Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
2.
Nat Microbiol ; 8(3): 424-440, 2023 03.
Article En | MEDLINE | ID: mdl-36759753

The molecular bases of how host genetic variation impacts the gut microbiome remain largely unknown. Here we used a genetically diverse mouse population and applied systems genetics strategies to identify interactions between host and microbe phenotypes including microbial functions, using faecal metagenomics, small intestinal transcripts and caecal lipids that influence microbe-host dynamics. Quantitative trait locus (QTL) mapping identified murine genomic regions associated with variations in bacterial taxa; bacterial functions including motility, sporulation and lipopolysaccharide production and levels of bacterial- and host-derived lipids. We found overlapping QTL for the abundance of Akkermansia muciniphila and caecal levels of ornithine lipids. Follow-up in vitro and in vivo studies revealed that A. muciniphila is a major source of these lipids in the gut, provided evidence that ornithine lipids have immunomodulatory effects and identified intestinal transcripts co-regulated with these traits including Atf3, which encodes for a transcription factor that plays vital roles in modulating metabolism and immunity. Collectively, these results suggest that ornithine lipids are potentially important for A. muciniphila-host interactions and support the role of host genetics as a determinant of responses to gut microbes.


Gastrointestinal Microbiome , Verrucomicrobia , Mice , Animals , Verrucomicrobia/genetics , Gastrointestinal Microbiome/genetics , Akkermansia/genetics , Phenotype
3.
JCI Insight ; 7(10)2022 05 23.
Article En | MEDLINE | ID: mdl-35603790

Insulin secretion from pancreatic ß cells is essential for glucose homeostasis. An insufficient response to the demand for insulin results in diabetes. We previously showed that ß cell-specific deletion of Zfp148 (ß-Zfp148KO) improves glucose tolerance and insulin secretion in mice. Here, we performed Ca2+ imaging of islets from ß­Zfp148KO and control mice fed both a chow and a Western-style diet. ß-Zfp148KO islets demonstrated improved sensitivity and sustained Ca2+ oscillations in response to elevated glucose levels. ß-Zfp148KO islets also exhibited elevated sensitivity to amino acid-induced Ca2+ influx under low glucose conditions, suggesting enhanced mitochondrial phosphoenolpyruvate-dependent (PEP-dependent), ATP-sensitive K+ channel closure, independent of glycolysis. RNA-Seq and proteomics of ß-Zfp148KO islets revealed altered levels of enzymes involved in amino acid metabolism (specifically, SLC3A2, SLC7A8, GLS, GLS2, PSPH, PHGDH, and PSAT1) and intermediary metabolism (namely, GOT1 and PCK2), consistent with altered PEP cycling. In agreement with this, ß-Zfp148KO islets displayed enhanced insulin secretion in response to l-glutamine and activation of glutamate dehydrogenase. Understanding pathways controlled by ZFP148 may provide promising strategies for improving ß cell function that are robust to the metabolic challenge imposed by a Western diet.


Insulin-Secreting Cells , Islets of Langerhans , Animals , Calcium/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucose/metabolism , Glutamine/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Nutrients , Transcription Factors/metabolism
4.
J Clin Invest ; 131(21)2021 11 01.
Article En | MEDLINE | ID: mdl-34491912

The transcription factor NFATC2 induces ß cell proliferation in mouse and human islets. However, the genomic targets that mediate these effects have not been identified. We expressed active forms of Nfatc2 and Nfatc1 in human islets. By integrating changes in gene expression with genomic binding sites for NFATC2, we identified approximately 2200 transcriptional targets of NFATC2. Genes induced by NFATC2 were enriched for transcripts that regulate the cell cycle and for DNA motifs associated with the transcription factor FOXP. Islets from an endocrine-specific Foxp1, Foxp2, and Foxp4 triple-knockout mouse were less responsive to NFATC2-induced ß cell proliferation, suggesting the FOXP family works to regulate ß cell proliferation in concert with NFATC2. NFATC2 induced ß cell proliferation in both mouse and human islets, whereas NFATC1 did so only in human islets. Exploiting this species difference, we identified approximately 250 direct transcriptional targets of NFAT in human islets. This gene set enriches for cell cycle-associated transcripts and includes Nr4a1. Deletion of Nr4a1 reduced the capacity of NFATC2 to induce ß cell proliferation, suggesting that much of the effect of NFATC2 occurs through its induction of Nr4a1. Integration of noncoding RNA expression, chromatin accessibility, and NFATC2 binding sites enabled us to identify NFATC2-dependent enhancer loci that mediate ß cell proliferation.


Cell Proliferation , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , NFATC Transcription Factors/metabolism , Response Elements , Transcription, Genetic , Animals , Humans , Mice, Knockout , NFATC Transcription Factors/genetics
5.
Nat Metab ; 2(10): 1149-1162, 2020 10.
Article En | MEDLINE | ID: mdl-32958938

Despite the crucial roles of lipids in metabolism, we are still at the early stages of comprehensively annotating lipid species and their genetic basis. Mass spectrometry-based discovery lipidomics offers the potential to globally survey lipids and their relative abundances in various biological samples. To discover the genetics of lipid features obtained through high-resolution liquid chromatography-tandem mass spectrometry, we analysed liver and plasma from 384 diversity outbred mice, and quantified 3,283 molecular features. These features were mapped to 5,622 lipid quantitative trait loci and compiled into a public web resource termed LipidGenie. The data are cross-referenced to the human genome and offer a bridge between genetic associations in humans and mice. Harnessing this resource, we used genome-lipid association data as an additional aid to identify a number of lipids, for example gangliosides through their association with B4galnt1, and found evidence for a group of sex-specific phosphatidylcholines through their shared locus. Finally, LipidGenie's ability to query either mass or gene-centric terms suggests acyl-chain-specific functions for proteins of the ABHD family.


Chromosome Mapping , Genome , Lipid Metabolism/genetics , Lipidomics , Lipids/chemistry , Lipids/genetics , Animals , Gangliosides/metabolism , Genome-Wide Association Study , Genotype , Humans , Hydrolases/genetics , Mice , Mice, Inbred C57BL , Phosphatidylcholines/metabolism , Phospholipases A2/genetics , Plasmids/genetics , Sex Characteristics
6.
PLoS Genet ; 15(8): e1008073, 2019 08.
Article En | MEDLINE | ID: mdl-31465442

The microbial communities that inhabit the distal gut of humans and other mammals exhibit large inter-individual variation. While host genetics is a known factor that influences gut microbiota composition, the mechanisms underlying this variation remain largely unknown. Bile acids (BAs) are hormones that are produced by the host and chemically modified by gut bacteria. BAs serve as environmental cues and nutrients to microbes, but they can also have antibacterial effects. We hypothesized that host genetic variation in BA metabolism and homeostasis influence gut microbiota composition. To address this, we used the Diversity Outbred (DO) stock, a population of genetically distinct mice derived from eight founder strains. We characterized the fecal microbiota composition and plasma and cecal BA profiles from 400 DO mice maintained on a high-fat high-sucrose diet for ~22 weeks. Using quantitative trait locus (QTL) analysis, we identified several genomic regions associated with variations in both bacterial and BA profiles. Notably, we found overlapping QTL for Turicibacter sp. and plasma cholic acid, which mapped to a locus containing the gene for the ileal bile acid transporter, Slc10a2. Mediation analysis and subsequent follow-up validation experiments suggest that differences in Slc10a2 gene expression associated with the different strains influences levels of both traits and revealed novel interactions between Turicibacter and BAs. This work illustrates how systems genetics can be utilized to generate testable hypotheses and provide insight into host-microbe interactions.


Bile Acids and Salts/metabolism , Biological Variation, Population/genetics , Gastrointestinal Microbiome/physiology , Organic Anion Transporters, Sodium-Dependent/genetics , Quantitative Trait Loci/genetics , Symporters/genetics , Akkermansia , Animals , Bile Acids and Salts/blood , Collaborative Cross Mice , Female , Firmicutes/growth & development , Male , Metabolic Networks and Pathways/genetics , Mice , Models, Animal , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Verrucomicrobia/growth & development
7.
J Clin Invest ; 129(10): 4419-4432, 2019 07 25.
Article En | MEDLINE | ID: mdl-31343992

Genetic susceptibility to type 2 diabetes is primarily due to ß-cell dysfunction. However, a genetic study to directly interrogate ß-cell function ex vivo has never been previously performed. We isolated 233,447 islets from 483 Diversity Outbred (DO) mice maintained on a Western-style diet, and measured insulin secretion in response to a variety of secretagogues. Insulin secretion from DO islets ranged >1,000-fold even though none of the mice were diabetic. The insulin secretory response to each secretagogue had a unique genetic architecture; some of the loci were specific for one condition, whereas others overlapped. Human loci that are syntenic to many of the insulin secretion QTL from mouse are associated with diabetes-related SNPs in human genome-wide association studies. We report on three genes, Ptpn18, Hunk and Zfp148, where the phenotype predictions from the genetic screen were fulfilled in our studies of transgenic mouse models. These three genes encode a non-receptor type protein tyrosine phosphatase, a serine/threonine protein kinase, and a Krϋppel-type zinc-finger transcription factor, respectively. Our results demonstrate that genetic variation in insulin secretion that can lead to type 2 diabetes is discoverable in non-diabetic individuals.


DNA-Binding Proteins/genetics , Genetic Loci , Insulin Secretion/genetics , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Transcription Factors/genetics , Animals , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Humans , Mice , Mice, Transgenic
8.
J Biol Chem ; 293(16): 5860-5877, 2018 04 20.
Article En | MEDLINE | ID: mdl-29496998

The mouse is a critical model in diabetes research, but most research in mice has been limited to a small number of mouse strains and limited genetic variation. Using the eight founder strains and both sexes of the Collaborative Cross (C57BL/6J (B6), A/J, 129S1/SvImJ (129), NOD/ShiLtJ (NOD), NZO/HILtJ (NZO), PWK/PhJ (PWK), WSB/EiJ (WSB), and CAST/EiJ (CAST)), we investigated the genetic dependence of diabetes-related metabolic phenotypes and insulin secretion. We found that strain background is associated with an extraordinary range in body weight, plasma glucose, insulin, triglycerides, and insulin secretion. Our whole-islet proteomic analysis of the eight mouse strains demonstrates that genetic background exerts a strong influence on the islet proteome that can be linked to the differences in diabetes-related metabolic phenotypes and insulin secretion. We computed protein modules consisting of highly correlated proteins that enrich for biological pathways and provide a searchable database of the islet protein expression profiles. To validate the data resource, we identified tyrosine hydroxylase (Th), a key enzyme in catecholamine synthesis, as a protein that is highly expressed in ß-cells of PWK and CAST islets. We show that CAST islets synthesize elevated levels of dopamine, which suppresses insulin secretion. Prior studies, using only the B6 strain, concluded that adult mouse islets do not synthesize l-3,4-dihydroxyphenylalanine (l-DOPA), the product of Th and precursor of dopamine. Thus, the choice of the CAST strain, guided by our islet proteomic survey, was crucial for these discoveries. In summary, we provide a valuable data resource to the research community, and show that proteomic analysis identified a strain-specific pathway by which dopamine synthesized in ß-cells inhibits insulin secretion.


Diabetes Mellitus, Type 2/metabolism , Dopamine/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Proteome/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Dopamine/genetics , Female , Genetic Variation , Glucagon/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Phenotype , Proteome/genetics , Proteomics
9.
Genetics ; 209(1): 335-356, 2018 05.
Article En | MEDLINE | ID: mdl-29567659

The majority of gene loci that have been associated with type 2 diabetes play a role in pancreatic islet function. To evaluate the role of islet gene expression in the etiology of diabetes, we sensitized a genetically diverse mouse population with a Western diet high in fat (45% kcal) and sucrose (34%) and carried out genome-wide association mapping of diabetes-related phenotypes. We quantified mRNA abundance in the islets and identified 18,820 expression QTL. We applied mediation analysis to identify candidate causal driver genes at loci that affect the abundance of numerous transcripts. These include two genes previously associated with monogenic diabetes (PDX1 and HNF4A), as well as three genes with nominal association with diabetes-related traits in humans (FAM83E, IL6ST, and SAT2). We grouped transcripts into gene modules and mapped regulatory loci for modules enriched with transcripts specific for α-cells, and another specific for δ-cells. However, no single module enriched for ß-cell-specific transcripts, suggesting heterogeneity of gene expression patterns within the ß-cell population. A module enriched in transcripts associated with branched-chain amino acid metabolism was the most strongly correlated with physiological traits that reflect insulin resistance. Although the mice in this study were not overtly diabetic, the analysis of pancreatic islet gene expression under dietary-induced stress enabled us to identify correlated variation in groups of genes that are functionally linked to diabetes-associated physiological traits. Our analysis suggests an expected degree of concordance between diabetes-associated loci in the mouse and those found in human populations, and demonstrates how the mouse can provide evidence to support nominal associations found in human genome-wide association mapping.


Genetic Association Studies , Islets of Langerhans/physiology , Quantitative Trait Loci , Quantitative Trait, Heritable , Alleles , Animals , Computational Biology/methods , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Genome-Wide Association Study/methods , Genotype , Glucagon-Secreting Cells/metabolism , Haplotypes , Humans , Mice , Somatostatin-Secreting Cells/metabolism , Transcriptome , Web Browser
10.
Cell Rep ; 18(7): 1739-1750, 2017 02 14.
Article En | MEDLINE | ID: mdl-28199845

Genetic variation drives phenotypic diversity and influences the predisposition to metabolic disease. Here, we characterize the metabolic phenotypes of eight genetically distinct inbred mouse strains in response to a high-fat/high-sucrose diet. We found significant variation in diabetes-related phenotypes and gut microbiota composition among the different mouse strains in response to the dietary challenge and identified taxa associated with these traits. Follow-up microbiota transplant experiments showed that altering the composition of the gut microbiota modifies strain-specific susceptibility to diet-induced metabolic disease. Animals harboring microbial communities with enhanced capacity for processing dietary sugars and for generating hydrophobic bile acids showed increased susceptibility to metabolic disease. Notably, differences in glucose-stimulated insulin secretion between different mouse strains were partially recapitulated via gut microbiota transfer. Our results suggest that the gut microbiome contributes to the genetic and phenotypic diversity observed among mouse strains and provide a link between the gut microbiome and insulin secretion.


Diabetes Mellitus/genetics , Diabetes Mellitus/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Insulin/metabolism , Microbiota/physiology , Animals , Bile Acids and Salts/metabolism , Diet, High-Fat/adverse effects , Genetic Variation/genetics , Genotype , Insulin Resistance/physiology , Male , Mice , Phenotype
11.
PLoS Genet ; 12(12): e1006466, 2016 Dec.
Article En | MEDLINE | ID: mdl-27935966

Human genome-wide association studies (GWAS) have shown that genetic variation at >130 gene loci is associated with type 2 diabetes (T2D). We asked if the expression of the candidate T2D-associated genes within these loci is regulated by a common locus in pancreatic islets. Using an obese F2 mouse intercross segregating for T2D, we show that the expression of ~40% of the T2D-associated genes is linked to a broad region on mouse chromosome (Chr) 2. As all but 9 of these genes are not physically located on Chr 2, linkage to Chr 2 suggests a genomic factor(s) located on Chr 2 regulates their expression in trans. The transcription factor Nfatc2 is physically located on Chr 2 and its expression demonstrates cis linkage; i.e., its expression maps to itself. When conditioned on the expression of Nfatc2, linkage for the T2D-associated genes was greatly diminished, supporting Nfatc2 as a driver of their expression. Plasma insulin also showed linkage to the same broad region on Chr 2. Overexpression of a constitutively active (ca) form of Nfatc2 induced ß-cell proliferation in mouse and human islets, and transcriptionally regulated more than half of the T2D-associated genes. Overexpression of either ca-Nfatc2 or ca-Nfatc1 in mouse islets enhanced insulin secretion, whereas only ca-Nfatc2 was able to promote ß-cell proliferation, suggesting distinct molecular pathways mediating insulin secretion vs. ß-cell proliferation are regulated by NFAT. Our results suggest that many of the T2D-associated genes are downstream transcriptional targets of NFAT, and may act coordinately in a pathway through which NFAT regulates ß-cell proliferation in both mouse and human islets.


Diabetes Mellitus, Type 2/genetics , Insulin/genetics , NFATC Transcription Factors/genetics , Animals , Cell Proliferation/genetics , Chromosome Mapping , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation , Genetic Linkage , Genome , Genome-Wide Association Study , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Obese , NFATC Transcription Factors/biosynthesis , Promoter Regions, Genetic
12.
Mol Cell ; 64(5): 982-992, 2016 12 01.
Article En | MEDLINE | ID: mdl-27889451

Histone-modifying enzymes regulate transcription and are sensitive to availability of endogenous small-molecule metabolites, allowing chromatin to respond to changes in environment. The gut microbiota produces a myriad of metabolites that affect host physiology and susceptibility to disease; however, the underlying molecular events remain largely unknown. Here we demonstrate that microbial colonization regulates global histone acetylation and methylation in multiple host tissues in a diet-dependent manner: consumption of a "Western-type" diet prevents many of the microbiota-dependent chromatin changes that occur in a polysaccharide-rich diet. Finally, we demonstrate that supplementation of germ-free mice with short-chain fatty acids, major products of gut bacterial fermentation, is sufficient to recapitulate chromatin modification states and transcriptional responses associated with colonization. These findings have profound implications for understanding the complex functional interactions between diet, gut microbiota, and host health.


Diet, Western , Epigenesis, Genetic , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Colon/enzymology , Colon/metabolism , DNA Methylation , Histones/genetics , Histones/metabolism , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Specificity
13.
Cell Cycle ; 15(23): 3191-3202, 2016 Dec.
Article En | MEDLINE | ID: mdl-27753532

Anti-silencing function 1 (ASF1) is a histone H3-H4 chaperone involved in DNA replication and repair, and transcriptional regulation. Here, we identify ASF1B, the mammalian paralog to ASF1, as a proliferation-inducing histone chaperone in human ß-cells. Overexpression of ASF1B led to distinct transcriptional signatures consistent with increased cellular proliferation and reduced cellular death. Using multiple methods of monitoring proliferation and mitotic progression, we show that overexpression of ASF1B is sufficient to induce human ß-cell proliferation. Co-expression of histone H3.3 further augmented ß-cell proliferation, whereas suppression of endogenous H3.3 attenuated the stimulatory effect of ASF1B. Using the histone binding-deficient mutant of ASF1B (V94R), we show that histone binding to ASF1B is required for the induction of ß-cell proliferation. In contrast to H3.3, overexpression of histone H3 variants H3.1 and H3.2 did not have an impact on ASF1B-mediated induction of proliferation. Our findings reveal a novel role of ASF1B in human ß-cell replication and show that ASF1B and histone H3.3A synergistically stimulate human ß-cell proliferation.


Cell Cycle Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Animals , Apoptosis/genetics , Cell Line , Cell Proliferation , DNA Damage/genetics , Gene Expression Regulation , Humans , Mice, Inbred C57BL , Mitosis/genetics , Protein Binding/genetics , Transcription, Genetic
14.
Cell Rep ; 16(2): 583-595, 2016 07 12.
Article En | MEDLINE | ID: mdl-27373151

We introduce neutron-encoded (NeuCode) amino acid labeling of mice as a strategy for multiplexed proteomic analysis in vivo. Using NeuCode, we characterize an inducible knockout mouse model of Bap1, a tumor suppressor and deubiquitinase whose in vivo roles outside of cancer are not well established. NeuCode proteomics revealed altered metabolic pathways following Bap1 deletion, including profound elevation of cholesterol biosynthetic machinery coincident with reduced expression of gluconeogenic and lipid homeostasis proteins in liver. Bap1 loss increased pancreatitis biomarkers and reduced expression of mitochondrial proteins. These alterations accompany a metabolic remodeling with hypoglycemia, hypercholesterolemia, hepatic lipid loss, and acinar cell degeneration. Liver-specific Bap1 null mice present with fully penetrant perinatal lethality, severe hypoglycemia, and hepatic lipid deficiency. This work reveals Bap1 as a metabolic regulator in liver and pancreas, and it establishes NeuCode as a reliable proteomic method for deciphering in vivo biology.


Proteomics/methods , Tumor Suppressor Proteins/physiology , Ubiquitin Thiolesterase/physiology , Animals , Hematopoiesis , Histones/metabolism , Isotope Labeling , Lipid Metabolism , Lysine/metabolism , Male , Metabolic Networks and Pathways , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Liver/metabolism , Pancreas/metabolism , Proteome/metabolism , Ubiquitination
15.
Genetics ; 201(3): 1253-62, 2015 Nov.
Article En | MEDLINE | ID: mdl-26385979

We surveyed gene expression in six tissues in an F2 intercross between mouse strains C57BL/6J (abbreviated B6) and BTBR T(+) tf/J (abbreviated BTBR) made genetically obese with the Leptin(ob) mutation. We identified a number of expression quantitative trait loci (eQTL) affecting the expression of numerous genes distal to the locus, called trans-eQTL hotspots. Some of these trans-eQTL hotspots showed effects in multiple tissues, whereas some were specific to a single tissue. An unusually large number of transcripts (∼8% of genes) mapped in trans to a hotspot on chromosome 6, specifically in pancreatic islets. By considering the first two principal components of the expression of genes mapping to this region, we were able to convert the multivariate phenotype into a simple Mendelian trait. Fine mapping the locus by traditional methods reduced the QTL interval to a 298-kb region containing only three genes, including Slco1a6, one member of a large family of organic anion transporters. Direct genomic sequencing of all Slco1a6 exons identified a nonsynonymous coding SNP that converts a highly conserved proline residue at amino acid position 564 to serine. Molecular modeling suggests that Pro564 faces an aqueous pore within this 12-transmembrane domain-spanning protein. When transiently overexpressed in HEK293 cells, BTBR organic anion transporting polypeptide (OATP)1A6-mediated cellular uptake of the bile acid taurocholic acid (TCA) was enhanced compared to B6 OATP1A6. Our results suggest that genetic variation in Slco1a6 leads to altered transport of TCA (and potentially other bile acids) by pancreatic islets, resulting in broad gene regulation.


Islets of Langerhans/metabolism , Organic Anion Transporters/genetics , Amino Acid Substitution , Animals , Carrier Proteins , Gene Expression Regulation , HEK293 Cells , Humans , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Quantitative Trait Loci , Taurocholic Acid/metabolism
16.
J Am Chem Soc ; 136(37): 12848-51, 2014 Sep 17.
Article En | MEDLINE | ID: mdl-25191938

Glucagon-like peptide-1 (GLP-1) is a natural agonist for GLP-1R, a G protein-coupled receptor (GPCR) on the surface of pancreatic ß cells. GLP-1R agoinsts are attractive for treatment of type 2 diabetes, but GLP-1 itself is rapidly degraded by peptidases in vivo. We describe a design strategy for retaining GLP-1-like activity while engendering prolonged activity in vivo, based on strategic replacement of native α residues with conformationally constrained ß-amino acid residues. This backbone-modification approach may be useful for developing stabilized analogues of other peptide hormones.


Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Receptors, Glucagon/agonists , Amino Acid Sequence , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Humans , Mice , Molecular Sequence Data , Protein Stability
17.
J Biol Chem ; 289(36): 25276-86, 2014 Sep 05.
Article En | MEDLINE | ID: mdl-25002582

The abundance and functional activity of proteins involved in the formation of the SNARE complex are tightly regulated for efficient exocytosis. Tomosyn proteins are negative regulators of exocytosis. Tomosyn causes an attenuation of insulin secretion by limiting the formation of the SNARE complex. We hypothesized that glucose-dependent stimulation of insulin secretion from ß-cells must involve reversing the inhibitory action of tomosyn. Here, we show that glucose increases tomosyn protein turnover. Within 1 h of exposure to 15 mM glucose, ~50% of tomosyn was degraded. The degradation of tomosyn in response to high glucose was blocked by inhibitors of the proteasomal pathway. Using (32)P labeling and mass spectrometry, we showed that tomosyn-2 is phosphorylated in response to high glucose, phorbol esters, and analogs of cAMP, all key insulin secretagogues. We identified 11 phosphorylation sites in tomosyn-2. Site-directed mutagenesis was used to generate phosphomimetic (Ser → Asp) and loss-of-function (Ser → Ala) mutants. The Ser → Asp mutant had enhanced protein turnover compared with the Ser → Ala mutant and wild type tomosyn-2. Additionally, the Ser → Asp tomosyn-2 mutant was ineffective at inhibiting insulin secretion. Using a proteomic screen for tomosyn-2-binding proteins, we identified Hrd-1, an E3-ubiquitin ligase. We showed that tomosyn-2 ubiquitination is increased by Hrd-1, and knockdown of Hrd-1 by short hairpin RNA resulted in increased abundance in tomosyn-2 protein levels. Taken together, our results reveal a mechanism by which enhanced phosphorylation of a negative regulator of secretion, tomosyn-2, in response to insulin secretagogues targets it to degradation by the Hrd-1 E3-ubiquitin ligase.


Insulin-Secreting Cells/metabolism , Insulin/metabolism , R-SNARE Proteins/metabolism , Serine/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Binding Sites/genetics , Cell Line, Tumor , Cells, Cultured , Glucose/pharmacology , HEK293 Cells , Humans , Immunoblotting , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Models, Molecular , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Structure, Tertiary , Proteolysis/drug effects , R-SNARE Proteins/chemistry , R-SNARE Proteins/genetics , RNA Interference , Serine/chemistry , Serine/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
18.
Diabetes ; 63(11): 3805-14, 2014 Nov.
Article En | MEDLINE | ID: mdl-24969106

We previously demonstrated that micro-RNAs (miRNAs) 132 and 212 are differentially upregulated in response to obesity in two mouse strains that differ in their susceptibility to obesity-induced diabetes. Here we show the overexpression of miRNAs 132 and 212 enhances insulin secretion (IS) in response to glucose and other secretagogues including nonfuel stimuli. We determined that carnitine acyl-carnitine translocase (CACT; Slc25a20) is a direct target of these miRNAs. CACT is responsible for transporting long-chain acyl-carnitines into the mitochondria for ß-oxidation. Small interfering RNA-mediated knockdown of CACT in ß-cells led to the accumulation of fatty acyl-carnitines and enhanced IS. The addition of long-chain fatty acyl-carnitines promoted IS from rat insulinoma ß-cells (INS-1) as well as primary mouse islets. The effect on INS-1 cells was augmented in response to suppression of CACT. A nonhydrolyzable ether analog of palmitoyl-carnitine stimulated IS, showing that ß-oxidation of palmitoyl-carnitine is not required for its stimulation of IS. These studies establish a link between miRNA-dependent regulation of CACT and fatty acyl-carnitine-mediated regulation of IS.


Carnitine Acyltransferases/metabolism , Glucose/pharmacology , Insulin/metabolism , MicroRNAs/genetics , Animals , Carnitine Acyltransferases/genetics , Cell Line , Down-Regulation/drug effects , Insulin Secretion , Mice , Rats
19.
PLoS Genet ; 8(12): e1003107, 2012.
Article En | MEDLINE | ID: mdl-23236292

Complex diseases result from molecular changes induced by multiple genetic factors and the environment. To derive a systems view of how genetic loci interact in the context of tissue-specific molecular networks, we constructed an F2 intercross comprised of >500 mice from diabetes-resistant (B6) and diabetes-susceptible (BTBR) mouse strains made genetically obese by the Leptin(ob/ob) mutation (Lep(ob)). High-density genotypes, diabetes-related clinical traits, and whole-transcriptome expression profiling in five tissues (white adipose, liver, pancreatic islets, hypothalamus, and gastrocnemius muscle) were determined for all mice. We performed an integrative analysis to investigate the inter-relationship among genetic factors, expression traits, and plasma insulin, a hallmark diabetes trait. Among five tissues under study, there are extensive protein-protein interactions between genes responding to different loci in adipose and pancreatic islets that potentially jointly participated in the regulation of plasma insulin. We developed a novel ranking scheme based on cross-loci protein-protein network topology and gene expression to assess each gene's potential to regulate plasma insulin. Unique candidate genes were identified in adipose tissue and islets. In islets, the Alzheimer's gene App was identified as a top candidate regulator. Islets from 17-week-old, but not 10-week-old, App knockout mice showed increased insulin secretion in response to glucose or a membrane-permeant cAMP analog, in agreement with the predictions of the network model. Our result provides a novel hypothesis on the mechanism for the connection between two aging-related diseases: Alzheimer's disease and type 2 diabetes.


Alzheimer Disease , Amyloid Precursor Protein Secretases , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulin , Adipose Tissue/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Glucose/metabolism , Humans , Insulin/blood , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Leptin/genetics , Mice , Mice, Knockout , Mice, Obese/genetics , Protein Interaction Maps
20.
J Lipid Res ; 53(8): 1493-501, 2012 Aug.
Article En | MEDLINE | ID: mdl-22628617

Nonalchoholic fatty liver disease (NAFLD) is the most common cause of liver dysfunction and is associated with metabolic diseases, including obesity, insulin resistance, and type 2 diabetes. We mapped a quantitative trait locus (QTL) for NAFLD to chromosome 17 in a cross between C57BL/6 (B6) and BTBR mouse strains made genetically obese with the Lep(ob/ob) mutation. We identified Tsc2 as a gene underlying the chromosome 17 NAFLD QTL. Tsc2 functions as an inhibitor of mammalian target of rapamycin, which is involved in many physiological processes, including cell growth, proliferation, and metabolism. We found that Tsc2(+/-) mice have increased lipogenic gene expression in the liver in an insulin-dependent manner. The coding single nucleotide polymorphism between the B6 and BTBR strains leads to a change in the ability to inhibit the expression of lipogenic genes and de novo lipogenesis in AML12 cells and to promote the proliferation of Ins1 cells. This difference is due to a different affinity of binding to Tsc1, which affects the stability of Tsc2.


Fatty Liver/genetics , Quantitative Trait Loci/genetics , Tumor Suppressor Proteins/genetics , Alleles , Animals , Cell Proliferation , Chromosomes, Mammalian/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation , Insulin-Secreting Cells/pathology , Lipogenesis/genetics , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease , Species Specificity , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism
...