55 results on '"Rabaglia ME"'
Search Results
2. Linoleic acid absorption in children with cystic fibrosis
- Author
-
Welch Nn, Rabaglia Me, Dupont J, and Chase Hp
- Subjects
Male ,Time Factors ,Cystic Fibrosis ,Linoleic acid ,medicine.medical_treatment ,Cystic fibrosis ,chemistry.chemical_compound ,Antacid ,medicine ,Ingestion ,Humans ,Food science ,Cimetidine ,Child ,Safflower Oil ,chemistry.chemical_classification ,Triglyceride ,Dose-Response Relationship, Drug ,business.industry ,Gastroenterology ,Fatty acid ,Monoglyceride ,medicine.disease ,Monoacylglycerol Lipases ,Enzymes ,chemistry ,Linoleic Acids ,Pediatrics, Perinatology and Child Health ,Female ,Antacids ,business ,medicine.drug - Abstract
When safflower oil (triglyceride) was consumed without pancreatic enzymes by children with cystic fibrosis (CF), there was no rise in mean plasma linoleic acid levels over the next 4 h. When linoleic acid monoglyceride (LAM) was consumed, the increase in plasma linoleic acid levels was significantly greater than for safflower oil at 2 (p less than 0.02), 3 (p less than 0.01), and 4 h (p less than 0.01). When free fatty acid (hydrolyzed safflower oil) was ingested, there was almost no increase in plasma linoleic acid levels in CF or control children. The absorption of linoleic acid from triglyceride, but not from LAM, was greater when the CF children also took pancreatic enzymes. Three children with CF had greater increases in plasma linoleic acid levels following ingestion of safflower oil when they took antacid and cimetidine with their pancreatic capsules, compared to when they only took the pancreatic capsules.
- Published
- 1982
3. Author Correction: Genetic mapping of microbial and host traits reveals production of immunomodulatory lipids by Akkermansia muciniphila in the murine gut.
- Author
-
Zhang Q, Linke V, Overmyer KA, Traeger LL, Kasahara K, Miller IJ, Manson DE, Polaske TJ, Kerby RL, Kemis JH, Trujillo EA, Reddy TR, Russell JD, Schueler KL, Stapleton DS, Rabaglia ME, Seldin M, Gatti DM, Keele GR, Pham DT, Gerdt JP, Vivas EI, Lusis AJ, Keller MP, Churchill GA, Blackwell HE, Broman KW, Attie AD, Coon JJ, and Rey FE
- Published
- 2023
- Full Text
- View/download PDF
4. Genetic mapping of microbial and host traits reveals production of immunomodulatory lipids by Akkermansia muciniphila in the murine gut.
- Author
-
Zhang Q, Linke V, Overmyer KA, Traeger LL, Kasahara K, Miller IJ, Manson DE, Polaske TJ, Kerby RL, Kemis JH, Trujillo EA, Reddy TR, Russell JD, Schueler KL, Stapleton DS, Rabaglia ME, Seldin M, Gatti DM, Keele GR, Pham DT, Gerdt JP, Vivas EI, Lusis AJ, Keller MP, Churchill GA, Blackwell HE, Broman KW, Attie AD, Coon JJ, and Rey FE
- Subjects
- Mice, Animals, Akkermansia genetics, Phenotype, Verrucomicrobia genetics, Gastrointestinal Microbiome genetics
- Abstract
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., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
5. β Cell-specific deletion of Zfp148 improves nutrient-stimulated β cell Ca2+ responses.
- Author
-
Emfinger CH, de Klerk E, Schueler KL, Rabaglia ME, Stapleton DS, Simonett SP, Mitok KA, Wang Z, Liu X, Paulo JA, Yu Q, Cardone RL, Foster HR, Lewandowski SL, Perales JC, Kendziorski CM, Gygi SP, Kibbey RG, Keller MP, Hebrok M, Merrins MJ, and Attie AD
- Subjects
- Animals, Calcium metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Glucose metabolism, Glutamine metabolism, Mice, Nutrients, Transcription Factors metabolism, Insulin-Secreting Cells metabolism, Islets of Langerhans metabolism
- Abstract
Insulin secretion from pancreatic β cells is essential for glucose homeostasis. An insufficient response to the demand for insulin results in diabetes. We previously showed that β cell-specific deletion of Zfp148 (β-Zfp148KO) improves glucose tolerance and insulin secretion in mice. Here, we performed Ca2+ imaging of islets from β‑Zfp148KO and control mice fed both a chow and a Western-style diet. β-Zfp148KO islets demonstrated improved sensitivity and sustained Ca2+ oscillations in response to elevated glucose levels. β-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 β-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, β-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 β cell function that are robust to the metabolic challenge imposed by a Western diet.
- Published
- 2022
- Full Text
- View/download PDF
6. Identification of direct transcriptional targets of NFATC2 that promote β cell proliferation.
- Author
-
Simonett SP, Shin S, Herring JA, Bacher R, Smith LA, Dong C, Rabaglia ME, Stapleton DS, Schueler KL, Choi J, Bernstein MN, Turkewitz DR, Perez-Cervantes C, Spaeth J, Stein R, Tessem JS, Kendziorski C, Keleş S, Moskowitz IP, Keller MP, and Attie AD
- Subjects
- Animals, Humans, Mice, Knockout, NFATC Transcription Factors genetics, Mice, Cell Proliferation, Gene Expression Regulation, Insulin-Secreting Cells metabolism, NFATC Transcription Factors metabolism, Response Elements, Transcription, Genetic
- Abstract
The transcription factor NFATC2 induces β 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 β cell proliferation, suggesting the FOXP family works to regulate β cell proliferation in concert with NFATC2. NFATC2 induced β 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 β 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 β cell proliferation.
- Published
- 2021
- Full Text
- View/download PDF
7. A large-scale genome-lipid association map guides lipid identification.
- Author
-
Linke V, Overmyer KA, Miller IJ, Brademan DR, Hutchins PD, Trujillo EA, Reddy TR, Russell JD, Cushing EM, Schueler KL, Stapleton DS, Rabaglia ME, Keller MP, Gatti DM, Keele GR, Pham D, Broman KW, Churchill GA, Attie AD, and Coon JJ
- Subjects
- Animals, Gangliosides metabolism, Genome-Wide Association Study, Genotype, Humans, Hydrolases genetics, Mice, Mice, Inbred C57BL, Phosphatidylcholines metabolism, Phospholipases A2 genetics, Plasmids genetics, Sex Characteristics, Chromosome Mapping, Genome, Lipid Metabolism genetics, Lipidomics, Lipids chemistry, Lipids genetics
- Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
8. Genetic determinants of gut microbiota composition and bile acid profiles in mice.
- Author
-
Kemis JH, Linke V, Barrett KL, Boehm FJ, Traeger LL, Keller MP, Rabaglia ME, Schueler KL, Stapleton DS, Gatti DM, Churchill GA, Amador-Noguez D, Russell JD, Yandell BS, Broman KW, Coon JJ, Attie AD, and Rey FE
- Subjects
- 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, Bile Acids and Salts metabolism, Biological Variation, Population genetics, Gastrointestinal Microbiome physiology, Organic Anion Transporters, Sodium-Dependent genetics, Quantitative Trait Loci genetics, Symporters genetics
- Abstract
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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
9. Gene loci associated with insulin secretion in islets from non-diabetic mice.
- Author
-
Keller MP, Rabaglia ME, Schueler KL, Stapleton DS, Gatti DM, Vincent M, Mitok KA, Wang Z, Ishimura T, Simonett SP, Emfinger CH, Das R, Beck T, Kendziorski C, Broman KW, Yandell BS, Churchill GA, and Attie AD
- Subjects
- Animals, Diabetes Mellitus, Type 2 genetics, Genetic Predisposition to Disease, Humans, Mice, Mice, Transgenic, DNA-Binding Proteins genetics, Genetic Loci, Insulin Secretion genetics, Protein Serine-Threonine Kinases genetics, Protein Tyrosine Phosphatases, Non-Receptor genetics, Transcription Factors genetics
- Abstract
Genetic susceptibility to type 2 diabetes is primarily due to β-cell dysfunction. However, a genetic study to directly interrogate β-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.
- Published
- 2019
- Full Text
- View/download PDF
10. Genetic Drivers of Pancreatic Islet Function.
- Author
-
Keller MP, Gatti DM, Schueler KL, Rabaglia ME, Stapleton DS, Simecek P, Vincent M, Allen S, Broman AT, Bacher R, Kendziorski C, Broman KW, Yandell BS, Churchill GA, and Attie AD
- Subjects
- 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, Genetic Association Studies, Islets of Langerhans physiology, Quantitative Trait Loci, Quantitative Trait, Heritable
- Abstract
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 β-cell-specific transcripts, suggesting heterogeneity of gene expression patterns within the β-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., (Copyright © 2018 by the Genetics Society of America.)
- Published
- 2018
- Full Text
- View/download PDF
11. Islet proteomics reveals genetic variation in dopamine production resulting in altered insulin secretion.
- Author
-
Mitok KA, Freiberger EC, Schueler KL, Rabaglia ME, Stapleton DS, Kwiecien NW, Malec PA, Hebert AS, Broman AT, Kennedy RT, Keller MP, Coon JJ, and Attie AD
- Subjects
- Animals, Diabetes Mellitus, Type 2 genetics, Dopamine genetics, Female, Genetic Variation, Glucagon metabolism, Male, Mice, Inbred C57BL, Mice, Inbred NOD, Phenotype, Proteome genetics, Proteomics, Diabetes Mellitus, Type 2 metabolism, Dopamine metabolism, Insulin Secretion, Islets of Langerhans metabolism, Proteome metabolism
- Abstract
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 β-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 β-cells inhibits insulin secretion., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
12. Host Genotype and Gut Microbiome Modulate Insulin Secretion and Diet-Induced Metabolic Phenotypes.
- Author
-
Kreznar JH, Keller MP, Traeger LL, Rabaglia ME, Schueler KL, Stapleton DS, Zhao W, Vivas EI, Yandell BS, Broman AT, Hagenbuch B, Attie AD, and Rey FE
- Subjects
- Animals, Bile Acids and Salts metabolism, Diet, High-Fat adverse effects, Genetic Variation genetics, Genotype, Insulin Resistance physiology, Male, Mice, Phenotype, Diabetes Mellitus genetics, Diabetes Mellitus microbiology, Gastrointestinal Microbiome physiology, Gastrointestinal Tract microbiology, Insulin metabolism, Microbiota physiology
- Abstract
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., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
13. The Transcription Factor Nfatc2 Regulates β-Cell Proliferation and Genes Associated with Type 2 Diabetes in Mouse and Human Islets.
- Author
-
Keller MP, Paul PK, Rabaglia ME, Stapleton DS, Schueler KL, Broman AT, Ye SI, Leng N, Brandon CJ, Neto EC, Plaisier CL, Simonett SP, Kebede MA, Sheynkman GM, Klein MA, Baliga NS, Smith LM, Broman KW, Yandell BS, Kendziorski C, and Attie AD
- Subjects
- 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, Diabetes Mellitus, Type 2 genetics, Insulin genetics, NFATC Transcription Factors genetics
- Abstract
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 β-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 β-cell proliferation, suggesting distinct molecular pathways mediating insulin secretion vs. β-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 β-cell proliferation in both mouse and human islets., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
14. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues.
- Author
-
Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett-Wilt GA, Rabaglia ME, Keller MP, Attie AD, Rey FE, and Denu JM
- Subjects
- 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, Diet, Western, Epigenesis, Genetic, Fatty Acids, Volatile metabolism, Gastrointestinal Microbiome physiology
- Abstract
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., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
15. Histone chaperone ASF1B promotes human β-cell proliferation via recruitment of histone H3.3.
- Author
-
Paul PK, Rabaglia ME, Wang CY, Stapleton DS, Leng N, Kendziorski C, Lewis PW, Keller MP, and Attie AD
- Subjects
- Animals, Apoptosis genetics, Cell Line, Cell Proliferation, DNA Damage genetics, Gene Expression Regulation, Humans, Mice, Inbred C57BL, Mitosis genetics, Protein Binding genetics, Transcription, Genetic, Cell Cycle Proteins metabolism, Histone Chaperones metabolism, Histones metabolism, Insulin-Secreting Cells cytology, Insulin-Secreting Cells metabolism
- Abstract
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 β-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 β-cell proliferation. Co-expression of histone H3.3 further augmented β-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 β-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 β-cell replication and show that ASF1B and histone H3.3A synergistically stimulate human β-cell proliferation.
- Published
- 2016
- Full Text
- View/download PDF
16. NeuCode Proteomics Reveals Bap1 Regulation of Metabolism.
- Author
-
Baughman JM, Rose CM, Kolumam G, Webster JD, Wilkerson EM, Merrill AE, Rhoads TW, Noubade R, Katavolos P, Lesch J, Stapleton DS, Rabaglia ME, Schueler KL, Asuncion R, Domeyer M, Zavala-Solorio J, Reich M, DeVoss J, Keller MP, Attie AD, Hebert AS, Westphall MS, Coon JJ, Kirkpatrick DS, and Dey A
- Subjects
- 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, Proteomics methods, Tumor Suppressor Proteins physiology, Ubiquitin Thiolesterase physiology
- Abstract
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., (Copyright © 2016. Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
17. Identification of the Bile Acid Transporter Slco1a6 as a Candidate Gene That Broadly Affects Gene Expression in Mouse Pancreatic Islets.
- Author
-
Tian J, Keller MP, Oler AT, Rabaglia ME, Schueler KL, Stapleton DS, Broman AT, Zhao W, Kendziorski C, Yandell BS, Hagenbuch B, Broman KW, and Attie AD
- Subjects
- Amino Acid Substitution, Animals, Carrier Proteins, Gene Expression Regulation, HEK293 Cells, Humans, Membrane Glycoproteins, Mice, Mice, Inbred C57BL, Quantitative Trait Loci, Taurocholic Acid metabolism, Islets of Langerhans metabolism, Organic Anion Transporters genetics
- Abstract
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., (Copyright © 2015 by the Genetics Society of America.)
- Published
- 2015
- Full Text
- View/download PDF
18. Downregulation of carnitine acyl-carnitine translocase by miRNAs 132 and 212 amplifies glucose-stimulated insulin secretion.
- Author
-
Soni MS, Rabaglia ME, Bhatnagar S, Shang J, Ilkayeva O, Mynatt R, Zhou YP, Schadt EE, Thornberry NA, Muoio DM, Keller MP, and Attie AD
- Subjects
- Animals, Carnitine Acyltransferases genetics, Cell Line, Down-Regulation drug effects, Insulin Secretion, Mice, Rats, Carnitine Acyltransferases metabolism, Glucose pharmacology, Insulin metabolism, MicroRNAs genetics
- Abstract
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 β-oxidation. Small interfering RNA-mediated knockdown of CACT in β-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 β-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 β-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., (© 2014 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.)
- Published
- 2014
- Full Text
- View/download PDF
19. A potent α/β-peptide analogue of GLP-1 with prolonged action in vivo.
- Author
-
Johnson LM, Barrick S, Hager MV, McFedries A, Homan EA, Rabaglia ME, Keller MP, Attie AD, Saghatelian A, Bisello A, and Gellman SH
- Subjects
- 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, Glucagon-Like Peptide 1 analogs & derivatives, Glucagon-Like Peptide 1 pharmacology, Receptors, Glucagon agonists
- Abstract
Glucagon-like peptide-1 (GLP-1) is a natural agonist for GLP-1R, a G protein-coupled receptor (GPCR) on the surface of pancreatic β 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 β-amino acid residues. This backbone-modification approach may be useful for developing stabilized analogues of other peptide hormones.
- Published
- 2014
- Full Text
- View/download PDF
20. Phosphorylation and degradation of tomosyn-2 de-represses insulin secretion.
- Author
-
Bhatnagar S, Soni MS, Wrighton LS, Hebert AS, Zhou AS, Paul PK, Gregg T, Rabaglia ME, Keller MP, Coon JJ, and Attie AD
- Subjects
- 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, Insulin metabolism, Insulin-Secreting Cells metabolism, R-SNARE Proteins metabolism, Serine metabolism
- Abstract
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 β-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., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
21. Tsc2, a positional candidate gene underlying a quantitative trait locus for hepatic steatosis.
- Author
-
Wang CY, Stapleton DS, Schueler KL, Rabaglia ME, Oler AT, Keller MP, Kendziorski CM, Broman KW, Yandell BS, Schadt EE, and Attie AD
- Subjects
- 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, Fatty Liver genetics, Quantitative Trait Loci genetics, Tumor Suppressor Proteins genetics
- Abstract
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.
- Published
- 2012
- Full Text
- View/download PDF
22. Integrative analysis of a cross-loci regulation network identifies App as a gene regulating insulin secretion from pancreatic islets.
- Author
-
Tu Z, Keller MP, Zhang C, Rabaglia ME, Greenawalt DM, Yang X, Wang IM, Dai H, Bruss MD, Lum PY, Zhou YP, Kemp DM, Kendziorski C, Yandell BS, Attie AD, Schadt EE, and Zhu J
- Subjects
- Adipose Tissue metabolism, Animals, Gene Expression Profiling, Gene Regulatory Networks, Glucose metabolism, Humans, Insulin Secretion, Islets of Langerhans metabolism, Leptin genetics, Mice, Mice, Knockout, Mice, Obese genetics, Protein Interaction Maps, Alzheimer Disease genetics, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases deficiency, Amyloid Precursor Protein Secretases genetics, Amyloid Precursor Protein Secretases metabolism, Diabetes Mellitus, Experimental genetics, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Type 2 blood, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 metabolism, Insulin blood, Insulin genetics, Insulin metabolism
- Abstract
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., Competing Interests: CZ, DMG, I-MW, HD, Y-PZ, and DMK work for Merck.
- Published
- 2012
- Full Text
- View/download PDF
23. Positional cloning of a type 2 diabetes quantitative trait locus; tomosyn-2, a negative regulator of insulin secretion.
- Author
-
Bhatnagar S, Oler AT, Rabaglia ME, Stapleton DS, Schueler KL, Truchan NA, Worzella SL, Stoehr JP, Clee SM, Yandell BS, Keller MP, Thurmond DC, and Attie AD
- Subjects
- 8-Bromo Cyclic Adenosine Monophosphate pharmacology, Adaptor Proteins, Vesicular Transport, Animals, Chromosome Mapping, Cloning, Molecular, Disease Models, Animal, Genetic Predisposition to Disease, Glucose analysis, HEK293 Cells, Humans, Hypoglycemia genetics, Insulin Secretion, Islets of Langerhans metabolism, Islets of Langerhans pathology, Leptin genetics, Leptin metabolism, Mice, Mice, Inbred C57BL, Polymorphism, Single Nucleotide, Qa-SNARE Proteins genetics, Qa-SNARE Proteins metabolism, Quantitative Trait Loci genetics, Rats, SNARE Proteins metabolism, Syntaxin 1 genetics, Syntaxin 1 metabolism, Diabetes Mellitus, Type 2 genetics, Insulin metabolism, R-SNARE Proteins genetics, R-SNARE Proteins metabolism
- Abstract
We previously mapped a type 2 diabetes (T2D) locus on chromosome 16 (Chr 16) in an F2 intercross from the BTBR T (+) tf (BTBR) Lep(ob/ob) and C57BL/6 (B6) Lep(ob/ob) mouse strains. Introgression of BTBR Chr 16 into B6 mice resulted in a consomic mouse with reduced fasting plasma insulin and elevated glucose levels. We derived a panel of sub-congenic mice and narrowed the diabetes susceptibility locus to a 1.6 Mb region. Introgression of this 1.6 Mb fragment of the BTBR Chr 16 into lean B6 mice (B6.16(BT36-38)) replicated the phenotypes of the consomic mice. Pancreatic islets from the B6.16(BT36-38) mice were defective in the second phase of the insulin secretion, suggesting that the 1.6 Mb region encodes a regulator of insulin secretion. Within this region, syntaxin-binding protein 5-like (Stxbp5l) or tomosyn-2 was the only gene with an expression difference and a non-synonymous coding single nucleotide polymorphism (SNP) between the B6 and BTBR alleles. Overexpression of the b-tomosyn-2 isoform in the pancreatic β-cell line, INS1 (832/13), resulted in an inhibition of insulin secretion in response to 3 mM 8-bromo cAMP at 7 mM glucose. In vitro binding experiments showed that tomosyn-2 binds recombinant syntaxin-1A and syntaxin-4, key proteins that are involved in insulin secretion via formation of the SNARE complex. The B6 form of tomosyn-2 is more susceptible to proteasomal degradation than the BTBR form, establishing a functional role for the coding SNP in tomosyn-2. We conclude that tomosyn-2 is the major gene responsible for the T2D Chr 16 quantitative trait locus (QTL) we mapped in our mouse cross. Our findings suggest that tomosyn-2 is a key negative regulator of insulin secretion., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2011
- Full Text
- View/download PDF
24. Loss of PDGF-B activity increases hepatic vascular permeability and enhances insulin sensitivity.
- Author
-
Raines SM, Richards OC, Schneider LR, Schueler KL, Rabaglia ME, Oler AT, Stapleton DS, Genové G, Dawson JA, Betsholtz C, and Attie AD
- Subjects
- Animals, Blood Glucose metabolism, Glucose Tolerance Test, Insulin Resistance, Insulin Secretion, Leptin genetics, Leptin metabolism, Liver blood supply, Mice, Mice, Transgenic, Obesity genetics, Obesity metabolism, Proto-Oncogene Proteins c-sis genetics, Receptor, Platelet-Derived Growth Factor beta metabolism, Signal Transduction, Capillary Permeability physiology, Insulin metabolism, Liver metabolism, Pericytes metabolism, Proto-Oncogene Proteins c-sis metabolism
- Abstract
Hepatic vasculature is not thought to pose a permeability barrier for diffusion of macromolecules from the bloodstream to hepatocytes. In contrast, in extrahepatic tissues, the microvasculature is critically important for insulin action, because transport of insulin across the endothelial cell layer is rate limiting for insulin-stimulated glucose disposal. However, very little is known concerning the role in this process of pericytes, the mural cells lining the basolateral membrane of endothelial cells. PDGF-B is a growth factor involved in the recruitment and function of pericytes. We studied insulin action in mice expressing PDGF-B lacking the proteoglycan binding domain, producing a protein with a partial loss of function (PDGF-B(ret/ret)). Insulin action was assessed through measurements of insulin signaling and insulin and glucose tolerance tests. PDGF-B deficiency enhanced hepatic vascular transendothelial transport. One outcome of this change was an increase in hepatic insulin signaling. This correlated with enhanced whole body glucose homeostasis and increased insulin clearance from the circulation during an insulin tolerance test. In obese mice, PDGF-B deficiency was associated with an 80% reduction in fasting insulin and drastically reduced insulin secretion. These mice did not have significantly higher glucose levels, reflecting a dramatic increase in insulin action. Our findings show that, despite already having a high permeability, hepatic transendothelial transport can be further enhanced. To the best of our knowledge, this is the first study to connect PDGF-B-induced changes in hepatic sinusoidal transport to changes in insulin action, demonstrating a link between PDGF-B signaling and insulin sensitivity.
- Published
- 2011
- Full Text
- View/download PDF
25. BTBR Ob/Ob mutant mice model progressive diabetic nephropathy.
- Author
-
Hudkins KL, Pichaiwong W, Wietecha T, Kowalewska J, Banas MC, Spencer MW, Mühlfeld A, Koelling M, Pippin JW, Shankland SJ, Askari B, Rabaglia ME, Keller MP, Attie AD, and Alpers CE
- Subjects
- Animals, Disease Models, Animal, Disease Progression, Female, Fibrosis, Galectin 3 analysis, Insulin Resistance, Kidney pathology, Male, Mice, Mice, Inbred C57BL, Mice, Obese, Podocytes pathology, Diabetic Nephropathies etiology
- Abstract
There remains a need for robust mouse models of diabetic nephropathy (DN) that mimic key features of advanced human DN. The recently developed mouse strain BTBR with the ob/ob leptin-deficiency mutation develops severe type 2 diabetes, hypercholesterolemia, elevated triglycerides, and insulin resistance, but the renal phenotype has not been characterized. Here, we show that these obese, diabetic mice rapidly develop morphologic renal lesions characteristic of both early and advanced human DN. BTBR ob/ob mice developed progressive proteinuria beginning at 4 weeks. Glomerular hypertrophy and accumulation of mesangial matrix, characteristic of early DN, were present by 8 weeks, and glomerular lesions similar to those of advanced human DN were present by 20 weeks. By 22 weeks, we observed an approximately 20% increase in basement membrane thickness and a >50% increase in mesangial matrix. Diffuse mesangial sclerosis (focally approaching nodular glomerulosclerosis), focal arteriolar hyalinosis, mesangiolysis, and focal mild interstitial fibrosis were present. Loss of podocytes was present early and persisted. In summary, BTBR ob/ob mice develop a constellation of abnormalities that closely resemble advanced human DN more rapidly than most other murine models, making this strain particularly attractive for testing therapeutic interventions.
- Published
- 2010
- Full Text
- View/download PDF
26. FoxM1 is up-regulated by obesity and stimulates beta-cell proliferation.
- Author
-
Davis DB, Lavine JA, Suhonen JI, Krautkramer KA, Rabaglia ME, Sperger JM, Fernandez LA, Yandell BS, Keller MP, Wang IM, Schadt EE, and Attie AD
- Subjects
- Adult, Animals, Cell Cycle, Cell Proliferation, Cell Separation, Female, Forkhead Box Protein M1, Forkhead Transcription Factors metabolism, Humans, Insulin metabolism, Insulin Secretion, Male, Mice, Middle Aged, Tissue Donors, Forkhead Transcription Factors genetics, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells pathology, Obesity genetics, Obesity pathology, Up-Regulation genetics
- Abstract
beta-Cell mass expansion is one mechanism by which obese animals compensate for insulin resistance and prevent diabetes. FoxM1 is a transcription factor that can regulate the expression of multiple cell cycle genes and is necessary for the maintenance of adult beta-cell mass, beta-cell proliferation, and glucose homeostasis. We hypothesized that FoxM1 is up-regulated by nondiabetic obesity and initiates a transcriptional program leading to beta-cell proliferation. We performed gene expression analysis on islets from the nondiabetic C57BL/6 Leptin(ob/ob) mouse, the diabetic BTBR Leptin(ob/ob) mouse, and an F2 Leptin(ob/ob) population derived from these strains. We identified obesity-driven coordinated up-regulation of islet Foxm1 and its target genes in the nondiabetic strain, correlating with beta-cell mass expansion and proliferation. This up-regulation was absent in the diabetic strain. In the F2 Leptin(ob/ob) population, increased expression of Foxm1 and its target genes segregated with higher insulin and lower glucose levels. We next studied the effects of FOXM1b overexpression on isolated mouse and human islets. We found that FoxM1 stimulated mouse and human beta-cell proliferation by activating many cell cycle phases. We asked whether FOXM1 expression is also responsive to obesity in human islets by collecting RNA from human islet donors (body mass index range: 24-51). We found that the expression of FOXM1 and its target genes is positively correlated with body mass index. Our data suggest that beta-cell proliferation occurs in adult obese humans in an attempt to expand beta-cell mass to compensate for insulin resistance, and that the FoxM1 transcriptional program plays a key role in this process.
- Published
- 2010
- Full Text
- View/download PDF
27. Cholecystokinin is up-regulated in obese mouse islets and expands beta-cell mass by increasing beta-cell survival.
- Author
-
Lavine JA, Raess PW, Stapleton DS, Rabaglia ME, Suhonen JI, Schueler KL, Koltes JE, Dawson JA, Yandell BS, Samuelson LC, Beinfeld MC, Davis DB, Hellerstein MK, Keller MP, and Attie AD
- Subjects
- Animals, Cell Count, Cell Survival genetics, Cells, Cultured, Cholecystokinin metabolism, Diabetes Mellitus etiology, Diabetes Mellitus genetics, Insulin Resistance genetics, Insulin-Secreting Cells metabolism, Islets of Langerhans pathology, Male, Mice, Mice, Inbred C57BL, Mice, Obese, Mice, Transgenic, Obesity complications, Obesity metabolism, Organ Size genetics, Up-Regulation, Cholecystokinin genetics, Insulin-Secreting Cells pathology, Islets of Langerhans metabolism, Obesity genetics, Obesity pathology
- Abstract
An absolute or functional deficit in beta-cell mass is a key factor in the pathogenesis of diabetes. We model obesity-driven beta-cell mass expansion by studying the diabetes-resistant C57BL/6-Leptin(ob/ob) mouse. We previously reported that cholecystokinin (Cck) was the most up-regulated gene in obese pancreatic islets. We now show that islet cholecystokinin (CCK) is up-regulated 500-fold by obesity and expressed in both alpha- and beta-cells. We bred a null Cck allele into the C57BL/6-Leptin(ob/ob) background and investigated beta-cell mass and metabolic parameters of Cck-deficient obese mice. Loss of CCK resulted in decreased islet size and reduced beta-cell mass through increased beta-cell death. CCK deficiency and decreased beta-cell mass exacerbated fasting hyperglycemia and reduced hyperinsulinemia. We further investigated whether CCK can directly affect beta-cell death in cell culture and isolated islets. CCK was able to directly reduce cytokine- and endoplasmic reticulum stress-induced cell death. In summary, CCK is up-regulated by islet cells during obesity and functions as a paracrine or autocrine factor to increase beta-cell survival and expand beta-cell mass to compensate for obesity-induced insulin resistance.
- Published
- 2010
- Full Text
- View/download PDF
28. Contamination with E1A-positive wild-type adenovirus accounts for species-specific stimulation of islet cell proliferation by CCK: a cautionary note.
- Author
-
Lavine JA, Raess PW, Davis DB, Rabaglia ME, Presley BK, Keller MP, Beinfeld MC, Kopin AS, Newgard CB, and Attie AD
- Subjects
- Adenoviridae chemistry, Adenoviridae isolation & purification, Adenoviridae metabolism, Adenovirus E1A Proteins chemistry, Adenovirus E1A Proteins genetics, Animals, Base Sequence, Cholecystokinin genetics, Genetic Vectors, Humans, Islets of Langerhans metabolism, Mice, Molecular Sequence Data, Protein Precursors genetics, RNA, Messenger chemistry, RNA, Messenger metabolism, Rats, Reverse Transcriptase Polymerase Chain Reaction, Sequence Alignment, Species Specificity, Adenoviridae genetics, Adenovirus E1A Proteins metabolism, Cell Proliferation, Cholecystokinin metabolism, Islets of Langerhans cytology, Protein Precursors metabolism, Transfection
- Abstract
We have previously reported that adenovirus-mediated expression of preprocholecystokin (CCK) stimulates human and mouse islet cell proliferation. In follow-up studies, we became concerned that the CCK adenovirus might have been contaminated with a wild-type E1A-containing adenovirus. Here we show conclusively that the proliferative effects reported in the original paper in mouse and human islets were not due to CCK expression but rather to a contaminating E1A-expressing wild-type adenovirus. We also show, however, that CCK expression does have a proliferative effect in rat islets. We hope that our report of the steps taken to detect the wild-type virus contamination, and purification of the contributing viral stocks, will be helpful to other investigators, and that our experience will serve as a cautionary tale for use of adenovirus vectors, especially for studies on cellular replication.
- Published
- 2010
- Full Text
- View/download PDF
29. Obesity and genetics regulate microRNAs in islets, liver, and adipose of diabetic mice.
- Author
-
Zhao E, Keller MP, Rabaglia ME, Oler AT, Stapleton DS, Schueler KL, Neto EC, Moon JY, Wang P, Wang IM, Lum PY, Ivanovska I, Cleary M, Greenawalt D, Tsang J, Choi YJ, Kleinhanz R, Shang J, Zhou YP, Howard AD, Zhang BB, Kendziorski C, Thornberry NA, Yandell BS, Schadt EE, and Attie AD
- Subjects
- Animals, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 metabolism, Disease Models, Animal, Female, Gene Dosage, Gene Expression Profiling, Humans, Male, Mice, Mice, Obese, MicroRNAs metabolism, Obesity metabolism, Adipose Tissue metabolism, Gene Expression Regulation, Islets of Langerhans metabolism, Liver metabolism, MicroRNAs genetics, Obesity genetics
- Abstract
Type 2 diabetes results from severe insulin resistance coupled with a failure of b cells to compensate by secreting sufficient insulin. Multiple genetic loci are involved in the development of diabetes, although the effect of each gene on diabetes susceptibility is thought to be small. MicroRNAs (miRNAs) are noncoding 19-22-nucleotide RNA molecules that potentially regulate the expression of thousands of genes. To understand the relationship between miRNA regulation and obesity-induced diabetes, we quantitatively profiled approximately 220 miRNAs in pancreatic islets, adipose tissue, and liver from diabetes-resistant (B6) and diabetes-susceptible (BTBR) mice. More than half of the miRNAs profiled were expressed in all three tissues, with many miRNAs in each tissue showing significant changes in response to genetic obesity. Furthermore, several miRNAs in each tissue were differentially responsive to obesity in B6 versus BTBR mice, suggesting that they may be involved in the pathogenesis of diabetes. In liver there were approximately 40 miRNAs that were downregulated in response to obesity in B6 but not BTBR mice, indicating that genetic differences between the mouse strains play a critical role in miRNA regulation. In order to elucidate the genetic architecture of hepatic miRNA expression, we measured the expression of miRNAs in genetically obese F2 mice. Approximately 10% of the miRNAs measured showed significant linkage (miR-eQTLs), identifying loci that control miRNA abundance. Understanding the influence that obesity and genetics exert on the regulation of miRNA expression will reveal the role miRNAs play in the context of obesity-induced type 2 diabetes.
- Published
- 2009
- Full Text
- View/download PDF
30. Overexpression of pre-pro-cholecystokinin stimulates beta-cell proliferation in mouse and human islets with retention of islet function.
- Author
-
Lavine JA, Raess PW, Davis DB, Rabaglia ME, Presley BK, Keller MP, Beinfeld MC, Kopin AS, Newgard CB, and Attie AD
- Subjects
- Adenoviridae genetics, Animals, Cells, Cultured, Cholecystokinin metabolism, Cholecystokinin physiology, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Cyclins genetics, Cyclins metabolism, Cytomegalovirus genetics, Genetic Vectors, Humans, Insulin-Secreting Cells metabolism, Islets of Langerhans metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Protein Precursors metabolism, Protein Precursors physiology, RNA, Messenger metabolism, Transfection, Up-Regulation, Cell Proliferation, Cholecystokinin genetics, Insulin-Secreting Cells physiology, Islets of Langerhans physiology, Protein Precursors genetics
- Abstract
Type 1 and type 2 diabetes result from a deficit in insulin production and beta-cell mass. Methods to expand beta-cell mass are under intensive investigation for the treatment of type 1 and type 2 diabetes. We tested the hypothesis that cholecystokinin (CCK) can promote beta-cell proliferation. We treated isolated mouse and human islets with an adenovirus containing the CCK cDNA (AdCMV-CCK). We measured [(3)H]thymidine and BrdU incorporation into DNA and additionally, performed flow cytometry analysis to determine whether CCK overexpression stimulates beta-cell proliferation. We studied islet function by measuring glucose-stimulated insulin secretion and investigated the cell cycle regulation of proliferating beta-cells by quantitative RT-PCR and Western blot analysis. Overexpression of CCK stimulated [(3)H]thymidine incorporation into DNA 5.0-fold and 15.8-fold in mouse and human islets, respectively. AdCMV-CCK treatment also stimulated BrdU incorporation into DNA 10-fold and 21-fold in mouse and human beta-cells, respectively. Glucose-stimulated insulin secretion was unaffected by CCK expression. Analysis of cyclin and cdk mRNA and protein abundance revealed that CCK overexpression increased cyclin A, cyclin B, cyclin E, cdk1, and cdk2 with no change in cyclin D1, cyclin D2, cyclin D3, cdk4, or cdk6 in mouse and human islets. Additionally, AdCMV-CCK treatment of CCK receptor knockout and wild-type mice resulted in equal [(3)H]thymidine incorporation. CCK is a beta-cell proliferative factor that is effective in both mouse and human islets. CCK triggers beta-cell proliferation without disrupting islet function, up-regulates a distinct set of cell cycle regulators in islets, and signals independently of the CCK receptors.
- Published
- 2008
- Full Text
- View/download PDF
31. A gene expression network model of type 2 diabetes links cell cycle regulation in islets with diabetes susceptibility.
- Author
-
Keller MP, Choi Y, Wang P, Davis DB, Rabaglia ME, Oler AT, Stapleton DS, Argmann C, Schueler KL, Edwards S, Steinberg HA, Chaibub Neto E, Kleinhanz R, Turner S, Hellerstein MK, Schadt EE, Yandell BS, Kendziorski C, and Attie AD
- Subjects
- Adipose Tissue cytology, Aging, Animals, Cell Proliferation, Diabetes Mellitus, Type 2 metabolism, Glucose metabolism, Insulin metabolism, Insulin-Secreting Cells cytology, Insulin-Secreting Cells pathology, Male, Mice, Models, Genetic, Obesity pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Transcription, Genetic, Cell Cycle, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 pathology, Gene Expression Regulation, Genetic Predisposition to Disease, Islets of Langerhans pathology
- Abstract
Insulin resistance is necessary but not sufficient for the development of type 2 diabetes. Diabetes results when pancreatic beta-cells fail to compensate for insulin resistance by increasing insulin production through an expansion of beta-cell mass or increased insulin secretion. Communication between insulin target tissues and beta-cells may initiate this compensatory response. Correlated changes in gene expression between tissues can provide evidence for such intercellular communication. We profiled gene expression in six tissues of mice from an obesity-induced diabetes-resistant and a diabetes-susceptible strain before and after the onset of diabetes. We studied the correlation structure of mRNA abundance and identified 105 co-expression gene modules. We provide an interactive gene network model showing the correlation structure between the expression modules within and among the six tissues. This resource also provides a searchable database of gene expression profiles for all genes in six tissues in lean and obese diabetes-resistant and diabetes-susceptible mice, at 4 and 10 wk of age. A cell cycle regulatory module in islets predicts diabetes susceptibility. The module predicts islet replication; we found a strong correlation between (2)H(2)O incorporation into islet DNA in vivo and the expression pattern of the cell cycle module. This pattern is highly correlated with that of several individual genes in insulin target tissues, including Igf2, which has been shown to promote beta-cell proliferation, suggesting that these genes may provide a link between insulin resistance and beta-cell proliferation.
- Published
- 2008
- Full Text
- View/download PDF
32. Loss of stearoyl-CoA desaturase-1 improves insulin sensitivity in lean mice but worsens diabetes in leptin-deficient obese mice.
- Author
-
Flowers JB, Rabaglia ME, Schueler KL, Flowers MT, Lan H, Keller MP, Ntambi JM, and Attie AD
- Subjects
- Animals, Blood Glucose metabolism, Gene Expression Regulation, Enzymologic, Glucose Clamp Technique, Glucose Tolerance Test, Insulin blood, Insulin metabolism, Insulin Secretion, Islets of Langerhans cytology, Islets of Langerhans metabolism, Leptin genetics, Lipoprotein Lipase metabolism, Mice, Mice, Knockout, Polymerase Chain Reaction, Stearoyl-CoA Desaturase deficiency, Stearoyl-CoA Desaturase genetics, Thinness, Islets of Langerhans physiology, Leptin deficiency, Stearoyl-CoA Desaturase metabolism
- Abstract
The lipogenic gene stearoyl-CoA desaturase (SCD)1 appears to be a promising new target for obesity-related diabetes, as mice deficient in this enzyme are resistant to diet- and leptin deficiency-induced obesity. The BTBR mouse strain replicates many features of insulin resistance found in humans with excess visceral adiposity. Using the hyperinsulinemic-euglycemic clamp technique, we determined that insulin sensitivity was improved in heart, soleus muscle, adipose tissue, and liver of BTBR SCD1-deficient mice. We next determined whether SCD1 deficiency could prevent diabetes in leptin-deficient BTBR mice. Loss of SCD1 in leptin(ob/ob) mice unexpectedly accelerated the progression to severe diabetes; 6-week fasting glucose increased approximately 70%. In response to a glucose challenge, Scd1(-/-) leptin(ob/ob) mice had insufficient insulin secretion, resulting in glucose intolerance. A morphologically distinct class of islets isolated from the Scd1(-/-) leptin(ob/ob) mice had reduced insulin content and increased triglycerides, free fatty acids, esterified cholesterol, and free cholesterol and also a much higher content of saturated fatty acids. We believe the accumulation of lipid is due to an upregulation of lipoprotein lipase (20-fold) and Cd36 (167-fold) and downregulation of lipid oxidation genes in this class of islets. Therefore, although loss of Scd1 has beneficial effects on adiposity, this benefit may come at the expense of beta-cells, resulting in an increased risk of diabetes.
- Published
- 2007
- Full Text
- View/download PDF
33. Positional cloning of Sorcs1, a type 2 diabetes quantitative trait locus.
- Author
-
Clee SM, Yandell BS, Schueler KM, Rabaglia ME, Richards OC, Raines SM, Kabara EA, Klass DM, Mui ET, Stapleton DS, Gray-Keller MP, Young MB, Stoehr JP, Lan H, Boronenkov I, Raess PW, Flowers MT, and Attie AD
- Subjects
- Animals, Cloning, Molecular, Fluorescent Antibody Technique, Glucose Tolerance Test, Insulin blood, Insulin metabolism, Insulin Secretion, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Diabetes Mellitus, Type 2 genetics, Quantitative Trait Loci, Receptors, Cell Surface genetics
- Abstract
We previously mapped the type 2 diabetes mellitus-2 locus (T2dm2), which affects fasting insulin levels, to distal chromosome 19 in a leptin-deficient obese F2 intercross derived from C57BL/6 (B6) and BTBR T+ tf/J (BTBR) mice. Introgression of a 7-Mb segment of the B6 chromosome 19 into the BTBR background (strain 1339A) replicated the reduced insulin linked to T2dm2. The 1339A mice have markedly impaired insulin secretion in vivo and disrupted islet morphology. We used subcongenic strains derived from 1339A to localize the T2dm2 quantitative trait locus (QTL) to a 242-kb segment comprising the promoter, first exon and most of the first intron of the Sorcs1 gene. This was the only gene in the 1339A strain for which we detected amino acid substitutions and expression level differences between mice carrying B6 and BTBR alleles of this insert, thereby identifying variation within the Sorcs1 gene as underlying the phenotype associated with the T2dm2 locus. SorCS1 binds platelet-derived growth factor, a growth factor crucial for pericyte recruitment to the microvasculature, and may thus have a role in expanding or maintaining the islet vasculature. Our identification of the Sorcs1 gene provides insight into the pathway underlying the pathophysiology of obesity-induced type 2 diabetes mellitus.
- Published
- 2006
- Full Text
- View/download PDF
34. Alpha-Ketoisocaproate-induced hypersecretion of insulin by islets from diabetes-susceptible mice.
- Author
-
Rabaglia ME, Gray-Keller MP, Frey BL, Shortreed MR, Smith LM, and Attie AD
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Glutamate Dehydrogenase metabolism, Glutamic Acid metabolism, Hyperinsulinism metabolism, Insulin blood, Insulin Resistance physiology, Insulin Secretion, Islets of Langerhans cytology, Leptin physiology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Species Specificity, Transaminases metabolism, Diabetes Mellitus, Type 2 metabolism, Insulin metabolism, Islets of Langerhans metabolism, Keto Acids metabolism, Ketoglutaric Acids metabolism
- Abstract
Most patients at risk for developing type 2 diabetes are hyperinsulinemic. Hyperinsulinemia may be a response to insulin resistance, but another possible abnormality is insulin hypersecretion. BTBR mice are insulin resistant and hyperinsulinemic. When the leptin(ob) mutation is introgressed into BTBR mice, they develop severe diabetes. We compared the responsiveness of lean B6 and BTBR mouse islets to various insulin secretagogues. The transamination product of leucine, alpha-ketoisocaproate (KIC), elicited a dramatic insulin secretory response in BTBR islets. The KIC response was blocked by methyl-leucine or aminooxyacetate, inhibitors of branched-chain amino transferase. When dimethylglutamate was combined with KIC, the fractional insulin secretion was identical in islets from both mouse strains, predicting that the amine donor is rate-limiting for KIC-induced insulin secretion. Consistent with this prediction, glutamate levels were higher in BTBR than in B6 islets. The transamination product of glutamate, alpha-ketoglutarate, elicited insulin secretion equally from B6 and BTBR islets. Thus formation of alpha-ketoglutarate is a requisite step in the response of mouse islets to KIC. alpha-Ketoglutarate can be oxidized to succinate. However, succinate does not stimulate insulin secretion in mouse islets. Our data suggest that alpha-ketoglutarate may directly stimulate insulin secretion and that increased formation of alpha-ketoglutarate leads to hyperinsulinemia.
- Published
- 2005
- Full Text
- View/download PDF
35. Increased insulin translation from an insulin splice-variant overexpressed in diabetes, obesity, and insulin resistance.
- Author
-
Minn AH, Lan H, Rabaglia ME, Harlan DM, Peculis BA, Attie AD, and Shalev A
- Subjects
- 5' Untranslated Regions, Animals, Base Sequence, Blotting, Northern, Cell Line, Cloning, Molecular, Cytoplasm metabolism, Exons, Glucose metabolism, Humans, Insulin genetics, Introns, Islets of Langerhans metabolism, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Nucleic Acid Conformation, Oligonucleotides chemistry, RNA chemistry, RNA metabolism, Reverse Transcriptase Polymerase Chain Reaction, Subcellular Fractions, Time Factors, Transfection, Alternative Splicing, Diabetes Mellitus, Type 2 genetics, Insulin metabolism, Insulin Resistance, Obesity genetics, Protein Biosynthesis
- Abstract
Type 2 diabetes occurs when pancreatic beta-cells become unable to compensate for the underlying insulin resistance. Insulin secretion requires beta-cell insulin stores to be replenished by insulin biosynthesis, which is mainly regulated at the translational level. Such translational regulation often involves the 5'-untranslated region. Recently, we identified a human insulin splice-variant (SPV) altering only the 5'-untranslated region and conferring increased translation efficiency. We now describe a mouse SPV (mSPV) that is found in the cytoplasm and exhibits increased translation efficiency resulting in more normal (prepro)insulin protein per RNA. The RNA stability of mSPV is not increased, but the predicted secondary RNA structure is altered, which may facilitate translation. To determine the role of mSPV in insulin resistance and diabetes, mSPV expression was measured by quantitative real-time RT-PCR in islets from three diabetic and/or insulin-resistant, obese and nonobese, mouse models (BTBRob/ob, C57BL/6ob/ob, and C57BL/6azip). Interestingly, mSPV expression was significantly higher in all diabetic/insulin-resistant mice compared with wild-type littermates and was dramatically induced in primary mouse islets incubated at high glucose. This raises the possibility that the mSPV may represent a compensatory beta-cell mechanism to enhance insulin biosynthesis when insulin requirements are elevated by hyperglycemia/insulin resistance.
- Published
- 2005
- Full Text
- View/download PDF
36. Distinguishing covariation from causation in diabetes: a lesson from the protein disulfide isomerase mRNA abundance trait.
- Author
-
Lan H, Rabaglia ME, Schueler KL, Mata C, Yandell BS, and Attie AD
- Subjects
- Animals, Diabetes Mellitus enzymology, Disease Models, Animal, Female, Genetic Predisposition to Disease genetics, Male, Mice, Mice, Obese, Reproducibility of Results, Reverse Transcriptase Polymerase Chain Reaction, Diabetes Mellitus etiology, Diabetes Mellitus genetics, Protein Disulfide-Isomerases genetics, Quantitative Trait Loci genetics, RNA, Messenger genetics
- Abstract
Protein disulfide isomerase (Pdi) is reported to be an insulin-regulated gene whose expression level is increased in the livers of rats with streptozotocin-induced diabetes. We found that Pdi mRNA is approximately 20-fold more abundant in the diabetes-susceptible BTBR mouse strain relative to the diabetes-resistant C56BL/6 (B6) strain. A genetic analysis was carried out to determine whether there is a causal relationship between elevated Pdi expression and diabetes phenotype in BTBR-ob/ob mice. We mapped Pdi mRNA abundance as a quantitative trait in 108 (B6 x BTBR)F(2)-ob/ob mice segregating for diabetes. We detected a single linkage at the telomeric end of chromosome 11, where the Pdi gene itself resides (logarithm of odds score >30.0). No linkage was detected for the Pdi mRNA trait in the regions where we have previously identified quantitative trait loci for diabetes traits. Sequencing of the Pdi promoter and cDNA revealed several single nucleotide polymorphisms between these two mouse strains. We conclude that in our experimental model, elevated Pdi expression is cis regulated and is not linked to diabetes susceptibility. Genetic analysis is a powerful tool for distinguishing covariation from causation in expression array studies of disease traits.
- Published
- 2004
- Full Text
- View/download PDF
37. Gene expression profiles of nondiabetic and diabetic obese mice suggest a role of hepatic lipogenic capacity in diabetes susceptibility.
- Author
-
Lan H, Rabaglia ME, Stoehr JP, Nadler ST, Schueler KL, Zou F, Yandell BS, and Attie AD
- Subjects
- Adipose Tissue metabolism, Animals, Diabetes Mellitus, Type 2 genetics, Fatty Liver genetics, Female, Gluconeogenesis genetics, Islets of Langerhans metabolism, Liver enzymology, Mice, Mice, Inbred C57BL, Mice, Obese, Muscle, Skeletal metabolism, Oligonucleotide Array Sequence Analysis, RNA, Messenger analysis, Reverse Transcriptase Polymerase Chain Reaction, Triglycerides blood, Triglycerides metabolism, Diabetes Mellitus genetics, Gene Expression Profiling, Genetic Predisposition to Disease, Lipids biosynthesis, Liver metabolism, Obesity genetics
- Abstract
Obesity is a strong risk factor for the development of type 2 diabetes. We have previously reported that in adipose tissue of obese (ob/ob) mice, the expression of adipogenic genes is decreased. When made genetically obese, the BTBR mouse strain is diabetes susceptible and the C57BL/6J (B6) strain is diabetes resistant. We used DNA microarrays and RT-PCR to compare the gene expression in BTBR-ob/ob versus B6-ob/ob mice in adipose tissue, liver, skeletal muscle, and pancreatic islets. Our results show: 1) there is an increased expression of genes involved in inflammation in adipose tissue of diabetic mice; 2) lipogenic gene expression was lower in adipose tissue of diabetes-susceptible mice, and it continued to decrease with the development of diabetes, compared with diabetes-resistant obese mice; 3) hepatic expression of lipogenic enzymes was increased and the hepatic triglyceride content was greatly elevated in diabetes-resistant obese mice; 4) hepatic expression of gluconeogenic genes was suppressed at the prediabetic stage but not at the onset of diabetes; and 5) genes normally not expressed in skeletal muscle and pancreatic islets were expressed in these tissues in the diabetic mice. We propose that increased hepatic lipogenic capacity protects the B6-ob/ob mice from the development of type 2 diabetes.
- Published
- 2003
- Full Text
- View/download PDF
38. Normal Akt/PKB with reduced PI3K activation in insulin-resistant mice.
- Author
-
Nadler ST, Stoehr JP, Rabaglia ME, Schueler KL, Birnbaum MJ, and Attie AD
- Subjects
- Adipocytes enzymology, Animals, Blotting, Western, Enzyme Activation physiology, Female, Male, Mice, Mice, Inbred Strains, Phosphorylation, Proto-Oncogene Proteins c-akt, Receptor, Insulin metabolism, Insulin Resistance physiology, Phosphatidylinositol 3-Kinases metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins
- Abstract
Insulin stimulates muscle and adipose tissue to absorb glucose through a signaling cascade that is incompletely understood. Insulin resistance, the inability of insulin to appropriately stimulate glucose uptake, is a hallmark of type 2 diabetes mellitus. The development of experimental systems that model human insulin resistance is important in elucidating the defects responsible for the development of type 2 diabetes. When two strains of mice, BTBR and C57BL/6J (B6), are crossed, the resultant male offspring (BtB6) demonstrate insulin resistance in muscle tissue. Here, we report an insulin resistance phenotype in adipose tissue from lean, nondiabetic BtB6 mice similar to that observed in human muscle. Adipocytes isolated from insulin-resistant male mice display 65% less insulin-stimulated glucose uptake compared with insulin-sensitive female mice. Similarly, adipocytes from insulin-resistant mice have diminished insulin-stimulated IRS-1 phosphorylation and phosphatidylinositol 3-kinase (PI3K) activation. However, normal activation of protein kinase B (Akt/PKB) by insulin is observed. Thus BtB6 mice demonstrate the dissociation of insulin-stimulated PI3K activity and Akt/PKB activation and represent a useful model to investigate the causes of insulin resistance in humans.
- Published
- 2001
- Full Text
- View/download PDF
39. Genetic obesity unmasks nonlinear interactions between murine type 2 diabetes susceptibility loci.
- Author
-
Stoehr JP, Nadler ST, Schueler KL, Rabaglia ME, Yandell BS, Metz SA, and Attie AD
- Subjects
- Alleles, Animals, Blood Glucose analysis, Diabetes Mellitus genetics, Diabetes Mellitus pathology, Diabetes Mellitus, Type 2 pathology, Fasting, Hyperinsulinism genetics, Immunohistochemistry, Insulin analysis, Insulin blood, Insulin Resistance genetics, Islets of Langerhans chemistry, Islets of Langerhans pathology, Lod Score, Male, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Mice, Obese, Diabetes Mellitus, Type 2 genetics, Genetic Predisposition to Disease, Obesity genetics
- Abstract
Nonlinear interactions between obesity and genetic risk factors are thought to determine susceptibility to type 2 diabetes. We used genetic obesity as a tool to uncover latent differences in diabetes susceptibility between two mouse strains, C57BL/6J (B6) and BTBR. Although both BTBR and B6 lean mice are euglycemic and glucose tolerant, lean BTBR x B6 F1 male mice are profoundly insulin resistant. We hypothesized that the genetic determinants of the insulin resistance syndrome might also predispose genetically obese mice to severe diabetes. Introgressing the ob allele into BTBR revealed large differences in diabetes susceptibility between the strain backgrounds. In a population of F2-ob/ob mice segregating for BTBR and B6 alleles, we observed large variation in pancreatic compensation for the underlying insulin resistance. We also detected two loci that substantially modify diabetes severity, and a third locus that strongly links to fasting plasma insulin levels. Amplification of the genetic signal from these latent diabetes susceptibility alleles in F2-ob/ob mice permitted discovery of an interaction between the two loci that substantially increased the risk of severe type 2 diabetes.
- Published
- 2000
- Full Text
- View/download PDF
40. A defect late in stimulus-secretion coupling impairs insulin secretion in Goto-Kakizaki diabetic rats.
- Author
-
Metz SA, Meredith M, Vadakekalam J, Rabaglia ME, and Kowluru A
- Subjects
- Animals, Disease Models, Animal, Humans, Insulin Secretion, Purine Nucleotides metabolism, Rats, Rats, Inbred Strains, Rats, Wistar, Secretory Rate, Signal Transduction physiology, Type C Phospholipases metabolism, Diabetes Mellitus, Experimental physiopathology, Diabetes Mellitus, Type 2 physiopathology, Insulin metabolism, Islets of Langerhans metabolism
- Abstract
A widely accepted genetically determined rodent model for human type 2 diabetes is the Goto-Kakizaki (GK) rat; however, the lesion(s) in the pancreatic islets of these rats has not been identified. Herein, intact islets from GK rats (aged 8-14 weeks) were studied, both immediately after isolation and after 18 h in tissue culture. Despite intact contents of insulin and protein, GK islets had markedly deficient insulin release in response to glucose, as well as to pure mitochondrial fuels or a non-nutrient membrane-depolarizing stimulus (40 mmol/l K+). In contrast, mastoparan (which activates GTP-binding proteins [GBPs]) completely circumvented any secretory defect. Basal and stimulated levels of adenine and guanine nucleotides, the activation of phospholipase C by Ca2+ or glucose, the secretory response to pertussis toxin, and the activation of selected low-molecular weight GBPs were not impaired. Defects were found, however, in the autophosphorylation and catalytic activity of cytosolic nucleoside diphosphokinase (NDPK), which may provide compartmentalized GTP pools to activate G-proteins; a deficient content of phosphoinositides was also detected. These studies identify novel, heretofore unappreciated, defects late in signal transduction in the islets of our colony of GK rats, possibly occurring at the site of activation by NDPK of a mastoparan-sensitive G-protein-dependent step in exocytosis.
- Published
- 1999
- Full Text
- View/download PDF
41. Prolonged depletion of guanosine triphosphate induces death of insulin-secreting cells by apoptosis.
- Author
-
Li G, Segu VB, Rabaglia ME, Luo RH, Kowluru A, and Metz SA
- Subjects
- Animals, Apoptosis drug effects, Bacterial Toxins pharmacology, GTP-Binding Proteins physiology, Insulin Secretion, Islets of Langerhans metabolism, Lovastatin pharmacology, Mitosis physiology, Mycophenolic Acid pharmacology, Rats, Rats, Sprague-Dawley, Time Factors, Apoptosis physiology, Bacterial Proteins, Guanosine Triphosphate deficiency, Insulin metabolism, Islets of Langerhans physiology
- Abstract
Inhibitors of IMP dehydrogenase, such as mycophenolic acid (MPA) and mizoribine, which deplete cellular GTP, are used clinically as immunosuppressive drugs. The prolonged effect of such agents on insulin-secreting beta-cells (HIT-T15 and INS-1) was investigated. Both MPA and mizoribine inhibited mitogenesis, as reflected by [3H]thymidine incorporation. Cell number, DNA and protein contents, and cell (metabolic) viability were decreased by about 30%, 60%, and 80% after treatment of HIT cells with clinically relevant concentrations (e.g. 1 microg/ml) of MPA for 1, 2, and 4 days, respectively. Mizoribine (48 h) similarly induced the death of HIT cells. INS-1 cells also were damaged by prolonged MPA treatment. MPA-treated HIT cells displayed a strong and localized staining with a DNA-binding dye (propidium iodide), suggesting condensation and fragmentation of DNA, which were confirmed by detection of DNA laddering in multiples of about 180 bp. DNA fragmentation was observed after 24-h MPA treatment and was dose dependent (29%, 49%, and 70% of cells were affected after 48-h exposure to 1, 3, and 10 microg/ml MPA, respectively). Examination of MPA-treated cells by electron microscopy revealed typical signs of apoptosis: condensed and marginated chromatin, apoptotic bodies, cytosolic vacuolization, and loss of microvilli. MPA-induced cell death was almost totally prevented by supplementation with guanosine, but not with adenosine or deoxyguanosine, indicating a specific effect of GTP depletion. An inhibitor of protein isoprenylation (lovastatin, 10-100 microM for 2-3 days) induced cell death and DNA degradation similar to those induced by sustained GTP depletion, suggesting a mediatory role of posttranslationally modified GTP-binding proteins. Indeed, impeding the function of G proteins of the Rho family (via glucosylation using Clostridium difficile toxin B), although not itself inducing apoptosis, potentiated cell death induced by MPA or lovastatin. These findings indicate that prolonged depletion of GTP induces beta-cell death compatible with apoptosis; this probably involves a direct impairment of GTP-dependent RNA-primed DNA synthesis, but also appears to be modulated by small GTP-binding proteins. Treatment of intact adult rat islets (the beta-cells of which replicate slowly) induced a modest, but definite, death by apoptosis over 1- to 3-day periods. Thus, more prolonged use of the new generation of immunosuppressive agents exemplified by MPA might have deleterious effects on the survival of islet or pancreas grafts.
- Published
- 1998
- Full Text
- View/download PDF
42. Evidence for differential roles of the Rho subfamily of GTP-binding proteins in glucose- and calcium-induced insulin secretion from pancreatic beta cells.
- Author
-
Kowluru A, Li G, Rabaglia ME, Segu VB, Hofmann F, Aktories K, and Metz SA
- Subjects
- ADP Ribose Transferases pharmacology, Adenosine Diphosphate Ribose metabolism, Animals, Bacterial Toxins pharmacology, Enterotoxins pharmacology, Glycosylation, Insulin Secretion, Male, Potassium pharmacology, Rats, Rats, Sprague-Dawley, rhoB GTP-Binding Protein, Bacterial Proteins, Botulinum Toxins, Calcium metabolism, GTP Phosphohydrolases physiology, GTP-Binding Proteins physiology, Glucose pharmacology, Insulin metabolism, Membrane Proteins physiology
- Abstract
We utilized clostridial toxins (with known specificities for inhibition of GTPases) to ascertain the contribution of candidate GTPases in physiologic insulin secretion from beta cells. Exposure of normal rat islets or isolated beta (HIT-T15) cells to Clostridium difficile toxins A and B catalyzed the glucosylation (and thereby the inactivation) of Rac, Cdc42, and Rho endogenous to beta cells; concomitantly, either toxin reduced glucose- or potassium-induced insulin secretion from rat islets and HIT cells. Treatment of beta cells with Clostridium sordellii lethal toxin (LT; which modified only Ras, Rap, and Rac) also reduced glucose- or potassium-induced secretion. However, clostridial toxin C3-exoenzyme (which ADP-ribosylates and inactivates only Rho) was without any effect on either glucose- or potassium-induced insulin secretion. These data suggest that Cdc42, Rac, Ras, and/or Rap (but not Rho) may be needed for glucose- or potassium-mediated secretion. The effects of these toxins appear to be specific on stimulus-secretion coupling, since no difference in metabolic viability (assessed colorimetrically by quantitating the conversion of the tetrazolium salt into a formazan in a reduction reaction driven by nutrient metabolism) was demonstrable between control and toxin (A or LT)-treated beta cells. Toxin (A or LT) treatment also did not alter glucose- or potassium-mediated rises in cytosolic free calcium concentrations ([Ca2+]i), suggesting that these GTPases are involved in steps distal to elevations in [Ca2+]i. Recent findings indicate that the carboxyl methylation of Cdc42 is stimulated by only glucose, whereas that of Rap (Kowluru et al., J Clin Invest 98: 540-555, 1996) and Rac (present study) are regulated by glucose or potassium. Together, these findings provide direct evidence, for the first time, that the Rho subfamily of GTPases plays a key regulatory role(s) in insulin secretion, and they suggest that Cdc42 may be required for early steps in glucose stimulation of insulin release, whereas Rap and/or Rac may be required for a later step(s) in the stimulus-secretion coupling cascade (i.e. Ca2+-induced exocytosis of insulin).
- Published
- 1997
- Full Text
- View/download PDF
43. Interleukin-1 beta inhibits phospholipase C and insulin secretion at sites apart from KATP channel.
- Author
-
Vadakekalam J, Rabaglia ME, and Metz SA
- Subjects
- Animals, Cell Polarity drug effects, Cells, Cultured, Glucose pharmacology, Glyburide pharmacology, Humans, Insulin Secretion, Islets of Langerhans drug effects, Kinetics, Male, Phosphatidylinositols metabolism, Potassium Channels drug effects, Rats, Rats, Sprague-Dawley, Recombinant Proteins pharmacology, Type C Phospholipases antagonists & inhibitors, Insulin metabolism, Interleukin-1 pharmacology, Islets of Langerhans physiology, Potassium pharmacology, Potassium Channels physiology, Potassium Channels, Inwardly Rectifying, Type C Phospholipases metabolism
- Abstract
Although interleukin-1 beta (IL-1 beta) reduces pancreatic islet content of ATP and GTP, the distal events that mediate its inhibitory effects on insulin secretion remain poorly understood. Herein, the activation of phospholipase C (PLC) was quantified during islet perifusions. An 18-h exposure to IL-1 beta (100 pM) totally vitiated activation of PLC induced by glucose, an effect that requires ATP and GTP and closure of the ATP-dependent K+ (KATP) channel. Surprisingly, however, when islets were depolarized directly using either of two agonists, glyburide (which does not act via generation of purine nucleotides) or 40 mM K+ (which acts distal to KATP channel), PLC and insulin secretion were again obliterated by IL-1 beta. IL-1 beta also reduced the labeling of phosphoinositide substrates; however, this effect was insufficient to explain the inhibition of PLC, since the effects on substrate labeling, but not on PLC, were prevented by coprovision of guanosine or adenosine. Furthermore, when IL-1 beta-treated islets were exposed to 100 microM carbachol (which activates PLC partially independent of extracellular Ca2+), the effects were still obliterated by IL-1 beta. These data (together with the finding that IL-1 beta inhibited Ca(2+)-induced insulin release) suggest that, in addition to its effects on ATP synthesis and thereby on the KATP channel, IL-1 beta has at least two undescribed, distal effects to block both PLC as well as Ca(2+)-induced exocytosis. The latter correlated best with IL-1 beta's effect to impede phosphoinositide synthesis, since it also was reversed by guanosine or adenosine.
- Published
- 1997
- Full Text
- View/download PDF
44. Roles of GTP and phospholipase C in the potentiation of Ca(2+)-induced insulin secretion by glucose in rat pancreatic islets.
- Author
-
Vadakekalam J, Rabaglia ME, and Metz SA
- Subjects
- Animals, Culture Techniques, Diazoxide pharmacology, Drug Synergism, Enzyme Activation, Insulin Secretion, Islets of Langerhans drug effects, Male, Phosphatidylinositols metabolism, Potassium Channels drug effects, Rats, Rats, Sprague-Dawley, Calcium metabolism, Glucose pharmacology, Guanosine Triphosphate metabolism, Insulin metabolism, Islets of Langerhans metabolism, Type C Phospholipases metabolism
- Abstract
Glucose can augment insulin secretion independently of K+ channel closure, provided cytoplasmic free Ca2+ concentration is elevated. A role for phospholipase C (PLC) in this phenomenon has been both claimed and refuted. Recently, we have shown a role for GTP in the secretory effect of glucose as well as in glucose-induced PLC activation, using islets pre-treated with GTP synthesis inhibitors such as mycophenolic acid (MPA). Therefore, in the current studies, we examined first, whether glucose augments Ca(2+)-induced PLC activation and second, whether GTP is required for this effect, when K+(ATP) channels are kept open using diazoxide. Isolated rat islets pre-labeled with [3H]myo-inositol were studied with or without first priming with glucose. There was a 98% greater augmentation of insulin secretion by 16.7 mM glucose (in the presence of diazoxide and 40 mM K+) in primed islets; however, the ability of high glucose to augment PLC activity bore no relationship to the secretory response. MPA markedly inhibited PLC in both conditions; however, insulin secretion was only inhibited (by 46%) in primed islets. None of these differences were attributable to alterations in labeling of phosphoinositides or levels of GTP or ATP. These data indicate that an adequate level of GTP is critical for glucose's potentiation of Ca(2+)-induced insulin secretion in primed islets but that PLC activation can clearly be dissociated from insulin secretion and therefore cannot be the major cause of glucose's augmentation of Ca(2+)-induced insulin secretion.
- Published
- 1997
- Full Text
- View/download PDF
45. Glucose- and GTP-dependent stimulation of the carboxyl methylation of CDC42 in rodent and human pancreatic islets and pure beta cells. Evidence for an essential role of GTP-binding proteins in nutrient-induced insulin secretion.
- Author
-
Kowluru A, Seavey SE, Li G, Sorenson RL, Weinhaus AJ, Nesher R, Rabaglia ME, Vadakekalam J, and Metz SA
- Subjects
- Acetylcysteine analogs & derivatives, Acetylcysteine pharmacology, Animals, Blotting, Western, Cell Cycle Proteins analysis, Cell Line, Cells, Cultured, Enzyme Inhibitors pharmacology, GTP-Binding Proteins analysis, Guanosine 5'-O-(3-Thiotriphosphate) pharmacology, Humans, Insulin Secretion, Insulinoma physiopathology, Islets of Langerhans drug effects, Kinetics, Male, Methylation, Pancreatic Neoplasms physiopathology, Potassium pharmacology, Protein Methyltransferases antagonists & inhibitors, Rats, Rats, Sprague-Dawley, cdc42 GTP-Binding Protein, Cell Cycle Proteins metabolism, GTP-Binding Proteins metabolism, Glucose pharmacology, Guanosine Triphosphate pharmacology, Insulin metabolism, Islets of Langerhans physiology, Protein Methyltransferases metabolism
- Abstract
Several GTP-binding proteins (G-proteins) undergo post-translational modifications (isoprenylation and carboxyl methylation) in pancreatic beta cells. Herein, two of these were identified as CDC42 and rap 1, using Western blotting and immunoprecipitation. Confocal microscopic data indicated that CDC42 is localized only in islet endocrine cells but not in acinar cells of the pancreas. CDC42 undergoes a guanine nucleotide-specific membrane association and carboxyl methylation in normal rat islets, human islets, and pure beta (HIT or INS-1) cells. GTPgammaS-dependent carboxyl methylation of a 23-kD protein was also demonstrable in secretory granule fractions from normal islets or beta cells. AFC (a specific inhibitor of prenyl-cysteine carboxyl methyl transferases) blocked the carboxyl methylation of CDC42 in five types of insulin-secreting cells, without blocking GTPgammaS-induced translocation, implying that methylation is a consequence (not a cause) of transfer to membrane sites. High glucose (but not a depolarizing concentration of K+) induced the carboxyl methylation of CDC42 in intact cells, as assessed after specific immunoprecipitation. This effect was abrogated by GTP depletion using mycophenolic acid and was restored upon GTP repletion by coprovision of guanosine. In contrast, although rap 1 was also carboxyl methylated, it was not translocated to the particulate fraction by GTPgammaS; furthermore, its methylation was also stimulated by 40 mM K+ (suggesting a role which is not specific to nutrient stimulation). AFC also impeded nutrient-induced (but not K+-induced) insulin secretion from islets and beta cells under static or perifusion conditions, whereas an inactive structural analogue of AFC failed to inhibit insulin release. These effects were reproduced not only by S-adenosylhomocysteine (another methylation inhibitor), but also by GTP depletion. Thus, the glucose- and GTP-dependent carboxyl methylation of G-proteins such as CDC42 is an obligate step in the stimulus-secretion coupling of nutrient-induced insulin secretion, but not in the exocytotic event itself. Furthermore, AFC blocked glucose-activated phosphoinositide turnover, which may provide a partial biochemical explanation for its effect on secretion, and implies that certain G-proteins must be carboxyl methylated for their interaction with signaling effector molecules, a step which can be regulated by intracellular availability of GTP.
- Published
- 1996
- Full Text
- View/download PDF
46. Role for GTP in glucose-induced phospholipase C activation in pancreatic islets.
- Author
-
Vadakekalam J, Rabaglia ME, Chen QH, and Metz SA
- Subjects
- Animals, Calcium physiology, Carbachol pharmacology, Enzyme Activation drug effects, Guanine metabolism, Hydrolysis drug effects, Inositol Phosphates metabolism, Male, Mycophenolic Acid pharmacology, Phosphatidylinositols agonists, Phosphatidylinositols metabolism, Phospholipids metabolism, Rats, Rats, Sprague-Dawley, Glucose pharmacology, Guanosine Triphosphate physiology, Islets of Langerhans drug effects, Islets of Langerhans enzymology, Type C Phospholipases metabolism
- Abstract
We have previously demonstrated a permissive role for GTP in insulin secretion; in the current studies, we examined the effect of GTP on phospholipase C (PLC) activation to explore one possible mechanism for that observation. In rat islets preexposed to the GTP synthesis inhibitors mycophenolic acid (MPA) or mizoribine (MZ), PLC activation induced by 16.7 mM glucose (or by 20 mM alpha-ketoisocaproic acid) was inhibited 63% without altering the labeling of phosphoinositide substrates. Provision of guanine, which normalizes islet GTP content and insulin release, prevented the inhibition of PLC by MPA. Glucose-induced phosphoinositide hydrolysis was blocked by removal of extracellular Ca2+ or by diazoxide. PLC induced directly by Ca2+ influx (i.e., 40 mM K+) was reduced 42% in MPA-pretreated islets but without inhibition of the concomitant insulin release. These data indicate that glucose-induced PLC activation largely reflects Ca2+ entry and demonstrate (for the first time in intact cells) that adequate GTP is necessary for glucose (and Ca(2+)-)-induced PLC activation but not for maximal Ca(2+)-induced exocytosis.
- Published
- 1996
- Full Text
- View/download PDF
47. Carboxylmethylation of the catalytic subunit of protein phosphatase 2A in insulin-secreting cells: evidence for functional consequences on enzyme activity and insulin secretion.
- Author
-
Kowluru A, Seavey SE, Rabaglia ME, Nesher R, and Metz SA
- Subjects
- Animals, Binding, Competitive, Catalysis, Enzyme Inhibitors pharmacology, Esterases antagonists & inhibitors, Ethers, Cyclic pharmacology, GTP-Binding Proteins metabolism, Glucose pharmacology, Insulin Secretion, Islets of Langerhans metabolism, Keto Acids pharmacology, Lactones pharmacology, Male, Methylation, Molecular Weight, Okadaic Acid, Peptide Fragments metabolism, Phosphoprotein Phosphatases antagonists & inhibitors, Protein Phosphatase 2, Rats, Rats, Sprague-Dawley, S-Adenosylmethionine metabolism, Insulin metabolism, Islets of Langerhans enzymology, Phosphoprotein Phosphatases metabolism
- Abstract
We report the carboxylmethylation of a 36-kDa protein in intact normal rat islets and clonal beta (INS-1) cells. This protein was predominantly cytosolic. Its carboxylmethylation, as assessed by vapor phase equilibration assay, was resistant to inhibition by N-acetyl-S-trans, trans-farnesyl-L-cysteine, a competitive substrate for cysteine methyl transferases. These data suggest that the methylated C-terminal amino acid is not cysteine. The methylated protein was identified as the catalytic subunit of protein phosphatase 2A (PP2Ac) by immunoblotting. The carboxylmethylation of the PP2Ac increased its catalytic activity, suggesting a key role in the functional regulation of PP2A. Therefore, we studied okadaic acid, a selective inhibitor of PP2A that acts by an unknown mechanism. Okadaic acid (but not 1-nor-okadaone, its inactive analog) inhibited (Ki = 10 nM) the carboxylmethylation of PP2Ac and phosphatase activity in the cytosolic fraction (from normal rat islets and clonal beta-cells) as well as in intact rat islets. Furthermore, methylated PP2Ac underwent rapid demethylation (t 1/2 = 40 min) catalyzed by a methyl esterase localized in islet homogenates. Ebelactone, a purported inhibitor of methyl esterases, significantly delayed (> 200 min) the demethylation of PP2Ac. Furthermore, ebelactone reversibly inhibited glucose- and ketoisocaproate-induced insulin secretion from normal rat islets. These data identify, for the first time, a methylation-demethylation cycle for PP2Ac in the beta-cell and suggest a key functional relationship between PP2A activity and the carboxylmethylation of its catalytic subunit. These findings thus suggest a negative modulatory role for PP2A in nutrient-induced insulin exocytosis.
- Published
- 1996
- Full Text
- View/download PDF
48. Evidence of a role for GTP in the potentiation of Ca(2+)-induced insulin secretion by glucose in intact rat islets.
- Author
-
Meredith M, Rabaglia ME, and Metz SA
- Subjects
- ATP-Binding Cassette Transporters, Adenosine Triphosphate metabolism, Animals, Biological Transport, Active drug effects, Calcium pharmacology, Diazoxide pharmacology, Drug Interactions, Insulin Secretion, Islets of Langerhans metabolism, KATP Channels, Keto Acids pharmacology, Male, Mitochondria drug effects, Mycophenolic Acid pharmacology, Potassium Channels metabolism, Potassium Channels, Inwardly Rectifying, Purine Nucleotides metabolism, Rats, Rats, Sprague-Dawley, Secretory Rate drug effects, Succinates pharmacology, Glucose pharmacology, Guanosine Triphosphate physiology, Insulin metabolism, Islets of Langerhans drug effects, Potassium Channels drug effects, Signal Transduction drug effects
- Abstract
Glucose initiates insulin secretion by closing K(+)-ATP channels, leading to Ca2+ influx (E1); it also potentiates Ca(2+)-induced secretion (E2) when the K(+)-ATP channel is kept open using diazoxide and depolarizing concentrations of K+ are provided. To examine the roles of purine nucleotides in E2, we compared the effects of glucose to those of the mitochondrial fuel monomethylsuccinate. Either agonist could induce E2 accompanied by significant increases in ATP, ATP/ADP ratio, and GTP/GDP ratio; GTP increased significantly only with glucose. Mycophenolic acid (MPA), an inhibitor of cytosolic GTP synthesis, markedly inhibited glucose-induced E2 (either in perifusions or in static incubations) and decreased GTP and the GTP/GDP ratio, but did not alter the ATP/ADP ratio. Provision of guanine (but not adenine) reversed these changes pari passu. In contrast, MPA had no effect on succinate-induced E2, despite generally similar changes in nucleotides. A similar lack of effect of MPA on E2 was seen with a second mitochondrial fuel, alpha-ketoisocaproic acid (KIC). However, in the absence of diazoxide and K+, MPA blunted the secretory effects of either glucose, succinate, or KIC. These studies suggest that GTP plays a role in both glucose and succinate or KIC-induced insulin secretion at a step dependent on mitochondrial metabolism and the K(+)-ATP channel. In addition to mitochondrial effects, glucose appears to have extramitochondrial effects important to its potentiation of Ca(2+)-induced insulin secretion that are also dependent on GTP.
- Published
- 1995
- Full Text
- View/download PDF
49. Non-specific stimulatory effects of mastoparan on pancreatic islet nucleoside diphosphokinase activity: dissociation from insulin secretion.
- Author
-
Kowluru A, Seavey SE, Rabaglia ME, and Metz SA
- Subjects
- Animals, Enzyme Activation drug effects, Insulin Secretion, Intercellular Signaling Peptides and Proteins, Islets of Langerhans enzymology, Male, Peptides, Rats, Rats, Sprague-Dawley, Insulin metabolism, Islets of Langerhans drug effects, Nucleoside-Diphosphate Kinase metabolism, Wasp Venoms pharmacology
- Abstract
We examined whether mastoparan (MAS)-induced insulin secretion might involve the activation of nucleoside diphosphokinase (NDP kinase), which catalyzes the conversion of GDP to GTP, a known permissive factor for insulin secretion. MAS and MAS 7 (which activate GTP-binding proteins), but not MAS 17 (an inactive analog), stimulated insulin secretion from normal rat islets. In contrast to their specific effects on insulin secretion, MAS, MAS 7 and MAS 17 each stimulated formation of the phosphoenzyme-intermediate of NDP kinase, as well as its catalytic activity. These effects were mimicked by several cationic drugs. Thus, caution is indicated in using MAS to study cellular regulation, since some of its effects appear to be non-specific, and may be due, in part, to its amphiphilic, cationic nature.
- Published
- 1995
- Full Text
- View/download PDF
50. Subcellular localization and kinetic characterization of guanine nucleotide binding proteins in normal rat and human pancreatic islets and transformed beta cells.
- Author
-
Kowluru A, Rabaglia ME, Muse KE, and Metz SA
- Subjects
- Adult, Animals, Cell Line, Transformed, Cricetinae, Cytoplasmic Granules enzymology, Female, GTP Phosphohydrolases chemistry, GTP-Binding Proteins chemistry, Humans, Islets of Langerhans enzymology, Islets of Langerhans ultrastructure, Magnesium pharmacology, Male, Middle Aged, Rats, Rats, Sprague-Dawley, Cytoplasmic Granules chemistry, GTP Phosphohydrolases analysis, GTP-Binding Proteins analysis, Islets of Langerhans chemistry
- Abstract
The subcellular localization and the kinetics of the GTPase activities of monomeric and heterotrimeric GTP-binding proteins were investigated in normal rat and human pancreatic islets and were compared to those obtained using a transformed hamster beta cell line (HIT cells). The [alpha-32P]GTP overlay technique revealed the presence of at least four low-molecular-mass proteins (approx. 20-27 kDa) in normal rat islets, which were enriched in the secretory granule fraction compared to the membrane fraction (with little abundance of these proteins in the cytosolic fraction). In contrast, in HIT cells, these proteins (at least six) were predominantly cytosolic. Three of these proteins were immunologically identified as rab3A, rac2, and CDC42Hs in islets as well as in HIT cells. In addition, pertussis toxin augmented the ribosylation of at least one heterotrimeric G-protein of about 39 kDa (probably G(i) and/or G(o)) in the membrane and secretory granule fractions of normal rat and human islets, whereas at least three such Ptx substrates (36-39 kDa) were found in HIT cell membranes. Kinetic activities revealed the presence of at least three such activities (Km for GTP of 372 nM, 2.2 microM, and 724 microM) in islet homogenates which were differentially distributed in various subcellular fractions; similar activities were also demonstrable in HIT cell homogenates. Thus, these studies demonstrate the presence of both monomeric G-proteins intrinsic to the secretory granules of normal rat islets which can be ascribed to beta cells; since these G-proteins are regulated by insulinotropic lipids (as described in the accompanying article), such proteins may couple the activation of phospholipases (endogenous to islets) to the exocytotic secretion of insulin. These findings also suggest that caution is necessary in extrapolating data concerning G-proteins from cultured, transformed beta cell lines to the physiology of normal islets, in view of both qualitative and quantitative differences between the two preparations.
- Published
- 1994
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.