Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Nat Med ; 29(11): 2939-2953, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37903863

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) is the commonest cause of chronic liver disease worldwide and represents an unmet precision medicine challenge. We established a retrospective national cohort of 940 histologically defined patients (55.4% men, 44.6% women; median body mass index 31.3; 32% with type 2 diabetes) covering the complete MASLD severity spectrum, and created a secure, searchable, open resource (SteatoSITE). In 668 cases and 39 controls, we generated hepatic bulk RNA sequencing data and performed differential gene expression and pathway analysis, including exploration of gender-specific differences. A web-based gene browser was also developed. We integrated histopathological assessments, transcriptomic data and 5.67 million days of time-stamped longitudinal electronic health record data to define disease-stage-specific gene expression signatures, pathogenic hepatic cell subpopulations and master regulator networks associated with adverse outcomes in MASLD. We constructed a 15-gene transcriptional risk score to predict future hepatic decompensation events (area under the receiver operating characteristic curve 0.86, 0.81 and 0.83 for 1-, 3- and 5-year risk, respectively). Additionally, thyroid hormone receptor beta regulon activity was identified as a critical suppressor of disease progression. SteatoSITE supports rational biomarker and drug development and facilitates precision medicine approaches for patients with MASLD.


Subject(s)
Diabetes Mellitus, Type 2 , Fatty Liver , Metabolic Diseases , Male , Humans , Female , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Retrospective Studies , Body Mass Index
2.
Methods Mol Biol ; 2076: 109-117, 2020.
Article in English | MEDLINE | ID: mdl-31586324

ABSTRACT

Gene expression analysis by microarray and more recently by next-generation sequencing has become a core part of biomedical research and its value can be seen in thousands of research papers. A successful gene expression experiment needs to be augmented by specialized data mining techniques if the data are to be fully exploited. Here, tools that concentrate on three areas-gene enrichment analysis, literature mining, and transcription factor binding site analysis-are described for the novice user of microarray and next generation sequencing technologies. The focus of this chapter is on free, publicly available, web-based tools.


Subject(s)
Data Mining , Diabetes Mellitus, Type 2/genetics , Gene Expression Profiling , Transcriptome , Computational Biology/methods , Data Mining/methods , Databases, Genetic , Diabetes Mellitus, Type 2/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans
4.
Endocr Connect ; 6(7): 446-457, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28720595

ABSTRACT

Chronic ACTH exposure is associated with adrenal hypertrophy and steroidogenesis. The underlying molecular processes in mice have been analysed by microarray, histological and immunohistochemical techniques. Synacthen infused for 2 weeks markedly increased adrenal mass and plasma corticosterone levels. Microarray analysis found greater than 2-fold changes in expression of 928 genes (P < 0.001; 397 up, 531 down). These clustered in pathways involved in signalling, sterol/lipid metabolism, cell proliferation/hypertrophy and apoptosis. Signalling genes included some implicated in adrenal adenomas but also upregulated genes associated with cyclic AMP and downregulated genes associated with aldosterone synthesis. Sterol metabolism genes were those promoting cholesterol supply (Scarb1, Sqle, Apoa1) and disposal (Cyp27a1, Cyp7b1). Oil red O staining showed lipid depletion consistent with reduced expression of genes involved in lipid synthesis. Genes involved in steroidogenesis (Star, Cyp11a1, Cyp11b1) were modestly affected (P < 0.05; <1.3-fold). Increased Ki67, Ccna2, Ccnb2 and Tk1 expression complemented immunohistochemical evidence of a 3-fold change in cell proliferation. Growth arrest genes, Cdkn1a and Cdkn1c, which are known to be active in hypertrophied cells, were increased >4-fold and cross-sectional area of fasciculata cells was 2-fold greater. In contrast, genes associated with apoptosis (eg Casp12, Clu,) were downregulated and apoptotic cells (Tunel staining) were fewer (P < 0.001) and more widely distributed throughout the cortex. In summary, long-term steroidogenesis with ACTH excess is sustained by genes controlling cholesterol supply and adrenal mass. ACTH effects on adrenal morphology and genes controlling cell hypertrophy, proliferation and apoptosis suggest the involvement of different cell types and separate molecular pathways.

5.
Stem Cells Transl Med ; 6(4): 1202-1206, 2017 04.
Article in English | MEDLINE | ID: mdl-28186687

ABSTRACT

The ability to identify and stratify patients that will respond to specific therapies has been transformational in a number of disease areas, particularly oncology. It is anticipated that this will also be the case for cell-based therapies, particularly in complex and heterogeneous diseases such as rheumatoid arthritis (RA). Recently, clinical results with expanded allogenic adipose-derived mesenchymal stem cells (eASCs) have indicated clinical efficacy in highly refractory RA patients. In this study, we set out to determine if circulating microRNAs (miRNAs) could be identified as potential biomarkers associated with response to eASCs in these RA patients. The miRNA expression profiles of pre-treatment plasma samples from responder and nonresponder patients were determined using microarrays. Ten miRNAs were identified that were differentially expressed in the responder group as compared to the nonresponder group. To confirm the differential expression of these 10 miRNA biomarkers, they were further assayed by quantitative reverse-transcriptase polymerase chain reaction (QRT-PCR). From this analysis, three miRNAs, miR-26b-5p, miR-487b-3p and miR-495-3p, were confirmed as being statistically significantly upregulated in the responder group as compared with the nonresponder group. Receiver operating characteristic analysis confirmed their diagnostic potential. These miRNAs could represent novel candidate stratification biomarkers associated with RA patient response to eASCs and are worthy of further clinical validation. Stem Cells Translational Medicine 2017;6:1202-1206.


Subject(s)
Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/therapy , Biomarkers/blood , Mesenchymal Stem Cells/cytology , MicroRNAs/blood , Cell- and Tissue-Based Therapy , Gene Expression Profiling , Humans , Mesenchymal Stem Cells/physiology , ROC Curve
7.
Nat Med ; 22(7): 771-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27270587

ABSTRACT

The discovery of genetic mechanisms for resistance to obesity and diabetes may illuminate new therapeutic strategies for the treatment of this global health challenge. We used the polygenic 'lean' mouse model, which has been selected for low adiposity over 60 generations, to identify mitochondrial thiosulfate sulfurtransferase (Tst; also known as rhodanese) as a candidate obesity-resistance gene with selectively increased expression in adipocytes. Elevated adipose Tst expression correlated with indices of metabolic health across diverse mouse strains. Transgenic overexpression of Tst in adipocytes protected mice from diet-induced obesity and insulin-resistant diabetes. Tst-deficient mice showed markedly exacerbated diabetes, whereas pharmacological activation of TST ameliorated diabetes in mice. Mechanistically, TST selectively augmented mitochondrial function combined with degradation of reactive oxygen species and sulfide. In humans, TST mRNA expression in adipose tissue correlated positively with insulin sensitivity in adipose tissue and negatively with fat mass. Thus, the genetic identification of Tst as a beneficial regulator of adipocyte mitochondrial function may have therapeutic significance for individuals with type 2 diabetes.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Mitochondria/metabolism , Obesity/genetics , Thiosulfate Sulfurtransferase/genetics , Animals , Cell Differentiation , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Gene Knock-In Techniques , Glucose Clamp Technique , Glucose Tolerance Test , Humans , Mice , Mice, Inbred Strains , Mice, Transgenic , Models, Animal , Molecular Targeted Therapy , Obesity/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Thiosulfate Sulfurtransferase/metabolism
8.
Lipids Health Dis ; 14: 86, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26256740

ABSTRACT

BACKGROUND: Cardiovascular disease (CVD) remains the major cause of excess mortality in patients with non-alcoholic fatty liver disease (NAFLD). The aim of this study was to investigate the individual contribution of NAFLD to CVD risk factors in the absence of pathogenic influences from other comorbidities often found in NAFLD patients, by using an established in-vitro model of hepatic steatosis. METHODS: Histopathological events in non-alcoholic fatty liver disease were recapitulated by focused metabolic nutrient overload of hepatoblastoma C3A cells, using oleate-treated-cells and untreated controls for comparison. Microarray and proteomic data from cell culture experiments were integrated into a custom-built systems biology database and proteogenomics analysis performed. Candidate genes with significant dysregulation and concomitant changes in protein abundance were identified and STRING association and enrichment analysis performed to identify putative pathogenic pathways. RESULTS: The search strategy yielded 3 candidate genes that were specifically and significantly up-regulated in nutrient-overloaded cells compared to untreated controls: fibrinogen alpha chain (2.2 fold), fibrinogen beta chain (2.3 fold) and fibrinogen gamma chain (2.1 fold) (all rank products pfp <0.05). Fibrinogen alpha and gamma chain also demonstrated significant concomitant increases in protein abundance (3.8-fold and 2.0-fold, respectively, p <0.05). CONCLUSIONS: In-vitro modelling of NAFLD and reactive oxygen species formation in nutrient overloaded C3A cells, in the absence of pathogenic influences from other comorbidities, suggests that NAFLD is an isolated determinant of CVD. Nutrient overload-induced up-regulation of all three fibrinogen component subunits of the coagulation cascade provides a possible mechanism to explain the excess CVD mortality observed in NAFLD patients.


Subject(s)
Cardiovascular Diseases/etiology , Fibrinogen/biosynthesis , Models, Biological , Non-alcoholic Fatty Liver Disease/metabolism , Cell Line, Tumor , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Genetic Association Studies , Humans , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Oligonucleotide Array Sequence Analysis , Proteomics , Risk Factors , Signal Transduction , Up-Regulation
9.
Curr Biol ; 25(5): 577-88, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25702581

ABSTRACT

BACKGROUND: Cells undergoing apoptosis are known to modulate their tissue microenvironments. By acting on phagocytes, notably macrophages, apoptotic cells inhibit immunological and inflammatory responses and promote trophic signaling pathways. Paradoxically, because of their potential to cause death of tumor cells and thereby militate against malignant disease progression, both apoptosis and tumor-associated macrophages (TAMs) are often associated with poor prognosis in cancer. We hypothesized that, in progression of malignant disease, constitutive loss of a fraction of the tumor cell population through apoptosis could yield tumor-promoting effects. RESULTS: Here, we demonstrate that apoptotic tumor cells promote coordinated tumor growth, angiogenesis, and accumulation of TAMs in aggressive B cell lymphomas. Through unbiased "in situ transcriptomics" analysis-gene expression profiling of laser-captured TAMs to establish their activation signature in situ-we show that these cells are activated to signal via multiple tumor-promoting reparatory, trophic, angiogenic, tissue remodeling, and anti-inflammatory pathways. Our results also suggest that apoptotic lymphoma cells help drive this signature. Furthermore, we demonstrate that, upon induction of apoptosis, lymphoma cells not only activate expression of the tumor-promoting matrix metalloproteinases MMP2 and MMP12 in macrophages but also express and process these MMPs directly. Finally, using a model of malignant melanoma, we show that the oncogenic potential of apoptotic tumor cells extends beyond lymphoma. CONCLUSIONS: In addition to its profound tumor-suppressive role, apoptosis can potentiate cancer progression. These results have important implications for understanding the fundamental biology of cell death, its roles in malignant disease, and the broader consequences of apoptosis-inducing anti-cancer therapy.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Neoplastic/physiology , Lymphoma, B-Cell/physiopathology , Phagocytes/physiology , Signal Transduction/physiology , Tumor Microenvironment/physiology , Analysis of Variance , Cell Proliferation/physiology , Fluorescence , Gene Expression Profiling , Histological Techniques , Humans , Kaplan-Meier Estimate , Macrophages/physiology , Matrix Metalloproteinases/metabolism , Melanoma, Experimental/physiopathology , Neovascularization, Pathologic/physiopathology
10.
Respir Res ; 16: 1, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25567521

ABSTRACT

BACKGROUND: Chronic Obstructive Pulmonary Disease (COPD) has significant systemic effects beyond the lungs amongst which muscle wasting is a prominent contributor to exercise limitation and an independent predictor of morbidity and mortality. The molecular mechanisms leading to skeletal muscle dysfunction/wasting are not fully understood and are likely to be multi-factorial. The need to develop therapeutic strategies aimed at improving skeletal muscle dysfunction/wasting requires a better understanding of the molecular mechanisms responsible for these abnormalities. Microarrays are powerful tools that allow the investigation of the expression of thousands of genes, virtually the whole genome, simultaneously. We aim at identifying genes and molecular pathways involved in skeletal muscle wasting in COPD. METHODS: We assessed and compared the vastus lateralis transcriptome of COPD patients with low fat free mass index (FFMI) as a surrogate of muscle mass (COPDL) (FEV1 30 ± 3.6%pred, FFMI 15 ± 0.2 Kg.m(-2)) with patients with COPD and normal FFMI (COPDN) (FEV1 44 ± 5.8%pred, FFMI 19 ± 0.5 Kg.m(-2)) and a group of age and sex matched healthy controls (C) (FEV1 95 ± 3.9%pred, FFMI 20 ± 0.8 Kg.m(-2)) using Agilent Human Whole Genome 4x44K microarrays. The altered expression of several of these genes was confirmed by real time TaqMan PCR. Protein levels of P21 were assessed by immunoblotting. RESULTS: A subset of 42 genes was differentially expressed in COPDL in comparison to both COPDN and C (PFP < 0.05; -1.5 ≥ FC ≥ 1.5). The altered expression of several of these genes was confirmed by real time TaqMan PCR and correlated with different functional and structural muscle parameters. Five of these genes (CDKN1A, GADD45A, PMP22, BEX2, CGREF1, CYR61), were associated with cell cycle arrest and growth regulation and had been previously identified in studies relating muscle wasting and ageing. Protein levels of CDKN1A, a recognized marker of premature ageing/cell cycle arrest, were also found to be increased in COPDL. CONCLUSIONS: This study provides evidence of differentially expressed genes in peripheral muscle in COPD patients corresponding to relevant biological processes associated with skeletal muscle wasting and provides potential targets for future therapeutic interventions to prevent loss of muscle function and mass in COPD.


Subject(s)
Adiposity , Cell Cycle Proteins/genetics , Gene Expression Profiling/methods , Muscular Atrophy/genetics , Oligonucleotide Array Sequence Analysis , Pulmonary Disease, Chronic Obstructive/genetics , Quadriceps Muscle/chemistry , RNA, Messenger/genetics , Aged , Blotting, Western , Case-Control Studies , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Genetic Markers , Genome-Wide Association Study , Humans , Male , Muscular Atrophy/diagnosis , Muscular Atrophy/physiopathology , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/physiopathology , Quadriceps Muscle/pathology , Quadriceps Muscle/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome
11.
Nephrology (Carlton) ; 19(12): 802-13, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25196678

ABSTRACT

AIMS: Regression of albuminuria and renal fibrosis occurs in patients with diabetic nephropathy (DN) following tight control of blood glucose and blood pressure, however the pathways that promote regression remain poorly understood and we wished to characterize these using a rodent model. METHODS: Diabetes was induced with streptozotocin in Cyp1a1mRen2 rats and hypertension was generated by inducing renin transgene expression with dietary indole-3-carbinol (I-3-C) for 28 weeks. At this point an 'injury cohort' was culled, while in a 'reversal cohort' glycaemia was tightly controlled using insulin implants and blood pressure normalized by withdrawing dietary I-3-C for a further 8 weeks. Pathways activated during and following reversal of diabetes and hypertension were assessed by microarray profiling. RESULTS: Tight control of blood glucose and blood pressure reduced albuminuria and renal hypertrophy, but had no impact on renal fibrosis. 85 genes were up-regulated specifically during the injury phase, including genes encoding multiple myofibroblast and extracellular matrix (ECM) proteins. Conversely, 314 genes remained persistently elevated during reversal including genes linked to innate/adaptive immunity, phagocytosis, lysosomal processing and degradative metalloproteinases (MMPs). Despite increased MMP gene expression, MMP activity was suppressed during both injury and reversal, in association with up-regulation of tissue inhibitor of metalloproteinase-1 (TIMP-1) protein. Physical separation of the TIMP-1/MMP complexes during zymography of tissue homogenate restored MMP activity. CONCLUSION: Normalization of blood glucose and pressure ameliorates albuminuria and inhibits excess ECM production, however persistent TIMP-1 expression hinders attempts at ECM remodelling. Therapies which counteract the action of TIMPs may accelerate scar resolution.


Subject(s)
Blood Glucose/drug effects , Blood Pressure , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Extracellular Matrix/metabolism , Hypertension/physiopathology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Kidney/drug effects , Albuminuria/metabolism , Albuminuria/pathology , Albuminuria/physiopathology , Albuminuria/prevention & control , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cytochrome P-450 CYP1A1/genetics , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/blood , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Extracellular Matrix/genetics , Fibrosis , Gene Expression Profiling/methods , Hypertension/genetics , Indoles , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Rats, Transgenic , Renin/genetics , Renin/metabolism , Time Factors , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
12.
PLoS One ; 9(4): e94680, 2014.
Article in English | MEDLINE | ID: mdl-24732937

ABSTRACT

Maternal obesity is linked with increased adverse pregnancy outcomes for both mother and child. The metabolic impact of excessive fat within the context of pregnancy is not fully understood. We used a mouse model of high fat (HF) feeding to induce maternal obesity to identify adipose tissue-mediated mechanisms driving metabolic dysfunction in pregnant and non-pregnant obese mice. As expected, chronic HF-feeding for 12 weeks preceding pregnancy increased peripheral (subcutaneous) and visceral (mesenteric) fat mass. However, unexpectedly at late gestation (E18.5) HF-fed mice exhibited a remarkable normalization of visceral but not peripheral adiposity, with a 53% reduction in non-pregnant visceral fat mass expressed as a proportion of body weight (P<0.001). In contrast, in control animals, pregnancy had no effect on visceral fat mass proportion. Obesity exaggerated glucose intolerance at mid-pregnancy (E14.5). However by E18.5, there were no differences, in glucose tolerance between obese and control mice. Transcriptomic analysis of visceral fat from HF-fed dams at E18.5 revealed reduced expression of genes involved in de novo lipogenesis (diacylglycerol O-acyltransferase 2--Dgat2) and inflammation (chemokine C-C motif ligand 20--Ccl2) and upregulation of estrogen receptor α (ERα) compared to HF non pregnant. Attenuation of adipose inflammation was functionally confirmed by a 45% reduction of CD11b+CD11c+ adipose tissue macrophages (expressed as a proportion of all stromal vascular fraction cells) in HF pregnant compared to HF non pregnant animals (P<0.001). An ERα selective agonist suppressed both de novo lipogenesis and expression of lipogenic genes in adipocytes in vitro. These data show that, in a HF model of maternal obesity, late gestation is associated with amelioration of visceral fat hypertrophy, inflammation and glucose intolerance, and suggest that these effects are mediated in part by elevated visceral adipocyte ERα signaling.


Subject(s)
Adipocytes/cytology , Adiposity , Estrogens/metabolism , Intra-Abdominal Fat/metabolism , Mice, Obese , Adipocytes/metabolism , Animals , Female , Glucose/metabolism , Glucose Tolerance Test , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Pregnancy , Pregnancy, Animal , Receptors, Estrogen/metabolism , Signal Transduction , Time Factors , Transcriptome
13.
J Cell Sci ; 126(Pt 20): 4769-81, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23943867

ABSTRACT

Mitochondria are the main producers of ATP, the principal energy source of the cell, and reactive oxygen species (ROS), important signaling molecules. Mitochondrial morphogenesis and function depend on a hierarchical network of mechanisms in which proteases appear to be center stage. The invadolysin gene encodes an essential conserved metalloproteinase of the M8 family that is necessary for mitosis and cell migration during Drosophila development. We previously demonstrated that invadolysin is found associated with lipid droplets in cells. Here, we present data demonstrating that invadolysin interacts physically with three mitochondrial ATP synthase subunits. Our studies have focused on the genetic phenotypes of invadolysin and bellwether, the Drosophila homolog of ATP synthase α, mutants. The invadolysin mutation presents defects in mitochondrial physiology similar to those observed in bellwether mutants. The invadolysin and bellwether mutants have parallel phenotypes that affect lipid storage and mitochondrial electron transport chain activity, which result in a reduction in ATP production and an accumulation of ROS. As a consequence, invadolysin mutant larvae show lower energetic status and higher oxidative stress. Our data demonstrate an essential role for invadolysin in mitochondrial function that is crucial for normal development and survival.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Metalloendopeptidases/physiology , Metalloproteases/metabolism , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Profiling , Mass Spectrometry , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Metalloproteases/genetics , Mitochondria/genetics , Mitochondria/metabolism , Reactive Oxygen Species
14.
Eur Heart J ; 34(13): 993-1001, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23161703

ABSTRACT

AIMS: A pooled analysis of 14 genome-wide association studies revealed 23 susceptibility loci for coronary artery disease (CAD), thereby providing the most comprehensive genetic blueprint of CAD susceptibility. Here, we evaluated whether these 23 loci also predispose to recurrent myocardial infarction (MI) or cardiac death following an acute coronary syndrome (ACS). METHODS AND RESULTS: A total of 2099 ACS patients enrolled in the Global Registry of Acute Coronary Events (GRACE) UK-Belgian study were prospectively followed for a median of 5 years (1668 days). C-allele carriers of the rs579459 variant, which is located upstream of the ABO gene and correlates with blood group A, were independently associated with recurrent MI [multivariable-adjusted hazard ratio (HR) 2.25, CI = 1.37-3.71; P = 0.001] and with recurrent MI or cardiac death [multivariable-adjusted (HR) 1.80, CI = 1.09-2.95; P = 0.021] within 5 years after an index ACS. The association of rs579459 was replicated in 1250 Polish patients with 6 months follow-up after an index ACS [multivariable-adjusted (HR) 2.70, CI = 1.26-5.82; P = 0.011 for recurrent MI]. Addition of rs579459 to a prediction model of 17 clinical risk factors improved risk classification for recurrent MI or cardiac death at 6 months as calculated by the integrated discrimination improvement method (P = 0.037), but not by C-statistics (P = 0.096). CONCLUSION: In this observational study, rs579459 was independently associated with adverse cardiac outcome after ACS. A weak improvement in clinical risk prediction was also observed, suggesting that rs579459 should be further tested as a potentially relevant contributor to risk prediction models for adverse outcome following ACS.


Subject(s)
Coronary Artery Disease/genetics , Death, Sudden, Cardiac/etiology , Genetic Predisposition to Disease/genetics , Myocardial Infarction/genetics , ABO Blood-Group System/genetics , Aged , Belgium/epidemiology , Coronary Artery Disease/mortality , Death, Sudden, Cardiac/epidemiology , Female , Gene Frequency , Genetic Loci/genetics , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Myocardial Infarction/mortality , Phenotype , Poland/epidemiology , Prognosis , Prospective Studies , Recurrence , Registries , Risk Factors , United Kingdom/epidemiology
15.
Proc Natl Acad Sci U S A ; 109(46): E3186-95, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23100531

ABSTRACT

Although macrophages are widely recognized to have a profibrotic role in inflammation, we have used a highly tractable CCl(4)-induced model of reversible hepatic fibrosis to identify and characterize the macrophage phenotype responsible for tissue remodeling: the hitherto elusive restorative macrophage. This CD11B(hi) F4/80(int) Ly-6C(lo) macrophage subset was most abundant in livers during maximal fibrosis resolution and represented the principle matrix metalloproteinase (MMP) -expressing subset. Depletion of this population in CD11B promoter-diphtheria toxin receptor (CD11B-DTR) transgenic mice caused a failure of scar remodeling. Adoptive transfer and in situ labeling experiments showed that these restorative macrophages derive from recruited Ly-6C(hi) monocytes, a common origin with profibrotic Ly-6C(hi) macrophages, indicative of a phenotypic switch in vivo conferring proresolution properties. Microarray profiling of the Ly-6C(lo) subset, compared with Ly-6C(hi) macrophages, showed a phenotype outside the M1/M2 classification, with increased expression of MMPs, growth factors, and phagocytosis-related genes, including Mmp9, Mmp12, insulin-like growth factor 1 (Igf1), and Glycoprotein (transmembrane) nmb (Gpnmb). Confocal microscopy confirmed the postphagocytic nature of restorative macrophages. Furthermore, the restorative macrophage phenotype was recapitulated in vitro by the phagocytosis of cellular debris with associated activation of the ERK signaling cascade. Critically, induced phagocytic behavior in vivo, through administration of liposomes, increased restorative macrophage number and accelerated fibrosis resolution, offering a therapeutic strategy to this orphan pathological process.


Subject(s)
Antigens, Ly/immunology , Carbon Tetrachloride Poisoning/immunology , Gene Expression Regulation/immunology , Liver Cirrhosis/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Antigens, Ly/genetics , CD11b Antigen/genetics , CD11b Antigen/immunology , Carbon Tetrachloride/toxicity , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/immunology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Macrophages/pathology , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase 12/immunology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Mice , Mice, Transgenic , Monocytes/pathology
16.
Open Biol ; 2(1): 110031, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22645656

ABSTRACT

Multi-cellular organisms need to successfully link cell growth and metabolism to environmental cues during development. Insulin receptor-target of rapamycin (InR-TOR) signalling is a highly conserved pathway that mediates this link. Herein, we describe poly, an essential gene in Drosophila that mediates InR-TOR signalling. Loss of poly results in lethality at the third instar larval stage, but only after a stage of extreme larval longevity. Analysis in Drosophila demonstrates that Poly and InR interact and that poly mutants show an overall decrease in InR-TOR signalling, as evidenced by decreased phosphorylation of Akt, S6K and 4E-BP. Metabolism is altered in poly mutants, as revealed by microarray expression analysis and a decreased triglyceride : protein ratio in mutant animals. Intriguingly, the cellular distribution of Poly is dependent on insulin stimulation in both Drosophila and human cells, moving to the nucleus with insulin treatment, consistent with a role in InR-TOR signalling. Together, these data reveal that Poly is a novel, conserved (from flies to humans) mediator of InR signalling that promotes an increase in cell growth and metabolism. Furthermore, homology to small subunits of Elongator demonstrates a novel, unexpected role for this complex in insulin signalling.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drosophila Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Ribonucleoproteins/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , HeLa Cells , Humans , Receptor Protein-Tyrosine Kinases/genetics , Ribonucleoproteins/genetics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
17.
Diabetes ; 61(3): 642-52, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22315313

ABSTRACT

Type 2 diabetes ultimately results from pancreatic ß-cell failure. Abnormally elevated intracellular regeneration of glucocorticoids by the enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) in fat or liver may underlie pathophysiological aspects of the metabolic syndrome. Elevated 11ß-HSD1 is also found in pancreatic islets of obese/diabetic rodents and is hypothesized to suppress insulin secretion and promote diabetes. To define the direct impact of elevated pancreatic ß-cell 11ß-HSD1 on insulin secretion, we generated ß-cell-specific, 11ß-HSD1-overexpressing (MIP-HSD1) mice on a strain background prone to ß-cell failure. Unexpectedly, MIP-HSD1(tg/+) mice exhibited a reversal of high fat-induced ß-cell failure through augmentation of the number and intrinsic function of small islets in association with induction of heat shock, protein kinase A, and extracellular signal-related kinase and p21 signaling pathways. 11ß-HSD1(-/-) mice showed mild ß-cell impairment that was offset by improved glucose tolerance. The benefit of higher ß-cell 11ß-HSD1 exhibited a threshold because homozygous MIP-HSD1(tg/tg) mice and diabetic Lep(db/db) mice with markedly elevated ß-cell 11ß-HSD1 levels had impaired basal ß-cell function. Optimal elevation of ß-cell 11ß-HSD1 represents a novel biological mechanism supporting compensatory insulin hypersecretion rather than exacerbating metabolic disease. These findings have immediate significance for current therapeutic strategies for type 2 diabetes.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , Diabetes Mellitus, Type 2/prevention & control , Diet, High-Fat/adverse effects , Insulin-Secreting Cells/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Diabetes Mellitus/etiology , Extracellular Signal-Regulated MAP Kinases/physiology , Glucose Tolerance Test , Insulin/metabolism , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Palmitic Acid/pharmacology
18.
J Am Soc Nephrol ; 23(3): 405-11, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22193383

ABSTRACT

Rodent models exhibit only the earliest features of human diabetic nephropathy, which limits our ability to investigate new therapies. Hypertension is a prerequisite for advanced diabetic nephropathy in humans, so its rarity in typical rodent models may partly explain their resistance to nephropathy. Here, we used the Cyp1a1mRen2 rat, in which the murine renin-2 gene is incorporated under the Cytochrome P4501a1 promoter. In this transgenic strain, administration of low-dose dietary indole-3-carbinol induces moderate hypertension. In the absence of hypertension, streptozotocin-induced diabetes resulted in a 14-fold increase in albuminuria but only mild changes in histology and gene expression despite 28 weeks of marked hyperglycemia. In the presence of induced hypertension, hyperglycemia resulted in a 500-fold increase in albuminuria, marked glomerulosclerosis and tubulointerstitial fibrosis, and induction of many of the same pathways that are upregulated in the tubulointerstitium in human diabetic nephropathy. In conclusion, although induction of diabetes alone in rodents has limited utility to model human diabetic nephropathy, renin-dependent hypertension and hyperglycemia synergize to recapitulate many of the clinical, histological, and gene expression changes observed in humans.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Disease Models, Animal , Hyperglycemia/complications , Hypertension/complications , Renin , Albuminuria/etiology , Albuminuria/urine , Animals , Comorbidity , Cytochrome P-450 CYP1A1/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/epidemiology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/urine , Fibrosis , Humans , Hyperglycemia/chemically induced , Hyperglycemia/epidemiology , Hypertension/chemically induced , Hypertension/epidemiology , Indoles/adverse effects , Kidney Tubules/pathology , Mice , Rats , Rats, Transgenic , Renin/genetics , Streptozocin/adverse effects
19.
PLoS One ; 6(9): e23944, 2011.
Article in English | MEDLINE | ID: mdl-21915269

ABSTRACT

BACKGROUND: Obesity and metabolic syndrome results from a complex interaction between genetic and environmental factors. In addition to brain-regulated processes, recent genome wide association studies have indicated that genes highly expressed in adipose tissue affect the distribution and function of fat and thus contribute to obesity. Using a stratified transcriptome gene enrichment approach we attempted to identify adipose tissue-specific obesity genes in the unique polygenic Fat (F) mouse strain generated by selective breeding over 60 generations for divergent adiposity from a comparator Lean (L) strain. RESULTS: To enrich for adipose tissue obesity genes a 'snap-shot' pooled-sample transcriptome comparison of key fat depots and non adipose tissues (muscle, liver, kidney) was performed. Known obesity quantitative trait loci (QTL) information for the model allowed us to further filter genes for increased likelihood of being causal or secondary for obesity. This successfully identified several genes previously linked to obesity (C1qr1, and Np3r) as positional QTL candidate genes elevated specifically in F line adipose tissue. A number of novel obesity candidate genes were also identified (Thbs1, Ppp1r3d, Tmepai, Trp53inp2, Ttc7b, Tuba1a, Fgf13, Fmr) that have inferred roles in fat cell function. Quantitative microarray analysis was then applied to the most phenotypically divergent adipose depot after exaggerating F and L strain differences with chronic high fat feeding which revealed a distinct gene expression profile of line, fat depot and diet-responsive inflammatory, angiogenic and metabolic pathways. Selected candidate genes Npr3 and Thbs1, as well as Gys2, a non-QTL gene that otherwise passed our enrichment criteria were characterised, revealing novel functional effects consistent with a contribution to obesity. CONCLUSIONS: A focussed candidate gene enrichment strategy in the unique F and L model has identified novel adipose tissue-enriched genes contributing to obesity.


Subject(s)
Adipose Tissue/metabolism , Obesity/genetics , Transcriptome/genetics , 3T3-L1 Cells , Animals , Computational Biology , Fibroblast Growth Factors/genetics , Glycogen/metabolism , Membrane Glycoproteins/genetics , Mice , Oligonucleotide Array Sequence Analysis , Proteins/genetics , Quantitative Trait Loci/genetics , Real-Time Polymerase Chain Reaction , Receptors, Complement/genetics
20.
Diabetes ; 60(4): 1158-67, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21350084

ABSTRACT

OBJECTIVE: The study objective was to determine the key early mechanisms underlying the beneficial redistribution, function, and inflammatory profile of adipose tissue in 11ß-hydroxysteroid dehydrogenase type 1 knockout (11ß-HSD1(-/-)) mice fed a high-fat (HF) diet. RESEARCH DESIGN AND METHODS: By focusing on the earliest divergence in visceral adiposity, subcutaneous and visceral fat depots from 11ß-HSD1(-/-) and C57Bl/6J control mice fed an HF diet for 4 weeks were used for comparative microarray analysis of gene expression, and differences were validated with real-time PCR. Key changes in metabolic signaling pathways were confirmed using Western blotting/immunoprecipitation, and fat cell size was compared with the respective chow-fed control groups. Altered adipose inflammatory cell content and function after 4 weeks (early) and 18 weeks (chronic) of HF feeding was investigated using fluorescence (and magnetic)-activated cell sorting analysis, immunohistochemistry, and in situ hybridization. RESULTS: In subcutaneous fat, HF-fed 11ß-HSD1(-/-) mice showed evidence of enhanced insulin and ß-adrenergic signaling associated with accretion of smaller metabolically active adipocytes. In contrast, reduced 11ß-HSD1(-/-) visceral fat accumulation was characterized by maintained AMP kinase activation, not insulin sensitization, and higher adipocyte interleukin-6 release. Intracellular glucocorticoid deficiency was unexpectedly associated with suppressed inflammatory signaling and lower adipocyte monocyte chemoattractant protein-1 secretion with strikingly reduced cytotoxic T-cell and macrophage infiltration, predominantly in visceral fat. CONCLUSIONS: Our data define for the first time the novel and distinct depot-specific mechanisms driving healthier fat patterning and function as a result of reduced intra-adipose glucocorticoid levels.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Inflammation/metabolism , Obesity, Abdominal/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blotting, Western , Dietary Fats/adverse effects , Enzyme Activation/drug effects , Flow Cytometry , Glucocorticoids/pharmacology , Immunohistochemistry , In Situ Hybridization , Inflammation/genetics , Interleukin-6/pharmacology , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Mutant Strains , Obesity, Abdominal/chemically induced , Obesity, Abdominal/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...