Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Physiol Genomics ; 48(1): 21-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26508702

ABSTRACT

Protein-leucine supplement ingestion following strenuous endurance exercise accentuates skeletal-muscle protein synthesis and adaptive molecular responses, but the underlying transcriptome is uncharacterized. In a randomized single-blind triple-crossover design, 12 trained men completed 100 min of high-intensity cycling then ingested 70/15/180/30 g protein-leucine-carbohydrate-fat (15LEU), 23/5/180/30 g (5LEU), or 0/0/274/30 g (CON) beverages during the first 90 min of a 240 min recovery period. Vastus lateralis muscle samples (30 and 240 min postexercise) underwent transcriptome analysis by microarray followed by bioinformatic analysis. Gene expression was regulated by protein-leucine in a dose-dependent manner affecting the inflammatory response and muscle growth and development. At 30 min, 15LEU and 5LEU vs. CON activated transcriptome networks with gene-set functions involving cell-cycle arrest (Z-score 2.0-2.7, P < 0.01), leukocyte maturation (1.7, P = 0.007), cell viability (2.4, P = 0.005), promyogenic networks encompassing myocyte differentiation and myogenin (MYOD1, MYOG), and a proteinaceous extracellular matrix, adhesion, and development program correlated with plasma lysine, arginine, tyrosine, taurine, glutamic acid, and asparagine concentrations. High protein-leucine dose (15LEU-5LEU) activated an IL-1I-centered proinflammatory network and leukocyte migration, differentiation, and survival functions (2.0-2.6, <0.001). By 240 min, the protein-leucine transcriptome was anti-inflammatory and promyogenic (IL-6, NF- ß, SMAD, STAT3 network inhibition), with overrepresented functions including decreased leukocyte migration and connective tissue development (-1.8-2.4, P < 0.01), increased apoptosis of myeloid and muscle cells (2.2-3.0, P < 0.002), and cell metabolism (2.0-2.4, P < 0.01). The analysis suggests protein-leucine ingestion modulates inflammatory-myogenic regenerative processes during skeletal muscle recovery from endurance exercise. Further cellular and translational research is warranted to validate amino acid-mediated myeloid and myocellular mechanisms within skeletal-muscle functional plasticity.


Subject(s)
Exercise , Inflammation/genetics , Leucine/administration & dosage , Muscle Development/genetics , Muscle, Skeletal/pathology , Physical Endurance , Transcriptome/genetics , Adult , Gene Regulatory Networks , Humans , Male , Models, Biological , Multigene Family , Muscle, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Software
2.
Pediatr Res ; 75(1-1): 51-61, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24213624

ABSTRACT

BACKGROUND: Intrauterine growth restriction (IUGR) is a major risk factor for both perinatal and long-term morbidity. Bovine lactoferrin (bLf) is a major milk glycoprotein considered as a pleiotropic functional nutrient. The impact of maternal supplementation with bLf on IUGR-induced sequelae, including inadequate growth and altered cerebral development, remains unknown. METHODS: IUGR was induced through maternal dexamethasone infusion (100 µg/kg during last gestational week) in rats. Maternal supplementation with bLf (0.85% in food pellet) was provided during both gestation and lactation. Pup growth was monitored, and Pup brain metabolism and gene expression were studied using in vivo (1)H NMR spectroscopy, quantitative PCR, and microarray in the hippocampus at postnatal day (PND)7. RESULTS: Maternal bLf supplementation did not change gestational weight but increased the birth body weight of control pups (4%) with no effect on the IUGR pups. Maternal bLf supplementation allowed IUGR pups to recover a normalized weight at PND21 (weaning) improving catch-up growth. Significantly altered levels of brain metabolites (γ-aminobutyric acid, glutamate, N-acetylaspartate, and N-acetylaspartylglutamate) and transcripts (brain-derived neurotrophic factor (BDNF), divalent metal transporter 1 (DMT-1), and glutamate receptors) in IUGR pups were normalized with maternal bLf supplementation. CONCLUSION: Our data suggest that maternal bLf supplementation is a beneficial nutritional intervention able to revert some of the IUGR-induced sequelae, including brain hippocampal changes.


Subject(s)
Brain/drug effects , Dietary Supplements , Growth/drug effects , Lactoferrin/administration & dosage , Animals , Body Weight/drug effects , Brain/metabolism , Dexamethasone/administration & dosage , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/prevention & control , Gene Expression/drug effects , Lactation , Lactoferrin/pharmacology , Polymerase Chain Reaction , Pregnancy , Rats , Weight Gain/drug effects
3.
Mol Cell Proteomics ; 10(3): M110.005231, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21191033

ABSTRACT

Postnatal intestinal development is a very dynamic process characterized by substantial morphological changes that coincide with functional adaption to the nutritional change from a diet rich in fat (milk) to a diet rich in carbohydrates on from weaning. Time-resolved studies of intestinal development have so far been limited to investigation at the transcription level or to single or few proteins at a time. In the present study, we elucidate proteomic changes of primary intestinal epithelial cells from jejunum during early suckling (1-7 days of age), middle suckling (7-14 days), and weaning period (14-35 days) in mice, using a label-free proteomics approach. We show differential expression of 520 proteins during intestinal development and a pronounced change of the proteome during the middle suckling period and weaning. Proteins involved in several metabolic processes were found differentially expressed along the development. The temporal expression profiles of enzymes of the glycolysis were found to correlate with the increase in carbohydrate uptake at weaning, whereas the abundance changes of proteins involved in fatty acid metabolism as well as lactose metabolism indicated a nondiet driven preparation for the nutritional change at weaning. Further, we report the developmental abundance changes of proteins playing a vital role in the neonatal acquisition of passive immunity. In addition, different isoforms of several proteins were quantified, which may contribute to a better understanding of the roles of the specific isoforms in the small intestine. In summary, we provide a first, time-resolved proteome profile of intestinal epithelial cells along postnatal intestinal development.


Subject(s)
Intestinal Mucosa/metabolism , Intestines/growth & development , Proteome/metabolism , Proteomics/methods , Animals , Carbohydrate Metabolism , Databases, Protein , Epithelial Cells/metabolism , Fatty Acids/metabolism , Glycolysis , Intestinal Absorption , Intestines/enzymology , Isotope Labeling , Lipid Metabolism , Mice , Mice, Inbred C57BL , Peptides/metabolism , Time Factors
4.
Lipids Health Dis ; 12: 81, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23725086

ABSTRACT

BACKGROUND: Inflammatory bowel diseases (IBD) are chronic intestinal inflammatory diseases affecting about 1% of western populations. New eating behaviors might contribute to the global emergence of IBD. Although the immunoregulatory effects of omega-3 fatty acids have been well characterized in vitro, their role in IBD is controversial. METHODS: The aim of this study was to assess the impact of increased fish oil intake on colonic gene expression, eicosanoid metabolism and development of colitis in a mouse model of IBD. Rag-2 deficient mice were fed fish oil (FO) enriched in omega-3 fatty acids i.e. EPA and DHA or control diet for 4 weeks before colitis induction by adoptive transfer of naïve T cells and maintained in the same diet for 4 additional weeks. Onset of colitis was monitored by colonoscopy and further confirmed by immunological examinations. Whole genome expression profiling was made and eicosanoids were measured by HPLC-MS/MS in colonic samples. RESULTS: A significant reduction of colonic proinflammatory eicosanoids in FO fed mice compared to control was observed. However, neither alteration of colonic gene expression signature nor reduction in IBD scores was observed under FO diet. CONCLUSION: Thus, increased intake of dietary FO did not prevent experimental colitis.


Subject(s)
Colitis/diet therapy , Colitis/metabolism , Eicosanoids/metabolism , Fish Oils/pharmacology , Intestines/drug effects , Animals , Colitis/genetics , Colon/physiopathology , Cytokines/metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Fatty Acids, Omega-3/pharmacology , Fish Oils/chemistry , Gene Expression Regulation/drug effects , Inflammatory Bowel Diseases/etiology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology
5.
Physiol Genomics ; 43(17): 1004-20, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21730029

ABSTRACT

Postexercise protein feeding regulates the skeletal muscle adaptive response to endurance exercise, but the transcriptome guiding these adaptations in well-trained human skeletal muscle is uncharacterized. In a crossover design, eight cyclists ingested beverages containing protein, carbohydrate and fat (PTN: 0.4, 1.2, 0.2 g/kg, respectively) or isocaloric carbohydrate and fat (CON: 1.6, 0.2 g/kg) at 0 and 1 h following 100 min of cycling. Biopsies of the vastus lateralis were collected at 3 and 48 h following to determine the early and late transcriptome and regulatory signaling responses via microarray and immunoblot. The top gene ontology enriched by PTN were: muscle contraction, extracellular matrix--signaling and structure, and nucleoside, nucleotide, and nucleic acid metabolism (3 and 48 h); developmental processes, immunity, and defense (3 h); glycolysis, lipid and fatty acid metabolism (48 h). The transcriptome was also enriched within axonal guidance, actin cytoskeletal, Ca2+, cAMP, MAPK, and PPAR canonical pathways linking protein nutrition to exercise-stimulated signaling regulating extracellular matrix, slow-myofibril, and metabolic gene expression. At 3 h, PTN attenuated AMPKα1Thr172 phosphorylation but increased mTORC1Ser2448, rps6Ser240/244, and 4E-BP1-γ phosphorylation, suggesting increased translation initiation, while at 48 h AMPKα1Thr172 phosphorylation and PPARG and PPARGC1A expression increased, supporting the late metabolic transcriptome, relative to CON. To conclude, protein feeding following endurance exercise affects signaling associated with cell energy status and translation initiation and the transcriptome involved in skeletal muscle development, slow-myofibril remodeling, immunity and defense, and energy metabolism. Further research should determine the time course and posttranscriptional regulation of this transcriptome and the phenotype responding to chronic postexercise protein feeding.


Subject(s)
Dietary Proteins/pharmacology , Exercise/physiology , Muscle, Skeletal/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adult , Blood Glucose/metabolism , Electrophoresis, Polyacrylamide Gel , Glycogen/metabolism , Humans , Immunoblotting , Insulin/blood , Male , Mechanistic Target of Rapamycin Complex 1 , Models, Biological , Multiprotein Complexes , Muscle, Skeletal/drug effects , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Polymerase Chain Reaction , Proteins/genetics , Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases , Transcriptome/drug effects , Transcriptome/genetics
6.
FASEB J ; 24(12): 4948-59, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20724524

ABSTRACT

Recent studies showed that germ-free (GF) mice are resistant to obesity when consuming a high-fat, high-carbohydrate Western diet. However, it remains unclear what mechanisms are involved in the antiobesity phenotype and whether GF mice develop insulin resistance and dyslipidemia with high-fat (HF) feeding. In the present study, we compared the metabolic consequences of HF feeding on GF and conventional (conv) C57BL/6J mice. GF mice consumed fewer calories, excreted more fecal lipids, and weighed significantly less than conv mice. GF/HF animals also showed enhanced insulin sensitivity with improved glucose tolerance, reduced fasting and nonfasting insulinemia, and increased phospho-Akt((Ser-473)) in adipose tissue. In association with enhanced insulin sensitivity, GF/HF mice had reduced plasma TNF-α and total serum amyloid A concentrations. Reduced hypercholesterolemia, a moderate accretion of hepatic cholesterol, and an increase in fecal cholesterol excretion suggest an altered cholesterol metabolism in GF/HF mice. Pronounced nucleus SREBP2 proteins and up-regulation of cholesterol biosynthesis genes indicate that enhanced cholesterol biosynthesis contributed to the cholesterol homeostasis in GF/HF mice. Our results demonstrate that fewer calorie consumption and increased lipid excretion contributed to the obesity-resistant phenotype of GF/HF mice and reveal that insulin sensitivity and cholesterol metabolism are metabolic targets influenced by the gut microbiota.


Subject(s)
Cholesterol/metabolism , Dietary Fats/adverse effects , Insulin Resistance/physiology , Animals , Blotting, Western , Body Weight/physiology , Germ-Free Life , Glucose Tolerance Test , Glycogen/metabolism , Lipids/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
7.
Genomics ; 91(1): 70-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18035521

ABSTRACT

The intestinal mucus layer and endogenous microbiota are strongly intertwined and this contributes to the maintenance of the epithelial barrier and ultimately of gut homeostasis. To understand the molecular foundations of such relationship, we investigated if the nature of the microbiota transcriptionally regulates mucus layer composition in vivo. We found that the expression of mucins 1 to 4 and trefoil factor 3 was down-regulated in the ileum and colon of conventional and reconventionalized mice compared with germ-free animals. Conversely, very limited colon-restricted changes in transmembrane mucins were detected in mice colonized with human adult or baby microbiota. Moreover, by microarray analysis, the murine endogenous microbiota was found to modulate genes putatively involved in mucin secretion. These findings show that a well-established microbial community participates in the regulation of the gut mucus layer and that its composition and adequacy to the host are key factors in this process.


Subject(s)
Down-Regulation/physiology , Germ-Free Life/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mucins/biosynthesis , Adult , Animals , Colon/metabolism , Colon/microbiology , Gene Expression Profiling , Humans , Ileum/metabolism , Ileum/microbiology , Infant , Infant, Newborn , Male , Mice , Mice, Inbred C3H , Mucins/genetics , Oligonucleotide Array Sequence Analysis , Trefoil Factor-3
8.
Free Radic Biol Med ; 43(5): 752-62, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17664139

ABSTRACT

Two sodium-dependent vitamin C transporter isoforms (SVCT1 and SVCT2) were identified as ascorbic acid transporters, but their roles in skin have, as yet, not been elucidated. Here we analyze the expression and function of SVCTs in healthy human skin cells and skin tissues, and in UVB-induced cutaneous tissue injury. SVCT1 was primarily found in the epidermis expressed by keratinocytes, whereas SVCT2 expression was in the epidermis and dermis in keratinocytes, fibroblasts, and endothelial cells. Uptake experiments revealed that ascorbic acid affinity of SVCT1 was lower than SVCT2 (K(m)=75 muM and K(m)=44 muM, respectively), but maximal velocity was 9-times higher (36 nmol/min/well). In keratinocytes, SVCT1 was found to be responsible for vitamin C transport, although SVCT2 gene expression was higher. On UVB irradiation, SVCT1 mRNA expression in murine skin declined significantly in a time- and dose-dependent manner, whereas SVCT2 mRNA levels were unchanged. Furthermore, UVB irradiation of keratinocytes in vitro was accompanied by reduced ascorbic acid transport. In summary, these data indicate that the two vitamin C transporter isoforms fulfill specific functions in skin: SVCT1 is responsible for epidermal ascorbic acid supply, whereas SVCT2 mainly facilitates ascorbic acid transport in the dermal compartment. UVB-induced oxidative stress in mice resulted in depletion of SVCT1 mRNA levels and led to significantly decreased ascorbic acid uptake in keratinocytes, providing evidence on why ascorbic acid levels are decreased on UVB irradiation in vivo.


Subject(s)
Ascorbic Acid/pharmacology , Organic Anion Transporters, Sodium-Dependent/metabolism , Oxidative Stress/radiation effects , Skin/drug effects , Skin/metabolism , Symporters/metabolism , Ultraviolet Rays , Animals , Biopsy , Cells, Cultured , Gene Expression Regulation , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Kinetics , Mice , Mice, Inbred C57BL , Organic Anion Transporters, Sodium-Dependent/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , Sodium-Coupled Vitamin C Transporters , Symporters/genetics
9.
Gene ; 384: 145-53, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17052863

ABSTRACT

The present study examined time-dependent changes in the gene expression profile of long-term cultured human myotubes. Microarray transcriptional analysis was performed in a primary culture of differentiated myotubes from one subject over seven weeks. This analysis showed a main gradual fall in genes of the contractile apparatus, and a broad upregulation of genes involved in cell development and growth, followed by stress response and signal transduction. Glucose metabolism was also monitored, but no significant alterations in glucose uptake, oxidation or glycogen storage were observed. Mitochondrial membrane potential, or the amount of membrane lipid peroxides, remained similarly unchanged, nor was lactate dehydrogenase leakage observed. Time-dependent changes in eight genes were validated by real-time RT-PCR in primary cultured myotubes from four subjects, of similar age and isolated after equivalent replication cycles in vitro and differentiated over seven weeks. Insulin-like growth factor-binding protein 2 (IGFBP2), a modulator of the IGF signal, was upregulated. The antiapoptotic gene heat-shock 70-kd protein 2 (HSPA2) was induced, whereas the proapoptotic tumor necrosis factor receptor superfamily, member 25 (WSL-1) was suppressed. A decline in the muscle-specific gene M-cadherin and contraction genes, such as slow-twitch troponin I (TNNI1) and myosin heavy chain 2 (MYH2), myosin light chain 1 (MYL1) and myosin-binding protein H (MYBPH), which are expressed in adult fast-twitch muscle, was shown. In summary, these data demonstrate extensive downregulation of contractile genes and modulation of apoptosis-related genes, in favour of cell survival, during maintenance of cultured human myotubes.


Subject(s)
Apoptosis/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Adolescent , Biopsy , Cell Culture Techniques , Cell Survival/genetics , Cells, Cultured , Child , Down-Regulation , Gene Expression Profiling , Glucose/metabolism , Humans , Lipid Metabolism/genetics , Membrane Potential, Mitochondrial , Muscles/cytology , Muscles/pathology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Time
10.
Biochimie ; 88(3-4): 309-18, 2006.
Article in English | MEDLINE | ID: mdl-16298468

ABSTRACT

The liver X receptor (LXR) was demonstrated to play a key role in cholesterol metabolism in liver, intestine and macrophage. However, its function on the regulation of preadipocyte differentiation remains unclear since contradictory results were reported. The objective of the present study was to unravel the functionality of LXR in human preadipocytes. We show that the LXR agonist T0901317 strongly stimulated the expression of SREBP-1c and the lipogenic enzymes ACC-1, FAS and SCD-1 in both the human preadipose cell line Chub-S7 as well as human primary stromal vascular fraction (SVF) cells. The effects on gene expression were associated with the stimulation of de novo lipogenesis in both cell models, resulting in the induction of lipid accumulation. In contrast with a PPARgamma agonist (BRL49653), T0901317 enhanced only slightly the expression of PPARgamma dependent genes (PPARgamma, aP2 and adiponectin) in Chub-S7 cells and failed to change their expression in human SVF cells. These results show that LXR stimulated preferentially triglyceride accumulation in human preadipocytes via the induction of de novo lipogenesis, rather than activating the differentiation process through PPARgamma activation.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Lipogenesis , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/metabolism , Adipocytes/cytology , Biomarkers , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/pharmacology , Gene Expression Regulation , Humans , Liver X Receptors , Orphan Nuclear Receptors , PPAR gamma/metabolism , Stromal Cells/metabolism , Transcription Factors/metabolism , Triglycerides/biosynthesis , Triglycerides/metabolism
11.
Physiol Genomics ; 23(2): 235-45, 2005 Oct 17.
Article in English | MEDLINE | ID: mdl-16131529

ABSTRACT

The postnatal maturation of the gut, partially modulated by bacterial colonization, ends up in the establishment of an efficient barrier to luminal antigens and bacteria. The use of broad-spectrum antibiotics in pediatric practices alters the gut bacterial colonization and, consequently, may impair the maturation of the gut barrier function. To test this hypothesis, suckling Sprague-Dawley rats received a daily intragastric gavage of antibiotic (Clamoxyl; an amoxicillin-based commercial preparation) or saline solution from postnatal day 7 (d7) until d17 or d21. Luminal microbiota composition and global gene expression profile were analyzed on samples from small intestine and colon of each group. The treatment with Clamoxyl resulted in the almost-complete eradication of Lactobacillus in the whole intestine and in a drastic reduction of colonic total aerobic and anaerobic bacteria, in particular Enterobacteriacae and Enterococcus. The global gene expression analysis revealed that Clamoxyl affects the maturation process of 249 and 149 Affymetrix probe sets in the proximal and distal small intestine, respectively, and 163 probe sets in the colon. The expression of genes coding for Paneth cell products (defensins, matrilysin, and phospholipase A2) was significantly downregulated by the Clamoxyl treatment. A significant downregulation of major histocompatibility complex (MHC) class Ib and II genes, involved in antigen presentation, was also observed. Conversely, mast cell proteases expression was upregulated. These results suggest that early treatment with a large-spectrum antibiotic deeply affects the gut barrier function at the suckling-weaning interface, a period during which the gut is challenged by an array of novel food-borne antigens.


Subject(s)
Amoxicillin/pharmacology , Colon/drug effects , Gene Expression Regulation, Developmental/drug effects , Genes, Developmental/genetics , Intestinal Mucosa/metabolism , Intestines/drug effects , Transcription, Genetic/drug effects , Aging , Animals , Animals, Newborn , Antigen Presentation/genetics , Down-Regulation/genetics , Female , Intestines/growth & development , Intestines/microbiology , Male , Mast Cells/enzymology , Peptide Hydrolases/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/genetics
12.
BMC Genomics ; 6: 69, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15882471

ABSTRACT

BACKGROUND: The purpose of this work was to characterize the expression of drug and nutrient carriers along the anterior-posterior and crypt-villus axes of the intestinal epithelium and to study the validity of utilizing whole gut tissue rather than purified epithelial cells to examine regional variations in gene expression. RESULTS: We have characterized the mRNA expression profiles of 76 % of all currently known transporters along the anterior-posterior axis of the gut. This is the first study to describe the expression profiles of the majority of all known transporters in the intestine. The expression profiles of transporters, as defined according to the Gene Ontology consortium, were measured in whole tissue of the murine duodenum, jejunum, ileum and colon using high-density microarrays. For nine transporters (Abca1, Abcc1, Abcc3, Abcg8, Slc10a2, Slc28a2, Slc2a1, Slc34a2 and Slc5a8), the mRNA profiles were further measured by RT-PCR in laser micro-dissected crypt and villus epithelial cells corresponding to the aforementioned intestinal regions. With respect to differentially regulated transporters, the colon had a distinct expression profile from small intestinal segments. The majority (59 % for p cutoff < or = 0.05) of transporter mRNA levels were constant across the intestinal sections studied. For the transporter subclass "carrier activity", which contains the majority of known carriers for biologically active compounds, a significant change (p < or = 0.05) along the anterior-posterior axis was observed. CONCLUSION: All nine transporters examined in laser-dissected material demonstrated good replication of the region-specific profiles revealed by microarray. Furthermore, we suggest that the distribution characteristics of Slc5a8 along the intestinal tract render it a suitable candidate carrier for monocarboxylate drugs in the posterior portion of the intestine. Our findings also predict that there is a significant difference in the absorption of carrier-mediated compounds in the different intestinal segments. The most pronounced differences can be expected between the adjoining segments ileum and colon, but the differences between the other adjoining segments are not negligible. Finally, for the examined genes, profiles measured in whole intestinal tissue extracts are representative of epithelial cell-only gene expression.


Subject(s)
Gene Expression Profiling/methods , Intestinal Mucosa/metabolism , Intestines/pathology , Transcription, Genetic , Analysis of Variance , Animals , Biological Transport , Colon/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Ileum/metabolism , Lasers , Male , Mice , Microdissection , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
13.
BMC Bioinformatics ; 3: 17, 2002 Jun 21.
Article in English | MEDLINE | ID: mdl-12095422

ABSTRACT

BACKGROUND: The biomedical community is developing new methods of data analysis to more efficiently process the massive data sets produced by microarray experiments. Systematic and global mathematical approaches that can be readily applied to a large number of experimental designs become fundamental to correctly handle the otherwise overwhelming data sets. RESULTS: The gene selection model presented herein is based on the observation that: (1) variance of gene expression is a function of absolute expression; (2) one can model this relationship in order to set an appropriate lower fold change limit of significance; and (3) this relationship defines a function that can be used to select differentially expressed genes. The model first evaluates fold change (FC) across the entire range of absolute expression levels for any number of experimental conditions. Genes are systematically binned, and those genes within the top X% of highest FCs for each bin are evaluated both with and without the use of replicates. A function is fitted through the top X% of each bin, thereby defining a limit fold change. All genes selected by the 5% FC model lie above measurement variability using a within standard deviation (SDwithin) confidence level of 99.9%. Real time-PCR (RT-PCR) analysis demonstrated 85.7% concordance with microarray data selected by the limit function. CONCLUSION: The FC model can confidently select differentially expressed genes as corroborated by variance data and RT-PCR. The simplicity of the overall process permits selecting model limits that best describe experimental data by extracting information on gene expression patterns across the range of expression levels. Genes selected by this process can be consistently compared between experiments and enables the user to globally extract information with a high degree of confidence.


Subject(s)
Gene Expression Profiling/methods , Models, Genetic , Oligonucleotide Array Sequence Analysis/methods , Animals , Computational Biology/methods , Diet , Genes/genetics , Genetic Variation/genetics , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Organ Specificity/genetics , RNA, Complementary/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
14.
Physiol Genomics ; 17(1): 11-20, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14679303

ABSTRACT

The ATP-binding cassette (ABC) family of proteins comprise a group of membrane transporters involved in the transport of a wide variety of compounds, such as xenobiotics, vitamins, lipids, amino acids, and carbohydrates. Determining their regional expression patterns along the intestinal tract will further characterize their transport functions in the gut. The mRNA expression levels of murine ABC transporters in the duodenum, jejunum, ileum, and colon were examined using the Affymetrix MuU74v2 GeneChip set. Eight ABC transporters (Abcb2, Abcb3, Abcb9, Abcc3, Abcc6, Abcd1, Abcg5, and Abcg8) displayed significant differential gene expression along the intestinal tract, as determined by two statistical models (a global error assessment model and a classic ANOVA, both with a P < 0.01). Concordance with semiquantitative real-time PCR was high. Analyzing the promoters of the differentially expressed ABC transporters did not identify common transcriptional motifs between family members or with other genes; however, the expression profile for Abcb9 was highly correlated with fibulin-1, and both genes share a common complex promoter model involving the NFkappaB, zinc binding protein factor (ZBPF), GC-box factors SP1/GC (SP1F), and early growth response factor (EGRF) transcription binding motifs. The cellular location of another of the differentially expressed ABC transporters, Abcc3, was examined by immunohistochemistry. Staining revealed that the protein is consistently expressed in the basolateral compartment of enterocytes along the anterior-posterior axis of the intestine. Furthermore, the intensity of the staining pattern is concordant with the expression profile. This agrees with previous findings in which the mRNA, protein, and transport function of Abcc3 were increased in the rat distal intestine. These data reveal regional differences in gene expression profiles along the intestinal tract and demonstrate that a complete understanding of intestinal ABC transporter function can only be achieved by examining the physiologically distinct regions of the gut.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Genetic Variation/genetics , Intestinal Mucosa/metabolism , Intestines/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Animals , Colon/chemistry , Colon/metabolism , Computer Systems , DNA, Complementary/genetics , Gene Expression Profiling/methods , Gene Expression Profiling/statistics & numerical data , Gene Expression Profiling/veterinary , Immunohistochemistry/methods , Immunohistochemistry/veterinary , Intestinal Mucosa/chemistry , Intestine, Small/chemistry , Intestine, Small/metabolism , Male , Mice , Mice, Inbred ICR , Multidrug Resistance-Associated Proteins/biosynthesis , Multidrug Resistance-Associated Proteins/immunology , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Oligonucleotide Array Sequence Analysis/veterinary , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/statistics & numerical data , Polymerase Chain Reaction/veterinary , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis
15.
PLoS One ; 7(11): e49058, 2012.
Article in English | MEDLINE | ID: mdl-23139832

ABSTRACT

Consumption of low-carbohydrate, high-protein, high-fat diets lead to rapid weight loss but the cardioprotective effects of these diets have been questioned. We examined the impact of high-protein and high-fat diets on cholesterol metabolism by comparing the plasma cholesterol and the expression of cholesterol biosynthesis genes in the liver of mice fed a high-fat (HF) diet that has a high (H) or a low (L) protein-to-carbohydrate (P/C) ratio. H-P/C-HF feeding, compared with L-P/C-HF feeding, decreased plasma total cholesterol and increased HDL cholesterol concentrations at 4-wk. Interestingly, the expression of genes involved in hepatic steroid biosynthesis responded to an increased dietary P/C ratio by first down-regulation (2-d) followed by later up-regulation at 4-wk, and the temporal gene expression patterns were connected to the putative activity of SREBF1 and 2. In contrast, Cyp7a1, the gene responsible for the conversion of cholesterol to bile acids, was consistently up-regulated in the H-P/C-HF liver regardless of feeding duration. Over expression of Cyp7a1 after 2-d and 4-wk H-P/C-HF feeding was connected to two unique sets of transcription regulators. At both time points, up-regulation of the Cyp7a1 gene could be explained by enhanced activations and reduced suppressions of multiple transcription regulators. In conclusion, we demonstrated that the hypocholesterolemic effect of H-P/C-HF feeding coincided with orchestrated changes of gene expressions in lipid metabolic pathways in the liver of mice. Based on these results, we hypothesize that the cholesterol lowering effect of high-protein feeding is associated with enhanced bile acid production but clinical validation is warranted. (246 words).


Subject(s)
Cholesterol/metabolism , Diet, High-Fat , Dietary Carbohydrates/pharmacology , Dietary Proteins/pharmacology , Animals , Cholesterol 7-alpha-Hydroxylase/metabolism , Dyslipidemias/complications , Dyslipidemias/metabolism , Dyslipidemias/pathology , Energy Intake/drug effects , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Models, Biological , Obesity/complications , Obesity/metabolism , Obesity/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Steroids/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Time Factors , Weight Gain/drug effects
16.
Mol Immunol ; 48(9-10): 1091-101, 2011 May.
Article in English | MEDLINE | ID: mdl-21367460

ABSTRACT

A complex interplay between the microbiota and the host immune system is evidenced to shape the immune system throughout life, but little is known about the microbial effect on key players of the adaptive immune system, the B2 B cells. In the presented study, we have evaluated the effect of commensal bacteria on B cell ontogeny and function, with the focus on B2 B cells of spleen and Peyer's patches. We have compared germ-free mice to mice that are exposed to a normal complex bacterial community from the day of birth and combined classical immunological assessment with advanced genome-wide expression profiling. Despite a preservation of all B cell subsets and phenotype, our results show that microbiota strongly impact mucosal B cell physiology and lead to higher serum Ig concentrations. We show that this microbial influence comprises downregulation of transcription factors involved in early B cell activation steps and upregulation of genes and proteins involved in later stages of B cell response. In summary, we show an influence of the gut microbiota on function of mucosal B2 B cells, involving mechanisms downstream of B cell activation and proliferation.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Gastrointestinal Tract/microbiology , Metagenome/immunology , Animals , Biomarkers/metabolism , Cell Proliferation , Colony Count, Microbial , Gastrointestinal Tract/immunology , Gene Expression Profiling , Gene Expression Regulation , Immunoglobulins/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocyte Activation/immunology , Mice , Peyer's Patches/cytology , Peyer's Patches/immunology , Reproducibility of Results , Spleen/cytology , Spleen/immunology
17.
Mol Nutr Food Res ; 55(11): 1603-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21957032

ABSTRACT

SCOPE: Weight maintenance after intended weight loss is a challenge in an obesogenic environment. In a large multicentre dietary intervention study (DiOGenes), it has recently been demonstrated that a high-protein/low-glycaemic index (HP/LGI) diet was slightly more efficient in maintaining weight loss than low-protein/LGI or high-GI (LP/LGI or HGI) diets. Here, we use a proteomic approach to assess the molecular mechanisms behind this positive effect. METHODS AND RESULTS: A subset of the most successful (weight loser, n=12) and unsuccessful (weight re-gainer, n=12) individuals consuming the LGI diets with either high- or low-protein content (HP or LP/LGI), following an initial calorie deficit run-in weight loss phase, were analyzed at the plasma protein level. Proteomic analysis revealed 18 proteins regulated after 6 months of the dietary weight maintenance phase. Furthermore, 12 proteins were significantly regulated as a function of success rate under an HP diet, arising as candidate biomarkers of mechanisms of successful weight maintenance under an HP/LGI diet. Pregnancy-zone protein (PZP) and protein S (PROS1) were revealed as novel biomarkers of weight maintenance showing opposite effects. CONCLUSION: Semantic network analysis of the 12 regulated proteins revealed that under an HP/LGI an anti-atherogenic effect and alterations of fat metabolism were associated with the success of maintaining the initial weight loss.


Subject(s)
Diet, Protein-Restricted , Diet, Reducing , Dietary Proteins/administration & dosage , Glycemic Index , Overweight/blood , Overweight/prevention & control , Adult , Biomarkers/blood , Blood Proteins/analysis , Body Mass Index , Cohort Studies , Europe , Family Health , Female , Follow-Up Studies , Genetic Predisposition to Disease , Humans , Obesity/blood , Obesity/diet therapy , Obesity/genetics , Obesity/prevention & control , Overweight/diet therapy , Overweight/genetics , Pregnancy Proteins/blood , Protein S , Secondary Prevention , Weight Loss
18.
Exp Gerontol ; 44(3): 208-18, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19049860

ABSTRACT

Engineering of fetal tissue has a high potential for the treatment of acute and chronic wounds of the skin in humans as these cells have high expansion capacity under simple culture conditions and one organ donation can produce Master Cell Banks which can fabricate over 900 million biological bandages (9 x 12cm). In a Phase 1 clinical safety study, cases are presented for the treatment of therapy resistant leg ulcers. All eight patients, representing 13 ulcers, tolerated multiple treatments with fetal biological bandages showing no negative secondary effects and repair processes similar to that seen in 3rd degree burns. Differential gene profiling using Affymetrix gene chips (analyzing 12,500 genes) were accomplished on these banked fetal dermal skin cells compared to banked dermal skin cells of an aged donor in order to point to potential indicators of wound healing. Families of genes involved in cell adhesion and extracellular matrix, cell cycle, cellular signaling, development and immune response show significant differences in regulation between banked fetal and those from banked old skin cells: with approximately 47.0% of genes over-expressed in fetal fibroblasts. It is perhaps these differences which contribute to efficient tissue repair seen in the clinic with fetal cell therapy.


Subject(s)
Fibroblasts/cytology , Leg Ulcer/therapy , Multigene Family/genetics , Skin Transplantation/methods , Skin/cytology , Wound Healing/genetics , Adult , Aged , Aged, 80 and over , Analysis of Variance , Biological Dressings , Cell Line , Cells, Cultured , Female , Fetus/cytology , Foreskin/cytology , Gene Expression Profiling/methods , Gene Expression Profiling/statistics & numerical data , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Polymerase Chain Reaction , RNA/isolation & purification , Skin/embryology , Tissue Engineering/methods
19.
Am J Physiol Gastrointest Liver Physiol ; 291(1): G63-72, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16455785

ABSTRACT

Using mice deficient in hepatic cytochrome P-450 oxidoreductase (POR), which disables the liver cytochrome P-450 system, we examined the metabolism and biological response of the anticarcinogenic flavonoid, quercetin. Profiling circulating metabolites revealed similar profiles over 72 h in wild-type (WT) and POR-null (KO) mice, showing that hepatic P450 and reduced biliary secretion do not affect quercetin metabolism. Transcriptional profiling at 24 h revealed that two- to threefold more genes responded significantly to quercetin in WT compared with KO in the jejunum, ileum, colon, and liver, suggesting that hepatic P450s mediate many of the biological effects of quercetin, such as immune function, estrogen receptor signaling, and lipid, glutathione, purine, and amino acid metabolism, even though quercetin metabolism is not modified. The functional interpretation of expression data in response to quercetin (single dose of 7 mg/animal) revealed a molecular relationship between the liver and jejunum. In WT animals, amino acid and sterol metabolism was predominantly modulated in the liver, fatty acid metabolism response was shared between the liver and jejunum, and glutathione metabolism was modulated in the small intestine. In contrast, KO animals do not regulate amino acid metabolism in the liver or small intestine, they share the control of fatty acid metabolism between the liver and jejunum, and regulation of sterol metabolism is shifted from the liver to the jejunum and that of glutathione metabolism from the jejunum to the liver. This demonstrates that the quercetin-mediated regulation of these biological functions in extrahepatic tissues is dependent on the functionality of the liver POR. In conclusion, using a systems biology approach to explore the contribution of hepatic phase 1 detoxification on quercetin metabolism demonstrated the resiliency and adaptive capacity of a biological organism in dealing with a bioactive nutrient when faced with a tissue-specific molecular dysfunction.


Subject(s)
Homeostasis/physiology , Jejunum/physiology , Liver/physiology , NADPH-Ferrihemoprotein Reductase/metabolism , Quercetin/administration & dosage , Transcription Factors/metabolism , Transcription, Genetic/physiology , Animals , Dose-Response Relationship, Drug , Homeostasis/drug effects , Jejunum/drug effects , Liver/drug effects , Male , Mice , Mice, Knockout , NADPH-Ferrihemoprotein Reductase/genetics , Transcription, Genetic/drug effects
20.
Gastroenterology ; 129(1): 321-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16012957

ABSTRACT

BACKGROUND & AIMS: Regulation of gene expression in the follicle-associated epithelium (FAE) over Peyer's patches is largely unknown. CCL20, a chemokine that recruits immature dendritic cells, is one of the few FAE-specific markers described so far. Lymphotoxin beta (LTalpha1beta2) expressed on the membrane of immune cells triggers CCL20 expression in enterocytes. In this study, we measured expression profiles of LTalpha1beta2-treated intestinal epithelial cells and selected CCL20 -coregulated genes to identify new FAE markers. METHODS: Genomic profiles of T84 and Caco-2 cell lines treated with either LTalpha1beta2, flagellin, or tumor necrosis factor alpha were measured using the Affymetrix GeneChip U133A. Clustering analysis was used to select CCL20 -coregulated genes, and laser dissection microscopy and real-time polymerase chain reaction on human biopsy specimens was used to assess the expression of the selected markers. RESULTS: Applying a 2-way analysis of variance, we identified regulated genes upon the different treatments. A subset of genes involved in inflammation and related to the nuclear factor kappaB pathway was coregulated with CCL20 . Among these genes, the antiapoptotic factor TNFAIP3 was highly expressed in the FAE. CCL23 , which was not coregulated in vitro with CCL20 , was also specifically expressed in the FAE. CONCLUSIONS: We have identified 2 novel human FAE specifically expressed genes. Most of the CCL20 -coregulated genes did not show FAE-specific expression, suggesting that other signaling pathways are critical to modulate FAE-specific gene expression.


Subject(s)
Biomarkers , Genomics/methods , Intestinal Mucosa/physiology , Microdissection , Polymerase Chain Reaction , Caco-2 Cells , Chemokines, CC/genetics , DNA-Binding Proteins , Flagellin/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lasers , Lymphotoxin-alpha/genetics , Lymphotoxin-beta , Membrane Proteins/genetics , Multigene Family , Nuclear Proteins , Phenotype , Proteins/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL