma and chronic obstructive pulmonary disease. In these patients, LUCAT1 levels were elevated and in both diseases, positively correlated with disease severity. Collectively, these studies define a key molecular mechanism of LUCAT1-dependent immune regulation through post-transcriptional regulation of mRNAs highlighting its role in the regulation of inflammatory disease.
- Published
- 2022
- Full Text
- View/download PDF
5. Genetic and epigenetic regulation of Catechol-O-methyltransferase in relation to inflammation in chronic fatigue syndrome and Fibromyalgia
- Author
-
Andrea Polli, Jolien Hendrix, Kelly Ickmans, Jelena Bakusic, Manosij Ghosh, Dora Monteyne, Brigitte Velkeniers, Bram Bekaert, Jo Nijs, Lode Godderis, Physiotherapy, Human Physiology and Anatomy, Brussels Heritage Lab, Pain in Motion, Faculty of Physical Education and Physical Therapy, Movement and Sport Sciences, Physical Medicine and Rehabilitation, Movement and Nutrition for Health and Performance, Internal Medicine, and Clinical sciences
- Subjects
Fibromyalgia ,BLOOD ,Interleukin-6/metabolism ,Pain ,Physical Therapy, Sports Therapy and Rehabilitation ,Catechol O-Methyltransferase/genetics ,Research & Experimental Medicine ,Catechol O-Methyltransferase ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Transforming Growth Factor beta ,Inflammation/genetics ,Internal Medicine ,Genetics ,Humans ,Pain/genetics ,Tumor Necrosis Factor-alpha/metabolism ,Inflammation ,TEMPORAL SUMMATION ,Fatigue Syndrome, Chronic ,Science & Technology ,DNA methylation ,Tumor Necrosis Factor-alpha ,Interleukin-6 ,PAIN ,General Medicine ,Polymorphism, Single Nucleotide/genetics ,catechol-O-methyltransferase (COMT) ,Fatigue Syndrome, Chronic/genetics ,CYTOKINE ,Fibromyalgia/genetics ,Anesthesiology and Pain Medicine ,Medicine, Research & Experimental ,Case-Control Studies ,CELLS ,Cytokines ,Epigenetics ,Life Sciences & Biomedicine ,Transforming Growth Factor beta/metabolism - Abstract
Background Catechol-O-methyltransferase (COMT) has been shown to influence clinical pain, descending modulation, and exercise-induced symptom worsening. COMT regulates nociceptive processing and inflammation, key pathophysiological features of Chronic Fatigue Syndrome and Fibromyalgia (CFS/FM). We aimed to determine the interactions between genetic and epigenetic mechanisms regulating COMT and its influence on inflammatory markers and symptoms in patients with CFS/FM. Methods. A case-control study with repeated-measures design was used to reduce the chance of false positive and increase the power of our findings. Fifty-four participants (28 patients with CFS/FM and 26 controls) were assessed twice within 4 days. The assessment included clinical questionnaires, neurophysiological assessment (pain thresholds, temporal summation, and conditioned pain modulation), and blood withdrawal in order to assess rs4818, rs4633, and rs4680 COMT polymorphisms and perform haplotype estimation, DNA methylation in the COMT gene (both MB-COMT and S-COMT promoters), and cytokine expression (TNF-α, IFN-γ, IL-6, and TGF-β). Results. COMT haplotypes were associated with DNA methylation in the S-COMT promoter, TGF-β expression, and symptoms. However, this was not specific for one condition. Significant between-group differences were found for increased DNA methylation in the MB-COMT promoter and decreased IFN-γ expression in patients. Discussion Our results are consistent with basic and clinical research, providing interesting insights into genetic-epigenetic regulatory mechanisms. MB-COMT DNA methylation might be an independent factor contributing to the pathophysiology of CFS/FM. Further research on DNA methylation in complex conditions such as CFS/FM is warranted. We recommend future research to employ a repeated-measure design to control for biomarkers variability and within-subject changes.
- Published
- 2022
6. DNA methylation signature of chronic low-grade inflammation and its role in cardio-respiratory diseases
- Author
-
Wielscher, Matthias, Mandaviya, Pooja R, Kuehnel, Brigitte, Joehanes, Roby, Mustafa, Rima, Robinson, Oliver, Zhang, Yan, Bodinier, Barbara, Walton, Esther, Mishra, Pashupati P, Schlosser, Pascal, Wilson, Rory, Tsai, Pei-Chien, Palaniswamy, Saranya, Marioni, Riccardo E, Fiorito, Giovanni, Cugliari, Giovanni, Karhunen, Ville, Ghanbari, Mohsen, Psaty, Bruce M, Loh, Marie, Bis, Joshua C, Lehne, Benjamin, Sotoodehnia, Nona, Deary, Ian J, Chadeau-Hyam, Marc, Brody, Jennifer A, Cardona, Alexia, Selvin, Elizabeth, Smith, Alicia K, Miller, Andrew H, Torres, Mylin A, Marouli, Eirini, Gào, Xin, Van Meurs, Joyce BJ, Graf-Schindler, Johanna, Rathmann, Wolfgang, Koenig, Wolfgang, Peters, Annette, Weninger, Wolfgang, Farlik, Matthias, Zhang, Tao, Chen, Wei, Xia, Yujing, Teumer, Alexander, Nauck, Matthias, Grabe, Hans J, Doerr, Macus, Lehtimäki, Terho, Guan, Weihua, Milani, Lili, Tanaka, Toshiko, Fisher, Krista, Waite, Lindsay L, Kasela, Silva, Vineis, Paolo, Verweij, Niek, Van Der Harst, Pim, Iacoviello, Licia, Sacerdote, Carlotta, Panico, Salvatore, Krogh, Vittorio, Tumino, Rosario, Tzala, Evangelia, Matullo, Giuseppe, Hurme, Mikko A, Raitakari, Olli T, Colicino, Elena, Baccarelli, Andrea A, Kähönen, Mika, Herzig, Karl-Heinz, Li, Shengxu, BIOS Consortium, Conneely, Karen N, Kooner, Jaspal S, Köttgen, Anna, Heijmans, Bastiaan T, Deloukas, Panos, Relton, Caroline, Ong, Ken K, Bell, Jordana T, Boerwinkle, Eric, Elliott, Paul, Brenner, Hermann, Beekman, Marian, Levy, Daniel, Waldenberger, Melanie, Chambers, John C, Dehghan, Abbas, Järvelin, Marjo-Riitta, Wielscher, Matthias [0000-0003-4138-1383], Mustafa, Rima [0000-0003-2623-5337], Robinson, Oliver [0000-0002-4735-0468], Bodinier, Barbara [0000-0002-0781-3624], Walton, Esther [0000-0002-0935-2200], Schlosser, Pascal [0000-0002-8460-0462], Wilson, Rory [0000-0001-6135-3764], Palaniswamy, Saranya [0000-0003-4145-540X], Karhunen, Ville [0000-0001-6064-1588], Ghanbari, Mohsen [0000-0002-9476-7143], Psaty, Bruce M [0000-0002-7278-2190], Loh, Marie [0000-0003-3626-8466], Bis, Joshua C [0000-0002-3409-1110], Chadeau-Hyam, Marc [0000-0001-8341-5436], Brody, Jennifer A [0000-0001-8509-148X], Cardona, Alexia [0000-0002-7877-5565], Smith, Alicia K [0000-0002-8537-5156], Miller, Andrew H [0000-0001-8260-7997], Marouli, Eirini [0000-0001-6179-1609], Gào, Xin [0000-0003-0108-6961], Koenig, Wolfgang [0000-0002-2064-9603], Peters, Annette [0000-0001-6645-0985], Farlik, Matthias [0000-0003-0698-2992], Chen, Wei [0000-0003-1566-7943], Xia, Yujing [0000-0003-0850-5591], Teumer, Alexander [0000-0002-8309-094X], Nauck, Matthias [0000-0002-6678-7964], Doerr, Macus [0000-0001-7471-475X], Lehtimäki, Terho [0000-0002-2555-4427], Milani, Lili [0000-0002-5323-3102], Fisher, Krista [0000-0002-3521-0599], van der Harst, Pim [0000-0002-2713-686X], Krogh, Vittorio [0000-0003-0122-8624], Herzig, Karl-Heinz [0000-0003-4460-2604], Li, Shengxu [0000-0001-9210-7283], Köttgen, Anna [0000-0002-4671-3714], Heijmans, Bastiaan T [0000-0001-5918-0534], Deloukas, Panos [0000-0001-9251-070X], Ong, Ken K [0000-0003-4689-7530], Bell, Jordana T [0000-0002-3858-5986], Elliott, Paul [0000-0002-7511-5684], Brenner, Hermann [0000-0002-6129-1572], Beekman, Marian [0000-0003-0585-6206], Waldenberger, Melanie [0000-0003-0583-5093], Chambers, John C [0000-0003-0209-4541], Dehghan, Abbas [0000-0001-6403-016X], Järvelin, Marjo-Riitta [0000-0002-2149-0630], Apollo - University of Cambridge Repository, Maastricht Centre for Systems Biology, RS: FSE MaCSBio, Tampere University, Clinical Medicine, Department of Clinical Chemistry, BioMediTech, Department of Clinical Physiology and Nuclear Medicine, Ong, Kenneth [0000-0003-4689-7530], Internal Medicine, and Epidemiology
- Subjects
obesity ,BLOOD ,45/61 ,PREDICTION ,NF-KAPPA-B ,BIOS consortium ,General Physics and Astronomy ,General Biochemistry, Genetics and Molecular Biology ,38 ,Low grade inflammation ,MARKERS ,SDG 3 - Good Health and Well-being ,Inflammation/genetics ,risk factors ,Medicine ,Humans ,TRANSCRIPTION ,Nucleotide Motifs ,C-Reactive Protein ,CpG Islands ,DNA Methylation ,Inflammation ,EPIGENOME-WIDE ASSOCIATION ,DNA methylation ,Multidisciplinary ,business.industry ,article ,631/208/176/1988 ,692/163/2743/393 ,Cardiorespiratory fitness ,General Chemistry ,CpG Islands/genetics ,LIFETIME RISK ,GENOME ,BODY-MASS INDEX ,Immunology ,3111 Biomedicine ,C-Reactive Protein/genetics ,business ,692/499 ,DNA Methylation/genetics - Abstract
Chronic inflammation, marked by C-reactive protein, has been associated with changes in methylation, but the causal relationship is unclear. Here, the authors perform a Epigenome-wide association meta-analysis for C-reactive protein levels and find that these methylation changes are likely the consequence of inflammation and could contribute to disease.We performed a multi-ethnic Epigenome Wide Association study on 22,774 individuals to describe the DNA methylation signature of chronic low-grade inflammation as measured by C-Reactive protein (CRP). We find 1,511 independent differentially methylated loci associated with CRP. These CpG sites show correlation structures across chromosomes, and are primarily situated in euchromatin, depleted in CpG islands. These genomic loci are predominantly situated in transcription factor binding sites and genomic enhancer regions. Mendelian randomization analysis suggests altered CpG methylation is a consequence of increased blood CRP levels. Mediation analysis reveals obesity and smoking as important underlying driving factors for changed CpG methylation. Finally, we find that an activated CpG signature significantly increases the risk for cardiometabolic diseases and COPD.
- Published
- 2022
- Full Text
- View/download PDF
7. PPARs as Key Mediators in the Regulation of Metabolism and Inflammation
- Author
-
Manuel Vazquez-Carrera and Walter Wahli
- Subjects
Peroxisome Proliferator-Activated Receptors ,Receptors, Cytoplasmic and Nuclear ,Àcids grassos ,Nuclear receptors (Biochemistry) ,Metabolisme cel·lular ,Catalysis ,Inorganic Chemistry ,Transcription factors ,Humans ,Physical and Theoretical Chemistry ,Fatty acids ,Molecular Biology ,Spectroscopy ,Biochemical genetics ,Inflammation ,Cell metabolism ,Organic Chemistry ,Inflammation/genetics ,Lipid Metabolism ,Peroxisome Proliferator-Activated Receptors/metabolism ,Receptors, Cytoplasmic and Nuclear/genetics ,Receptors, Cytoplasmic and Nuclear/metabolism ,Transcription Factors/metabolism ,General Medicine ,Genètica bioquímica ,Inflamació ,Computer Science Applications ,Factors de transcripció ,Receptors nuclears (Bioquímica) ,Transcription Factors - Abstract
Nuclear receptors (NRs) form a large family of ligand-dependent transcription factors that control the expression of a multitude of genes involved in diverse, vital biological processes. Three of these receptors, the peroxisome proliferator-activated receptors (PPARs), were discovered in the 1990s and play key roles in the regulation of cellular differentiation, embryonic development, cellular and whole-body metabolism, inflammation, and tumorigenesis in higher organisms. PPARs are activated not only by fatty acids and their derivatives, some of which also signal through membrane receptors, but also by many plant- and marine-derived natural ligands. Furthermore, drugs that target PPARs, such as fibrates and thiazolidinediones, have been developed to treat metabolic diseases. The 'classic' molecular mode of action of PPARs in the control of physiological and metabolic processes is via their direct binding, as PPAR:retinoid X receptor (RXR) heterodimers, to peroxisome proliferator response elements (PPREs) in the regulatory regions of target genes. The activity of PPARs can also be modulated by posttranslational modifications and their transcriptional regulatory capacity may present a circadian pattern, depending on their expression and the availability of ligands.
- Published
- 2022
8. Genetic analysis of over half a million people characterises C-reactive protein loci
- Author
-
Saredo Said, Raha Pazoki, Ville Karhunen, Urmo Võsa, Symen Ligthart, Barbara Bodinier, Fotios Koskeridis, Paul Welsh, Behrooz Z. Alizadeh, Daniel I. Chasman, Naveed Sattar, Marc Chadeau-Hyam, Evangelos Evangelou, Marjo-Riitta Jarvelin, Paul Elliott, Ioanna Tzoulaki, Abbas Dehghan, Life Course Epidemiology (LCE), and Real World Studies in PharmacoEpidemiology, -Genetics, -Economics and -Therapy (PEGET)
- Subjects
Inflammation ,Male ,Multidisciplinary ,General Physics and Astronomy ,Single Nucleotide ,General Chemistry ,Chronic inflammation ,Mendelian Randomization Analysis ,Genome-wide association studies ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Data processing ,C-Reactive Protein ,Genetic Loci ,Inflammation/genetics ,Humans ,Human medicine ,Polymorphism ,Phenomics ,C-Reactive Protein/genetics ,Genome-Wide Association Study - Abstract
Data availability: The summary statistics of the CHARGE CRP GWAS used in this study is publicly available from the IEU open GWAS project accession code ieu-b-35 (Trait: C-Reactive protein level - IEU Open GWAS project (mrcieu.ac.uk)). The derived CRP GWAS meta-analysis summary statistics generated in this study has been deposited in the GWAS catalogue under accession code GCST00186 (https://www.ebi.ac.uk/gwas/). Human genome assembly GRCh37 (hg19) from Genome Reference Consortium https://www.sanger.ac.uk/data/genome-reference-consortium/). Copyright © The Author(s) 2022. Chronic low-grade inflammation is linked to a multitude of chronic diseases. We report the largest genome-wide association study (GWAS) on C-reactive protein (CRP), a marker of systemic inflammation, in UK Biobank participants (N = 427,367, European descent) and the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium (total N = 575,531 European descent). We identify 266 independent loci, of which 211 are not previously reported. Gene-set analysis highlighted 42 gene sets associated with CRP levels (p ≤ 3.2 ×10−6) and tissue expression analysis indicated a strong association of CRP related genes with liver and whole blood gene expression. Phenome-wide association study identified 27 clinical outcomes associated with genetically determined CRP and subsequent Mendelian randomisation analyses supported a causal association with schizophrenia, chronic airway obstruction and prostate cancer. Our findings identified genetic loci and functional properties of chronic low-grade inflammation and provided evidence for causal associations with a range of diseases. UK Dementia Research Institute at Imperial College, which receives its funding from UK DRI Ltd. (funded by the UK Medical Research Council, Alzheimer’s Society and Alzheimer’s Research UK) and the British Heart Foundation Centre for Research Excellence at Imperial College London and the National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London. S.S. received funding from the Medical Research Council – Public Health England (MRC-PHE) Centre for Environment and Health awarded studentship, of which funding is derived from the MRC Industrial Strategy Fund. I.T and F.K. have received funding from the Hellenic Foundation for Research and Innovation (HFRI) and the General Secretariat for Research and Technology (GSRT), under grant agreement No 1312. R.P. holds a fellowship supported by Rutherford Fund from Medical Research Council (MR/R0265051/1 and MR/R0265051/2). V.K. is funded by the European Union’s Horizon 2020 research and innovation program under the Marie Sklodowska-Curie grant (721567).
- Published
- 2022
9. Gut microbial DNA and immune checkpoint gene Vsig4/CRIg are key antagonistic players in healthy aging and age-associated development of hypertension and diabetes
- Author
-
Matthew A. Liu, Shandy Shahabi, Suborno Jati, Kechun Tang, Hong Gao, Zhongmou Jin, Wyatt Miller, Frédéric A. Meunier, Wei Ying, Geert van den Bogaart, Gourisankar Ghosh, Sushil K. Mahata, and Molecular Immunology
- Subjects
DNA, Bacterial ,Aging ,hypertension ,Endocrinology, Diabetes and Metabolism ,Knockout ,Clinical Sciences ,Insulin Resistance/genetics ,Cardiovascular ,Mice ,insulin resistance ,Inflammation/genetics ,Genetics ,Diabetes Mellitus ,2.1 Biological and endogenous factors ,Animals ,Aetiology ,pancreastatin ,Mice, Knockout ,Inflammation ,Nutrition and Dietetics ,diabetes ,Bacterial ,catestatin ,DNA ,Gastrointestinal Microbiome ,healthy aging ,Hypertension ,Chromogranin A ,Insulin Resistance ,Hypertension/genetics - Abstract
AimsAging is associated with the development of insulin resistance and hypertension which may stem from inflammation induced by accumulation of toxic bacterial DNA crossing the gut barrier. The aim of this study was to identify factors counter-regulating these processes. Taking advantage of the Chromogranin A (CgA) knockout (CgA-KO) mouse as a model for healthy aging, we have identified Vsig4 (V-set and immunoglobulin domain containing 4) as the critical checkpoint gene in offsetting age-associated hypertension and diabetes.Methods and ResultsThe CgA-KO mice display two opposite aging phenotypes: hypertension but heightened insulin sensitivity at young age, whereas the blood pressure normalizes at older age and insulin sensitivity further improves. In comparison, aging WT mice gradually lost glucose tolerance and insulin sensitivity and developed hypertension. The gut barrier, compromised in aging WT mice, was preserved in CgA KO mice leading to major 35-fold protection against bacterial DNA-induced inflammation. Similarly, RNA sequencing showed increased expression of the Vsig4 gene (which removes bacterial DNA) in the liver of 2-yr-old CgA-KO mice, which may account for the very low accumulation of microbial DNA in the heart. The reversal of hypertension in aging CgA-KO mice likely stems from (i) low accumulation of microbial DNA, (ii) decreased spillover of norepinephrine in the heart and kidneys, and (iii) reduced inflammation.ConclusionWe conclude that healthy aging relies on protection from bacterial DNA and the consequent low inflammation afforded by CgA-KO. Vsig4 also plays a crucial role in “healthy aging” by counteracting age-associated insulin resistance and hypertension.
- Published
- 2022
10. VEXAS Syndrome: A Call for Diagnostic Awareness Based on a Case Series of Seven Patients.
- Author
-
R Pinto F, Lamas A, G Oliveira D, E Oliveira M, and Faria R
- Subjects
- Humans, Mutation, Research
- Published
- 2023
- Full Text
- View/download PDF
11. Transcriptional profile of cytokines, regulatory mediators and tlr in mesenchymal stromal cells after inflammatory signaling and cell-passaging
- Author
-
Robim Marcelino Rodrigues, Makram Merimi, Vera Rogiers, Philippe Lewalle, Laurence Lagneaux, Dhouha Daassi, Rahma Melki, Karolien Buyl, Hassan Fahmi, Mehdi Najar, Joery De Kock, Tamara Vanhaecke, Department of Bio-engineering Sciences, Pharmaceutical and Pharmacological Sciences, Experimental in vitro toxicology and dermato-cosmetology, and Vriendenkring VUB
- Subjects
0301 basic medicine ,Regulatory mediators ,TLR 1 ,Transcription, Genetic ,Mesenchymal Stem Cells/metabolism ,Mesenchymal stromal cells ,Informatique appliquée logiciel ,Cell-passage ,Toll-Like Receptors/genetics ,Physico-chimie générale ,0302 clinical medicine ,Inflammation/genetics ,Biology (General) ,Spectroscopy ,education.field_of_study ,Cytokines/genetics ,Brief Report ,Toll-Like Receptors ,General Medicine ,Down-Regulation/genetics ,Computer Science Applications ,Cell biology ,Up-Regulation ,Transcriptional profile ,Chemistry ,030220 oncology & carcinogenesis ,Cytokines ,Tumor necrosis factor alpha ,medicine.symptom ,mesenchymal stromal cells ,Signal Transduction ,QH301-705.5 ,Population ,Subcutaneous Fat ,Down-Regulation ,Inflammation ,Biology ,Chimie inorganique ,Gene Expression Regulation/genetics ,Catalysis ,CCL5 ,Inorganic Chemistry ,Up-Regulation/genetics ,03 medical and health sciences ,Cell Proliferation/genetics ,Downregulation and upregulation ,cell-passage ,TLR ,medicine ,Humans ,Spectroscopie [état condense] ,Physical and Theoretical Chemistry ,transcriptional profile ,education ,Molecular Biology ,QD1-999 ,regulatory mediators ,Cell Proliferation ,Transcription, Genetic/genetics ,Organic Chemistry ,Mesenchymal stem cell ,Biologie moléculaire ,Mesenchymal Stem Cells ,Chimie théorique ,cytokines ,Chimie organique ,030104 developmental biology ,Spectroscopie [électromagnétisme, optique, acoustique] ,Gene Expression Regulation ,Cell culture ,Signal Transduction/genetics ,Catalyses hétérogène et homogène ,Subcutaneous Fat/metabolism - Abstract
Adult human subcutaneous adipose tissue (AT) harbors a rich population of mesenchymal stromal cells (MSCs) that are of interest for tissue repair. For this purpose, it is of utmost importance to determine the response of AT-MSCs to proliferative and inflammatory signals within the damaged tissue. We have characterized the transcriptional profile of cytokines, regulatory mediators and Toll-like receptors (TLR) relevant to the response of MSCs. AT-MSCs constitutively present a distinct profile for each gene and differentially responded to inflammation and cell-passaging. Inflammation leads to an upregulation of IL-6, IL-8, IL-1β, TNFα and CCL5 cytokine expression. Inflammation and cell-passaging increased the expression of HGF, IDO1, PTGS1, PTGS2 and TGFβ. The expression of the TLR pattern was differentially modulated with TLR 1, 2, 3, 4, 9 and 10 being increased, whereas TLR 5 and 6 downregulated. Functional enrichment analysis demonstrated a complex interplay between cytokines, TLR and regulatory mediators central for tissue repair. This profiling highlights that following a combination of inflammatory and proliferative signals, the sensitivity and responsive capacity of AT-MSCs may be significantly modified. Understanding these transcriptional changes may help the development of novel therapeutic approaches., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2021
12. The lung environment controls alveolar macrophage metabolism and responsiveness in type 2 inflammation
- Author
-
Freya R. Svedberg, Tracy Hussell, Christopher M. Evans, Peter C. Cook, Maryam Clausen, Sheila Brown, Maria Z Krauss, Gareth J. Howell, Richard K. Grencis, Danen Cunoosamy, Alasdair Ivens, Jens Madsen, Laura Campbell, Howard Clark, Andrew S. MacDonald, David J. Thornton, Catherine Sharpe, and Tara E. Sutherland
- Subjects
Lydia Becker Institute ,medicine.medical_treatment ,Immunology ,Mice, Transgenic ,Inflammation ,Larva/immunology ,Article ,Peritoneal cavity ,ResearchInstitutes_Networks_Beacons/lydia_becker_institute_of_immunology_and_inflammation ,Inflammation/genetics ,Macrophages, Alveolar ,medicine ,Animals ,Immunology and Allergy ,Macrophage ,Glucose homeostasis ,Lung ,Strongylida Infections ,Mice, Knockout ,Mice, Inbred BALB C ,Strongylida Infections/genetics ,Chemistry ,Surfactant protein D ,Interleukin-4/genetics ,Macrophage Activation ,Pulmonary Surfactant-Associated Protein D ,Mucin-5B/genetics ,Nippostrongylus/immunology ,Mucin-5B ,Pulmonary Surfactant-Associated Protein D/genetics ,Mice, Inbred C57BL ,Macrophage Activation/genetics ,medicine.anatomical_structure ,Cytokine ,Lung/immunology ,Larva ,Alveolar macrophage ,Macrophages, Alveolar/immunology ,Interleukin-4 ,Nippostrongylus ,medicine.symptom - Abstract
Fine control of macrophage activation is needed to prevent inflammatory disease, particularly at barrier sites such as the lungs.However, the dominant mechanisms that regulate the activation of pulmonary macrophages during inflammation are poorly understood. We found that alveolar macrophages (AlvMs) were much less able to respond to the canonical type 2 cytokine IL-4, which underpins allergic disease and parasitic worm infections, than macrophages from lung tissue or the peritoneal cavity. We found that the hyporesponsiveness of AlvMs to IL-4 depended upon the lung environment but was independent of the host microbiota or the lung extracellular matrix components surfactant protein D (SP-D) and mucin 5b (Muc5b). AlvMs showedseverely dysregulated metabolism relative to that of cavity macrophages. After removal from the lungs, AlvMs regained responsiveness to IL-4 in a glycolysis-dependent manner. Thus, impaired glycolysis in the pulmonary niche regulates AlvM responsiveness during type 2 inflammation.
- Published
- 2019
- Full Text
- View/download PDF
13. Micronuclei as biomarkers of DNA damage, aneuploidy, inducers of chromosomal hypermutation and as sources of pro-inflammatory DNA in humans
- Author
-
Stefano Bonassi, Claudia Bolognesi, Siegfried Knasmueller, Micheline Kirsch-Volders, Nina Holland, Michael Fenech, Fenech, Michael, Knasmueller, Siegfried, Bolognesi, Claudia, Holland, Nina, Bonassi, Stefano, Kirsch-Volders, Micheline, Cell Genetics, and Biology
- Subjects
0301 basic medicine ,Genome instability ,Genetic Markers ,DNA damage ,Health, Toxicology and Mutagenesis ,Aneuploidy ,Biology ,Chromosomal Instability/genetics ,03 medical and health sciences ,0302 clinical medicine ,Chromosome instability ,Chromosomal Instability ,Inflammation/genetics ,Genetics ,medicine ,Humans ,micronucleus ,aneuploidy ,human ,Mitosis ,Micronuclei, Chromosome-Defective ,Inflammation ,Genetic Markers/genetics ,Micronucleus Tests ,Chromosome ,medicine.disease ,Chromatin ,Cell biology ,030104 developmental biology ,inflammation ,030220 oncology & carcinogenesis ,micronuclei ,Micronucleus test - Abstract
Micronuclei (MNi) are among the most widely studied biomarkers of DNA damage and chromosomal instability in humans. They originate from chromosome fragments or intact chromosomes that are not included in daughter nuclei during mitosis. The main reasons for their formation are a lack of functional centromere in the chromosome fragments or whole chromosomes or defects in one or more of the proteins of the mitotic system that, consequently, fails to segregate chromosomes properly. Assays have been developed to measure MNi in peripheral blood lymphocytes, red blood cells as well as various types of epithelial cells such as buccal, nasal, urothelial and cervical cells. Some of the assays have been further developed into micronucleus (MN) cytome assays to include additional nuclear anomalies, cell death and nuclear division biomarkers. In addition, the use of molecular probes has been adopted widely for the purpose of understanding the mechanistic origin of MNi. MN assays in humans are used for the purpose of investigating the genotoxic effects of adverse environmental, life-style and occupational factors, genetic susceptibility to DNA damage, and for determining risk of accelerated aging and diseases affected by genomic instability such as developmental defects and cancer. The emerging new knowledge showing that chromosomes trapped in MNi can undergo a high rate of fragmentation and become massively re-arranged have highlighted the possibility that MN formation is not only a biomarker of induced DNA damage but also a mechanism that drives hypermutation. Furthermore, another line of recent research showed that DNA and chromatin leaking from disrupted MNi triggers the innate immune cGAS-STING mechanism that promotes inflammation which can cause a wide-range of age-related diseases if left unresolved. For these reasons, MN assays in humans have become an increasingly important biomarker of disease initiation and progression across all life-stages. Refereed/Peer-reviewed
- Published
- 2020
14. CARD14 Gain-of-Function Mutation Alone Is Sufficient to Drive IL-23/IL-17–Mediated Psoriasiform Skin Inflammation In Vivo
- Author
-
Deepa Mohanan, Stephan Nobbe, Lars E. French, Caroline Ospelt, Mark Mellett, Emmanuel Contassot, Barbara Meier, Phil F. Cheng, Margot Thome, Betina Kiefer, Rebekka Schairer, Takashi K. Satoh, University of Zurich, and Mellett, Mark
- Subjects
Keratinocytes ,0301 basic medicine ,Chemokine ,1303 Biochemistry ,medicine.medical_treatment ,DNA Mutational Analysis ,610 Medicine & health ,Inflammation ,Dermatology ,Biology ,Interleukin-23 ,Biochemistry ,Proinflammatory cytokine ,2708 Dermatology ,1307 Cell Biology ,Mice ,03 medical and health sciences ,Animals ,CARD Signaling Adaptor Proteins/genetics ,CARD Signaling Adaptor Proteins/metabolism ,Cells, Cultured ,Cytokines/metabolism ,DNA/genetics ,Disease Models, Animal ,Female ,Gain of Function Mutation ,Guanylate Kinases/genetics ,Guanylate Kinases/metabolism ,Humans ,Inflammation/genetics ,Inflammation/metabolism ,Inflammation/pathology ,Interleukin-17/metabolism ,Interleukin-23/metabolism ,Keratinocytes/metabolism ,Keratinocytes/pathology ,Membrane Proteins ,Psoriasis/genetics ,Psoriasis/metabolism ,Psoriasis/pathology ,Psoriasis ,1312 Molecular Biology ,Interleukin 23 ,medicine ,Molecular Biology ,Interleukin-17 ,10051 Rheumatology Clinic and Institute of Physical Medicine ,10177 Dermatology Clinic ,DNA ,Cell Biology ,medicine.disease ,CARD Signaling Adaptor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Immunology ,biology.protein ,Cytokines ,Interleukin 17 ,medicine.symptom ,Keratinocyte ,Guanylate Kinases - Abstract
Rare autosomal dominant mutations in the gene encoding the keratinocyte signaling molecule CARD14, have been associated with an increased susceptibility to psoriasis, but the physiological impact of CARD14 gain-of-function mutations remains to be fully determined in vivo. Here, we report that heterozygous mice harboring a CARD14 gain-of-function mutation (Card14ΔE138) spontaneously develop a chronic psoriatic phenotype with characteristic scaling skin lesions, epidermal thickening, keratinocyte hyperproliferation, hyperkeratosis, and immune cell infiltration. Affected skin of these mice is characterized by elevated expression of anti-microbial peptides, chemokines, and cytokines (including T helper type 17 cell-signature cytokines) and an immune infiltrate rich in neutrophils, myeloid cells, and T cells, reminiscent of human psoriatic skin. Disease pathogenesis was driven by the IL-23/IL-17 axis, and neutralization of IL-23p19, the key cytokine in maintaining T helper type 17 cell polarization, significantly reduced skin lesions and the expression of antimicrobial peptides and proinflammatory cytokines. Therefore, hyperactivation of CARD14 alone is sufficient to orchestrate the complex immunopathogenesis that drives T helper type 17-mediated psoriasis skin disease in vivo.
- Published
- 2018
- Full Text
- View/download PDF
15. A dualistic model of primary anal canal adenocarcinoma with distinct cellular origins, etiologies, inflammatory microenvironments and mutational signatures: implications for personalised medicine
- Author
-
Philippe Delvenne, Frédéric Lambert, Nathalie Piazzon, Séverine Valmary-Degano, Elodie Hendrick, Vincent Bours, Aurélie Poncin, Jean-Luc Prétet, Christiane Mougin, Lucine Vuitton, Karin Segers, Olivier Peulen, David Guenat, Patrick Roncarati, Franck Monnien, Benjamin Koopmansch, Diane Bruyère, Pascale Hubert, Laurence de Leval, William Penny, Michael Herfs, Alizée Lebeau, Christopher P. Crum, and Charles M. Quick
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Adult ,Male ,Cancer Research ,Programmed Cell Death 1 Receptor ,Adenocarcinoma/genetics ,Adenocarcinoma/pathology ,Aged ,Aged, 80 and over ,Anus Neoplasms/genetics ,Anus Neoplasms/pathology ,B7-H1 Antigen/genetics ,ErbB Receptors/genetics ,Female ,Gene Expression Regulation, Neoplastic/drug effects ,Humans ,Inflammation/genetics ,Inflammation/pathology ,Kaplan-Meier Estimate ,Lymphocytes, Tumor-Infiltrating/pathology ,Middle Aged ,Mutation ,Precision Medicine ,Prognosis ,Programmed Cell Death 1 Receptor/genetics ,Tumor Microenvironment/genetics ,Adenocarcinoma ,medicine.disease_cause ,Article ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,Tumor Microenvironment ,Medicine ,Gastrointestinal cancer ,Inflammation ,business.industry ,FOXP3 ,Anal canal ,medicine.disease ,Anus Neoplasms ,Anal canal adenocarcinoma ,3. Good health ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Anal gland ,Cancer research ,KRAS ,business - Abstract
Background Primary adenocarcinoma of the anal canal is a rare and aggressive gastrointestinal disease with unclear pathogenesis. Because of its rarity, no clear clinical practice guideline has been defined and a targeted therapeutic armamentarium has yet to be developed. The present article aimed at addressing this information gap by in-depth characterising the anal glandular neoplasms at the histologic, immunologic, genomic and epidemiologic levels. Methods In this multi-institutional study, we first examined the histological features displayed by each collected tumour (n = 74) and analysed their etiological relationship with human papillomavirus (HPV) infection. The intratumoural immune cell subsets (CD4, CD8, Foxp3), the expression of immune checkpoints (PD-1, PD-L1), the defect in mismatch repair proteins and the mutation analysis of multiple clinically relevant genes in the gastrointestinal cancer setting were also determined. Finally, the prognostic significance of each clinicopathological variable was assessed. Results Phenotypic analysis revealed two region-specific subtypes of anal canal adenocarcinoma. The significant differences in the HPV status, density of tumour-infiltrating lymphocytes, expression of immune checkpoints and mutational profile of several targetable genes further supported the separation of these latter neoplasms into two distinct entities. Importantly, anal gland/transitional-type cancers, which poorly respond to standard treatments, displayed less mutations in downstream effectors of the EGFR signalling pathway (i.e., KRAS and NRAS) and demonstrated a significantly higher expression of the immune inhibitory ligand-receptor pair PD-1/PD-L1 compared to their counterparts arising from the colorectal mucosa. Conclusions Taken together, the findings reported in the present article reveal, for the first time, that glandular neoplasms of the anal canal arise by HPV-dependent or independent pathways. These etiological differences leads to both individual immune profiles and mutational landscapes that can be targeted for therapeutic benefits.
- Published
- 2018
16. The Multifaceted S100A4 Protein in Cancer and Inflammation
- Author
-
Ambartsumian, Noona, Klingelhöfer, Jörg, Grigorian, Mariam, Ambartsumian, Noona, Klingelhöfer, Jörg, and Grigorian, Mariam
- Abstract
The metastasis-promoting S100A4 protein, a member of the S100 family, has recently been discovered as a potent factor implicated in various inflammation-associated diseases. S100A4 is involved in a range of biological functions such as angiogenesis, cell differentiation, apoptosis, motility, and invasion. Moreover, S100A4 is also a potent trigger of inflammatory processes and induces the release of cytokines and growth factors under different pathological conditions.Indeed, the release of S100A4 upon stress and mainly its pro-inflammatory role emerges as the most decisive activity in disease development, such as rheumatoid arthritis (RA), systemic sclerosis (SSc) allergy, psoriasis, and cancer. In the scope of this review, we will focus on the role of S100A4 as a mediator of pro-inflammatory pathways and its associated biological processes involved in the pathogenesis of various human noncommunicable diseases (NCDs) including cancer.
- Published
- 2019
17. Age-related macular degeneration and the complement system
- Author
-
Khandhadia, S., Cipriani, V., Yates, J.R.W., and Lotery, A.J.
- Subjects
- *
RETINAL degeneration , *DISEASES in older people , *VISION disorders , *GENE expression , *OXIDATIVE stress , *BLINDNESS - Abstract
Abstract: Age-related macular degeneration (AMD) is the leading cause of blindness in the developed world. It is a complex multifactorial disease, and despite new advances in treatment, many patients still succumb to visual impairment. The complement pathway has been implicated in the pathogenesis of many diseases, and recently variants in several genes encoding complement pathway proteins have been associated with AMD. Complement proteins have been found in histological specimens of eyes with AMD. Altered levels of both intrinsic complement proteins and activated products have been found in the circulation of patients with AMD. Complement activation may be triggered by oxidative stress, resulting from retinal exposure to incoming light; indeed an inter-play between these two pathological processes seems to exist. Finally, complement inhibitors are currently being evaluated in clinical trials. This article reviews the role of the complement system in AMD, and the potential of complement inhibition in preventing the devastating blindness resulting from this disease. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
18. Human adipose tissue H3K4me3 histone mark in adipogenic, lipid metabolism and inflammatory genes is positively associated with BMI and HOMA-IR
- Author
-
Pierre-Damien Denechaud, Francisco J. Tinahones, Daniel Castellano-Castillo, Wilfredo Oliva-Olivera, Lluis Fajas, María Isabel Queipo-Ortuño, Isabel Moreno-Indias, Fernando Cardona, Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria [Malaga, Espagne], Instituto de Investigación Biomédica de Málaga [Malaga, Espagne] (IBIMA), Universidad de Málaga [Málaga] = University of Málaga [Málaga]-Universidad de Málaga [Málaga] = University of Málaga [Málaga], Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición [Madrid, Espagne] (CIBERobn), Center for Integrative Genomics - Institute of Bioinformatics, Génopode (CIG), Swiss Institute of Bioinformatics [Lausanne] (SIB), Université de Lausanne (UNIL)-Université de Lausanne (UNIL), Department of Physiology [Lausanne, Suisse], Université de Lausanne (UNIL), Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM), Unidad de Gestión Clínica Intercentro de Oncología Médica del Hospital Virgen de la Victoria [Malaga, Espagne], Maria Isabel Queipo-Ortuño was supported by the 'Programa Nicolas Monarde', Consejeria de Salud, Junta de Andalucia, co-funded by the Fondo Europeo de Desarrollo Regional-FEDER (C-0030-2018). Daniel Castellano-Castillo was supported by a grant 'FPU' (FPU13/04211) and a fellowship 'Estancias breves FPU', Ministerio de Educacion, Cultura y Deporte cofunded by the Fondo Europeo de Desarrollo Regional-FEDER EST15/00657. Isabel Moreno-Indias was supported by a ‘‘Miguel Servet Type I’’ contract from the Instituto de Salud Carlos III (CP16/00163). Maria Isabel Queipo-Ortuño acknowledges support from the 'Miguel Servet Type II' program (CPI18/00003). Fernando Cardona acknowledges support from the 'Programa Nicolas Monarde', Consejeria de Salud, Junta de Andalucia C-0032-2016, and the Instituto de Salud Carlos III co-founded by Fondo Europeo de Desarrollo Regional - FEDER, PI11/ 02518, PI14/00082, Madrid Spain., Bodescot, Myriam, Université de Lausanne = University of Lausanne (UNIL)-Université de Lausanne = University of Lausanne (UNIL), and Université de Lausanne = University of Lausanne (UNIL)
- Subjects
0301 basic medicine ,Male ,Physiology ,Adipose tissue ,Gene Expression ,Biochemistry ,Body Mass Index ,Epigenesis, Genetic ,Histones ,0302 clinical medicine ,Immune Physiology ,Medicine and Health Sciences ,Promoter Regions, Genetic ,2. Zero hunger ,Regulation of gene expression ,[SDV.MHEP.EM] Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,Innate Immune System ,Multidisciplinary ,Adipogenesis ,biology ,Organic Compounds ,Monosaccharides ,[SDV.MHEP.EM]Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,Middle Aged ,Lipids ,Chromatin ,Chemistry ,Histone ,Cholesterol ,Physiological Parameters ,Adipose Tissue ,030220 oncology & carcinogenesis ,DNA methylation ,Physical Sciences ,Cytokines ,Medicine ,lipids (amino acids, peptides, and proteins) ,Female ,Anatomy ,Inflammation Mediators ,Research Article ,Adult ,medicine.medical_specialty ,Adipose Tissue/metabolism ,Adipose Tissue/pathology ,Chromatin/genetics ,DNA Methylation ,Gene Expression Regulation ,Histones/genetics ,Humans ,Inflammation/genetics ,Inflammation Mediators/metabolism ,Insulin Resistance ,Lipid Metabolism/genetics ,Lipoproteins ,Science ,Immunology ,Carbohydrates ,03 medical and health sciences ,Internal medicine ,DNA-binding proteins ,medicine ,Genetics ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Epigenetics ,Inflammation ,Body Weight ,Organic Chemistry ,Chemical Compounds ,Biology and Life Sciences ,Proteins ,nutritional and metabolic diseases ,Promoter ,Molecular Development ,Lipid Metabolism ,030104 developmental biology ,Endocrinology ,Biological Tissue ,Glucose ,Immune System ,biology.protein ,Chromatin immunoprecipitation ,Developmental Biology - Abstract
IntroductionAdipose tissue is considered an important metabolic tissue, in charge of energy storage as well as being able to act in systemic homeostasis and inflammation. Epigenetics involves a series of factors that are important for gene regulation or for chromatin structure, mostly DNA methylation and histone-tail modifications, which can be modified by environmental conditions (nutrition, lifestyle, smoking…). Since metabolic diseases like obesity and diabetes are closely related to lifestyle and nutrition, epigenetic deregulation could play an important role in the onset of these diseases and vice versa. However, little is known about histone marks in human adipose tissue. In a previous work, we developed a protocol for chromatin immunoprecipitation (ChIP) of frozen human adipose tissue. By using this method, this study investigates, for the first time, the H3K4 trimethylation (H3K4me3) mark (open chromatin) on the promoter of several factors involved in adipogenesis, lipid metabolism and inflammation in visceral adipose tissue (VAT) from human subjects with different degrees of body mass index (BMI) and metabolic disease.MethodologyVAT was collected and frozen at -80°C. 100 mg VAT samples were fixed in 0.5% formaldehyde and homogenized. After sonication, the sheared chromatin was immune-precipitated with an anti-H3K4me3 antibody linked to magnetic beads and purified. H3K4me3 enrichment was analyzed by qPCR for LEP, LPL, SREBF2, SCD1, PPARG, IL6, TNF and E2F1 promoters. mRNA extraction on the same samples was performed to quantify gene expression of these genes.ResultsH3K4me3 was enriched at the promoter of E2F1, LPL, SREBF2, SCD1, PPARG and IL6 in lean normoglycemic compared to morbid obese subjects with prediabetes. Accordingly H3K4me3 mark enrichment at E2F1, LPL, SREBF2, SCD1, PPARG and IL6 promoters was positively correlated with the BMI and the HOMA-IR. Regression analysis showed a strong relationship between the BMI with H3K4me3 at the promoter of E2F1 and LPL, and with mRNA levels of LEP and SCD. In the case of HOMA-IR, the regression analysis showed associations with H3K4me3 enrichment at the promoter of SCD1 and IL6, and with the mRNA of LEP and SCD1. Moreover H3K4me3 at the E2F1 promoter was positively associated to E2F1 mRNA levels.ConclusionsH3K4me3 enrichment in the promoter of LEP, LPL, SREBF2, SCD1, PPARG, IL6, TNF and E2F1 is directly associated with increasing BMI and metabolic deterioration. The H3k4me3 mark could be regulating E3F1 mRNA levels in adipose tissue, while no associations between the promoter enrichment of this mark and mRNA levels existed for the other genes studied.
- Published
- 2019
- Full Text
- View/download PDF
19. Whole Blood Gene Expression Profiling in patients undergoing colon cancer surgery identifies differential expression of genes involved in immune surveillance, inflammation and carcinogenesis
- Author
-
Hans Carl Hasselbalch, Mads Thomassen, Lasse Kjær, Mark Burton, Torben A Kruse, Ismail Gögenur, Sara Kehlet Watt, and Vibe Skov
- Subjects
0301 basic medicine ,Laparoscopic surgery ,Male ,medicine.medical_specialty ,Angiogenesis ,Colorectal cancer ,Carcinogenesis ,medicine.medical_treatment ,Inflammation ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Inflammation/genetics ,Gene expression ,Colonic Neoplasms/blood ,medicine ,Biomarkers, Tumor ,Humans ,Immunologic Surveillance ,Aged ,Aged, 80 and over ,Perioperative period ,business.industry ,Gene Expression Profiling ,Immunosuppression ,General Medicine ,medicine.disease ,Prognosis ,Surgery ,Gene expression profiling ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Female ,Laparoscopy ,Carcinogenesis/genetics ,medicine.symptom ,Biomarkers, Tumor/genetics ,business ,Immunologic Surveillance/genetics - Abstract
Introduction Cancer surgery may represent a potential risk of enhanced growth and metastatic ability of residual cancer cells due to post-operative immune dysfunction. This study identifies changes in transcription of genes involved in immune surveillance, immune suppression and carcinogenesis in the post-operative period of laparoscopic colon-cancer surgery within an ERAS regime. Methods Patients undergoing elective, curatively intended laparoscopic surgery for colon cancer stage I-III UICC were included in the study. Patients followed standard of care in an ERAS setting. Whole blood gene expression profiling (WBGP) was performed on the day prior to surgery and 1, 2, 3 and 10–14 days after surgery. Samples were collected in Paxgene tubes and Labeled cDNA was fragmented and hybridized to Affymetrix GeneChip™ 2.0. Results were corrected for multiple hypothesis testing using the false discovery rate. Pathway analysis was performed through the Molecular Signature Database. Paired fold changes of gene expression were calculated for post-operative compared to pre-operative samples. A mixed effect model was used to test differential gene expression by repeated-measures ANOVA. Results WBGP of 33,804 genes at five timepoints in six patients showed 302 significantly differentially expressed genes between samples from the day prior to surgery and the day after surgery. Pathway gene enrichment analysis showed a downregulation of immunologically relevant pathways. There was a significant downregulation of genes involved in T-cell receptor signaling, antigen presentation and NK-cell activity after surgery. Furthermore, there was an upregulation of cytokines related to metastatic ability, growth and angiogenesis. Conclusion Whole blood gene expression profiling revealed dysregulation of genes involved in immune surveillance, inflammation and carcinogenesis, after laparoscopic colon cancer surgery.
- Published
- 2018
- Full Text
- View/download PDF
20. The role of Fibroblast growth factor binding protein 1 in skin carcinogenesis and inflammation
- Author
-
Dirk H. Busch, Jan Rozman, Robert Brommage, Helmut Fuchs, Raffi Bekeredjian, Lillian Garrett, Sabine M. Hölter, Anton Wellstein, Julia Calzada-Wack, Holger Maier, T. Klopstock, Ralph Steinkamp, Yong Gu Lee, Johannes Beckers, Marion Horsch, Patrick James Beck, Martin Hrabé de Angelis, Khalid Garman, Carsten B. Schmidt-Weber, Annemarie Zimprich, Claudia Stoeger, Marcel O. Schmidt, L. Becker, Eckhard Wolf, Thure Adler, Jochen Graw, Chong Zuo, Ildiko Racz, Irina Treise, Andreas Zimmer, Alexandra Vernaleken, Juan Antonio Aguilar-Pimentel, Wolfgang Hans, Wolfgang Wurst, Mingjun Tan, Stefanie Leuchtenberger, Christoph Lengger, Martin Klingenspor, Patricia da Silva-Buttkus, Elena Tassi, Anna T. Riegel, Valerie Gailus-Durner, Birgit Rathkolb, Manuela A. Östereicher, Markus Ollert, Kristin Moreth, Frauke Neff, and Oana V. Amarie
- Subjects
0301 basic medicine ,Male ,Carrier Proteins/genetics ,Pathology ,Neoplasms, Experimental/chemically induced ,Skin Neoplasms ,Angiogenesis ,Carcinogenesis ,Fibroblast growth factor ,Biochemistry ,Mice ,Wound Healing/physiology ,Bone Marrow ,Inflammation/genetics ,Fibroblast growth factor binding ,Bone Marrow Transplantation ,Skin ,Mice, Knockout ,integumentary system ,Intracellular Signaling Peptides and Proteins ,Fgf ,Inflammation ,Wound Healing ,Up-Regulation ,medicine.anatomical_structure ,Knockout mouse ,Intercellular Signaling Peptides and Proteins ,Tetradecanoylphorbol Acetate ,Female ,Carcinogenesis/pathology ,Tetradecanoylphorbol Acetate/toxicity ,medicine.medical_specialty ,Dermatology ,Biology ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,Fibroblast ,Molecular Biology ,Matrigel ,Bone Marrow/metabolism ,Papilloma ,Cell Biology ,Squamous cell skin cancer ,Neoplasms, Experimental ,Skin Neoplasms/chemically induced ,Water Loss, Insensible ,Mice, Inbred C57BL ,030104 developmental biology ,Skin/drug effects ,Cancer research ,Carcinogens ,Papilloma/chemically induced ,Wound healing ,Carrier Proteins ,Carcinogens/toxicity - Abstract
Fibroblast growth factor-binding protein 1 (FGFBP1) is a secreted chaperone that mobilizes paracrine-acting FGFs, stored in the extracellular matrix, and presents them to their cognate receptors. FGFBP1 enhances FGF signaling including angiogenesis during cancer progression and is upregulated in various cancers. Here we evaluated the contribution of endogenous FGFBP1 to a wide range of organ functions as well as to skin pathologies using Fgfbp1-knockout mice. Relative to wild-type littermates, knockout mice showed no gross pathologies. Still, in knockout mice a significant thickening of the epidermis associated with a decreased transepidermal water loss and increased proinflammatory gene expression in the skin was detected. Also, skin carcinogen challenge by 7,12-dimethylbenz[a]anthracene/12-O-tetradecanoyl-phorbol-13-acetate resulted in delayed and reduced papillomatosis in knockout mice. This was paralleled by delayed healing of skin wounds and reduced angiogenic sprouting in subcutaneous matrigel plugs. Heterozygous green fluorescent protein (GFP)–knock-in mice revealed rapid induction of gene expression during papilloma induction and during wound healing. Examination of wild-type skin grafted onto Fgfbp1 GFP–knock-in reporter hosts and bone marrow transplants from the GFP-reporter model into wild-type hosts revealed that circulating Fgfbp1-expressing cells migrate into healing wounds. We conclude that tissue-resident and circulating Fgfbp1-expressing cells modulate skin carcinogenesis and inflammation.
- Published
- 2018
- Full Text
- View/download PDF
21. Gene expression response in peripheral blood cells of petroleum workers exposed to sub-ppm benzene levels
- Author
-
Bjørn Tore Gjertsen, Nancy B. Hopf, Rita Holdhus, Anne-Kristin Stavrum, Katarina Mariann Jørgensen, Ellen Færgestad Mosleth, Kristian Hovde Liland, and Jorunn Kirkeleit
- Subjects
0301 basic medicine ,Adult ,Male ,Benzene/adverse effects ,Gene Expression ,Inflammation/genetics ,Occupational Exposure/analysis ,Oil and Gas Industry ,benzene ,gene expression ,immune response ,inflammation ,leukaemia risk ,offshore ,petroleum industry ,Health, Toxicology and Mutagenesis ,lcsh:Medicine ,Biology ,Article ,Andrology ,03 medical and health sciences ,chemistry.chemical_compound ,Immune system ,Occupational Exposure ,Gene expression ,Humans ,Longitudinal Studies ,Benzene ,Gene ,Whole blood ,Inflammation ,Blood Cells ,Norway ,lcsh:R ,Public Health, Environmental and Occupational Health ,Middle Aged ,Crude oil ,Peripheral blood ,030104 developmental biology ,chemistry ,Gene Expression Regulation ,biology.protein ,Environmental Pollutants ,Female ,Antibody - Abstract
Altered gene expression in pathways relevant to leukaemogenesis, as well as reduced levels of circulating lymphocytes, have been reported in workers that were exposed to benzene concentrations below 1 ppm. In this study, we analysed whole blood global gene expression patterns in a worker cohort with altered levels of T cells and immunoglobulins IgM and IgA at three time points, pre-shift, post-shift (after three days), and post-recovery (12 hours later). Eight benzene exposed tank workers performing maintenance work in crude oil cargo tanks with a mean benzene exposure of 0.3 ppm (range 0.1&ndash, 0.5 ppm) and five referents considered to be unexposed were examined by gene expression arrays. By using our data as independent validation, we reanalysed selected genes that were reported to be altered from previous studies of workers being exposed to sub-ppm benzene levels Four out of six genes previously proposed as marker genes in chronically exposed workers separated benzene exposed workers from unexposed referents (CLEC5, ACSL1, PRG2, IFNB1). Even better separation of benzene exposed workers and referents was observed for short-term exposure for genes in the Jak-STAT pathway, particularly elevated expression of IL6 and reduced expression of IL19.
- Published
- 2018
22. Life-course socioeconomic status and DNA methylation of genes regulating inflammation
- Author
-
Paolo Vineis, Domenico Palli, Maria Concetta Giurdanella, Fred Paccaud, Raphaële Castagné, Giovanna Masala, Gianluca Campanella, Marc Chadeau-Hyam, Amalia Mattiello, Salvatore Panico, Silvia Stringhini, Rachel S. Kelly, Rosario Tumino, Carlotta Sacerdote, Karin van Veldhoven, Sara Grioni, Claudia Agnoli, Silvia Polidoro, Valentina Gallo, Campanella G., Stringhini, Silvia, Polidoro, Silvia, Sacerdote, Carlotta, Kelly, Rachel S, van Veldhoven, Karin, Agnoli, Claudia, Grioni, Sara, Tumino, Rosario, Giurdanella, Maria Concetta, Panico, Salvatore, Mattiello, Amalia, Palli, Domenico, Masala, Giovanna, Gallo, Valentina, Castagné, Raphaële, Paccaud, Fred, Campanella, Gianluca, Chadeau Hyam, Marc, and Vineis, Paolo
- Subjects
Male ,Candidate gene ,Epidemiology ,Context (language use) ,Genome-wide association study ,Biology ,Social Environment ,NFATC Transcription Factor ,03 medical and health sciences ,0302 clinical medicine ,Inflammation/genetics ,Humans ,Prospective Studies ,030212 general & internal medicine ,Epigenetics ,NFATC Transcription Factors/genetics ,Multivariate Analysi ,Gene ,030304 developmental biology ,life course ,Inflammation ,0303 health sciences ,DNA methylation ,NFATC Transcription Factors ,social sciences ,General Medicine ,Methylation ,DNA Methylation ,Middle Aged ,Healthy Volunteer ,Healthy Volunteers ,3. Good health ,Prospective Studie ,Italy ,Social Class ,IL1A ,socioeconomic statu ,Multivariate Analysis ,Immunology ,Female ,Genome-Wide Association Study ,Human - Abstract
BACKGROUND: In humans, low socioeconomic status (SES) across the life course is associated with greater diurnal cortisol production, increased inflammatory activity and higher circulating antibodies for several pathogens, all suggesting a dampened immune response. Recent evidence suggests that DNA methylation of pro-inflammatory genes may be implicated in the biological embedding of the social environment. METHODS: The present study examines the association between life-course SES and DNA methylation of candidate genes, selected on the basis of their involvement in SES-related inflammation, in the context of a genome-wide methylation study. Participants were 857 healthy individuals sampled from the EPIC Italy prospective cohort study. RESULTS: Indicators of SES were associated with DNA methylation of genes involved in inflammation. NFATC1, in particular, was consistently found to be less methylated in individuals with low vs high SES, in a dose-dependent manner. IL1A, GPR132 and genes belonging to the MAPK family were also less methylated among individuals with low SES. In addition, associations were found between SES and CXCL2 and PTGS2, but these genes were consistently more methylated among low SES individuals. CONCLUSIONS: Our findings support the hypothesis that the social environment leaves an epigenetic signature in cells. Although the functional significance of SES-related DNA methylation is still unclear, we hypothesize that it may link SES to chronic disease risk.
- Published
- 2015
- Full Text
- View/download PDF
23. Genetic immune and inflammatory markers associated with diabetes in solid organ transplant recipients
- Author
-
Lina Quteineh, Frederik Vandenberghe, Roger Lehmann, Nicolas J. Mueller, Pierre-Yves Bochud, Agnieszka Wójtowicz, Mike Recher, Paul Mohacsi, Jean-François Dufour, Manuel Pascual, Paola M. Soccal, Séverine Crettol, Zoltán Kutalik, Isabelle Binet, Jean-Pierre Venetz, Peter Vollenweider, Dela Golshayan, Christian van Delden, Oriol Manuel, Pedro Marques-Vidal, Jürg Steiger, Christoph Hess, Chin B. Eap, University of Zurich, Eap, Chin B, and Swiss Transplant Cohort Study
- Subjects
Male ,2747 Transplantation ,medicine.medical_treatment ,030230 surgery ,Gastroenterology ,Organ transplantation ,10234 Clinic for Infectious Diseases ,Diabetes mellitus genetics ,0302 clinical medicine ,600 Technology ,Odds Ratio ,Immunology and Allergy ,2736 Pharmacology (medical) ,ddc:576.5 ,Pharmacology (medical) ,Prospective Studies ,610 Medicine & health ,Prospective cohort study ,ddc:615 ,education.field_of_study ,ddc:617 ,Homozygote ,Immunosuppression ,Middle Aged ,2723 Immunology and Allergy ,Female ,Immunosuppressive Agents ,Switzerland ,Adult ,medicine.medical_specialty ,Heterozygote ,Adolescent ,Population ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Young Adult ,Internal medicine ,Diabetes mellitus ,medicine ,Aged ,Diabetes Mellitus/etiology ,Diabetes Mellitus/genetics ,Diabetes Mellitus/immunology ,Gene-Environment Interaction ,Humans ,Immunosuppressive Agents/therapeutic use ,Inflammation/genetics ,Inflammation/immunology ,Organ Transplantation ,Switzerland/epidemiology ,Transplant Recipients ,clinical research/practice ,diabetes ,genetics ,new onset/posttransplant ,Diabetes Mellitus ,education ,Immunosuppression Therapy ,Inflammation ,Transplantation ,business.industry ,Odds ratio ,medicine.disease ,business - Abstract
New-onset diabetes mellitus after transplantation (NODAT) is a complication following solid organ transplantation (SOT) and may be related to immune or inflammatory responses. We investigated whether single nucleotide polymorphisms (SNPs) within 158 immune- or inflammation-related genes contribute to NODAT in SOT recipients. The association between 263 SNPs and NODAT was investigated in a discovery sample of SOT recipients from the Swiss Transplant Cohort Study (STCS, n 1 = 696). Positive results were tested in a first STCS replication sample (n 2 = 489) and SNPs remaining significant after multiple test corrections were tested in a second SOT replication sample (n 3 = 156). Associations with diabetic traits were further tested in several large general population-based samples (n > 480 000). Only SP110 rs2114592C>T remained associated with NODAT in the STCS replication sample. Carriers of rs2114592-TT had 9.9 times (95% confidence interval [CI]: 3.22-30.5, P = .00006) higher risk for NODAT in the combined STCS samples (n = 1184). rs2114592C>T was further associated with NODAT in the second SOT sample (odds ratio: 4.8, 95% CI: 1.55-14.6, P = .006). On the other hand, SP110 rs2114592C>T was not associated with diabetic traits in population-based samples, suggesting a specific gene-environment interaction, possibly due to the use of specific medications (ie, immunosuppressants) in transplant patients and/or to the illness that may unmask the gene effect.
- Published
- 2017
24. Mitochondrial DNA Double-Strand Breaks in Oligodendrocytes Cause Demyelination, Axonal Injury, and CNS Inflammation
- Author
-
Roberta Brambilla, Mehran Taherian, Dmitry Ivanov, Han Gao, Galina Dvoriantchikova, Milena F. Pinto, Carlos T. Moraes, Stephanie McCarthy, Shreyans Patel, Kenji F. Tanaka, Claudia V. Pereira, Shaffiat Karmally, and Pernille M. Madsen
- Subjects
0301 basic medicine ,Central Nervous System ,Male ,Axonal loss ,Mitochondrion ,DNA, Mitochondrial/genetics ,Nerve Degeneration/genetics ,Mice ,0302 clinical medicine ,Inflammation/genetics ,DNA Breaks, Double-Stranded ,Research Articles ,General Neuroscience ,Experimental autoimmune encephalomyelitis ,Mitochondria ,Oligodendroglia/pathology ,Oligodendroglia ,medicine.anatomical_structure ,Locomotion/physiology ,Central Nervous System/pathology ,Female ,Demyelination ,Locomotion ,Mitochondrial DNA ,Encephalomyelitis, Autoimmune, Experimental ,Encephalomyelitis, Autoimmune, Experimental/genetics ,Demyelinating Diseases/genetics ,Mice, Transgenic ,Biology ,Neuroprotection ,DNA, Mitochondrial ,Multiple sclerosis ,03 medical and health sciences ,medicine ,Animals ,Animal model ,Oxidative phosphorylation ,Remyelination ,Inflammation ,medicine.disease ,Oligodendrocyte ,Axons ,Mice, Inbred C57BL ,030104 developmental biology ,Axons/pathology ,Nerve Degeneration ,Neuroscience ,030217 neurology & neurosurgery ,Demyelinating Diseases - Abstract
Mitochondrial dysfunction has been implicated in the pathophysiology of neurodegenerative disorders, including multiple sclerosis (MS). To date, the investigation of mitochondrial dysfunction in MS has focused exclusively on neurons, with no studies exploring whether dysregulation of mitochondrial bioenergetics and/or genetics in oligodendrocytes might be associated with the etiopathogenesis of MS and other demyelinating syndromes. To address this question, we established a mouse model where mitochondrial DNA (mtDNA) double-strand breaks (DSBs) were specifically induced in myelinating oligodendrocytes (PLP:mtPstI mice) by expressing a mitochondrial-targeted endonuclease, mtPstI, starting at 3 weeks of age. In both female and male mice, DSBs of oligodendroglial mtDNA caused impairment of locomotor function, chronic demyelination, glial activation, and axonal degeneration, which became more severe with time of induction. In addition, after short transient induction of mtDNA DSBs, PLP:mtPstI mice showed an exacerbated response to experimental autoimmune encephalomyelitis. Together, our data demonstrate that mtDNA damage can cause primary oligodendropathy, which in turn triggers demyelination, proving PLP:mtPstI mice to be a useful tool to study the pathological consequences of mitochondrial dysfunction in oligodendrocytes. In addition, the demyelination and axonal loss displayed by PLP:mtPstI mice recapitulate some of the key features of chronic demyelinating syndromes, including progressive MS forms, which are not accurately reproduced in the models currently available. For this reason, the PLP:mtPstI mouse represents a unique and much needed platform for testing remyelinating therapies.SIGNIFICANCE STATEMENTIn this study, we show that oligodendrocyte-specific mitochondrial DNA double-strand breaks in PLP:mtPstI mice cause oligodendrocyte death and demyelination associated with axonal damage and glial activation. Hence, PLP:mtPstI mice represent a unique tool to study the pathological consequences of mitochondrial dysfunction in oligodendrocytes, as well as an ideal platform to test remyelinating and neuroprotective agents.
- Published
- 2017
- Full Text
- View/download PDF
25. CCR6 selectively promotes monocyte mediated inflammation and atherogenesis in mice
- Author
-
Helga D. Manthey, Martin Busch, Christian Weber, Alma Zernecke, Jaroslav Pelisek, Ela Karshovska, Miriam Koch, Stefanie Barnsteiner, Rory R. Koenen, Hans-Henning Eckstein, Clément Cochain, and Sweena M. Chaudhari
- Subjects
0301 basic medicine ,Chemokine ,T-Lymphocytes ,C-C chemokine receptor type 6 ,Macrophages/immunology ,030204 cardiovascular system & hematology ,Inbred C57BL ,Monocytes ,Chemokine receptor ,Mice ,0302 clinical medicine ,Cell Movement ,Inflammation/genetics ,Receptors ,Receptors, LDL/genetics ,Cells, Cultured ,Mice, Knockout ,Cultured ,biology ,hemic and immune systems ,Hematology ,medicine.anatomical_structure ,Disease Susceptibility ,medicine.symptom ,Atherosclerosis/immunology ,Receptors, CCR6 ,Endothelium ,Cell Adhesion/genetics ,Cells ,Knockout ,Receptors, CCR6/genetics ,Cell Surface/metabolism ,LDL/genetics ,Receptors, Cell Surface ,Inflammation ,chemical and pharmacologic phenomena ,Diet, High-Fat ,03 medical and health sciences ,Immune system ,Cell Adhesion ,medicine ,Cell Movement/genetics ,Animals ,Receptors, Cell Surface/metabolism ,Animal ,Macrophages ,Monocyte ,Monocytes/immunology ,T-Lymphocytes/immunology ,Atherosclerosis ,Diet ,Mice, Inbred C57BL ,CCL20 ,Disease Models, Animal ,High-Fat ,030104 developmental biology ,Receptors, LDL ,Immunology ,Disease Models ,biology.protein ,CCR6/genetics - Abstract
SummaryThe chemokine receptor CCR6 is expressed by various cell subsets implicated in atherogenesis, such as monocytes, Th17 and regulatory T cells. In order to further define the role of CCR6 in atherosclerosis, CCR6-deficient (Ccr6 -/-) mice were crossed with low-density lipoprotein receptor-deficient (Ldlr -/-) mice to generate atherosclerosis-prone mice deficient in CCR6. Compared to Ldlr -/- controls, atherosclerotic burden in the aortic sinus and aorta were reduced in Ccr6 -/- Ldlr -/- mice fed a high fat diet, associated with a profound depression in lesional macrophage accumulation. Local and systemic distributions of T cells, including frequencies of Th1, Th17 and regulatory T cells were unaltered. In contrast, circulating counts of both Gr-1high and Gr1low monocytes were reduced in Ccr6 -/- Ldlr -/- mice. Moreover, CCR6 was revealed to promote monocyte adhesion to inflamed endothelium in vitro and leukocyte adhesion to carotid arteries in vivo. Finally, CCR6 selectively recruited monocytes but not T cells in an acute inflammatory air pouch model. We here show that CCR6 functions on multiple levels and regulates the mobilisation, adhesion and recruitment of monocytes/macrophages to the inflamed vessel, thereby promoting atherosclerosis, but is dispensable for hypercholesterolaemia-associated adaptive immune priming. Targeting CCR6 or its ligand CCL20 may therefore be a promising therapeutic strategy to alleviate atherosclerosis.Note: The review process for this manuscript was fully handled by G. Y. H. Lip, Editor in Chief.
- Published
- 2013
- Full Text
- View/download PDF
26. Biological marks of early-life socioeconomic experience is detected in the adult inflammatory transcriptome
- Author
-
Castagné, Raphaële, Kelly-Irving, Michelle, Campanella, Gianluca, Guida, Florence, Krogh, Vittorio, Palli, Domenico, Panico, Salvatore, Sacerdote, Carlotta, Tumino, Rosario, Kleinjans, Jos, de Kok, Theo, Kyrtopoulos, Soterios A, Lang, Thierry, Stringhini, Silvia, Vermeulen, Roel, Vineis, Paolo, Delpierre, Cyrille, Chadeau-Hyam, Marc, Castagné, Raphaële, Kelly Irving, Michelle, Campanella, Gianluca, Guida, Florence, Krogh, Vittorio, Palli, Domenico, Panico, Salvatore, Sacerdote, Carlotta, Tumino, Rosario, Kleinjans, Jo, de Kok, Theo, Kyrtopoulos, Soterios A, Lang, Thierry, Stringhini, Silvia, Vermeulen, Roel, Vineis, Paolo, Delpierre, Cyrille, Chadeau Hyam, Marc, RS: MHeNs - R3 - Neuroscience, RS: GROW - R1 - Prevention, Toxicogenomics, RS: FSE MaCSBio, RS: FPN MaCSBio, RS: FHML MaCSBio, Epidémiologie et analyses en santé publique : risques, maladies chroniques et handicaps (LEASP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées, Commission of the European Communities, Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM), LS IRAS EEPI GRA (Gezh.risico-analyse), dIRAS RA-2, and dIRAS RA-I&I RA
- Subjects
Inflammation ,Adult ,Male ,Gene Expression Profiling ,[SDV]Life Sciences [q-bio] ,Biomarkers/metabolism ,Female ,Genome-Wide Association Study ,Humans ,Inflammation/genetics ,Inflammation/metabolism ,Middle Aged ,Socioeconomic Factors ,Transcriptome ,Inflammation/genetics/metabolism ,Article ,Biomarkers ,ComputingMilieux_MISCELLANEOUS - Abstract
Consistent evidence is accumulating to link lower socioeconomic position (SEP) and poorer health, and the inflammatory system stands out as a potential pathway through which socioeconomic environment is biologically embedded. Using bloodderived genome-wide transcriptional profiles from 268 Italian participants of the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort, we evaluated the association between early life, young and later adulthood SEP and the expression of 845 genes involved in human inflammatory responses. These were examined individually and jointly using several inflammatory scores. Our results consistently show that participants whose father had a manual (as compared to nonmanual) occupation exhibit, later in life, a higher inflammatory score, hence indicating an overall increased level of expression for the selected inflammatory-related genes. Adopting a life course approach, these associations remained statistically significant upon adjustment for later-in-life socioeconomic experiences. Sensitivity analyses indicated that our findings were not affected by the way the inflammatory score was calculated, and were replicated in an independent study. Our study provides additional evidence that childhood SEP is associated with a sustainable upregulation of the inflammatory transcriptome, independently of subsequent socioeconomic experiences. Our results support the hypothesis that early social inequalities impacts adult physiology.
- Published
- 2016
- Full Text
- View/download PDF
27. Altered Prostasin (CAP1/Prss8) Expression Favors Inflammation and Tissue Remodeling in DSS-induced Colitis
- Author
-
Antoine Nobile, Sumedha Malsure, Olivier Bonny, Muriel Auberson, Edith Hummler, and Anna Keppner
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Colon ,medicine.medical_treatment ,Inflammation ,Inflammatory bowel disease ,03 medical and health sciences ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Genetic Predisposition to Disease ,Colitis ,Intestinal Mucosa ,Colitis/chemically induced ,Colitis/genetics ,Colon/metabolism ,Cytoskeletal Proteins/metabolism ,Dextran Sulfate ,Disease Models, Animal ,Inflammation/chemically induced ,Inflammation/genetics ,Intestinal Mucosa/metabolism ,Rats ,Serine Endopeptidases/metabolism ,Lamina propria ,business.industry ,PRSS8 ,Serine Endopeptidases ,Gastroenterology ,Histology ,Anatomy ,medicine.disease ,Ulcerative colitis ,digestive system diseases ,Cytoskeletal Proteins ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Endocrinology ,medicine.symptom ,business - Abstract
Inflammatory bowel diseases (IBD) including ulcerative colitis and Crohn's disease are diseases with impaired epithelial barrier function. We aimed to investigate whether mutated prostasin and thus, reduced colonic epithelial sodium channel activity predisposes to develop an experimentally dextran sodium sulfate (DSS)-induced colitis. Wildtype, heterozygous (fr/+), and homozygous (fr/fr) prostasin-mutant rats were treated 7 days with DSS followed by 7 days of recovery and analyzed with respect to histology, clinicopathological parameters, inflammatory marker mRNA transcript expression, and sodium transporter protein expression. In this study, a more detailed analysis on rat fr/fr colons revealed reduced numbers of crypt and goblet cells, and local angiodysplasia, as compared with heterozygous (fr/+) and wildtype littermates. Following 2% DSS treatment for 7 days followed by 7 days recovery, fr/fr animals lost body weight, and reached maximal diarrhea score and highest disease activity after only 3 days, and strongly increased cytokine levels. The histology score significantly increased in all groups, but fr/fr colons further displayed pronounced histological alterations with near absence of goblet cells, rearrangement of the lamina propria, and presence of neutrophils, eosinophils, and macrophages. Additionally, fr/fr colons showed ulcerations and edemas that were absent in fr/+ and wildtype littermates. Following recovery, fr/fr rats reached, although significantly delayed, near-normal diarrhea score and disease activity, but exhibited severe architectural remodeling, despite unchanged sodium transporter protein expression. In summary, our results demonstrate a protective role of colonic prostasin expression against experimental colitis, and thus represent a susceptibility gene in the development of inflammatory bowel disease.
- Published
- 2016
28. TNF and ROS Crosstalk in Inflammation
- Author
-
Dirk Brenner, Catherine Dostert, Heiko Blaser, and Tak W. Mak
- Subjects
0301 basic medicine ,NADPH oxidase (NOX) ,MAP Kinase Kinase 4 ,Reactive Nitrogen Species/immunology ,Regulator ,Cellular homeostasis ,Lymphocytes/immunology ,Apoptosis ,chemistry.chemical_compound ,0302 clinical medicine ,Inflammation/genetics ,Homeostasis ,Lymphocytes ,chemistry.chemical_classification ,NF-kappa B/genetics ,NF-kappa B ,Reactive Nitrogen Species ,Cell biology ,030220 oncology & carcinogenesis ,Necroptosis ,Homeostasis/immunology ,Tumor necrosis factor alpha ,medicine.symptom ,Signal Transduction ,Cell Survival ,Inflammation ,Tumor necrosis factor (TNF) ,Biology ,MAP Kinase Kinase 4/genetics ,03 medical and health sciences ,Necrosis ,Necrosis/genetics ,medicine ,Humans ,Animals ,Reactive nitrogen species ,Reactive oxygen species ,Reactive Oxygen Species/immunology ,Innate immune system ,Signal Transduction/immunology ,Tumor Necrosis Factor-alpha ,Immunity ,Reactive oxygen species (ROS) ,Cell Biology ,Immunity, Innate ,030104 developmental biology ,chemistry ,Gene Expression Regulation ,Immunology ,Tumor Necrosis Factor-alpha/genetics ,Reactive Oxygen Species - Abstract
Tumor necrosis factor (TNF) is tremendously important for mammalian immunity and cellular homeostasis. The role of TNF as a master regulator in balancing cell survival, apoptosis and necroptosis has been extensively studied in various cell types and tissues. Although these findings have revealed much about the direct impact of TNF on the regulation of NF-κB and JNK, there is now rising interest in understanding the emerging function of TNF as a regulator of the generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS). In this review we summarize work aimed at defining the role of TNF in the control of ROS/RNS signaling that influences innate immune cells under both physiological and inflammatory conditions.
- Published
- 2016
- Full Text
- View/download PDF
29. Peptide- and Amine-Modified Glucan Particles for the Delivery of Therapeutic siRNA
- Author
-
Jessica Cohen, Pranitha Vangala, Joseph C. Yawe, Michael P. Czech, Shinya U. Amano, Yuefei Shen, Sarah M. Nicoloro, Myriam Aouadi, and Michaela Tencerova
- Subjects
0301 basic medicine ,Male ,siRNA delivery ,beta-Glucans ,Obesity/genetics ,Pharmaceutical Science ,Macrophages, Peritoneal/cytology ,Peptide ,Mice ,Drug Delivery Systems ,RNA interference ,Drug Discovery ,Inflammation/genetics ,Macrophage ,Amines ,RNA, Small Interfering ,Cells, Cultured ,chemistry.chemical_classification ,glucan particle ,beta-Glucans/chemistry ,Translation (biology) ,amphipathic peptide ,Biochemistry ,Molecular Medicine ,Proteoglycans ,medicine.symptom ,Osteopontin/antagonists & inhibitors ,Peptide Fragments/chemistry ,Endosome ,Inflammation ,macrophage ,Biology ,Article ,03 medical and health sciences ,small-molecule amine ,In vivo ,RNA interference therapy ,medicine ,Animals ,Humans ,Obesity ,Glucan ,Amines/chemistry ,Genetic Therapy ,Peptide Fragments ,Mice, Inbred C57BL ,030104 developmental biology ,RNA, Small Interfering/administration & dosage ,chemistry ,inflammation ,Macrophages, Peritoneal ,Osteopontin - Abstract
Translation of siRNA technology into the clinic is limited by the need for improved delivery systems that target specific cell types. Macrophages are particularly attractive targets for RNAi therapy because they promote pathogenic inflammatory responses in a number of important human diseases. We previously demonstrated that a multicomponent formulation of β-1,3-d-glucan-encapsulated siRNA particles (GeRPs) can specifically and potently silence genes in mouse macrophages. A major advance would be to simplify the GeRP system by reducing the number of delivery components, thus enabling more facile manufacturing and future commercialization. Here we report the synthesis and evaluation of a simplified glucan-based particle (GP) capable of delivering siRNA in vivo to selectively silence macrophage genes. Covalent attachment of small-molecule amines and short peptides containing weak bases to GPs facilitated electrostatic interaction of the particles with siRNA and aided in the endosomal release of siRNA by the proton-sponge effect. Modified GPs were nontoxic and were efficiently internalized by macrophages in vitro. When injected intraperitoneally (i.p.), several of the new peptide-modified GPs were found to efficiently deliver siRNA to peritoneal macrophages in lean, healthy mice. In an animal model of obesity-induced inflammation, i.p. administration of one of the peptide-modified GPs (GP-EP14) bound to siRNA selectively reduced the expression of target inflammatory cytokines in the visceral adipose tissue macrophages. Decreasing adipose tissue inflammation resulted in an improvement of glucose metabolism in these metabolically challenged animals. Thus, modified GPs represent a promising new simplified system for the efficient delivery of therapeutic siRNAs specifically to phagocytic cells in vivo for modulation of inflammation responses.
- Published
- 2016
- Full Text
- View/download PDF
30. Whole-Genome Saliva and Blood DNA Methylation Profiling in Individuals with a Respiratory Allergy
- Author
-
Wim Vanden Berghe, Sophie Traen, Katarzyna Szarc vel Szic, Greet Schoeters, Guy Van Camp, Gudrun Koppen, Patrick De Boever, Sabine A. S. Langie, Ken Declerck, LANGIE, Sabine, Szic, Katarzyna Szarc vel, Declerck, Ken, Traen, Sophie, Koppen, Gudrun, Van Camp, Guy, Schoeters, Greet, Vanden Berghe, Wim, and DE BOEVER, Patrick
- Subjects
0301 basic medicine ,Saliva ,Leukocytes, Mononuclear/metabolism ,Pulmonology ,Physiology ,lcsh:Medicine ,Artificial Gene Amplification and Extension ,Saliva/metabolism ,Biochemistry ,Polymerase Chain Reaction ,law.invention ,0302 clinical medicine ,law ,Animal Cells ,Inflammation/genetics ,Allergies ,Medicine and Health Sciences ,Cluster Analysis ,Gene Regulatory Networks ,Hypersensitivity/blood ,lcsh:Science ,Lung ,Polymerase chain reaction ,Multidisciplinary ,DNA methylation ,Methylation ,Hematology ,Chromatin ,Body Fluids ,Nucleic acids ,Blood ,030220 oncology & carcinogenesis ,Epigenetics ,Anatomy ,Cellular Types ,DNA modification ,Engineering sciences. Technology ,Chromatin modification ,DNA Methylation/genetics ,Signal Transduction ,Research Article ,Chromosome biology ,Adult ,Cell biology ,Lung/pathology ,Immunology ,Biology ,Research and Analysis Methods ,Peripheral blood mononuclear cell ,03 medical and health sciences ,Hypersensitivity ,Genetics ,Humans ,Molecular Biology Techniques ,Molecular Biology ,Inflammation ,Blood Cells ,Biology and life sciences ,Genome, Human ,lcsh:R ,Case-control study ,Reproducibility of Results ,Correction ,Sequence Analysis, DNA ,DNA ,Asthma ,030104 developmental biology ,Case-Control Studies ,Leukocytes, Mononuclear ,Pyrosequencing ,Signal Transduction/genetics ,lcsh:Q ,Clinical Immunology ,Gene expression ,Human medicine ,Clinical Medicine - Abstract
The etiology of respiratory allergies (RA) can be partly explained by DNA methylation changes caused by adverse environmental and lifestyle factors experienced early in life. Longitudinal, prospective studies can aid in the unravelment of the epigenetic mechanisms involved in the disease development. High compliance rates can be expected in these studies when data is collected using non-invasive and convenient procedures. Saliva is an attractive biofluid to analyze changes in DNA methylation patterns. We investigated in a pilot study the differential methylation in saliva of RA (n = 5) compared to healthy controls (n = 5) using the Illumina Methylation 450K BeadChip platform. We evaluated the results against the results obtained in mononuclear blood cells from the same individuals. Differences in methylation patterns from saliva and mononuclear blood cells were clearly distinguishable (P-Adj0.1), respectively, of which 216 were in common. The methylation levels of these sites were significantly correlated between blood and saliva. The absolute levels of methylation in blood and saliva were confirmed for 3 selected DMS in the PM20D1, STK32C, and FGFR2 genes using pyrosequencing analysis. The differential methylation could only be confirmed for DMS in PM20D1 and STK32C genes in saliva. We show that saliva can be used for genome-wide methylation analysis and that it is possible to identify DMS when comparing RA cases and healthy controls. The results were replicated in blood cells of the same individuals and confirmed by pyrosequencing analysis. This study provides proof-of-concept for the applicability of saliva-based whole-genome methylation analysis in the field of respiratory allergy. SL is the beneficiary of a post-doctoral grant from the AXA Research Fund and the Cefic-LRI Innovative Science Award 2013.
- Published
- 2016
- Full Text
- View/download PDF
31. Pannexin1 as mediator of inflammation and cell death
- Author
-
Elke Decrock, Marijke De Bock, Mathieu Vinken, Luc Leybaert, Michaël Maes, Joost Willebrords, Bruno Cogliati, Scott R. Johnstone, Sara Crespo Yanguas, Pharmaceutical and Pharmacological Sciences, Faculty of Medicine and Pharmacy, Toxicology, Dermato-cosmetology and Pharmacognosy, Connexin Signalling Research Group, Liver Connexin and Pannexin Research Group, and Experimental in vitro toxicology and dermato-cosmetology
- Subjects
0301 basic medicine ,Programmed cell death ,Connexins/chemistry ,Inflammasomes ,Inflammasomes/immunology ,Nerve Tissue Proteins ,Inflammation ,Signal transduction ,Cell Death/genetics ,Biology ,Pannexin ,Nerve Tissue Proteins/chemistry ,Connexins ,Article ,Connexon ,Cytokines/biosynthesis ,03 medical and health sciences ,Cytosol ,Mediator ,Cell Movement ,Cytosol/immunology ,Inflammation/genetics ,Leukocytes ,Autophagy ,Pyroptosis ,medicine ,Humans ,Animals ,PROTEÍNAS DE TRANSPORTE ,Leukocytes/immunology ,Molecular Biology ,Cell Death ,Cell Membrane ,apoptosis ,Cell Membrane/immunology ,Cell Biology ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,inflammation ,Cytokines ,medicine.symptom - Abstract
Pannexins form channels at the plasma membrane surface that establish a pathway for communication between the cytosol of individual cells and their extracellular environment. By doing so, pannexin signaling dictates several physiological functions, but equally underlies a number of pathological processes. Indeed, pannexin channels drive inflammation by assisting in the activation of inflammasomes, the release of pro-inflammatory cytokines, and the activation and migration of leukocytes. Furthermore, these cellular pores facilitate cell death, including apoptosis, pyroptosis and autophagy. The present paper reviews the roles of pannexin channels in inflammation and cell death. In a first part, a state-of-the-art overview of pannexin channel structure, regulation and function is provided. In a second part, the mechanisms behind their involvement in inflammation and cell death are discussed.
- Published
- 2016
32. Prostaglandin-dependent modulation of dopaminergic neurotransmission elicits inflammation-induced aversion in mice
- Author
-
Fritz, Michael, Klawonn, Anna M, Nilsson, Anna, Singh, Anand Kumar, Zajdel, Joanna, Wilhelms, Daniel Björk, Lazarus, Michael, Löfberg, Andreas, Jaarola, Maarit, Kugelberg, Unn Örtegren, Billiar, Timothy R, Hackam, David J, Sodhi, Chhinder P, Breyer, Matthew D, Jakobsson, Johan, Schwaninger, Markus, Schütz, Günther, Parkitna, Jan Rodriguez, Saper, Clifford B, Blomqvist, Anders, Engblom, David, Fritz, Michael, Klawonn, Anna M, Nilsson, Anna, Singh, Anand Kumar, Zajdel, Joanna, Wilhelms, Daniel Björk, Lazarus, Michael, Löfberg, Andreas, Jaarola, Maarit, Kugelberg, Unn Örtegren, Billiar, Timothy R, Hackam, David J, Sodhi, Chhinder P, Breyer, Matthew D, Jakobsson, Johan, Schwaninger, Markus, Schütz, Günther, Parkitna, Jan Rodriguez, Saper, Clifford B, Blomqvist, Anders, and Engblom, David
- Abstract
Systemic inflammation causes malaise and general feelings of discomfort. This fundamental aspect of the sickness response reduces the quality of life for people suffering from chronic inflammatory diseases and is a nuisance during mild infections like common colds or the flu. To investigate how inflammation is perceived as unpleasant and causes negative affect, we used a behavioral test in which mice avoid an environment that they have learned to associate with inflammation-induced discomfort. Using a combination of cell-type–specific gene deletions, pharmacology, and chemogenetics, we found that systemic inflammation triggered aversion through MyD88-dependent activation of the brain endothelium followed by COX1-mediated cerebral prostaglandin E2 (PGE2) synthesis. Further, we showed that inflammation-induced PGE2 targeted EP1 receptors on striatal dopamine D1 receptor–expressing neurons and that this signaling sequence induced aversion through GABA-mediated inhibition of dopaminergic cells. Finally, we demonstrated that inflammation-induced aversion was not an indirect consequence of fever or anorexia but that it constituted an independent inflammatory symptom triggered by a unique molecular mechanism. Collectively, these findings demonstrate that PGE2-mediated modulation of the dopaminergic motivational circuitry is a key mechanism underlying the negative affect induced by inflammation.
- Published
- 2016
33. Biological pathways and genetic variables involved in pain
- Subjects
Subfamily B ,Pain/drug therapy ,ATP Binding Cassette Transporter ,Cytochrome P-450 Enzyme System/genetics ,Inflammation/genetics ,Member 1/genetics ,Analgesics/pharmacology ,Humans ,Pain Perception ,Single Nucleotide ,Synaptic Transmission/genetics ,Polymorphism - Abstract
PURPOSE: This paper summarizes current knowledge of pain-related and analgesic-related pathways as well as genetic variations involved in pain perception and management.METHODS: The pain group of the GENEQOL Consortium was given the task of summarizing the current status of research on genetic variations in pain and analgesic efficacy. This review is neither exhaustive nor comprehensive; we focus primarily on single-nucleotide polymorphisms.RESULTS: Two categories of potential genetic pain-perception pathways were identified: neurotransmission modulators and mechanisms that affect inflammation. Four categories were identified for analgesic efficacy: genes related to receptor interaction, modulation of opioid effects, metabolism, and transport. Various genetic variations involved in these pathways are proposed as candidate genetic markers for pain perception and for individual sensitivity to analgesics.CONCLUSIONS: Candidate gene association studies have been used to provide evidence for the genetic modulation of pain perception and response to analgesics. However, the nature and range of genetic modulation of pain is not well addressed due to the limited number of patients and the limited number of genes and genetic variants investigated in studies to date. Moreover, personalized analgesic treatments will require a more complete understanding of the effects of genetic variants and gene-gene interactions in response to analgesics.
- Published
- 2010
34. Biological pathways and genetic variables involved in pain
- Author
-
Qiuling, Shi, Charles S, Cleeland, Pål, Klepstad, Christine, Miaskowski, Nancy L, Pedersen, Ailko H, Zwinderman, ANS - Amsterdam Neuroscience, Neurology, Genome Analysis, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, CCA -Cancer Center Amsterdam, Cell Biology and Histology, APH - Amsterdam Public Health, Medical Psychology, and Epidemiology and Data Science
- Subjects
Candidate gene ,ATP Binding Cassette Transporter ,Cytochrome P-450 Enzyme System/genetics ,Analgesic ,Member 1/genetics ,Analgesics/pharmacology ,Pain ,Bioinformatics ,Polymorphism, Single Nucleotide ,Synaptic Transmission ,Biological pathway ,Cytochrome P-450 Enzyme System ,Pain/drug therapy ,Inflammation/genetics ,Genetic variation ,medicine ,Humans ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Polymorphism ,Gene ,Genetic association ,Inflammation ,Analgesics ,business.industry ,ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics ,Public Health, Environmental and Occupational Health ,Pain Perception ,Single Nucleotide ,Subfamily B ,Opioid ,Genetic marker ,Synaptic Transmission/genetics ,business ,medicine.drug - Abstract
PURPOSE: This paper summarizes current knowledge of pain-related and analgesic-related pathways as well as genetic variations involved in pain perception and management.METHODS: The pain group of the GENEQOL Consortium was given the task of summarizing the current status of research on genetic variations in pain and analgesic efficacy. This review is neither exhaustive nor comprehensive; we focus primarily on single-nucleotide polymorphisms.RESULTS: Two categories of potential genetic pain-perception pathways were identified: neurotransmission modulators and mechanisms that affect inflammation. Four categories were identified for analgesic efficacy: genes related to receptor interaction, modulation of opioid effects, metabolism, and transport. Various genetic variations involved in these pathways are proposed as candidate genetic markers for pain perception and for individual sensitivity to analgesics.CONCLUSIONS: Candidate gene association studies have been used to provide evidence for the genetic modulation of pain perception and response to analgesics. However, the nature and range of genetic modulation of pain is not well addressed due to the limited number of patients and the limited number of genes and genetic variants investigated in studies to date. Moreover, personalized analgesic treatments will require a more complete understanding of the effects of genetic variants and gene-gene interactions in response to analgesics.
- Published
- 2010
- Full Text
- View/download PDF
35. Docosahexaenoic acid reduces amyloid-β(1-42) secretion in human AβPP-transfected CHO-cells by mechanisms other than inflammation related to PGE₂
- Subjects
food and beverages ,CHO Cells ,Cell Membrane/genetics ,Transfection/methods ,Cell Proliferation/drug effects ,Dose-Response Relationship ,Cricetulus ,Cricetinae ,Inflammation/genetics ,Animals ,Humans ,lipids (amino acids, peptides, and proteins) ,Peptide Fragments/antagonists & inhibitors ,Drug ,Docosahexaenoic Acids/pharmacology ,Amyloid beta-Peptides/antagonists & inhibitors ,Amyloid beta-Protein Precursor/antagonists & inhibitors ,Dinoprostone/antagonists & inhibitors - Abstract
The effect of supplementation with the omega 3 polyunsaturated fatty acid (n3 PUFA) docosahexaenoic acid (DHA) on membrane composition and amyloid-β₁₋₄₂ (Aβ₄₂) secretion was studied in human amyloid-β protein precursor-transfected Chinese Hamster Ovary (CHO) cells. Twenty-four hour incubation with a range of DHA concentrations resulted in a dose-dependent increase in membrane DHA and eicosapentaenoic acid content and a decrease in arachidonic acid content. In addition, DHA supplementation caused a dose-dependent reduction in the secreted Aβ₄₂ levels and resulted in a 4-8 fold decrease in extracellular prostaglandin E₂ (PGE₂) levels. Tocopherol, which was added to DHA to prevent oxidation, may have contributed to the effect of DHA, since it slightly decreased extracellular Aβ₄₂ and PGE₂ levels when given alone. The addition of selective COX2 inhibitors Celebrex and curcumin to the culture medium resulted in a significant and comparable inhibition of PGE₂ release, but did not inhibit Aβ₄₂ secretion, and even significantly increased Aβ₄₂ production in this cell system. Together, the present data show that, whereas both DHA and COX2 inhibitors may reduce PGE₂ production, only DHA in the presence of tocopherol significantly reduced Aβ₄₂ production and concurrently changed membrane lipid composition in CHO cells. It is concluded that in this in vitro setting DHA reduced Aβ₄₂ secretion through membrane-related, but not PGE₂-related mechanisms.
- Published
- 2010
36. Involvement of MicroRNAs in the Cytotoxic Effects Exerted by Proinflammatory Cytokines on Pancreatic -Cells
- Author
-
Amar Abderrahmani, Romano Regazzi, Sonia Gattesco, Paolo Meda, Aurore Britan, E. Roggli, and Nathalie Lin-Marq
- Subjects
Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Polymerase Chain Reaction ,Mice ,0302 clinical medicine ,Genes, Reporter ,Mice, Inbred NOD ,Insulin-Secreting Cells ,Gene expression ,Cytotoxic T cell ,Lymphocytes ,Luciferases/genetics ,Luciferases ,Cells, Cultured ,NOD mice ,Oligonucleotide Array Sequence Analysis ,0303 health sciences ,Cell Death ,Inflammation/genetics/physiopathology ,Reverse Transcriptase Polymerase Chain Reaction ,Animals ,Cell Line ,Cytokines/genetics ,Female ,Gene Expression Profiling ,Humans ,Inflammation/genetics ,Inflammation/physiopathology ,Insulin-Secreting Cells/cytology ,Insulin-Secreting Cells/physiology ,Lymphocytes/cytology ,Lymphocytes/immunology ,MicroRNAs/genetics ,medicine.anatomical_structure ,Cytokine ,Insulin-Secreting Cells/cytology/*physiology ,Cytokines ,Original Article ,Lymphocytes/cytology/immunology/physiology ,030209 endocrinology & metabolism ,Biology ,Proinflammatory cytokine ,03 medical and health sciences ,microRNA ,Internal Medicine ,medicine ,ddc:612 ,030304 developmental biology ,Inflammation ,Pancreatic islets ,medicine.disease ,MicroRNAs ,Islet Studies ,MicroRNAs/*genetics ,Immunology ,Cancer research ,Cytokines/*genetics ,Insulitis - Abstract
OBJECTIVE Pancreatic β-cells exposed to proinflammatory cytokines display alterations in gene expression resulting in defective insulin secretion and apoptosis. MicroRNAs are small noncoding RNAs emerging as key regulators of gene expression. Here, we evaluated the contribution of microRNAs to cytokine-mediated β-cell cytotoxicity. RESEARCH DESIGN AND METHODS We used global microarray profiling and real-time PCR analysis to detect changes in microRNA expression in β-cells exposed to cytokines and in islets of pre-diabetic NOD mice. We assessed the involvement of the microRNAs affected in cytokine-mediated β-cell failure by modifying their expression in insulin-secreting MIN6 cells. RESULTS We found that IL-1β and TNF-α induce the expression of miR-21, miR-34a, and miR-146a both in MIN6 cells and human pancreatic islets. We further show an increase of these microRNAs in islets of NOD mice during development of pre-diabetic insulitis. Blocking miR-21, miR-34a, or miR-146a function using antisense molecules did not restore insulin-promoter activity but prevented the reduction in glucose-induced insulin secretion observed upon IL-1β exposure. Moreover, anti–miR-34a and anti–miR-146a treatment protected MIN6 cells from cytokine-triggered cell death. CONCLUSIONS Our data identify miR-21, miR-34a, and miR-146a as novel players in β-cell failure elicited in vitro and in vivo by proinflammatory cytokines, notably during the development of peri-insulitis that precedes overt diabetes in NOD mice.
- Published
- 2010
- Full Text
- View/download PDF
37. Decreased age-related cardiac dysfunction, myocardial nitrative stress, inflammatory gene expression, and apoptosis in mice lacking fatty acid amide hydrolase
- Author
-
Zoltan Ungvari, Gorgy Hasko, Anna Csiszar, Partha Mukhopadhyay, Benjamin F. Cravatt, Mohamraj Rajesh, Sandor Batkai, Lucas Liaudet, and Pal Pacher
- Subjects
Aging ,Physiology ,Apoptosis ,Monocytes ,Pathogenesis ,Mice ,Ventricular Dysfunction, Left ,chemistry.chemical_compound ,Aging/genetics ,Aging/metabolism ,Amidohydrolases/deficiency ,Amidohydrolases/genetics ,Animals ,Arachidonic Acids/metabolism ,Cell Adhesion ,Cells, Cultured ,Coronary Vessels/cytology ,Coronary Vessels/metabolism ,Endothelial Cells/metabolism ,Gene Expression Regulation ,Humans ,Inflammation/enzymology ,Inflammation/genetics ,Intercellular Adhesion Molecule-1/metabolism ,Mice, Knockout ,Monocytes/metabolism ,Myocardium/enzymology ,Myocardium/metabolism ,NF-kappa B/metabolism ,Polyunsaturated Alkamides/metabolism ,Reactive Nitrogen Species/metabolism ,Receptors, Cannabinoid/metabolism ,Tumor Necrosis Factor-alpha/metabolism ,Vascular Cell Adhesion Molecule-1/metabolism ,Ventricular Dysfunction, Left/enzymology ,Ventricular Dysfunction, Left/genetics ,Fatty acid amide hydrolase ,Receptors, Cannabinoid ,Regulation of gene expression ,NF-kappa B ,Anandamide ,Intercellular Adhesion Molecule-1 ,Coronary Vessels ,Reactive Nitrogen Species ,lipids (amino acids, peptides, and proteins) ,Tumor necrosis factor alpha ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,psychological phenomena and processes ,medicine.medical_specialty ,Polyunsaturated Alkamides ,Vascular Cell Adhesion Molecule-1 ,Inflammation ,Arachidonic Acids ,Biology ,Article ,Amidohydrolases ,Physiology (medical) ,Internal medicine ,medicine ,Reactive nitrogen species ,Tumor Necrosis Factor-alpha ,Myocardium ,Endothelial Cells ,Lipid metabolism ,Endocrinology ,nervous system ,chemistry ,Immunology ,Endocannabinoids - Abstract
Recent studies have uncovered important cross talk between inflammation, generation of reactive oxygen and nitrogen species, and lipid metabolism in the pathogenesis of cardiovascular aging. Inhibition of the endocannabinoid anandamide metabolizing enzyme, the fatty acid amide hydrolase (FAAH), is emerging as a promising novel approach for the treatment of various inflammatory disorders. In this study, we have investigated the age-associated decline of cardiac function and changes in inflammatory gene expression, nitrative stress, and apoptosis in FAAH knockout (FAAH−/−) mice and their wild-type (FAAH+/+) littermates. Additionally, we have explored the effects of anandamide on TNF-α-induced ICAM-1 and VCAM-1 expression and monocyte-endothelial adhesion in human coronary artery endothelial cells (HCAECs). There was no difference in the cardiac function (measured by the pressure-volume conductance catheter system) between 2- to 3-mo-old (young) FAAH−/− and FAAH+/+ mice. In contrast, the aging-associated decline in cardiac function and increased myocardial gene expression of TNF-α, gp91phox, matrix metalloproteinase (MMP)-2, MMP-9, caspase-3 and caspase-9, myocardial inducible nitric oxide synthase protein expression, nitrotyrosine formation, poly (ADP-ribose)polymerase cleavage and caspase-3/9 activity, observed in 28- to 31-mo-old (aging) FAAH+/+ mice, were largely attenuated in knockouts. There was no difference in the myocardial cannabinoid CB1 and CB2 receptor gene expression between young and aging FAAH−/− and FAAH+/+ mice. Anandamide dose dependently attenuated the TNF-α-induced ICAM-1 and VCAM-1 expression, NF-κB activation in HCAECs, and the adhesion of monocytes to HCAECs in a CB1- and CB2-dependent manner. These findings suggest that pharmacological inhibition of FAAH may represent a novel protective strategy against chronic inflammation, oxidative/nitrative stress, and apoptosis associated with cardiovascular aging and atherosclerosis.
- Published
- 2007
- Full Text
- View/download PDF
38. In vitro and in vivo ocular biocompatibility of electrospun poly(ɛ-caprolactone) nanofibers
- Author
-
Da Silva, Gisele Rodrigues, Lima, Tadeu Henrique, Oréfice, Rodrigo Lambert, Fernandes-Cunha, Gabriella Maria, Silva-Cunha, Armando, Zhao, Min, and Behar-Cohen, Francine
- Subjects
Retina/metabolism ,Polyesters/adverse effects ,genetic structures ,Polyesters/pharmacology ,Inflammation/metabolism ,Cytokines/metabolism ,Gene Expression/drug effects ,Neuroglia/drug effects ,eye diseases ,Vitreous Body/drug effects ,Eye/cytology ,Retina/cytology ,Retina/drug effects ,Inflammation/genetics ,Nanofibers/adverse effects ,sense organs ,Cell Survival/drug effects ,Eye/drug effects - Abstract
Biocompatibility is a requirement for the development of nanofibers for ophthalmic applications. In this study, nanofibers were elaborated using poly(ε-caprolactone) via electrospinning. The ocular biocompatibility of this material was investigated. MIO-M1 and ARPE-19 cell cultures were incubated with nanofibers and cellular responses were monitored by viability and morphology. The in vitro biocompatibility revealed that the nanofibers were not cytotoxic to the ocular cells. These cells exposed to the nanofibers proliferated and formed an organized monolayer. ARPE-19 and MIO-M1 cells were capable of expressing GFAP, respectively, demonstrating their functionality. Nanofibers were inserted into the vitreous cavity of the rat's eye for 10days and the in vivo biocompatibility was investigated using Optical Coherence Tomography (OCT), histology and measuring the expression of pro-inflammatory genes (IL-1β, TNF-α, VEGF and iNOS) (real-time PCR). The OCT and the histological analyzes exhibited the preserved architecture of the tissues of the eye. The biomaterial did not elicit an inflammatory reaction and pro-inflammatory cytokines were not expressed by the retinal cells, and the other posterior tissues of the eye. Results from the biocompatibility studies indicated that the nanofibers exhibited a high degree of cellular biocompatibility and short-term intraocular tolerance, indicating that they might be applied as drug carrier for ophthalmic use.
- Published
- 2015
39. Increased Systemic and Plaque Inflammation in ABCA1 Mutation Carriers With Attenuation by Statins
- Author
-
Fleur M. van der Valk, Sonia Tolani, Erik S.G. Stroes, Andrea E. Bochem, Marit Westerterp, Alan R. Tall, Other departments, ACS - Amsterdam Cardiovascular Sciences, Vascular Medicine, Center for Liver, Digestive and Metabolic Diseases (CLDM), and Translational Immunology Groningen (TRIGR)
- Subjects
medicine.medical_specialty ,Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use ,ATP-Binding Cassette Transporters/genetics ,Lipoproteins ,Inflammation ,Biology ,medicine.disease_cause ,Cholesterol/blood ,Cytokines/blood ,Proinflammatory cytokine ,chemistry.chemical_compound ,Internal medicine ,Macrophages/metabolism ,Inflammation/genetics ,medicine ,Lipoproteins/blood ,polycyclic compounds ,Humans ,Plaque ,Apolipoproteins B ,Mutation ,Atherosclerotic/genetics ,medicine.diagnostic_test ,Cholesterol ,Macrophages ,Reverse cholesterol transport ,Biological Transport ,Plaque, Atherosclerotic/genetics ,medicine.disease ,Plaque, Atherosclerotic ,Apolipoproteins B/blood ,Endocrinology ,chemistry ,Positron emission tomography ,ABCA1 ,Immunology ,biology.protein ,Cytokines ,ATP-Binding Cassette Transporters ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Cardiology and Cardiovascular Medicine ,Dyslipidemia - Abstract
Objective— We previously demonstrated that subjects with functional ATP-binding cassette ( ABC ) A1 mutations have increased atherosclerosis, which has been attributed to the role of ABCA1 in reverse cholesterol transport. More recently, a proinflammatory effect of Abca1 deficiency was shown in mice, potentially contributing to atherogenesis. In this study, we investigated whether ABCA1 deficiency was associated with proinflammatory changes in humans. Approach and Results— Thirty-one heterozygous, 5 homozygous ABCA1 mutation carriers, and 21 matched controls were studied. 18 Fluorodeoxyglucose positron emission tomography with computed tomographic scanning was performed in a subset of carriers and controls to assess arterial wall inflammation (target:background ratio). Heterozygous ABCA1 mutation carriers had a 20% higher target:background ratio than in controls (target:background ratio; P =0.008). In carriers using statins (n=7), target:background ratio was 21% reduced than in nonstatin users (n=7; P =0.03). We then measured plasma cytokine levels. Tumor necrosis factor α, monocyte chemoattractant protein-1, and interleukin-6 levels were increased in heterozygous and homozygous ABCA1 mutation carriers. We isolated monocytes from carriers and controls and measured inflammatory gene expression. Only TNFα mRNA was increased in monocytes from heterozygous ABCA1 mutation carriers. Additional studies in THP-1 macrophages showed that both ABCA1 deficiency and lipoprotein-deficient plasma from ABCA1 mutation carriers increased inflammatory gene expression. Conclusions— Our data suggest a proinflammatory state in ABCA1 mutation carriers as reflected by an increased positron emission tomography–MRI signal in nonstatin using subjects, and increased circulating cytokines. The increased inflammation in ABCA1 mutation carriers seems to be attenuated by statins.
- Published
- 2015
40. Hierarchical modeling identifies novel lung cancer susceptibility variants in inflammation pathways among 10,140 cases and 11,012 controls
- Author
-
Heike Bickeböller, John S. Witte, Angela Risch, Albert Rosenberger, Simone Benhamou, Joachim Heinrich, Thomas Muley, Rayjean J. Hung, Gary Chen, Juan Pablo Lewinger, Shelley B. Bull, Christine Bouchardy, Darren R. Brenner, Paul Brennan, Paolo Boffetta, Christopher I. Amos, Margaret R. Spitz, John R. McLaughlin, Chu Chen, Gary E. Goodman, Brenner, D.R., Brennan, P., Boffetta, P., Amos, C.I., Spitz, M.R., Chen, C., Goodman, G., Heinrich, J., Bickeböller, H., Rosenberger, A., Risch, A., Muley, T., McLaughlin, J.R., Benhamou, S., Bouchardy, C., Lewinger, J.P., Witte, J.S., Chen, G., Bull, S., and Hung, R.J.
- Subjects
Male ,Lung Neoplasms ,Genome-wide association study ,0302 clinical medicine ,Gene Frequency ,Models ,Inflammation/genetics ,2.1 Biological and endogenous factors ,European Continental Ancestry Group/genetics ,Aetiology ,Lung ,Genetics (clinical) ,Cancer ,Genetics ,Genetics & Heredity ,0303 health sciences ,Likelihood Functions ,Lung Cancer ,Single Nucleotide ,Statistical ,Middle Aged ,Polymorphism, Single Nucleotide/genetics ,United States/epidemiology ,3. Good health ,Europe ,Genetic Variation/genetics ,030220 oncology & carcinogenesis ,Female ,Risk ,Lung Neoplasms/epidemiology/genetics ,Canada ,Genotype ,blood test, CD4, CD8, chemotherapy, Detection, EGFR, EPHX1, Erlotinib, Germline mutations, HER3, HER4, hierarchical modeling, Immune system, Inflammation, Lung Cancer, metastasis, Modeling, Mutations, Neuregulin 1, NRG1, Oncimmune, PAML, Phase 3 trial, Polymorphism, radiation therapy, radiosurgery, radiotherapy, rc, Response, SATB1, small cell lung cancer, Synovium, Temozolomide ,In silico ,Canada/epidemiology ,Human leukocyte antigen ,Biology ,Polymorphism, Single Nucleotide ,White People ,Europe/epidemiology ,Paediatrics and Reproductive Medicine ,03 medical and health sciences ,Complementary and Alternative Medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Polymorphism ,Lung cancer ,Allele frequency ,Alleles ,ddc:613 ,030304 developmental biology ,Inflammation ,Models, Statistical ,Whites ,Prevention ,Human Genome ,Genetic Variation ,medicine.disease ,Lung cancer susceptibility ,Human genetics ,United States ,Case-Control Studies ,Multiple comparisons problem ,Genome-Wide Association Study - Abstract
Recent evidence suggests that inflammation plays a pivotal role in the development of lung cancer. In this study, we used a two-stage approach to investigate associations between genetic variants in inflammation pathways and lung cancer risk based on genome-wide association study (GWAS) data. A total of 7,650 sequence variants from 720 genes relevant to inflammation pathways were identified using keyword and pathway searches from Gene Cards and Gene Ontology databases. In Stage 1, six GWAS datasets from the International Lung Cancer Consortium were pooled (4,441 cases and 5,094 controls of European ancestry), and a hierarchical modeling (HM) approach was used to incorporate prior information for each of the variants into the analysis. The prior matrix was constructed using (1) role of genes in the inflammation and immune pathways; (2) physical properties of the variants including the location of the variants, their conservation scores and amino acid coding; (3) LD with other functional variants and (4) measures of heterogeneity across the studies. HM affected the priority ranking of variants particularly among those having low prior weights, imprecise estimates and/or heterogeneity across studies. In Stage 2, we used an independent NCI lung cancer GWAS study (5,699 cases and 5,818 controls) for in silico replication. We identified one novel variant at the level corrected for multiple comparisons (rs2741354 in EPHX2 at 8q21.1 with p value = 7.4 × 10-6), and confirmed the associations between TERT (rs2736100) and the HLA region and lung cancer risk. HM allows for prior knowledge such as from bioinformatic sources to be incorporated into the analysis systematically, and it represents a complementary analytical approach to the conventional GWAS analysis. © 2013 Springer-Verlag Berlin Heidelberg.
- Published
- 2012
41. Erk5 Activation Elicits a Vasoprotective Endothelial Phenotype via Induction of Krüppel-like Factor 4 (KLF4)*
- Author
-
Marc Schmidt, Stephan Ludwig, Matthias Goebeler, Nicole M. Schmidt, Mirco Schmolke, Dorothee Viemann, Tobias Czymai, Désirée Spiering, Johannes Roth, and Nils Ohnesorge
- Subjects
Transcriptional Activation ,Endothelium ,Kruppel-Like Transcription Factors ,Biology ,MAP Kinase Kinase 5 ,Protective Agents ,Biochemistry ,Endothelial Cells/cytology/metabolism/physiology ,Kruppel-Like Factor 4 ,Inflammation/genetics ,medicine ,Humans ,Mitogen-Activated Protein Kinase 7/metabolism/physiology ,Molecular Biology ,Transcription factor ,Kruppel-Like Transcription Factors/genetics ,Cells, Cultured ,Mitogen-Activated Protein Kinase 7 ,Regulation of gene expression ,Inflammation ,Hemostasis ,Microarray analysis techniques ,MAP Kinase Kinase 5/metabolism ,Signal Transduction/physiology ,Endothelial Cells ,Cell Biology ,Phenotype ,Cell biology ,Vasoprotective ,Vasodilation/genetics ,Vasodilation ,medicine.anatomical_structure ,Gene Expression Regulation ,KLF4 ,Immunology ,Hemostasis/genetics ,Endothelium, Vascular ,Signal transduction ,Endothelium, Vascular/cytology/physiology ,Signal Transduction - Abstract
The MEK5/Erk5 MAPK cascade has recently been implicated in the regulation of endothelial integrity and represents a candidate pathway mediating the beneficial effects of laminar flow, a major factor preventing vascular dysfunction and disease. Here we expressed a constitutively active mutant of MEK5 (MEK5D) to study the transcriptional and functional responses to Erk5 activation in human primary endothelial cells. We provide evidence that constitutive Erk5 activation elicits an overall protective phenotype characterized by increased apoptosis resistance and a decreased angiogenic, migratory, and inflammatory potential. This is supported by bioinformatic microarray analysis, which uncovered a statistical overrepresentation of corresponding functional clusters as well as a significant induction of anti-thrombotic, hemostatic, and vasodilatory genes. We identify KLF4 as a novel Erk5 target and demonstrate a critical role of this transcription factor downstream of Erk5. We show that KLF4 expression largely reproduces the protective phenotype in endothelial cells, whereas KLF4 siRNA suppresses expression of various Erk5 targets. Additionally, we show that vasoprotective statins potently induce KLF4 and KLF4-dependent gene expression via activation of Erk5. Our data underscore a major protective function of the MEK5/Erk5/KLF4 module in ECs and implicate agonistic Erk5 activation as potential strategy for treatment of vascular diseases.
- Published
- 2010
42. Transcriptional signature of human macrophages exposed to the environmental contaminant benzo(a)pyrene
- Author
-
Serge Koscielny, Lydie Sparfel, Sophie Desmots, Olivier Fardel, Alexandre R.R. Pery, Magali Boize, Marie-Laure Pinel-Marie, Signalisation et Réponses aux Agents Infectieux et Chimiques (SeRAIC), Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES), Groupe d'Etude de la Reproduction Chez l'Homme et les Mammiferes (GERHM), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Rennes 1 (UR1), Institut National de l'Environnement Industriel et des Risques (INERIS), Service d'Hématologie, Immunologie et de Thérapie Cellulaire (HITC), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Hôpital Pontchaillou-CHU Pontchaillou [Rennes], Université de Rennes (UR), Université de Rennes (UR)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Université de Rennes (UR)-Hôpital Pontchaillou-CHU Pontchaillou [Rennes]
- Subjects
p53 ,Gene Expression Regulation/drug effects ,Messenger/metabolism ,Toxicology ,CXCR5 ,chemistry.chemical_compound ,Chemokine receptor ,0302 clinical medicine ,Macrophages/drug effects ,Cell Death/drug effects ,Environmental/toxicity ,Signal Transduction/drug effects ,Inflammation/genetics ,polycyclic compounds ,microarrays ,Cells, Cultured ,Genetics ,Tumor Necrosis Factor-alpha/metabolism ,0303 health sciences ,Cultured ,Cell Death ,biology ,aryl hydrocarbon receptor ,3. Good health ,Cell biology ,macrophages ,Benzo(a)pyrene ,Interleukin-8/metabolism ,030220 oncology & carcinogenesis ,[SDV.TOX]Life Sciences [q-bio]/Toxicology ,Signal transduction ,Signal Transduction ,Cell Survival/drug effects ,Inflammation/chemically induced ,Benzo(a)pyrene/toxicity ,Cell Survival ,Cells ,Repressor ,Cell Death/genetics ,03 medical and health sciences ,Macrophages/metabolism ,Gene silencing ,Humans ,RNA, Messenger ,Gene Silencing ,Carcinogen ,Tumor Suppressor Protein p53/metabolism ,030304 developmental biology ,Inflammation ,immunotoxicity ,Tumor Necrosis Factor-alpha ,Interleukin-8 ,Immunity ,Immunity/genetics ,Aryl hydrocarbon receptor ,Carcinogens, Environmental ,Gene Expression Regulation ,chemistry ,Immune System/drug effects ,Immunity/drug effects ,Immune System ,biology.protein ,Carcinogens ,RNA ,benzo(a)pyrene ,Tumor Suppressor Protein p53 - Abstract
International audience; Polycyclic aromatic hydrocarbons (PAHs) are widely distributed immunotoxic and carcinogenic environmental contaminants, known to affect macrophages. In order to identify their molecular targets in such cells, we have analyzed gene expression profile of primary human macrophages treated by the prototypical PAH benzo(a)pyrene (BaP), using pangenomic oligonucleotides microarrays. Exposure of macrophages to BaP for 8 and 24 h resulted in 96 and 1100 genes, differentially expressed by at least a twofold change factor, respectively. Some of these targets, including the chemokine receptor CXCR5, the G protein-coupled receptor 35 (GPR35), and the Ras regulator RASAL1, have not been previously shown to be affected by PAHs, in contrast to others, such as interleukin-1beta and the aryl hydrocarbon receptor (AhR) repressor. These BaP-mediated gene regulations were fully validated by reverse transcription-quantitative polymerase chain reaction assays for some selected genes. Their bioinformatic analysis indicated that biological functions linked to immunity, inflammation, and cell death were among the most affected by BaP in human macrophages and that the AhR and p53 signaling pathways were the most significant canonical pathways activated by the PAH. AhR and p53 implications were moreover fully confirmed by the prevention of BaP-related upregulation of some selected target genes by AhR silencing or the use of pifithrin-alpha, an inhibitor of PAH bioactivation-related DNA damage/p53 pathways. Overall, these data, through identifying genes and signaling pathways targeted by PAHs in human macrophages, may contribute to better understand the molecular basis of the immunotoxicity of these environmental contaminants.
- Published
- 2010
- Full Text
- View/download PDF
43. Leptin administration downregulates the increased expression levels of genes related to oxidative stress and inflammation in the skeletal muscle of ob/ob mice
- Author
-
Neira Sáinz, Victoria Catalán, Amaia Rodríguez, Beatriz Ramírez, Sara Becerril, Gema Frühbeck, and Javier Gómez-Ambrosi
- Subjects
Leptin ,Male ,medicine.medical_specialty ,Article Subject ,Thiobarbituric acid ,Immunology ,Molecular Sequence Data ,Down-Regulation ,Mice, Obese ,Inflammation ,Biology ,medicine.disease_cause ,Thiobarbituric Acid Reactive Substances ,chemistry.chemical_compound ,Mice ,Downregulation and upregulation ,In vivo ,Internal medicine ,Inflammation/genetics ,lcsh:Pathology ,medicine ,Animals ,Muscle, Skeletal ,Leptin Deficiency ,digestive, oral, and skin physiology ,Skeletal muscle ,Cell Biology ,Microarray Analysis ,Leptin/pharmacology ,Oxidative Stress ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Gene Expression Regulation ,Gene expression regulation/drug effects ,medicine.symptom ,Oxidative stress ,hormones, hormone substitutes, and hormone antagonists ,lcsh:RB1-214 ,Research Article - Abstract
Obese leptin-deficientob/obmice exhibit a low-grade chronic inflammation together with a low muscle mass. Our aim was to analyze the changes in muscle expression levels of genes related to oxidative stress and inflammatory responses in leptin deficiency and to identify the effect ofin vivoleptin administration.Ob/obmice were divided in three groups as follows: controlob/ob, leptin-treatedob/ob(1 mg/kg/d) and leptin pair-fedob/obmice. Gastrocnemius weight was lower in controlob/obthan in wild type mice (P<.01) exhibiting an increase after leptin treatment compared to control and pair-fed (P<.01)ob/obanimals. Thiobarbituric acid reactive substances, markers of oxidative stress, were higher in serum (P<.01) and gastrocnemius (P=.05) of controlob/obthan in wild type mice and were significantly decreased (P<.01) by leptin treatment. Leptin deficiency altered the expression of 1,546 genes, while leptin treatment modified the regulation of 1,127 genes with 86 of them being involved in oxidative stress, immune defense and inflammatory response. Leptin administration decreased the high expression ofCrybb1, Hspb3, Hspb7, Mt4, Cat, Rbm9, Serpinc1andSerpinb1aobserved in controlob/obmice, indicating that it improves inflammation and muscle loss.
- Published
- 2010
44. Docosahexaenoic acid reduces amyloid-β(1-42) secretion in human AβPP-transfected CHO-cells by mechanisms other than inflammation related to PGE₂
- Author
-
de Wilde, Martijn C, van der Beek, Eline M, Kiliaan, Amanda J, Leenders, Inge, Kuipers, Almar A M, Kamphuis, Patrick J, Broersen, Laus M, Center for Liver, Digestive and Metabolic Diseases (CLDM), and Reproductive Origins of Adult Health and Disease (ROAHD)
- Subjects
Dose-Response Relationship, Drug ,food and beverages ,CHO Cells ,Cell Membrane/genetics ,Transfection/methods ,Cell Proliferation/drug effects ,Cricetulus ,Cricetinae ,Inflammation/genetics ,Animals ,Humans ,lipids (amino acids, peptides, and proteins) ,Peptide Fragments/antagonists & inhibitors ,Docosahexaenoic Acids/pharmacology ,Amyloid beta-Peptides/antagonists & inhibitors ,Amyloid beta-Protein Precursor/antagonists & inhibitors ,Dinoprostone/antagonists & inhibitors - Abstract
The effect of supplementation with the omega 3 polyunsaturated fatty acid (n3 PUFA) docosahexaenoic acid (DHA) on membrane composition and amyloid-β₁₋₄₂ (Aβ₄₂) secretion was studied in human amyloid-β protein precursor-transfected Chinese Hamster Ovary (CHO) cells. Twenty-four hour incubation with a range of DHA concentrations resulted in a dose-dependent increase in membrane DHA and eicosapentaenoic acid content and a decrease in arachidonic acid content. In addition, DHA supplementation caused a dose-dependent reduction in the secreted Aβ₄₂ levels and resulted in a 4-8 fold decrease in extracellular prostaglandin E₂ (PGE₂) levels. Tocopherol, which was added to DHA to prevent oxidation, may have contributed to the effect of DHA, since it slightly decreased extracellular Aβ₄₂ and PGE₂ levels when given alone. The addition of selective COX2 inhibitors Celebrex and curcumin to the culture medium resulted in a significant and comparable inhibition of PGE₂ release, but did not inhibit Aβ₄₂ secretion, and even significantly increased Aβ₄₂ production in this cell system. Together, the present data show that, whereas both DHA and COX2 inhibitors may reduce PGE₂ production, only DHA in the presence of tocopherol significantly reduced Aβ₄₂ production and concurrently changed membrane lipid composition in CHO cells. It is concluded that in this in vitro setting DHA reduced Aβ₄₂ secretion through membrane-related, but not PGE₂-related mechanisms.
- Published
- 2010
45. Multifaceted roles of peroxisome proliferator-activated receptors (PPARs) at the cellular and whole organism levels
- Author
-
Yessoufou, A. and Wahli, W.
- Subjects
Animals ,Autoimmune Diseases/genetics ,Autoimmune Diseases/physiopathology ,Diabetes Mellitus, Type 2/genetics ,Diabetes Mellitus, Type 2/physiopathology ,Embryonic Development/genetics ,Embryonic Development/physiology ,Female ,Gene Expression Regulation/genetics ,Gene Expression Regulation/physiology ,Humans ,Hyperlipidemias/genetics ,Hyperlipidemias/physiopathology ,Inflammation/genetics ,Inflammation/physiopathology ,Liver/physiopathology ,Metabolic Syndrome X/genetics ,Metabolic Syndrome X/physiopathology ,Peroxisome Proliferator-Activated Receptors/genetics ,Peroxisome Proliferator-Activated Receptors/physiology ,Pregnancy ,Pregnancy Maintenance/genetics ,Pregnancy Maintenance/physiology ,Repressor Proteins/genetics ,Repressor Proteins/physiology ,Sex Characteristics ,Skin Diseases/genetics ,Skin Diseases/physiopathology ,Wound Healing/genetics ,Wound Healing/physiology - Abstract
Chronic disorders, such as obesity, diabetes, inflammation, non-alcoholic fatty liver disease and atherosclerosis, are related to alterations in lipid and glucose metabolism, in which peroxisome proliferator-activated receptors (PPAR)α, PPARβ/δ and PPARγ are involved. These receptors form a subgroup of ligand-activated transcription factors that belong to the nuclear hormone receptor family. This review discusses a selection of novel PPAR functions identified during the last few years. The PPARs regulate processes that are essential for the maintenance of pregnancy and embryonic development. Newly found hepatic functions of PPARα are the mediation of female-specific gene repression and the protection of the liver from oestrogen induced toxicity. PPARα also controls lipid catabolism and is the target of hypolipidaemic drugs, whereas PPARγ controls adipocyte differentiation and regulates lipid storage; it is the target for the insulin sensitising thiazolidinediones used to treat type 2 diabetes. Activation of PPARβ/δ increases lipid catabolism in skeletal muscle, the heart and adipose tissue. In addition, PPARβ/δ ligands prevent weight gain and suppress macrophage derived inflammation. In fact, therapeutic benefits of PPAR ligands have been confirmed in inflammatory and autoimmune diseases, such as encephalomyelitis and inflammatory bowel disease. Furthermore, PPARs promote skin wound repair. PPARα favours skin healing during the inflammatory phase that follows injury, whilst PPARβ/δ enhances keratinocyte survival and migration. Due to their collective functions in skin, PPARs represent a major research target for our understanding of many skin diseases. Taken altogether, these functions suggest that PPARs serve as physiological sensors in different stress situations and remain valuable targets for innovative therapies.
- Published
- 2010
46. Y1 receptor knockout increases nociception and prevents the anti-allodynic actions of NPY
- Author
-
K.E. Kuphal, B. Solway, Bradley K. Taylor, and T. Pedrazzini
- Subjects
Male ,medicine.medical_specialty ,Hot Temperature ,Endocrinology, Diabetes and Metabolism ,Analgesic ,Anti-Inflammatory Agents ,Pain ,Arginine ,Article ,Mice ,Internal medicine ,mental disorders ,medicine ,Animals ,Neuropeptide Y ,Pain Measurement ,Inflammation ,Mice, Knockout ,Analgesics ,Nutrition and Dietetics ,Behavior, Animal ,business.industry ,Antagonist ,Neuropeptide Y receptor ,humanities ,Receptors, Neuropeptide Y ,Disease Models, Animal ,Nociception ,Allodynia ,Endocrinology ,Analgesics/pharmacology ,Anti-Inflammatory Agents/pharmacology ,Arginine/administration & dosage ,Arginine/analogs & derivatives ,Behavior, Animal/drug effects ,Hot Temperature/adverse effects ,Hyperalgesia/drug therapy ,Hyperalgesia/genetics ,Inflammation/drug therapy ,Inflammation/genetics ,Neuropeptide Y/genetics ,Neuropeptide Y/pharmacology ,Pain/drug therapy ,Pain/genetics ,Pain Measurement/methods ,Pain Measurement/statistics & numerical data ,Receptors, Neuropeptide Y/physiology ,Sciatic Neuropathy/drug therapy ,Sciatic Neuropathy/genetics ,Hyperalgesia ,Neuropathic pain ,Sciatic nerve ,medicine.symptom ,Sciatic Neuropathy ,business - Abstract
Objective Recent pharmacologic studies in our laboratory have suggested that the spinal neuropeptide Y (NPY) Y1 receptor contributes to pain inhibition and to the analgesic effects of NPY. To rule out off-target effects, the present study used Y1-receptor–deficient (−/−) mice to further explore the contribution of Y1 receptors to pain modulation. Methods and results Y1−/− mice exhibited reduced latency in the hotplate test of acute pain and a longer-lasting heat allodynia in the complete Freund's adjuvant (CFA) model of inflammatory pain. Y1 deletion did not change CFA-induced inflammation. Upon targeting the spinal NPY systems with intrathecal drug delivery, NPY reduced tactile and heat allodynia in the CFA model and the partial sciatic nerve ligation model of neuropathic pain. Importantly, we show for the first time that NPY does not exert these anti-allodynic effects in Y1−/− mice. Furthermore, in nerve-injured CD1 mice, concomitant injection of the potent Y1 antagonist BIBO3304 prevented the anti-allodynic actions of NPY. Neither NPY nor BIBO3304 altered performance on the Rotorod test, arguing against an indirect effect of motor function. Conclusion The Y1 receptor contributes to pain inhibition and to the analgesic effects of NPY.
- Published
- 2007
47. Leptin administration downregulates the increased expression levels of genes related to oxidative stress and inflammation in the skeletal muscle of ob/ob mice
- Author
-
Sainz, N. (Neira)
- Subjects
- Gene expression regulation/drug effects, Inflammation/genetics, Leptin/pharmacology
- Abstract
Obese leptin-deficient ob/ob mice exhibit a low-grade chronic inflammation together with a low muscle mass. Our aim was to analyze the changes in muscle expression levels of genes related to oxidative stress and inflammatory responses in leptin deficiency and to identify the effect of in vivo leptin administration. Ob/ob mice were divided in three groups as follows: control ob/ob, leptintreated ob/ob (1 mg/kg/d) and leptin pair-fed ob/ob mice. Gastrocnemius weight was lower in control ob/ob than in wild type mice (P < .01) exhibiting an increase after leptin treatment compared to control and pair-fed (P < .01) ob/ob animals. Thiobarbituric acid reactive substances, markers of oxidative stress, were higher in serum (P < .01) and gastrocnemius (P = .05) of control ob/ob than in wild type mice and were significantly decreased (P < .01) by leptin treatment. Leptin deficiency altered the expression of 1,546 genes, while leptin treatment modified the regulation of 1,127 genes with 86 of them being involved in oxidative stress, immune defense and inflammatory response. Leptin administration decreased the high expression of Crybb1, Hspb3, Hspb7, Mt4, Cat, Rbm9, Serpinc1 and Serpinb1a observed in control ob/ob mice, indicating that it improves inflammation and muscle loss.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.