Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
JHEP Rep ; 5(5): 100693, 2023 May.
Article in English | MEDLINE | ID: mdl-37122688

ABSTRACT

Background & Aims: Non-alcoholic fatty liver disease (NAFLD) has a prevalence of ∼25% worldwide, with significant public health consequences yet few effective treatments. Human genetics can help elucidate novel biology and identify targets for new therapeutics. Genetic variants in mitochondrial amidoxime-reducing component 1 (MTARC1) have been associated with NAFLD and liver-related mortality; however, its pathophysiological role and the cell type(s) mediating these effects remain unclear. We aimed to investigate how MTARC1 exerts its effects on NAFLD by integrating human genetics with in vitro and in vivo studies of mARC1 knockdown. Methods: Analyses including multi-trait colocalisation and Mendelian randomisation were used to assess the genetic associations of MTARC1. In addition, we established an in vitro long-term primary human hepatocyte model with metabolic readouts and used the Gubra Amylin NASH (GAN)-diet non-alcoholic steatohepatitis mouse model treated with hepatocyte-specific N-acetylgalactosamine (GalNAc)-siRNA to understand the in vivo impacts of MTARC1. Results: We showed that genetic variants within the MTARC1 locus are associated with liver enzymes, liver fat, plasma lipids, and body composition, and these associations are attributable to the same causal variant (p.A165T, rs2642438 G>A), suggesting a shared mechanism. We demonstrated that increased MTARC1 mRNA had an adverse effect on these traits using Mendelian randomisation, implying therapeutic inhibition of mARC1 could be beneficial. In vitro mARC1 knockdown decreased lipid accumulation and increased triglyceride secretion, and in vivo GalNAc-siRNA-mediated knockdown of mARC1 lowered hepatic but increased plasma triglycerides. We found alterations in pathways regulating lipid metabolism and decreased secretion of 3-hydroxybutyrate upon mARC1 knockdown in vitro and in vivo. Conclusions: Collectively, our findings from human genetics, and in vitro and in vivo hepatocyte-specific mARC1 knockdown support the potential efficacy of hepatocyte-specific targeting of mARC1 for treatment of NAFLD. Impact and implications: We report that genetically predicted increases in MTARC1 mRNA associate with poor liver health. Furthermore, knockdown of mARC1 reduces hepatic steatosis in primary human hepatocytes and a murine NASH model. Together, these findings further underscore the therapeutic potential of targeting hepatocyte MTARC1 for NAFLD.

2.
J Am Soc Nephrol ; 30(6): 1109-1122, 2019 06.
Article in English | MEDLINE | ID: mdl-31085678

ABSTRACT

BACKGROUND: Studies have identified many common genetic associations that influence renal function and all-cause CKD, but these explain only a small fraction of variance in these traits. The contribution of rare variants has not been systematically examined. METHODS: We performed exome sequencing of 3150 individuals, who collectively encompassed diverse CKD subtypes, and 9563 controls. To detect causal genes and evaluate the contribution of rare variants we used collapsing analysis, in which we compared the proportion of cases and controls carrying rare variants per gene. RESULTS: The analyses captured five established monogenic causes of CKD: variants in PKD1, PKD2, and COL4A5 achieved study-wide significance, and we observed suggestive case enrichment for COL4A4 and COL4A3. Beyond known disease-associated genes, collapsing analyses incorporating regional variant intolerance identified suggestive dominant signals in CPT2 and several other candidate genes. Biallelic mutations in CPT2 cause carnitine palmitoyltransferase II deficiency, sometimes associated with rhabdomyolysis and acute renal injury. Genetic modifier analysis among cases with APOL1 risk genotypes identified a suggestive signal in AHDC1, implicated in Xia-Gibbs syndrome, which involves intellectual disability and other features. On the basis of the observed distribution of rare variants, we estimate that a two- to three-fold larger cohort would provide 80% power to implicate new genes for all-cause CKD. CONCLUSIONS: This study demonstrates that rare-variant collapsing analyses can validate known genes and identify candidate genes and modifiers for kidney disease. In so doing, these findings provide a motivation for larger-scale investigation of rare-variant risk contributions across major clinical CKD categories.


Subject(s)
Collagen Type IV/genetics , Exome Sequencing , Genetic Variation/genetics , Protein Kinases/genetics , Renal Insufficiency, Chronic/genetics , TRPP Cation Channels/genetics , Case-Control Studies , Female , Humans , Male , Prognosis , Protein Kinase D2 , Reference Values , Renal Insufficiency, Chronic/diagnosis
3.
N Engl J Med ; 380(2): 142-151, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30586318

ABSTRACT

BACKGROUND: Exome sequencing is emerging as a first-line diagnostic method in some clinical disciplines, but its usefulness has yet to be examined for most constitutional disorders in adults, including chronic kidney disease, which affects more than 1 in 10 persons globally. METHODS: We conducted exome sequencing and diagnostic analysis in two cohorts totaling 3315 patients with chronic kidney disease. We assessed the diagnostic yield and, among the patients for whom detailed clinical data were available, the clinical implications of diagnostic and other medically relevant findings. RESULTS: In all, 3037 patients (91.6%) were over 21 years of age, and 1179 (35.6%) were of self-identified non-European ancestry. We detected diagnostic variants in 307 of the 3315 patients (9.3%), encompassing 66 different monogenic disorders. Of the disorders detected, 39 (59%) were found in only a single patient. Diagnostic variants were detected across all clinically defined categories, including congenital or cystic renal disease (127 of 531 patients [23.9%]) and nephropathy of unknown origin (48 of 281 patients [17.1%]). Of the 2187 patients assessed, 34 (1.6%) had genetic findings for medically actionable disorders that, although unrelated to their nephropathy, would also lead to subspecialty referral and inform renal management. CONCLUSIONS: Exome sequencing in a combined cohort of more than 3000 patients with chronic kidney disease yielded a genetic diagnosis in just under 10% of cases. (Funded by the National Institutes of Health and others.).


Subject(s)
Exome , Genetic Predisposition to Disease , Mutation , Renal Insufficiency, Chronic/genetics , Sequence Analysis, DNA/methods , Adult , Aged , Cohort Studies , Genetic Variation , Humans , Male , Middle Aged , Renal Insufficiency, Chronic/ethnology , Young Adult
4.
Inflamm Bowel Dis ; 22(9): 2078-97, 2016 09.
Article in English | MEDLINE | ID: mdl-27542128

ABSTRACT

BACKGROUND: Beneficial roles for glucagon-like peptide 1 (GLP-1)/GLP-1R signaling have recently been described in diseases, where low-grade inflammation is a common phenomenon. We investigated the effects of GLP-1 in Brunner's glands and duodenum with abundant expression of GLP-1 receptors, as well as GLP-1 effect on colonic inflammation. METHODS: RNA from Brunner's glands of GLP-1R knockout and wild-type mice were subjected to full transcriptome profiling. Array results were validated by quantitative reverse transcription polymerase chain reaction in wild-type mice and compared with samples from inflammatory bowel disease (IBD) patients and controls. In addition, we performed a detailed investigation of the effects of exogenous liraglutide dosing in a T-cell driven adoptive transfer (AdTr) colitis mouse model. RESULTS: Analyses of the Brunner's gland transcriptomes of GLP-1R knockout and wild-type mice identified 722 differentially expressed genes. Upregulated transcripts after GLP-1 dosing included IL-33, chemokine ligand 20 (CCL20), and mucin 5b. Biopsies from IBD patients and controls, as well as data from the AdTr model, showed deregulated expression of GLP-1R, CCL20, and IL-33 in colon. Circulating levels of GLP-1 were found to be increased in mice with colitis. Finally, the colonic cytokine levels and disease scores of the AdTr model indicated reduced levels of colonic inflammation in liraglutide-dosed animals. CONCLUSIONS: We demonstrate that IL-33, GLP-1R, and CCL20 are deregulated in human IBD, and that prophylactic treatment with 0.6 mg/kg liraglutide improves disease in AdTr colitis. In addition, GLP-1 receptor agonists upregulate IL-33, mucin 5b, and CCL20 in murine Brunner's glands. Taken together, our data indicate that GLP-1 receptor agonists affect gut homeostasis in both proximal and distal parts of the gut.


Subject(s)
Brunner Glands/metabolism , Colitis/pathology , Colon/metabolism , Inflammatory Bowel Diseases/pathology , Liraglutide/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Chemokine CCL20/metabolism , Colitis/drug therapy , Female , Gene Expression , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Humans , Inflammation/pathology , Interleukin-33/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mucin-5B/metabolism , RNA, Messenger/analysis , Young Adult
5.
Drug Discov Today ; 20(10): 1176-81, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26050580

ABSTRACT

Recent groundbreaking work in genetics has identified thousands of small-effect genetic variants throughout the genome that are associated with almost all major diseases. These genome-wide association studies (GWAS) are often proposed as a source of future medical breakthroughs. However, with several notable exceptions, the journey from a small-effect genetic variant to a functional drug has proven arduous, and few examples of actual contributions to drug discovery exist. Here, we discuss novel approaches of overcoming this hurdle by using instead public genetics resources as a pragmatic guide alongside existing drug discovery methods. Our aim is to evaluate human genetic confidence as a rationale for drug target selection.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drug Design , Inflammation/drug therapy , Animals , Drug Discovery/methods , Genetic Variation , Genome-Wide Association Study/methods , Humans , Inflammation/genetics , Molecular Targeted Therapy
6.
Diabetes ; 63(11): 3785-97, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24947352

ABSTRACT

Upper- and lower-body fat depots exhibit opposing associations with obesity-related metabolic disease. We defined the relationship between DEXA-quantified fat depots and diabetes/cardiovascular risk factors in a healthy population-based cohort (n = 3,399). Gynoid fat mass correlated negatively with insulin resistance after total fat mass adjustment, whereas the opposite was seen for abdominal fat. Paired transcriptomic analysis of gluteal subcutaneous adipose tissue (GSAT) and abdominal subcutaneous adipose tissue (ASAT) was performed across the BMI spectrum (n = 49; 21.4-45.5 kg/m(2)). In both depots, energy-generating metabolic genes were negatively associated and inflammatory genes were positively associated with obesity. However, associations were significantly weaker in GSAT. At the systemic level, arteriovenous release of the proinflammatory cytokine interleukin-6 (n = 34) was lower from GSAT than ASAT. Isolated preadipocytes retained a depot-specific transcriptional "memory" of embryonic developmental genes and exhibited differential promoter DNA methylation of selected genes (HOTAIR, TBX5) between GSAT and ASAT. Short hairpin RNA-mediated silencing identified TBX5 as a regulator of preadipocyte proliferation and adipogenic differentiation in ASAT. In conclusion, intrinsic differences in the expression of developmental genes in regional adipocytes provide a mechanistic basis for diversity in adipose tissue (AT) function. The less inflammatory nature of lower-body AT offers insight into the opposing metabolic disease risk associations between upper- and lower-body obesity.


Subject(s)
Adipose Tissue/metabolism , Cardiovascular Diseases/metabolism , Obesity/metabolism , Abdominal Fat/metabolism , Adult , DNA Methylation , Female , Humans , Intra-Abdominal Fat/metabolism , Male , Middle Aged , Risk Factors , Subcutaneous Fat, Abdominal/metabolism , T-Box Domain Proteins/metabolism
7.
Mod Rheumatol ; 23(4): 729-40, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22872428

ABSTRACT

OBJECTIVES: Gene expression signatures can provide an unbiased view into the molecular changes underlying biologically and medically interesting phenotypes. We therefore initiated this study to identify signatures that would be of utility in studying rheumatoid arthritis (RA). METHODS: We used microarray profiling of peripheral blood mononuclear cells (PBMCs) in 30 RA patients to assess the effect of different biologic agent (biologics) treatments and to quantify the degree of a type-I interferon (IFN) signature in these patients. A numeric score was derived for the quantification step and applied to patients with RA. To further characterize the IFN response in our cohort, we employed type-I IFN treatment of PBMCs in vitro and in reporter assays. RESULTS: Profiling identified a subset of RA patients with upregulation of type-I IFN-regulated transcripts, thereby corroborating previous reports showing RA to be heterogeneous for an IFN component. A comparison of individuals currently untreated with a biologic with those treated with infliximab, tocilizumab, or abatacept suggested that each biologic induces a specific gene signature in PBMCs. CONCLUSIONS: It is possible to observe signs of type-I IFN pathway activation in a subset of clinically active RA patients without C-reactive protein elevation. Furthermore, biologics-specific gene signatures in patients with RA indicate that looking for a biologic-specific response pattern may be a potential future tool for predicting individual patient response.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/genetics , Biological Products/therapeutic use , Gene Expression Profiling , Interferon Type I/genetics , Abatacept , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Female , Humans , Immunoconjugates/therapeutic use , Infliximab , Interferon Type I/metabolism , Male , Middle Aged , Transcriptome , Treatment Outcome
8.
PLoS Genet ; 8(2): e1002505, 2012.
Article in English | MEDLINE | ID: mdl-22383892

ABSTRACT

Metabolic Syndrome (MetS) is highly prevalent and has considerable public health impact, but its underlying genetic factors remain elusive. To identify gene networks involved in MetS, we conducted whole-genome expression and genotype profiling on abdominal (ABD) and gluteal (GLU) adipose tissue, and whole blood (WB), from 29 MetS cases and 44 controls. Co-expression network analysis for each tissue independently identified nine, six, and zero MetS-associated modules of coexpressed genes in ABD, GLU, and WB, respectively. Of 8,992 probesets expressed in ABD or GLU, 685 (7.6%) were expressed in ABD and 51 (0.6%) in GLU only. Differential eigengene network analysis of 8,256 shared probesets detected 22 shared modules with high preservation across adipose depots (D(ABD-GLU) = 0.89), seven of which were associated with MetS (FDR P<0.01). The strongest associated module, significantly enriched for immune response-related processes, contained 94/620 (15%) genes with inter-depot differences. In an independent cohort of 145/141 twins with ABD and WB longitudinal expression data, median variability in ABD due to familiality was greater for MetS-associated versus un-associated modules (ABD: 0.48 versus 0.18, P = 0.08; GLU: 0.54 versus 0.20, P = 7.8×10(-4)). Cis-eQTL analysis of probesets associated with MetS (FDR P<0.01) and/or inter-depot differences (FDR P<0.01) provided evidence for 32 eQTLs. Corresponding eSNPs were tested for association with MetS-related phenotypes in two GWAS of >100,000 individuals; rs10282458, affecting expression of RARRES2 (encoding chemerin), was associated with body mass index (BMI) (P = 6.0×10(-4)); and rs2395185, affecting inter-depot differences of HLA-DRB1 expression, was associated with high-density lipoprotein (P = 8.7×10(-4)) and BMI-adjusted waist-to-hip ratio (P = 2.4×10(-4)). Since many genes and their interactions influence complex traits such as MetS, integrated analysis of genotypes and coexpression networks across multiple tissues relevant to clinical traits is an efficient strategy to identify novel associations.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Metabolic Syndrome/genetics , Body Mass Index , Chemokines/genetics , Female , Genetic Loci , Genome-Wide Association Study , HLA-DRB1 Chains/genetics , Humans , Intercellular Signaling Peptides and Proteins , Metabolic Syndrome/pathology , Organ Specificity , Phenotype , Quantitative Trait Loci
9.
Regul Pept ; 175(1-3): 21-9, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22252224

ABSTRACT

Of the mammalian species, only the GLP-1 receptors of rat and human origin have been described and characterized. Here, we report the cloning of the homologous GLP-1 receptors from mouse, rabbit, pig, cynomolgus monkey and chimp. The GLP-1 receptor is highly conserved across species, thus underlining the physiological importance of the peptide hormone and its receptor across a wide range of mammals. We expressed the receptors by stable transfection of BHK cells, both in cell lines with high expression levels of the cloned receptors, as well as in cell lines with lower expression levels, more comparable to endogenous expression of these receptors. High expression levels of cloned GLP-1 receptors markedly increased the potency of GLP-1 and other high affinity ligands, whereas the K(d) values were not affected. For a low affinity ligand like the ago-allosteric modulator Compound 2, expression levels of the human GLP-1 receptor were important for maximal efficacy as well as potency. The two natural metabolites of GLP-1, GLP-1(9-37) and GLP-1(9-36)amide were agonists when tested on a cell line with high expression of the recombinant human GLP-1 receptor, whereas they behaved as (low potent) antagonists on a cell line that expressed the receptor endogenously, as well as cells expressing a moderate level of the recombinant human GLP-1 receptor. The amide form was a more potent agonist than the free acid from. In conclusion, receptor expression level is an important parametre for selecting cell lines with cloned GLP-1 receptors for functional characterization of physiological and pharmaceutical ligands.


Subject(s)
Cell Membrane/metabolism , Glucagon-Like Peptide 1/pharmacology , Lung/metabolism , Receptors, Glucagon/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Glucagon-Like Peptide-1 Receptor , Humans , Immunoblotting , Lung/cytology , Lung/drug effects , Mice , Molecular Sequence Data , Rabbits , Rats , Sequence Homology, Amino Acid , Swine
10.
PLoS One ; 6(11): e27338, 2011.
Article in English | MEDLINE | ID: mdl-22102887

ABSTRACT

To understand how miRNAs contribute to the molecular phenotype of adipose tissues and related traits, we performed global miRNA expression profiling in subcutaneous abdominal and gluteal adipose tissue of 70 human subjects and characterised which miRNAs were differentially expressed between these tissues. We found that 12% of the miRNAs were significantly differentially expressed between abdominal and gluteal adipose tissue (FDR adjusted p<0.05) in the primary study, of which 59 replicated in a follow-up study of 40 additional subjects. Further, 14 miRNAs were found to be associated with metabolic syndrome case-control status in abdominal tissue and three of these replicated (primary study: FDR adjusted p<0.05, replication: p<0.05 and directionally consistent effect). Genome-wide genotyping was performed in the 70 subjects to enable miRNA expression quantitative trait loci (eQTL) analysis. Candidate miRNA eQTLs were followed-up in the additional 40 subjects and six significant, independent cis-located miRNA eQTLs (primary study: p<0.001; replication: p<0.05 and directionally consistent effect) were identified. Finally, global mRNA expression profiling was performed in both tissues to enable association analysis between miRNA and target mRNA expression levels. We find 22% miRNAs in abdominal and 9% miRNAs in gluteal adipose tissue with expression levels significantly associated with the expression of corresponding target mRNAs (FDR adjusted p<0.05). Taken together, our results indicate a clear difference in the miRNA molecular phenotypic profile of abdominal and gluteal adipose tissue, that the expressions of some miRNAs are influenced by cis-located genetic variants and that miRNAs are associated with expression levels of their predicted mRNA targets.


Subject(s)
Abdominal Fat/physiology , Biomarkers/metabolism , Buttocks/physiology , MicroRNAs/genetics , Quantitative Trait Loci , RNA, Messenger/genetics , Adult , Case-Control Studies , Female , Follow-Up Studies , Gene Expression Profiling , Genome-Wide Association Study , Genotype , Humans , Male , Metabolic Syndrome/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide/genetics
11.
Eur J Pharmacol ; 596(1-3): 173-9, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18761337

ABSTRACT

Balaglitazone is a novel thiazolidinedione in clinical development for the treatment of type 2 diabetes. Common side effects associated with PPARgamma receptor agonists are weight gain, oedema and adipogenesis. Balaglitazone is a selective partial PPARgamma agonist and it has been speculated that such compounds have a more favourable safety margin than full agonists. We have compared impact of equi-efficacious antihyperglycaemic doses of balaglitazone with full PPARgamma agonist rosiglitazone on body fluid accumulation, cardiac enlargement, and adipogenesis. Equi-efficacious antihyperglycaemic doses (ED(90)) of balaglitazone (3 mg/kg/day) and rosiglitazone (6 mg/kg/day) were determined in male diabetic db/db mice. In adult male rats treated for up to 42 days, feeding, drinking, anthropometry, and plasma volumes were measured. Total plasma volume was measured with dye dilution technique. Compared to vehicle, rosiglitazone consistently increased food intake throughout the 42 day treatment period. In contrast, balaglitazone increased food intake in the last week of the experiment. However, both rosiglitazone and balaglitazone increased water intake. After 42 days, rosiglitazone treated rats displayed significantly elevated adiposity. Rosiglitazone increased total blood and plasma volumes throughout the treatment. Twenty-one days of balaglitazone treatment had no significant impact on blood or plasma volumes, whilst 42 days of balaglitazone increased plasma volume but to a significantly lesser extent than seen for rosiglitazone (vehicle: 46.1+/-1.5; balaglitazone: 50.8+/-1.21; rosiglitazone: 54.6+/-1.6 ml/kg). Heart weight was significantly elevated only in rosiglitazone treated animals. At doses inducing comparable antihyperglycaemic control, the full PPARgamma agonist, rosiglitazone, induces more pronounced body fluid retention and heart enlargement than seen for the partial PPARgamma agonist, balaglitazone. Thus, partial agonists may pose safer alternative to current anti-diabetic therapy with full PPARgamma agonist.


Subject(s)
Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , PPAR gamma/agonists , Quinazolines/adverse effects , Quinazolines/pharmacology , Thiazolidinediones/adverse effects , Thiazolidinediones/pharmacology , Adipogenesis/drug effects , Adipose Tissue/drug effects , Animals , Blood Volume/drug effects , Body Weight/drug effects , Drinking/drug effects , Eating/drug effects , Heart/anatomy & histology , Heart/drug effects , Humans , Male , Mice , Organ Size/drug effects , PPAR gamma/genetics , Rats , Rosiglitazone
12.
Bioorg Med Chem Lett ; 17(11): 3198-202, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17379517

ABSTRACT

Computational analysis of the ligand binding pocket of the three PPAR receptor subtypes was utilized in the design of potent PPARalpha agonists. Optimum PPARalpha potency and selectivity were obtained with substituents having van der Waals volume around 260. Compound 6 had a PPARalpha potency of 0.002 microM and a selectivity ratio to PPARgamma and PPARdelta of 410 and 2000, respectively.


Subject(s)
Drug Design , PPAR alpha/agonists , Phenylpropionates/chemistry , Phenylpropionates/pharmacology , Animals , Computers , Crystallography , Ligands , PPAR alpha/chemistry , Phenylpropionates/chemical synthesis
13.
J Med Chem ; 50(7): 1495-503, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17343371

ABSTRACT

The aim was to identify a novel selective PPARdelta agonist with full efficacy on free fatty acid (FFA) oxidation in vitro and plasma lipid correction in vivo. Using the triple PPARalpha,gamma,delta agonist 1 as the structural starting point, we wanted to investigate the possibility of obtaining selective PPARdelta agonists by modifying only the acidic part of 1, while holding the lipophilic half of the molecule constant. The structure-activity relationship was guided by in vitro transactivation data using the human PPAR receptors, FFA oxidation efficacy performed in the rat muscle L6 cell line, and in vivo rat pharmacokinetic properties. Compound 7 ([4-[3,3-bis-(4-bromo-phenyl)-allylthio]-2-chloro-phenoxy]-acetic acid) was identified as a selective, partial agonist with good oral pharmacokinetic properties in rat. Chronic treatment of high fat fed ApoB100/CETP-Tgn mice with 7 corrected the plasma lipid parameters and improved insulin sensitivity. These data suggest that selective PPARdelta agonists have the potential to become a novel treatment of dyslipidemia.


Subject(s)
Allyl Compounds/chemical synthesis , Lipid Metabolism/drug effects , PPAR delta/agonists , Phenylacetates/chemical synthesis , Administration, Oral , Allyl Compounds/pharmacokinetics , Allyl Compounds/pharmacology , Animals , Apolipoprotein B-100/genetics , Binding Sites , Cell Line , Cholesterol Ester Transfer Proteins/genetics , Crystallography, X-Ray , Dietary Fats/administration & dosage , Fatty Acids, Nonesterified/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Models, Molecular , Muscle, Skeletal/cytology , Oxidation-Reduction , Phenylacetates/pharmacokinetics , Phenylacetates/pharmacology , Rats , Structure-Activity Relationship , Transcriptional Activation
14.
J Mol Endocrinol ; 37(2): 301-16, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17032746

ABSTRACT

To understand the molecular mechanisms regulating pancreatic endocrine development and function, pancreatic gene expression was compared between Ngn3-deficient mice and littermate controls on embryonic days 13 and 15. Microarray analysis identified 504 genes with significant differences in expression. Fifty-two of these showed at least twofold reduction in Ngn3 knockouts compared to controls. Many of them were previously described to be involved in endocrine development and function. Among the genes not previously characterized were Rhomboid veinlet-like 4, genes involved in tetrahydrobiopterin biosynthesis and the Iroquois-type homeobox gene Irx1, the latter was selected for further investigation. In situ hybridisation demonstrated that two Iroquois genes, Irx1 and Irx2, were expressed in pancreatic endoderm of wild-type, but not Ngn3 mutant embryos. Furthermore, ectopic Ngn3 induced prominent Irx2 expression in chicken endoderm. Co-labelling established that Irx1 and Irx2 mRNA is located to glucagon-, but not insulin- or somatostatin-producing cells in mice and chicken. These data suggest that Irx1 and Irx2 serve an evolutionary conserved role in the regulation of alpha-cell-specific gene expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Pancreas/embryology , Pancreas/metabolism , Animals , Chick Embryo , Cluster Analysis , Endoderm/metabolism , Gene Expression , Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Transcription Factors/metabolism
15.
J Proteome Res ; 5(7): 1751-62, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16823983

ABSTRACT

The Harderian gland is an orbital gland located behind the ocular bulb in most terrestrial vertebrates probably functioning for production of lipid secretion to protect the eye. We herein present a protein reference database of the rat Harderian gland that may serve as analytical tool for future proteomic work, report lipid and porphyrin handling cascades, address sequence conflicts and report structures that have not been so far described by proteomics methods.


Subject(s)
Harderian Gland/metabolism , Proteome/analysis , Rats/anatomy & histology , Amino Acid Sequence , Animals , Databases, Protein , Immunohistochemistry , Lipid Metabolism , Lipoproteins/chemistry , Male , Molecular Sequence Data , Peptide Mapping , Porphyrins/chemistry , Porphyrins/metabolism , Proteomics/methods , Rats/metabolism , Rats, Sprague-Dawley , Sequence Homology, Amino Acid , Uroporphyrinogen Decarboxylase/chemistry
16.
Ann N Y Acad Sci ; 1067: 448-53, 2006 May.
Article in English | MEDLINE | ID: mdl-16804025

ABSTRACT

Type 2 diabetes is a metabolic disease characterized by increased plasma glucose and insulin as well as dyslipidemia. If left untreated, chronic diseases will develop that are associated with neuropathic damage and higher mortality risk. Using a rational drug design, novel compounds have been developed that selectively activate the human PPAR receptors, leading to lessening of hyperglycemia and hyperinsulinemia as well as reduction of lipid levels in conjunction with an increase of the beneficial HDL-cholesterol. These PPAR agonists showed increased potency and efficacy compared to previously marketed insulin sensitizers. Lead compounds with desirable pharmacokinetic properties were chosen for further testing in several animal models. The in vivo activity of some synthetic ligands, capable of activating two or all three members of peroxisome proliferator-activated receptors (PPAR) family of receptors, suggested that they may have improved efficacy in type 2 diabetes therapy. Here, we briefly summarize the development of some novel PPAR agonists identified by our group in recent years.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Peroxisome Proliferator-Activated Receptors/agonists , Animals , Dyslipidemias/drug therapy , Humans , Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Models, Biological , Peroxisome Proliferator-Activated Receptors/classification , Peroxisome Proliferator-Activated Receptors/physiology
17.
Am J Physiol Endocrinol Metab ; 288(6): E1137-45, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15883192

ABSTRACT

Recent studies have suggested that sensory nerves may influence insulin secretion and action. The present study investigated the effects of resiniferatoxin (RTX) inactivation of sensory nerves (desensitization) on oral glucose tolerance, insulin secretion and whole body insulin sensitivity in the glucose intolerant, hyperinsulinemic, and insulin-resistant obese Zucker rat. After RTX treatment (0.05 mg/kg RTX sc given at ages 8, 10, and 12 wk), fasting plasma insulin was reduced (P < 0.0005), and oral glucose tolerance was improved (P < 0.005). Pancreas perfusion showed that baseline insulin secretion (7 mM glucose) was lower in RTX-treated rats (P = 0.01). Insulin secretory responsiveness to 20 mM glucose was enhanced in the perfused pancreas of RTX-treated rats (P < 0.005) but unaffected in stimulated, isolated pancreatic islets. At the peak of spontaneous insulin resistance in the obese Zucker rat, insulin sensitivity was substantially improved after RTX treatment, as evidenced by higher glucose infusion rates (GIR) required to maintain euglycemia during a hyperinsulinemic euglycemic (5 mU.kg(-1).min(-1)) clamp (GIR(60-120min): 5.97 +/- 0.62 vs. 11.65 +/- 0.83 mg.kg(-1).min(-1) in RTX-treated rats, P = 0.003). In conclusion, RTX treatment and, hence, sensory nerve desensitization of adult male obese Zucker rats improved oral glucose tolerance by enhancing insulin secretion, and, in particular, by improving insulin sensitivity.


Subject(s)
Diterpenes/pharmacology , Insulin/metabolism , Neurons, Afferent/drug effects , Neurotoxins/pharmacology , Pancreas/drug effects , Animals , Body Weight/drug effects , Body Weight/physiology , Drinking/drug effects , Eating/drug effects , Glucose/metabolism , Glucose Clamp Technique , Glucose Tolerance Test , In Vitro Techniques , Insulin/pharmacology , Insulin Resistance/physiology , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Neurons, Afferent/physiology , Pancreas/metabolism , Perfusion , Random Allocation , Rats , Rats, Zucker
18.
Scand J Gastroenterol ; 40(2): 198-205, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15764152

ABSTRACT

OBJECTIVE: Peroxisome proliferator-activated receptor (PPAR) ligands, widely used in type 2 diabetes treatment, have variably been shown to promote or prevent colon tumor formation in animal models and cell lines, but their role in normal human colon is unknown. The aim of this study was to determine PPAR expression and function in normal human colonic epithelial cells and tubular adenomas. MATERIAL AND METHODS: Short-term cultures of normal human colonic epithelial cells were established from biopsies obtained in 42 patients with normal colonoscopy. PPAR and adipophilin mRNA expression was assessed by real-time RT-PCR. PPARs were activated by ligands for PPAR alpha (Wy-14643), PPAR delta (GW-501516) and PPAR gamma (rosiglitazone or troglitazone). Cell viability was measured using the methyltetrazoleum assay, proliferation by thymidine incorporation, and DNA profiles by flow cytometry. PPAR mRNA levels in tubular adenomas or metaplastic polyps (n=12) were compared with those in controls. RESULTS: PPAR alpha and gamma were consistently expressed in normal colonocytes while no PPAR delta expression could be detected. PPAR gamma activation induced a 7.5-fold increase in adipophilin expression (a PPAR-activated gene). PPAR gamma activation had no effect on viability or DNA profiles, but led to a 25% significant decrease in cell proliferation. Finally, a selective and significant 2.5-fold decrease in PPAR alpha expression was observed in tubular adenomas, but not in metaplastic polyps, compared to controls. CONCLUSIONS: Our findings support the view that PPAR gamma ligands act as anti-proliferative agents rather than as promoters of tumorigenesis in normal human colon. Moreover, they raise interest in investigation of PPAR alpha as a therapeutic target to prevent adenoma formation.


Subject(s)
Adenoma/metabolism , Colonic Neoplasms/metabolism , Epithelial Cells/metabolism , Peroxisome Proliferator-Activated Receptors/biosynthesis , Adult , Aged , Aged, 80 and over , Cell Proliferation , Cell Survival/physiology , Colon , Female , Humans , Male , Middle Aged
19.
Bioorg Med Chem Lett ; 15(5): 1497-500, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15713415

ABSTRACT

A series of dimeric PPAR agonists were designed and tested for PPAR activity in vitro. The SAR showed that dimeric ligands with a common group or full dimeric ligands had retained or even increased PPARgamma potency. The dimeric agonist concept can be used to fine tune the subtype selectivity of PPAR agonists. The PPARgamma potency could, at least partly, be explained using molecular modeling.


Subject(s)
Peroxisome Proliferator-Activated Receptors/agonists , Dimerization , Drug Design , Humans , Ligands , Models, Molecular , Molecular Structure , Peroxisome Proliferator-Activated Receptors/chemistry , Structure-Activity Relationship
20.
Diabetes ; 53(10): 2603-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15448090

ABSTRACT

Several recent reports claim the generation of insulin-producing cells from embryonic stem cells via the differentiation of progenitors that express nestin. Here, we investigate further the properties of these insulin-containing cells. We find that although differentiated cells contain immunoreactive insulin, they do not contain proinsulin-derived C-peptide. Furthermore, we find variable insulin release from these cells upon glucose addition, but C-peptide release is never detected. In addition, many of the insulin-immunoreactive cells are undergoing apoptosis or necrosis. We further show that cells cultured in the presence of a phosphoinositide 3-kinase inhibitor, which previously was reported to facilitate the differentiation of insulin(+) cells, are not C-peptide immunoreactive but take up fluorescein isothiocyanate-labeled insulin from the culture medium. Together, these data suggest that nestin(+) progenitor cells give rise to a population of cells that contain insulin, not as a result of biosynthesis but from the uptake of exogenous insulin. We conclude that C-peptide biosynthesis and secretion should be demonstrated to claim insulin production from embryonic stem cell progeny.


Subject(s)
C-Peptide/metabolism , Cell Differentiation/physiology , Insulin/metabolism , Stem Cells/metabolism , Animals , Apoptosis/physiology , Artifacts , Cell Culture Techniques , Cell Differentiation/drug effects , Glucose/pharmacology , Humans , In Situ Nick-End Labeling , Insulin Secretion , Mice , Stem Cells/cytology , Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL