106 results on '"Shiota C"'
Search Results
2. Evidence for an endogenous regulator of beta-cell glucokinase from a naturally occurring glucokinase mutation (V62M) which causes maturity-onset diabetes of the young
- Author
-
Gloyn, A. L., Odili, S., Zelent, D., Buettger, C., Castledon, H. A. J., Steele, A. M., Stride, S., Shiota, C., Magnuson, M. A., Lorini, R., dʼAnnunzio, G., Grimsby, J., Grippo, J. F., Barbetti, F., Ellard, S., Hattersley, A. T., and Matschinsky, F. M.
- Published
- 2004
3. Molecular Genetics of Myelin Proteolipid Proteins and Their Expression in Normal and Jimpy Mice
- Author
-
Mikoshiba, K., Shiota, C., Ikenaka, K., and Kim, Seung U., editor
- Published
- 1989
- Full Text
- View/download PDF
4. Insights into biochemical and genetical basis of glucokinase activation from naturally occuring hypoglycaemia mutations
- Author
-
Gloyn, A., cees noordam, Willemsen, M., Ellard, S., Lam, W., Shiota, C., Magnuson, M., Matschinsky, F., and Hattersley, A.
- Published
- 2016
5. Avaliação sócio-ambiental da integração tecnológica Embrapa Pecuária Sudeste para produção leiteira na agricultura familiar
- Author
-
RODRIGUES, G. S., RODRIGUES, I. A., TUPY, O., CAMARGO, A. C. de, NOVO, A. L. M., BONADIO, L. F., TOKUDA, F. S., ANDRADE, E. F., SHIOTA, C. M., SILVA, R. A. da, GERALDO STACHETTI RODRIGUES, CNPMA, I. A. RODRIGUES, Embrapa Meio Ambiente, OSCAR TUPY, CPPSE, ARTUR CHINELATO DE CAMARGO, CPPSE, ANDRE LUIZ MONTEIRO NOVO, CPPSE, L. F. BONADIO, F. S. TOKUDA, Casa da Agricultura Pontes Gestal, E. F. ANDRADE, Casa da Agricultura em Cardoso, C. M. SHIOTA, and R. A. da. SILVA, Casa da Lavoura Valentim Gentil.
- Subjects
Tecnologia ,Gado leiteiro ,Avaliação de impacto ,Impacto Ambiental ,Adoção de Inovações ,Indicadores de sustentabilidade ,Metodologia ,Desenvolvimento Sustentável - Abstract
Duas características da pecuária leiteira podem ser destacadas na atualidade: a) o baixo valor do leite, que dificulta a adoção de tecnologias que favoreçam a produtividade e b) a concentração da produção leiteira em estabelecimentos pequenos, nos quais predomina a atividade em nível de subsistência, com mão-de-obra familiar e renda mensal que não ultrapassa um salário mínimo. Um projeto de pesquisa e transferência de tecnologia da Embrapa Pecuária Sudeste (São Carlos, Estado de São Paulo) vem sendo desenvolvido desde 1998 para melhoria desse cenário. Uma vez implantado o projeto em uma série de estabelecimentos na região de Votuporanga, Estado de São Paulo, desenvolveu-se este estudo, para avaliar os impactos sócio-ambientais da adoção da tecnologia. Os resultados indicam que o desempenho dos estabelecimentos melhora em função do tempo desde a adoção e que a tecnologia contribui positivamente para o desenvolvimento sustentável, sendo recomendada para transferência.
- Published
- 2006
6. Regulation of β Cell Proliferation by Targeting SMAD2
- Author
-
Gaffar, I., primary, Xiao, X., additional, El-Gohary, Y., additional, Wiersch, J., additional, Prasadan, K., additional, Guo, P., additional, Shiota, C., additional, and Gittes, G., additional
- Published
- 2014
- Full Text
- View/download PDF
7. The second activating glucokinase mutation (A456V): Implications for glucose homeostasis and diabetes therapy
- Author
-
Henrik Thybo Christesen, Jacobsen, B. B., Odili, S., Buettger, C., Cuesta-Munoz, A., Hansen, T., Brusgaard, K., Massa, O., Magnuson, M. A., Shiota, C., Matschinsky, F. M., and Barbetti, F.
- Subjects
Male ,Adolescent ,Diazoxide ,Infant, Newborn ,Mutation, Missense ,Settore MED/13 - Endocrinologia ,Enzyme Activation ,Kinetics ,Glucose ,Allosteric Regulation ,Glucokinase ,Infant, Small for Gestational Age ,Insulin Secretion ,Diabetes Mellitus ,Homeostasis ,Humans ,Insulin ,Phosphorylation ,Glutathione Transferase - Abstract
In this study, a second case of hyperinsulinemic hypoglycemia due to activation of glucokinase is reported. The 14-year-old proband had a history of neonatal hypoglycemia, treated with diazoxide. He was admitted with coma and convulsions due to nonketotic hypoglycemia. His BMI was 34 kg/m(2), and his fasting blood glucose ranged from 2.1 to 2.7 mmol/l, associated with inappropriately high serum levels of insulin, C-peptide, and proinsulin. An oral glucose tolerance test (OGTT) showed exaggerated responses of these peptides followed by profound hypoglycemia. Treatment with diazoxide and chlorothiazide was effective. His mother never had clinical hypoglycemic symptoms, even though her fasting blood glucose ranged from 2.9 to 3.5 mmol/l. Increases in serum insulin, C-peptide, and proinsulin in response to an OGTT suggested a lower threshold for glucose-stimulated insulin release (GSIR). Screening for mutations in candidate genes revealed a heterozygous glucokinase mutation in exon 10, substituting valine for alanine at codon 456 (A456V) in the proband and his mother. The purified recombinant glutathionyl S-transferase fusion protein of the A456V glucokinase revealed a decreased glucose S(0.5) (the concentration of glucose needed to achieve the half-maximal rate of phosphorylation) from 8.04 (wild-type) to 2.53 mmol/l. The mutant's Hill coefficient was decreased, and its maximal specific activity k(cat) was increased. Mathematical modeling predicted a markedly lowered GSIR threshold of 1.5 mmol/l. The theoretical and practical implications are manifold and significant.
- Published
- 2002
8. Dysregulation of the norepinephrine transporter sustains cortical hypodopaminergia and schizophrenialike behaviors in neuronal rictor null mice
- Author
-
Siuta, MA, Robertson, SD, Kocalis, H, Saunders, C, Gresch, PJ, Khatri, V, Shiota, C, Philip Kennedy, J, Lindsley, CW, Daws, LC, Polley, DB, Veenstra-Vanderweele, J, Stanwood, GD, Magnuson, MA, Niswender, KD, Galli, A, Siuta, MA, Robertson, SD, Kocalis, H, Saunders, C, Gresch, PJ, Khatri, V, Shiota, C, Philip Kennedy, J, Lindsley, CW, Daws, LC, Polley, DB, Veenstra-Vanderweele, J, Stanwood, GD, Magnuson, MA, Niswender, KD, and Galli, A
- Abstract
The mammalian target of rapamycin (mTOR) complex 2 (mTORC2) is a multimeric signaling unit that phosphorylates protein kinase B/Akt following hormonal and growth factor stimulation. Defective Akt phosphorylation at the mTORC2-catalyzed Ser473 site has been linked to schizophrenia. While human imaging and animal studies implicate a fundamental role for Akt signaling in prefrontal dopaminergic networks, the molecular mechanisms linking Akt phosphorylation to specific schizophrenia-related neurotransmission abnormalities have not yet been described. Importantly, current understanding of schizophrenia suggests that cortical decreases in DA neurotransmission and content, defined here as cortical hypodopaminergia, contribute to both the cognitive deficits and the negative symptoms characteristic of this disorder. We sought to identify a mechanism linking aberrant Akt signaling to these hallmarks of schizophrenia. We used conditional gene targeting in mice to eliminate the mTORC2 regulatory protein rictor in neurons, leading to impairments in neuronal Akt Ser473 phosphorylation. Rictor-null (KO) mice exhibit prepulse inhibition (PPI) deficits, a schizophrenia-associated behavior. In addition, they show reduced prefrontal dopamine (DA) content, elevated cortical norepinephrine (NE), unaltered cortical serotonin (5-HT), and enhanced expression of the NE transporter (NET). In the cortex, NET takes up both extracellular NE and DA. Thus, we propose that amplified NET function in rictor KO mice enhances accumulation of both NE and DA within the noradrenergic neuron. This phenomenon leads to conversion of DA to NE and ultimately supports both increased NE tissue content as well as a decrease in DA. In support of this hypothesis, NET blockade in rictor KO mice reversed cortical deficits in DA content and PPI, suggesting that dysregulation of DA homeostasis is driven by alteration in NET expression, which we show is ultimately influenced by Akt phosphorylation status. These data il
- Published
- 2010
9. Immunologic Regulation of Pancreatic Ductal Invasion of Islets
- Author
-
Wiersch, J., primary, Marr, M., additional, Gohary, Y., additional, Xiao, X., additional, Guo, P., additional, Prasadan, K., additional, Shiota, C., additional, Piganelli, J., additional, and Gittes, G., additional
- Published
- 2013
- Full Text
- View/download PDF
10. Pancreatic Duct Glands: Are They the Missing Pancreas Specific Stem Cells
- Author
-
El-Gohary, Y., primary, Xiao, X., additional, Guo, P., additional, Wiersch, J., additional, Prasadan, K., additional, Shiota, C., additional, and Gittes, G., additional
- Published
- 2013
- Full Text
- View/download PDF
11. Three-Dimensional Analysis of the Islet Vasculature
- Author
-
El-Gohary, Y., primary, Sims-Lucas, S., additional, Lath, N., additional, Tulachan, S., additional, Guo, P., additional, Xiao, X., additional, Welsh, C., additional, Paredes, J., additional, Wiersch, J., additional, Prasadan, K., additional, Shiota, C., additional, and Gittes, G.K., additional
- Published
- 2012
- Full Text
- View/download PDF
12. Pancreatic Ducts and Alpha Cells Contribute to New Beta Cells Under Normal Physiological Conditions and During Pancreatic Regeneration
- Author
-
El-Gohary, Y., primary, Tulachan, S., additional, Prasadan, K., additional, Shiota, C., additional, Guo, P., additional, and Gittes, G., additional
- Published
- 2012
- Full Text
- View/download PDF
13. Insights into the biochemical and genetic basis of glucokinase activation from naturally occurring hypoglycemia mutations.
- Author
-
Gloyn, A.L., Noordam, C., Willemsen, M.A.A.P., Ellard, S., Lam, W.W., Campbell, I.W., Midgley, P., Shiota, C., Buettger, C., Magnuson, M.A., Matschinsky, F.M., Hattersley, A.T., Gloyn, A.L., Noordam, C., Willemsen, M.A.A.P., Ellard, S., Lam, W.W., Campbell, I.W., Midgley, P., Shiota, C., Buettger, C., Magnuson, M.A., Matschinsky, F.M., and Hattersley, A.T.
- Abstract
Item does not contain fulltext, Glucokinase (GCK) is a key regulatory enzyme in the pancreatic beta-cell and catalyzes the rate-limiting step for beta-cell glucose metabolism. We report two novel GCK mutations (T65I and W99R) that have arisen de novo in two families with familial hypoglycemia. Insulin levels, although inappropriately high for the degree of hypoglycemia, remain regulated by fluctuations in glycemia, and pancreatic histology was normal. These mutations are within the recently identified heterotropic allosteric activator site in the theoretical model of human beta-cell glucokinase. Functional analysis of the purified recombinant glutathionyl S-transferase fusion proteins of T65I and W99R GCK revealed that the kinetic changes result in a relative increased activity index (a measure of the enzyme's phosphorylating potential) of 9.81 and 6.36, respectively, compared with wild-type. The predicted thresholds for glucose-stimulated insulin release using mathematical modeling were 3.1 (T65I) and 2.8 (W99R) mmol/l, which were in line with the patients' fasting glucose. In conclusion, we have identified two novel spontaneous GCK-activating mutations whose clinical phenotype clearly differs from mutations in ATP-sensitive K(+) channel genes. In vitro studies confirm the validity of structural and functional models of GCK and the putative allosteric activator site, which is a potential drug target for the treatment of type 2 diabetes.
- Published
- 2003
14. Glucokinase and the Regulation of Blood Sugar.
- Author
-
Gloyn, A.L., Odili, S., Buettger, C., Njølstad, P.R., Shiota, C., Magnuson, M.A., and Matschinsky, F.M.
- Published
- 2004
- Full Text
- View/download PDF
15. Characterization of glucokinase regulatory protein-deficient mice.
- Author
-
Grimsby, J, Coffey, J W, Dvorozniak, M T, Magram, J, Li, G, Matschinsky, F M, Shiota, C, Kaur, S, Magnuson, M A, and Grippo, J F
- Abstract
The glucokinase regulatory protein (GKRP) inhibits glucokinase competitively with respect to glucose by forming a protein-protein complex with this enzyme. The physiological role of GKRP in controlling hepatic glucokinase activity was addressed using gene targeting to disrupt GKRP gene expression. Heterozygote and homozygote knockout mice have a substantial decrease in hepatic glucokinase expression and enzymatic activity as measured at saturating glucose concentrations when compared with wild-type mice, with no change in basal blood glucose levels. Interestingly, when assayed under conditions to promote the association between glucokinase and GKRP, liver glucokinase activity in wild-type and null mice displayed comparable glucose phosphorylation capacities at physiological glucose concentrations (5 mM). Thus, despite reduced hepatic glucokinase expression levels in the null mice, glucokinase activity in the liver homogenates was maintained at nearly normal levels due to the absence of the inhibitory effects of GKRP. However, following a glucose tolerance test, the homozygote knockout mice show impaired glucose clearance, indicating that they cannot recruit sufficient glucokinase due to the absence of a nuclear reserve. These data suggest both a regulatory and a stabilizing role for GKRP in maintaining adequate glucokinase in the liver. Furthermore, this study provides evidence for the important role GKRP plays in acutely regulating of hepatic glucose metabolism.
- Published
- 2000
16. Nuclear import of hepatic glucokinase depends upon glucokinase regulatory protein, whereas export is due to a nuclear export signal sequence in glucokinase.
- Author
-
Shiota, C, Coffey, J, Grimsby, J, Grippo, J F, and Magnuson, M A
- Abstract
Hepatic glucokinase (GK) moves between the nucleus and cytoplasm in response to metabolic alterations. Here, using heterologous cell systems, we have found that at least two different mechanisms are involved in the intracellular movement of GK. In the absence of the GK regulatory protein (GKRP) GK resides only in the cytoplasm. However, in the presence of GKRP, GK moves to the nucleus and resides there in association with this protein until changes in the metabolic milieu prompt its release. GK does not contain a nuclear localization signal sequence and does not enter the nucleus in a GKRP-independent manner because cells treated with leptomycin B, a specific inhibitor of leucine-rich NES-dependent nuclear export, do not accumulate GK in the nucleus. Instead, entry of GK into the nucleus appears to occur via a piggy-back mechanism that involves binding to GKRP. Nuclear export of GK, which occurs after its release from GKRP, is due to a leucine-rich nuclear export signal within the protein ((300)ELVRLVLLKLV(310)). Thus, GKRP appears to function as both a nuclear chaperone and metabolic sensor and is a critical component of a hepatic GK translocation cycle for regulating the activity of this enzyme in response to metabolic alterations.
- Published
- 1999
17. Concentration of serum laminin and type IV collagen in liver diseases assayed by a sandwich enzyme-immunoassay using monoclonal antibodies
- Author
-
Yokoya, Y., Iwata, K., Muragaki, Y., and Shiota, C.
- Published
- 1992
- Full Text
- View/download PDF
18. Neonatal diabetes mellitus due to complete glucokinase deficiency.
- Author
-
Njølstad, P R, Søvik, O, Cuesta-Muñoz, A, Bjørkhaug, L, Massa, O, Barbetti, F, Undlien, D E, Shiota, C, Magnuson, M A, Molven, A, Matschinsky, F M, and Bell, G I
- Published
- 2001
19. The impact of glucagon to support postabsorptive glucose flux and glycemia in healthy rats and its attenuation in male Zucker diabetic fatty rats.
- Author
-
Estes SK, Shiota C, O'Brien TP, Printz RL, and Shiota M
- Subjects
- Animals, Male, Rats, Blood Glucose metabolism, Cyclic AMP, Glucagon metabolism, Glucose metabolism, Insulin metabolism, Rats, Zucker, Diabetes Mellitus, Type 2 metabolism, Hyperglycemia metabolism
- Abstract
Hyperglucagonemia is a hallmark of type 2 diabetes (T2DM), yet the role of elevated plasma glucagon (P-GCG) to promote excessive postabsorptive glucose production and contribute to hyperglycemia in patients with this disease remains debatable. We investigated the acute action of P-GCG to safeguard/support postabsorptive endogenous glucose production (EGP) and euglycemia in healthy Zucker control lean (ZCL) rats. Using male Zucker diabetic fatty (ZDF) rats that exhibit the typical metabolic disorders of human T2DM, such as excessive EGP, hyperglycemia, hyperinsulinemia, and hyperglucagonemia, we examined the ability of hyperglucagonemia to promote greater rates of postabsorptive EGP and hyperglycemia. Euglycemic or hyperglycemic basal insulin (INS-BC) and glucagon (GCG-BC) clamps were performed in the absence or during an acute setting of glucagon deficiency (GCG-DF, ∼10% of basal), either alone or in combination with insulin deficiency (INS-DF, ∼10% of basal). Glucose appearance, disappearance, and cycling rates were measured using [2-
3 H] and [3-3 H]-glucose. In ZCL rats, GCG-DF reduced the levels of hepatic cyclic AMP, EGP, and plasma glucose (PG) by 50%, 32%, and 50%, respectively. EGP fell in the presence GCG-DF and INS-BC, but under GCG-DF and INS-DF, EGP and PG increased two- and threefold, respectively. GCG-DF revealed the hyperglucagonemia present in ZDF rats lacked the ability to regulate hepatic intracellular cyclic AMP levels and glucose flux, since EGP and PG levels fell by only 10%. We conclude that the liver in T2DM suffers from resistance to all three major regulatory factors, glucagon, insulin, and glucose, thus leading to a loss of metabolic flexibility. NEW & NOTEWORTHY In postabsorptive state, basal plasma insulin (P-INS) and plasma glucose (PG) act dominantly to increase hepatic glucose cycling and reduce endogenous glucose production (EGP) and PG in healthy rats, which is only counteracted by the acute action of basal plasma glucagon (P-GCG) to support EGP and euglycemia. Hyperglucagonemia, a hallmark of type 2 diabetes (T2DM) present in Zucker diabetic fatty (ZDF) rats, is not the primary mediator of hyperglycemia and high EGP as commonly thought; instead, the liver is resistant to glucagon as well as insulin and glucose.- Published
- 2024
- Full Text
- View/download PDF
20. Oral Hypofunction and Risk of Weight Change among Independent Older Adults.
- Author
-
Shiota C, Kusama T, Takeuchi K, Kiuchi S, and Osaka K
- Subjects
- Humans, Aged, Follow-Up Studies, Oral Health, Weight Loss, Independent Living, Xerostomia epidemiology, Xerostomia etiology
- Abstract
Oral health is essential for nutritional status; however, little is known about its association with weight change. This study aimed to investigate whether the risk of weight change differs according to the presence of each important component of oral hypofunction (fewer remaining teeth, low chewing efficiency, swallowing problems, and xerostomia) among independent older adults. This was a three-year follow-up cohort study based on self-reported questionnaires. The participants were independent older adults aged ≥65 from the Japan Gerontological Evaluation Study (JAGES). We used >5% weight loss/gain during follow-up as the outcome variables, and the number of remaining teeth (≥20/10-19/0-9), the presence of chewing difficulty, swallowing problems, and xerostomia (yes/no) as the exposure variables. We fitted the Poisson regression model, including possible confounders to estimate the risk ratios (RRs) and 95% confidence intervals (CIs). For weight loss, RRs were significantly higher among those with 0-9 remaining teeth (RR = 1.17; 95% CI = 1.11-1.23), chewing difficulty (RR = 1.12; 95% CI = 1.07-1.16), and xerostomia (RR = 1.11; 95% CI = 1.06-1.16), but there was no significant association with swallowing problems (RR = 1.01; 95% CI = 0.97-1.06). For weight gain, we also found similar associations with oral hypofunction. Oral hypofunction among older adults could have non-negligible health impacts on nutritional status.
- Published
- 2023
- Full Text
- View/download PDF
21. Assessing Kidney Injury Induced by Mercuric Chloride in Guinea Pigs with In Vivo and In Vitro Experiments.
- Author
-
Goel H, Printz RL, Shiota C, Estes SK, Pannala V, AbdulHameed MDM, Shiota M, and Wallqvist A
- Subjects
- Rats, Male, Guinea Pigs, Animals, Kidney metabolism, Kidney Function Tests, Liver metabolism, Mercuric Chloride toxicity, Acute Kidney Injury metabolism
- Abstract
Acute kidney injury, which is associated with high levels of morbidity and mortality, affects a significant number of individuals, and can be triggered by multiple factors, such as medications, exposure to toxic chemicals or other substances, disease, and trauma. Because the kidney is a critical organ, understanding and identifying early cellular or gene-level changes can provide a foundation for designing medical interventions. In our earlier work, we identified gene modules anchored to histopathology phenotypes associated with toxicant-induced liver and kidney injuries. Here, using in vivo and in vitro experiments, we assessed and validated these kidney injury-associated modules by analyzing gene expression data from the kidneys of male Hartley guinea pigs exposed to mercuric chloride. Using plasma creatinine levels and cell-viability assays as measures of the extent of renal dysfunction under in vivo and in vitro conditions, we performed an initial range-finding study to identify the appropriate doses and exposure times associated with mild and severe kidney injuries. We then monitored changes in kidney gene expression at the selected doses and time points post-toxicant exposure to characterize the mechanisms of kidney injury. Our injury module-based analysis revealed a dose-dependent activation of several phenotypic cellular processes associated with dilatation, necrosis, and fibrogenesis that were common across the experimental platforms and indicative of processes that initiate kidney damage. Furthermore, a comparison of activated injury modules between guinea pigs and rats indicated a strong correlation between the modules, highlighting their potential for cross-species translational studies.
- Published
- 2023
- Full Text
- View/download PDF
22. Specific reprogramming of alpha cells to insulin-producing cells by short glucagon promoter-driven Pdx1 and MafA.
- Author
-
Guo P, Zhang T, Lu A, Shiota C, Huard M, Whitney KE, and Huard J
- Abstract
Endogenous reprogramming of pancreas-derived non-beta cells into insulin-producing cells is a promising approach to treat type 1 diabetes (T1D). One strategy that has yet to be explored is the specific delivery of insulin-producing essential genes, Pdx1 and MafA, to pancreatic alpha cells to reprogram the cells into insulin-producing cells in an adult pancreas. In this study, we used an alpha cell-specific glucagon (GCG) promoter to drive Pdx1 and MafA transcription factors to reprogram alpha cells to insulin-producing cells in chemically induced and autoimmune diabetic mice. Our results showed that a combination of a short glucagon-specific promoter with AAV serotype 8 (AAV8) can be used to successfully deliver Pdx1 and MafA to pancreatic alpha cells in the mouse pancreas. Pdx1 and MafA expression specifically in alpha cells were also able to correct hyperglycemia in both induced and autoimmune diabetic mice. With this technology, targeted gene specificity and reprogramming were accomplished with an alpha-specific promotor combined with an AAV-specific serotype and provide an initial basis to develop a novel therapy for the treatment of T1D., Competing Interests: The authors declare no competing interests., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
23. Genomics and metabolomics of early-stage thioacetamide-induced liver injury: An interspecies study between guinea pig and rat.
- Author
-
Schyman P, Printz RL, Pannala VR, AbdulHameed MDM, Estes SK, Shiota C, Boyd KL, Shiota M, and Wallqvist A
- Subjects
- Animals, Biomarkers metabolism, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury pathology, Disease Models, Animal, Gene Regulatory Networks, Guinea Pigs, Liver pathology, Male, Rats, Sprague-Dawley, Species Specificity, Time Factors, Rats, Chemical and Drug Induced Liver Injury genetics, Chemical and Drug Induced Liver Injury metabolism, Gene Expression Profiling, Liver metabolism, Metabolome, Metabolomics, Thioacetamide, Transcriptome
- Abstract
To study the complex processes involved in liver injuries, researchers rely on animal investigations, using chemically or surgically induced liver injuries, to extrapolate findings and infer human health risks. However, this presents obvious challenges in performing a detailed comparison and validation between the highly controlled animal models and development of liver injuries in humans. Furthermore, it is not clear whether there are species-dependent and -independent molecular initiating events or processes that cause liver injury before they eventually lead to end-stage liver disease. Here, we present a side-by-side study of rats and guinea pigs using thioacetamide to examine the similarities between early molecular initiating events during an acute-phase liver injury. We exposed Sprague Dawley rats and Hartley guinea pigs to a single dose of 25 or 100 mg/kg thioacetamide and collected blood plasma for metabolomic analysis and liver tissue for RNA-sequencing. The subsequent toxicogenomic analysis identified consistent liver injury trends in both genomic and metabolomic data within 24 and 33 h after thioacetamide exposure in rats and guinea pigs, respectively. In particular, we found species similarities in the key injury phenotypes of inflammation and fibrogenesis in our gene module analysis for liver injury phenotypes. We identified expression of several common genes (e.g., SPP1, TNSF18, SERPINE1, CLDN4, TIMP1, CD44, and LGALS3), activation of injury-specific KEGG pathways, and alteration of plasma metabolites involved in amino acid and bile acid metabolism as some of the key molecular processes that changed early upon thioacetamide exposure and could play a major role in the initiation of acute liver injury., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Toxicant-Induced Metabolic Alterations in Lipid and Amino Acid Pathways Are Predictive of Acute Liver Toxicity in Rats.
- Author
-
Pannala VR, Estes SK, Rahim M, Trenary I, O'Brien TP, Shiota C, Printz RL, Reifman J, Shiota M, Young JD, and Wallqvist A
- Subjects
- Acetaminophen pharmacology, Acetaminophen toxicity, Acute Disease, Animals, Biomarkers analysis, Biomarkers metabolism, Bromobenzenes pharmacology, Bromobenzenes toxicity, Carbon Tetrachloride pharmacology, Carbon Tetrachloride toxicity, Chemical and Drug Induced Liver Injury genetics, Chemical and Drug Induced Liver Injury metabolism, Chemical and Drug Induced Liver Injury pathology, Gene Expression Profiling, Hazardous Substances toxicity, Hepatocytes drug effects, Hepatocytes metabolism, Hepatocytes pathology, Lipid Metabolism genetics, Liver drug effects, Liver metabolism, Liver pathology, Male, Metabolic Networks and Pathways drug effects, Metabolic Networks and Pathways genetics, Metabolome genetics, Metabolomics, Prognosis, Rats, Rats, Sprague-Dawley, Transcriptome drug effects, Amino Acids metabolism, Chemical and Drug Induced Liver Injury diagnosis, Hazardous Substances pharmacology, Lipid Metabolism drug effects, Metabolome drug effects
- Abstract
Liver disease and disorders associated with aberrant hepatocyte metabolism can be initiated via drug and environmental toxicant exposures. In this study, we tested the hypothesis that gene and metabolic profiling can reveal commonalities in liver response to different toxicants and provide the capability to identify early signatures of acute liver toxicity. We used Sprague Dawley rats and three classical hepatotoxicants: acetaminophen (2 g/kg), bromobenzene (0.4 g/kg), and carbon tetrachloride (0.3 g/kg), to identify early perturbations in liver metabolism after a single acute exposure dose. We measured changes in liver genes and plasma metabolites at two time points (5 and 10 h) and used genome-scale metabolic models to identify commonalities in liver responses across the three toxicants. We found strong correlations for gene and metabolic profiles between the toxicants, indicative of similarities in the liver response to toxicity. We identified several injury-specific pathways in lipid and amino acid metabolism that changed similarly across the three toxicants. Our findings suggest that several plasma metabolites in lipid and amino acid metabolism are strongly associated with the progression of liver toxicity, and as such, could be targeted and clinically assessed for their potential as early predictors of acute liver toxicity.
- Published
- 2020
- Full Text
- View/download PDF
25. A toxicogenomic approach to assess kidney injury induced by mercuric chloride in rats.
- Author
-
Schyman P, Printz RL, AbdulHameed MDM, Estes SK, Shiota C, Shiota M, and Wallqvist A
- Subjects
- Animals, Aspartate Aminotransferases blood, Base Sequence, Biomarkers urine, Body Weight drug effects, Cell Adhesion Molecules metabolism, Cell Adhesion Molecules urine, Gene Expression drug effects, Male, Necrosis, Protein Folding drug effects, Rats, Rats, Sprague-Dawley, Kidney Diseases chemically induced, Kidney Diseases pathology, Mercuric Chloride toxicity, Toxicogenetics methods
- Abstract
Kidney injury caused by disease, trauma, environmental exposures, or drugs may result in decreased renal function, chronic kidney disease, or acute kidney failure. Diagnosis of kidney injury using serum creatinine levels, a common clinical test, only identifies renal dysfunction after the kidneys have undergone severe damage. Other indicators sensitive to kidney injury, such as the level of urine kidney injury molecule-1 (KIM-1), lack the ability to differentiate between injury phenotypes. To address early detection as well as detailed categorization of kidney-injury phenotypes in preclinical animal or cellular studies, we previously identified eight sets (modules) of co-expressed genes uniquely associated with kidney histopathology. Here, we used mercuric chloride (HgCl
2 )-a model nephrotoxicant-to chemically induce kidney injuries as monitored by KIM-1 levels in Sprague Dawley rats at two doses (0.25 or 0.50 mg/kg) and two exposure lengths (10 or 34 h). We collected whole transcriptome RNA-seq data derived from five animals at each dose and time point to perform a toxicogenomics analysis. Consistent with documented injury phenotypes for HgCl2 toxicity, our kidney-injury-module approach identified the onset of necrosis and dilation as early as 10 h after a dose of 0.50 mg/kg that produced only mild injury as judged by urinary KIM-1 excretion. The results of these animal studies highlight the potential of the kidney-injury-module approach to provide a sensitive and histopathology-specific readout of renal toxicity., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
26. Mechanism-based identification of plasma metabolites associated with liver toxicity.
- Author
-
Pannala VR, Estes SK, Rahim M, Trenary I, O'Brien TP, Shiota C, Printz RL, Reifman J, Oyama T, Shiota M, Young JD, and Wallqvist A
- Subjects
- Acetaminophen toxicity, Animals, Biomarkers blood, Biomarkers urine, Bromobenzenes toxicity, Chemical and Drug Induced Liver Injury diagnosis, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury urine, Gene Expression Profiling, Liver drug effects, Liver metabolism, Male, Metabolomics, Rats, Sprague-Dawley, Chemical and Drug Induced Liver Injury blood
- Abstract
Early diagnosis of liver injuries caused by drugs or occupational exposures is necessary to enable effective treatments and prevent liver failure. Whereas histopathology remains the gold standard for assessing hepatotoxicity in animals, plasma aminotransferase levels are the primary measures for monitoring liver dysfunction in humans. In this study, using Sprague Dawley rats, we investigated whether integrated analyses of transcriptomic and metabolomic data with genome-scale metabolic models (GSMs) could identify early indicators of injury and provide new insights into the mechanisms of hepatotoxicity. We obtained concurrent measurements of gene-expression changes in the liver and kidneys, and expression changes along with metabolic profiles in the plasma and urine, from rats 5 or 10 h after exposing them to one of two classical hepatotoxicants, acetaminophen (2 g/kg) or bromobenzene (0.4 g/kg). Global multivariate analyses revealed that gene-expression changes in the liver and metabolic profiles in the plasma and urine of toxicant-treated animals differed from those of controls, even at time points much earlier than changes detected by conventional markers of liver injury. Furthermore, clustering analysis revealed that both the gene-expression changes in the liver and the metabolic profiles in the plasma induced by the two hepatotoxicants were highly correlated, indicating commonalities in the liver toxicity response. Systematic GSM-based analyses yielded metabolites associated with the mechanisms of toxicity and identified several lipid and amino acid metabolism pathways that were activated by both toxicants and those uniquely activated by each. Our findings suggest that several metabolite alterations, which are strongly associated with the mechanisms of toxicity and occur within injury-specific pathways (e.g., of bile acid and fatty acid metabolism), could be targeted and clinically assessed for their potential as early indicators of liver damage., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
27. SMAD7 enhances adult β-cell proliferation without significantly affecting β-cell function in mice.
- Author
-
Sehrawat A, Shiota C, Mohamed N, DiNicola J, Saleh M, Kalsi R, Zhang T, Wang Y, Prasadan K, and Gittes GK
- Subjects
- Animals, Female, Glucose pharmacology, Male, Mice, Mice, Knockout, Signal Transduction, Sweetening Agents pharmacology, Transforming Growth Factor beta genetics, Cell Proliferation, Insulin physiology, Insulin Secretion drug effects, Insulin-Secreting Cells cytology, Insulin-Secreting Cells physiology, Smad7 Protein physiology, Transforming Growth Factor beta metabolism
- Abstract
The interplay between the transforming growth factor β (TGF-β) signaling proteins, SMAD family member 2 (SMAD2) and 3 (SMAD3), and the TGF-β-inhibiting SMAD, SMAD7, seems to play a vital role in proper pancreatic endocrine development and also in normal β-cell function in adult pancreatic islets. Here, we generated conditional SMAD7 knockout mice by crossing insulin1
Cre mice with SMAD7fx/fx mice. We also created a β cell-specific SMAD7-overexpressing mouse line by crossing insulin1Dre mice with HPRT-SMAD7/RosaGFP mice. We analyzed β-cell function in adult islets when SMAD7 was either absent or overexpressed in β cells. Loss of SMAD7 in β cells inhibited proliferation, and SMAD7 overexpression enhanced cell proliferation. However, alterations in basic glucose homeostasis were not detectable following either SMAD7 deletion or overexpression in β cells. Our results show that both the absence and overexpression of SMAD7 affect TGF-β signaling and modulates β-cell proliferation but does not appear to alter β-cell function. Reversible SMAD7 overexpression may represent an attractive therapeutic option to enhance β-cell proliferation without negative effects on β-cell function., (© 2020 Sehrawat et al.)- Published
- 2020
- Full Text
- View/download PDF
28. Cellular Autophagy in α Cells Plays a Role in the Maintenance of Islet Architecture.
- Author
-
Himuro M, Miyatsuka T, Suzuki L, Miura M, Katahira T, Goto H, Nishida Y, Sasaki S, Koike M, Shiota C, Gittes GK, Fujitani Y, and Watada H
- Abstract
Autophagy is known to play a pivotal role in intracellular quality control through the degradation of subcellular damaged organelles and components. Whereas autophagy is essential for maintaining β -cell function in pancreatic islets, it remains unclear as to how the cellular autophagy affects the homeostasis and function of glucagon-secreting α cells. To investigate the role of autophagy in α cells, we generated a mutant mouse model lacking Atg7 , a key molecule for autophagosome formation, specifically in α cells. Histological analysis demonstrated more glucagon-positive cells, with a multilayered structure, in the islets under Atg7 deficiency, although metabolic profiles, such as body weight, blood glucose, and plasma glucagon levels were comparable between Atg7 -deficient mice and control littermates. Consistent with our previous findings that Atg7 deficiency suppressed β -cell proliferation, cellular proliferation was suppressed in Atg7 -deficient α cells. These findings suggest that α -cell autophagy plays a role in maintaining α -cell area and normal islet architecture but appears to be dispensable for metabolic homeostasis., (Copyright © 2019 Endocrine Society.)
- Published
- 2019
- Full Text
- View/download PDF
29. Evidence of a developmental origin for β-cell heterogeneity using a dual lineage-tracing technology.
- Author
-
Chen C, Shiota C, Agostinelli G, Ridley D, Jiang Y, Ma J, Prasadan K, Xiao X, and Gittes GK
- Subjects
- Animals, Cell Differentiation genetics, Cell Separation methods, Cells, Cultured, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Insulin-Secreting Cells metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Molecular Imaging methods, Organogenesis genetics, Pancreas embryology, Pancreas growth & development, Pancreas metabolism, Stem Cells metabolism, Trans-Activators genetics, Trans-Activators metabolism, Transcription Factors genetics, Transcription Factors metabolism, Cell Lineage genetics, Cell Tracking methods, Insulin-Secreting Cells cytology, Pancreas cytology, Stem Cells cytology
- Abstract
The Cre/loxP system has been used extensively in mouse models with a limitation of one lineage at a time. Differences in function and other properties among populations of adult β-cells is termed β-cell heterogeneity, which was recently associated with diabetic phenotypes. Nevertheless, the presence of a developmentally derived β-cell heterogeneity is unclear. Here, we have developed a novel dual lineage-tracing technology, using a combination of two recombinase systems, Dre/RoxP and Cre/LoxP, to independently trace green fluorescent Pdx1-lineage cells and red fluorescent Ptf1a-lineage cells in the developing and adult mouse pancreas. We detected a few Pdx1
+ /Ptf1a- lineage cells in addition to the vast majority of Pdx1+ /Ptf1a+ lineage cells in the pancreas. Moreover, Pdx1+ /Ptf1a+ lineage β-cells had fewer Ki-67+ proliferating β-cells, and expressed higher mRNA levels of insulin, Glut2, Pdx1, MafA and Nkx6.1, but lower CCND1 and CDK4 levels, compared with Pdx1+ /Ptf1a- lineage β-cells. Furthermore, more TSQ-high, SSC-high cells were detected in the Pdx1+ Ptf1a+ lineage population than in the Pdx1+ Ptf1a- lineage population. Together, these data suggest that differential activation of Ptf1a in the developing pancreas may correlate with this β-cell heterogeneity., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2019. Published by The Company of Biologists Ltd.)- Published
- 2019
- Full Text
- View/download PDF
30. Islet α-cell Inflammation Induced By NF-κB inducing kinase (NIK) Leads to Hypoglycemia, Pancreatitis, Growth Retardation, and Postnatal Death in Mice.
- Author
-
Li X, Jia L, Chen X, Dong Y, Ren X, Dong Y, Chen Y, Xie L, Liu M, Shiota C, Gittes GK, Rui L, and Chen Z
- Subjects
- Animals, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Gene Expression Profiling, Glucagon-Secreting Cells drug effects, Growth Disorders complications, Growth Disorders pathology, Hypoglycemia complications, Hypoglycemia pathology, Immunohistochemistry, Mice, Pancreatitis complications, Pancreatitis pathology, Real-Time Polymerase Chain Reaction, NF-kappaB-Inducing Kinase, Death, Glucagon-Secreting Cells pathology, Growth Disorders physiopathology, Hypoglycemia physiopathology, Insulin-Secreting Cells pathology, Pancreatitis physiopathology, Protein Serine-Threonine Kinases metabolism
- Abstract
Islet α-cell dysfunction has been shown to contribute to type 2 diabetes; however, whether islet α-cell inflammation is involved in the occurrence of pancreatitis is largely unknown. The aims of this study were to investigate how NF-κB inducing kinase (NIK) regulates pancreatic α-cell function, both in vitro and in vivo , and to assess how islet α-cell inflammation induced by NIK affects the development of pancreatitis. Methods: We utilized adenovirus-mediated NIK overexpression, ELISA, qPCR, RNA-seq, and Western blot analyses to study the role of NIK in islet α cells in vitro . Islet α-cell-specific NIK overexpressing (α-NIK-OE) mice were generated, and pancreatic α/β-cell function and the occurrence of pancreatitis in these mice were assessed via ELISA, qPCR, and immunohistochemical analyses. Results: The LTβR/noncanonical NF-κB signaling pathway is present in islet α cells. Overexpression of NIK in αTC1-6 cells induces inflammation and cell death, contributing to a decrease in the expression and secretion of glucagon. Additionally, α-cell specific overexpression of NIK (α-NIK-OE) results in α-cell death, lower serum glucagon levels, and hypoglycemia in mice. Strikingly, α-NIK-OE mice also display a reduced β-cell mass, growth retardation, pancreatitis, and postnatal death. Conclusions: Islet α-cell specific overexpression of NIK results in islet α-cell dysfunction and causes islet β-cell death and pancreatitis, which are most likely due to paracrine secretion of cytokines and chemokines from islet α cells, thus leading to hypoglycemia, growth retardation, and postnatal death in mice., Competing Interests: Competing Interests: The authors have declared that no competing interest exists.
- Published
- 2018
- Full Text
- View/download PDF
31. Endogenous Reprogramming of Alpha Cells into Beta Cells, Induced by Viral Gene Therapy, Reverses Autoimmune Diabetes.
- Author
-
Xiao X, Guo P, Shiota C, Zhang T, Coudriet GM, Fischbach S, Prasadan K, Fusco J, Ramachandran S, Witkowski P, Piganelli JD, and Gittes GK
- Subjects
- Alloxan, Animals, Blood Glucose, Dependovirus metabolism, Gene Expression Profiling, Glucagon metabolism, Glucagon-Secreting Cells metabolism, Homeodomain Proteins metabolism, Humans, Hyperglycemia complications, Hyperglycemia pathology, Insulin metabolism, Insulin-Secreting Cells metabolism, Lectins, C-Type, Mice, Inbred C57BL, Mice, SCID, Receptors, Immunologic metabolism, Trans-Activators metabolism, Cellular Reprogramming, Diabetes Mellitus, Experimental pathology, Diabetes Mellitus, Experimental therapy, Diabetes Mellitus, Type 1 pathology, Diabetes Mellitus, Type 1 therapy, Genetic Therapy, Glucagon-Secreting Cells pathology, Insulin-Secreting Cells pathology
- Abstract
Successful strategies for treating type 1 diabetes need to restore the function of pancreatic beta cells that are destroyed by the immune system and overcome further destruction of insulin-producing cells. Here, we infused adeno-associated virus carrying Pdx1 and MafA expression cassettes through the pancreatic duct to reprogram alpha cells into functional beta cells and normalized blood glucose in both beta cell-toxin-induced diabetic mice and in autoimmune non-obese diabetic (NOD) mice. The euglycemia in toxin-induced diabetic mice and new insulin
+ cells persisted in the autoimmune NOD mice for 4 months prior to reestablishment of autoimmune diabetes. This gene therapy strategy also induced alpha to beta cell conversion in toxin-treated human islets, which restored blood glucose levels in NOD/SCID mice upon transplantation. Hence, this strategy could represent a new therapeutic approach, perhaps complemented by immunosuppression, to bolster endogenous insulin production. Our study thus provides a potential basis for further investigation in human type 1 diabetes., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
32. Gcg CreERT2 knockin mice as a tool for genetic manipulation in pancreatic alpha cells.
- Author
-
Shiota C, Prasadan K, Guo P, Fusco J, Xiao X, and Gittes GK
- Subjects
- Animals, Female, Glucagon blood, Glucagon-Like Peptide 1 blood, Immunohistochemistry, Male, Mice, Mice, Transgenic, Proglucagon genetics, Tamoxifen pharmacology, Glucagon-Secreting Cells metabolism, Proglucagon metabolism
- Abstract
Aims/hypothesis: The Cre/loxP system, which enables tissue-specific manipulation of genes, is widely used in mice for diabetes research. Our aim was to develop a new Cre-driver mouse line for the specific and efficient manipulation of genes in pancreatic alpha cells., Methods: A Gcg
CreERT2 knockin mouse, which expresses a tamoxifen-inducible form of Cre from the endogenous preproglucagon (Gcg) gene locus, was generated by homologous recombination. The new GcgCreERT2 mouse line was crossed to the Rosa26tdTomato (R26tdTomato ) Cre reporter mouse line in order to evaluate the tissue specificity, efficiency and tamoxifen dependency of GcgCreERT2 -mediated recombination. Cell types of pancreatic islets were identified using immunohistochemistry. Biochemical and physiological data, including blood glucose levels, plasma glucagon and glucagon-like peptide (GLP)-1 levels, and pancreatic glucagon content, were collected and used to assess the overall effect of Gcg gene targeting on GcgCreERT2/w heterozygous mice., Results: Tamoxifen-treated GcgCreERT2/w ;R26tdTomato/w mice displayed Cre reporter activity, i.e. expression of tdTomato red fluorescent protein (RFP) in all known cells that produce proglucagon-derived peptides. In the adult pancreas, RFP was detected in 94-97% of alpha cells, whereas it was detected in a negligible (~ 0.2%) proportion of beta cells. While more than 98% of cells labelled with tamoxifen-induced RFP were glucagon-positive cells, 14-25% of pancreatic polypeptide (PP)-positive cells were also positive for RFP, indicating the presence of glucagon/PP bihormonal cell population. Tamoxifen-independent expression of RFP occurred in approximately 6% of alpha cells. In contrast to alpha cells and GLP-1-producing neurons, in which RFP expression persisted for at least 5 months after tamoxifen administration (presumably due to rare neogenesis in these cell types in adulthood), nearly half of RFP-positive intestinal L cells were replaced with RFP-negative L cells over the first 2 weeks after tamoxifen administration. Heterozygous GcgCreERT2/w mice showed reduced Gcg mRNA levels in islets, but maintained normal levels of pancreatic and plasma glucagon. The mice did not exhibit any detectable baseline physiological abnormalities, at least in young adulthood., Conclusions/interpretation: The newly developed GcgCreERT2 knockin mouse shows faithful expression of CreERT2 in pancreatic alpha cells, intestinal L cells and GLP-1-producing neurons. This mouse line will be particularly useful for manipulating genes in alpha cells, due to highly specific and efficient CreERT2 -mediated recombination in this cell type in the pancreas.- Published
- 2017
- Full Text
- View/download PDF
33. SMAD3/Stat3 Signaling Mediates β-Cell Epithelial-Mesenchymal Transition in Chronic Pancreatitis-Related Diabetes.
- Author
-
Xiao X, Fischbach S, Zhang T, Chen C, Sheng Q, Zimmerman R, Patnaik S, Fusco J, Ming Y, Guo P, Shiota C, Prasadan K, Gangopadhyay N, Husain SZ, Dong H, and Gittes GK
- Subjects
- Animals, Apoptosis physiology, Diabetes Mellitus, Type 1 metabolism, Diabetes Mellitus, Type 1 pathology, Epithelial-Mesenchymal Transition drug effects, Epithelial-Mesenchymal Transition physiology, Insulin-Secreting Cells drug effects, Insulin-Secreting Cells metabolism, Male, Mice, Mice, Inbred C57BL, Pancreatitis, Chronic pathology, STAT3 Transcription Factor genetics, Signal Transduction drug effects, Signal Transduction genetics, Smad3 Protein genetics, Transforming Growth Factor beta1 pharmacology, Pancreatitis, Chronic metabolism, STAT3 Transcription Factor metabolism, Smad3 Protein metabolism
- Abstract
Many patients with chronic pancreatitis develop diabetes (chronic pancreatitis-related diabetes [CPRD]) through an undetermined mechanism. Here we used long-term partial pancreatic duct ligation (PDL) as a model to study CPRD. We found that long-term PDL induced significant β-cell dedifferentiation, followed by a time-dependent decrease in functional β-cell mass-all specifically in the ligated tail portion of the pancreas (PDL-tail). High levels of transforming growth factor β1 (TGFβ1) were detected in the PDL-tail and were mainly produced by M2 macrophages at the early stage and by activated myofibroblasts at the later stage. Loss of β-cell mass was then found to result from TGFβ1-triggered epithelial-mesenchymal transition (EMT) by β-cells, rather than resulting directly from β-cell apoptosis. Mechanistically, TGFβ1-treated β-cells activated expression of the EMT regulator gene Snail in a SMAD3/Stat3-dependent manner. Moreover, forced expression of forkhead box protein O1 (FoxO1), an antagonist for activated Stat3, specifically in β-cells ameliorated β-cell EMT and β-cell loss and prevented the onset of diabetes in mice undergoing PDL. Together, our data suggest that chronic pancreatitis may trigger TGFβ1-mediated β-cell EMT to lead to CPRD, which could substantially be prevented by sustained expression of FoxO1 in β-cells., (© 2017 by the American Diabetes Association.)
- Published
- 2017
- Full Text
- View/download PDF
34. Forkhead Box Protein 1 (FoxO1) Inhibits Accelerated β Cell Aging in Pancreas-specific SMAD7 Mutant Mice.
- Author
-
Xiao X, Chen C, Guo P, Zhang T, Fischbach S, Fusco J, Shiota C, Prasadan K, Dong H, and Gittes GK
- Subjects
- Animals, Cell Proliferation, Cellular Senescence, Diabetes Mellitus genetics, Diabetes Mellitus pathology, Forkhead Box Protein O1 genetics, Insulin-Secreting Cells metabolism, Male, Mice, Mice, Knockout, Mice, SCID, Mutation, Pancreas metabolism, Pancreas pathology, Protein Transport, Smad7 Protein genetics, Diabetes Mellitus metabolism, Forkhead Box Protein O1 metabolism, Insulin-Secreting Cells pathology, Smad7 Protein metabolism
- Abstract
The mechanisms underlying the effects of exocrine dysfunction on the development of diabetes remain largely unknown. Here we show that pancreatic depletion of SMAD7 resulted in age-dependent increases in β cell dysfunction with accelerated glucose intolerance, followed by overt diabetes. The accelerated β cell dysfunction and loss of proliferation capacity, two features of β cell aging, appeared to be non-cell-autonomous, secondary to the adjacent exocrine failure as a "bystander effect." Increased Forkhead box protein 1 (FoxO1) acetylation and nuclear retention was followed by progressive FoxO1 loss in β cells that marked the onset of diabetes. Moreover, forced FoxO1 expression in β cells prevented β cell dysfunction and loss in this model. Thus, we present a model of accelerated β cell aging that may be useful for studying the mechanisms underlying β cell failure in diabetes. Moreover, we provide evidence highlighting a critical role of FoxO1 in maintaining β cell identity in the context of SMAD7 failure., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
35. Epidermal Growth Factor Receptor Signaling Regulates β Cell Proliferation in Adult Mice.
- Author
-
Song Z, Fusco J, Zimmerman R, Fischbach S, Chen C, Ricks DM, Prasadan K, Shiota C, Xiao X, and Gittes GK
- Subjects
- Animals, Cyclin D1 genetics, Cyclin D1 metabolism, ErbB Receptors genetics, Mice, Mice, Transgenic, Cell Proliferation physiology, ErbB Receptors metabolism, Insulin-Secreting Cells metabolism, Signal Transduction physiology
- Abstract
A thorough understanding of the signaling pathways involved in the regulation of β cell proliferation is an important initial step in restoring β cell mass in the diabetic patient. Here, we show that epidermal growth factor receptor 1 (EGFR) was significantly up-regulated in the islets of C57BL/6 mice after 50% partial pancreatectomy (PPx), a model for workload-induced β cell proliferation. Specific deletion of EGFR in the β cells of adult mice impaired β cell proliferation at baseline and after 50% PPx, suggesting that the EGFR signaling pathway plays an essential role in adult β cell proliferation. Further analyses showed that β cell-specific depletion of EGFR resulted in impaired expression of cyclin D1 and impaired suppression of p27 after PPx, both of which enhance β cell proliferation. These data highlight the importance of EGFR signaling and its downstream signaling cascade in postnatal β cell growth., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
36. A synopsis of factors regulating beta cell development and beta cell mass.
- Author
-
Prasadan K, Shiota C, Xiangwei X, Ricks D, Fusco J, and Gittes G
- Subjects
- Animals, Humans, Intercellular Signaling Peptides and Proteins metabolism, Organ Size, Signal Transduction, Transcription Factors metabolism, Transforming Growth Factor beta metabolism, Insulin-Secreting Cells cytology, Insulin-Secreting Cells metabolism
- Abstract
The insulin-secreting beta cells in the endocrine pancreas regulate blood glucose levels, and loss of functional beta cells leads to insulin deficiency, hyperglycemia (high blood glucose) and diabetes mellitus. Current treatment strategies for type-1 (autoimmune) diabetes are islet transplantation, which has significant risks and limitations, or normalization of blood glucose with insulin injections, which is clearly not ideal. The type-1 patients can lack insulin counter-regulatory mechanism; therefore, hypoglycemia is a potential risk. Hence, a cell-based therapy offers a better alternative for the treatment of diabetes. Past research was focused on attempting to generate replacement beta cells from stem cells; however, recently there has been an increasing interest in identifying mechanisms that will lead to the conversion of pre-existing differentiated endocrine cells into beta cells. The goal of this review is to provide an overview of several of the key factors that regulate new beta cell formation (neogenesis) and beta cell proliferation.
- Published
- 2016
- Full Text
- View/download PDF
37. Unilateral mandibular condylar osteochondroma treated with ipsilateral condylectomy and contralateral ramus osteotomy.
- Author
-
Tanaka E, Shiota C, Sato M, Fujihara S, Kondoh T, and Kuroda S
- Subjects
- Adult, Female, Humans, Mandibular Neoplasms pathology, Osteochondroma pathology, Mandibular Condyle surgery, Mandibular Neoplasms surgery, Osteochondroma surgery, Osteotomy, Sagittal Split Ramus
- Abstract
We successfully treated a 32-year-old woman who had facial asymmetry and unilateral mandibular condylar osteochondroma using ipsilateral mandibular condylectomy and contralateral ramus osteotomy. Mirror image analysis with a noncontact 3-dimensional image scanner showed that the soft tissue on the deviated side was protruded more than 5.50 mm compared with the nondeviated side. The patient was diagnosed as having facial asymmetry with a skeletal Class III jaw-base relationship caused by unilateral mandibular condylar osteochondroma. After 18 months of preoperative orthodontic treatment, an ipsilateral condylectomy and a contralateral sagittal split ramus osteotomy were performed. As the result of postoperative orthodontic treatment for 20 months, an ideal occlusion with a Class I molar relationship and an adequate interincisal relationship was achieved. Facial asymmetry and mandibular protrusion were dramatically improved, and the total differences between the deviated and nondeviated sides were decreased to less than 1.11 mm. The acceptable occlusion and the symmetric face were maintained throughout the 1-year retention period. Our results indicated stability after condylectomy without condylar reconstruction in a patient with unilateral condylar osteochondroma., (Copyright © 2016 American Association of Orthodontists. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
38. Transient Suppression of TGFβ Receptor Signaling Facilitates Human Islet Transplantation.
- Author
-
Xiao X, Fischbach S, Song Z, Gaffar I, Zimmerman R, Wiersch J, Prasadan K, Shiota C, Guo P, Ramachandran S, Witkowski P, and Gittes GK
- Subjects
- Animals, Benzamides pharmacology, Blood Glucose metabolism, Blotting, Western, C-Peptide metabolism, Cell Proliferation drug effects, Cells, Cultured, Dioxoles pharmacology, Female, Humans, Insulin blood, Insulin metabolism, Insulin-Secreting Cells drug effects, Insulin-Secreting Cells metabolism, Islets of Langerhans drug effects, Mice, Inbred NOD, Mice, SCID, Microscopy, Confocal, Protein Serine-Threonine Kinases antagonists & inhibitors, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta antagonists & inhibitors, Signal Transduction drug effects, Transplantation, Heterologous, Islets of Langerhans metabolism, Islets of Langerhans Transplantation methods, Protein Serine-Threonine Kinases metabolism, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction physiology
- Abstract
Although islet transplantation is an effective treatment for severe diabetes, its broad application is greatly limited due to a shortage of donor islets. Suppression of TGFβ receptor signaling in β-cells has been shown to increase β-cell proliferation in mice, but has not been rigorously examined in humans. Here, treatment of human islets with a TGFβ receptor I inhibitor, SB-431542 (SB), significantly improved C-peptide secretion by β-cells, and significantly increased β-cell number by increasing β-cell proliferation. In addition, SB increased cell-cycle activators and decreased cell-cycle suppressors in human β-cells. Transplantation of SB-treated human islets into diabetic immune-deficient mice resulted in significant improvement in blood glucose control, significantly higher serum and graft insulin content, and significantly greater increases in β-cell proliferation in the graft, compared with controls. Thus, our data suggest that transient suppression of TGFβ receptor signaling may improve the outcome of human islet transplantation, seemingly through increasing β-cell number and function.
- Published
- 2016
- Full Text
- View/download PDF
39. PNA lectin for purifying mouse acinar cells from the inflamed pancreas.
- Author
-
Xiao X, Fischbach S, Fusco J, Zimmerman R, Song Z, Nebres P, Ricks DM, Prasadan K, Shiota C, Husain SZ, and Gittes GK
- Subjects
- Acinar Cells metabolism, Animals, Flow Cytometry methods, Mice, Pancreas pathology, Acinar Cells cytology, Cell Separation methods, Pancreas cytology, Pancreatitis pathology, Peanut Agglutinin metabolism
- Abstract
Better methods for purifying human or mouse acinar cells without the need for genetic modification are needed. Such techniques would be advantageous for the specific study of certain mechanisms, such as acinar-to-beta-cell reprogramming and pancreatitis. Ulex Europaeus Agglutinin I (UEA-I) lectin has been used to label and isolate acinar cells from the pancreas. However, the purity of the UEA-I-positive cell fraction has not been fully evaluated. Here, we screened 20 widely used lectins for their binding specificity for major pancreatic cell types, and found that UEA-I and Peanut agglutinin (PNA) have a specific affinity for acinar cells in the mouse pancreas, with minimal affinity for other major pancreatic cell types including endocrine cells, duct cells and endothelial cells. Moreover, PNA-purified acinar cells were less contaminated with mesenchymal and inflammatory cells, compared to UEA-I purified acinar cells. Thus, UEA-I and PNA appear to be excellent lectins for pancreatic acinar cell purification. PNA may be a better choice in situations where mesenchymal cells or inflammatory cells are significantly increased in the pancreas, such as type 1 diabetes, pancreatitis and pancreatic cancer.
- Published
- 2016
- Full Text
- View/download PDF
40. Intraislet Pancreatic Ducts Can Give Rise to Insulin-Positive Cells.
- Author
-
El-Gohary Y, Wiersch J, Tulachan S, Xiao X, Guo P, Rymer C, Fischbach S, Prasadan K, Shiota C, Gaffar I, Song Z, Galambos C, Esni F, and Gittes GK
- Subjects
- Adolescent, Age Factors, Animals, Cadaver, Child, Preschool, Female, Humans, Infant, Insulin-Secreting Cells physiology, Islets of Langerhans growth & development, Islets of Langerhans physiology, Luminescent Proteins genetics, Luminescent Proteins metabolism, Male, Mice, Mutant Strains, Mice, Transgenic, Mutant Proteins genetics, Mutant Proteins metabolism, Pancreatectomy, Pancreatic Ducts growth & development, Pancreatic Ducts physiology, Protein Serine-Threonine Kinases genetics, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta genetics, Regeneration, Red Fluorescent Protein, Cell Transdifferentiation, Insulin biosynthesis, Insulin-Secreting Cells cytology, Islets of Langerhans cytology, Pancreatic Ducts cytology, Protein Serine-Threonine Kinases metabolism, Receptors, Transforming Growth Factor beta metabolism
- Abstract
A key question in diabetes research is whether new β-cells can be derived from endogenous, nonendocrine cells. The potential for pancreatic ductal cells to convert into β-cells is a highly debated issue. To date, it remains unclear what anatomical process would result in duct-derived cells coming to exist within preexisting islets. We used a whole-mount technique to directly visualize the pancreatic ductal network in young wild-type mice, young humans, and wild-type and transgenic mice after partial pancreatectomy. Pancreatic ductal networks, originating from the main ductal tree, were found to reside deep within islets in young mice and humans but not in mature mice or humans. These networks were also not present in normal adult mice after partial pancreatectomy, but TGF-β receptor mutant mice demonstrated formation of these intraislet duct structures after partial pancreatectomy. Genetic and viral lineage tracings were used to determine whether endocrine cells were derived from pancreatic ducts. Lineage tracing confirmed that pancreatic ductal cells can typically convert into new β-cells in normal young developing mice as well as in adult TGF-β signaling mutant mice after partial pancreatectomy. Here the direct visual evidence of ducts growing into islets, along with lineage tracing, not only represents strong evidence for duct cells giving rise to β-cells in the postnatal pancreas but also importantly implicates TGF-β signaling in this process.
- Published
- 2016
- Full Text
- View/download PDF
41. Prevention of skeletal muscle atrophy in vitro using anti-ubiquitination oligopeptide carried by atelocollagen.
- Author
-
Kawai N, Hirasaka K, Maeda T, Haruna M, Shiota C, Ochi A, Abe T, Kohno S, Ohno A, Teshima-Kondo S, Mori H, Tanaka E, and Nikawa T
- Subjects
- Animals, Culture Media, Serum-Free, Insulin Receptor Substrate Proteins metabolism, Insulin-Like Growth Factor I metabolism, Mice, Muscle Fibers, Skeletal drug effects, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal drug effects, Proteolysis drug effects, Proto-Oncogene Proteins c-cbl, Rats, Signal Transduction drug effects, Collagen pharmacology, Muscle, Skeletal pathology, Muscular Atrophy prevention & control, Oligopeptides pharmacology, Ubiquitination drug effects
- Abstract
Skeletal muscle atrophy occurs when the rate of protein degradation exceeds that of protein synthesis in various catabolic conditions, such as fasting, disuse, aging, and chronic diseases. Insulin-like growth factor-1 (IGF-1) signaling stimulates muscle growth and suppresses muscle protein breakdown. In atrophied muscles, ubiquitin ligase, Cbl-b, increases and stimulates the ubiquitination and degradation of IRS-1, an intermediate in IGF-1 signaling pathway, resulting in IGF-1 resistance. In this study, we evaluated the efficacy of atelocollagen (ATCOL)-transported anti-ubiquitination oligopeptide (Cblin: Cbl-b inhibitor) (consisting of tyrosine phosphorylation domain of IRS-1) in starved C2C12 myotubes. The amount of IRS-1 protein was lower in starved versus unstarved myotubes. The Cblin-ATCOL complex inhibited IRS-1 degradation in a concentration-dependent manner. Myotubes incubated with Cblin-ATCOL complex showed significant resistance to starvation-induced atrophy (p<0.01). Furthermore, the Cblin-ATCOL complex significantly inhibited any decrease in Akt phosphorylation (p<0.01) and localization of FOXO3a to the nucleus in starved myotubes. These results suggest that Cblin prevented starvation-induced C2C12 myotube atrophy by maintaining the IGF-1/Akt/FOXO signaling. Therefore, attachment of anti-ubiquitination oligopeptide, Cblin, to ATCOL enhances its delivery to myotubes and could be a potentially effective strategy in the treatment of atrophic myopathies., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
42. Flavones Inhibit LPS-Induced Atrogin-1/MAFbx Expression in Mouse C2C12 Skeletal Myotubes.
- Author
-
Shiota C, Abe T, Kawai N, Ohno A, Teshima-Kondo S, Mori H, Terao J, Tanaka E, and Nikawa T
- Subjects
- Animals, Apigenin therapeutic use, Cell Line, Flavones pharmacology, Flavones therapeutic use, Inflammation metabolism, Inflammation prevention & control, Luteolin therapeutic use, MAP Kinase Signaling System, Mice, Muscle Fibers, Skeletal drug effects, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal cytology, Muscle, Skeletal metabolism, Muscular Atrophy chemically induced, Muscular Atrophy prevention & control, Phosphorylation, Phytotherapy, Plant Extracts therapeutic use, Signal Transduction, Apigenin pharmacology, Luteolin pharmacology, Muscle Proteins metabolism, Muscle, Skeletal drug effects, Muscular Atrophy metabolism, Plant Extracts pharmacology, SKP Cullin F-Box Protein Ligases metabolism
- Abstract
Muscle atrophy is a complex process that occurs as a consequence of various stress events. Muscle atrophy-associated genes (atrogenes) such as atrogin-1/MAFbx and MuRF-1 are induced early in the atrophy process, and the increase in their expression precedes the loss of muscle weight. Although antioxidative nutrients suppress atrogene expression in skeletal muscle cells, the inhibitory effects of flavonoids on inflammation-induced atrogin-1/MAFbx expression have not been clarified. Here, we investigated the inhibitory effects of flavonoids on lipopolysaccharide (LPS)-induced atrogin-1/MAFbx expression. We examined whether nine flavonoids belonging to six flavonoid categories inhibited atrogin-1/MAFbx expression in mouse C2C12 myotubes. Two major flavones, apigenin and luteolin, displayed potent inhibitory effects on atrogin-1/MAFbx expression. The pretreatment with apigenin and luteolin significantly prevented the decrease in C2C12 myotube diameter caused by LPS stimulation. Importantly, the pretreatment of LPS-stimulated myoblasts with these flavones significantly inhibited LPS-induced JNK phosphorylation in C2C12 myotubes, resulting in the significant suppression of atrogin-1/MAFbx promoter activity. These results suggest that apigenin and luteolin, prevent LPS-mediated atrogin-1/MAFbx expression through the inhibition of the JNK signaling pathway in C2C12 myotubes. Thus, these flavones, apigenin and luteolin, may be promising agents to prevent LPS-induced muscle atrophy.
- Published
- 2015
- Full Text
- View/download PDF
43. Pancreatic cell tracing, lineage tagging and targeted genetic manipulations in multiple cell types using pancreatic ductal infusion of adeno-associated viral vectors and/or cell-tagging dyes.
- Author
-
Xiao X, Guo P, Prasadan K, Shiota C, Peirish L, Fischbach S, Song Z, Gaffar I, Wiersch J, El-Gohary Y, Husain SZ, and Gittes GK
- Subjects
- Animals, Cell Lineage, Dimethylamines administration & dosage, Female, Genetic Techniques, Laparotomy, Male, Mice, Inbred C57BL, Mice, Transgenic, Pancreatic Ducts physiology, Promoter Regions, Genetic, RNA, Small Interfering, Transduction, Genetic, Transgenes, Coloring Agents administration & dosage, Dependovirus genetics, Genetic Vectors, Pancreas cytology, Pancreatic Ducts surgery
- Abstract
Genetic manipulations, with or without lineage tracing for specific pancreatic cell types, are very powerful tools for studying diabetes, pancreatitis and pancreatic cancer. Nevertheless, the use of Cre/loxP systems to conditionally activate or inactivate the expression of genes in a cell type- and/or temporal-specific manner is not applicable to cell tracing and/or gene manipulations in more than one lineage at a time. Here we report a technique that allows efficient delivery of dyes for cell tagging into the mouse pancreas through the duct system, and that also delivers viruses carrying transgenes or siRNA under a specific promoter. When this technique is applied in genetically modified mice, it enables the investigator to perform either double lineage tracing or cell lineage tracing combined with gene manipulation in a second lineage. The technique requires <40 min.
- Published
- 2014
- Full Text
- View/download PDF
44. Barrier function of the coelomic epithelium in the developing pancreas.
- Author
-
Guo P, Preuett B, Krishna P, Xiao X, Shiota C, Wiersch J, Gaffar I, Tulachan S, El-Gohary Y, Song Z, and Gittes G
- Subjects
- Activin Receptors metabolism, Animals, Epithelium embryology, Mesoderm embryology, Mice, Organ Culture Techniques, Signal Transduction physiology, Organogenesis physiology, Pancreas embryology
- Abstract
Tight spatial regulation of extracellular morphogen signaling within the close confines of a developing embryo is critical for proper organogenesis. Given the complexity of extracellular signaling in developing organs, together with the proximity of adjacent organs that use disparate signaling pathways, we postulated that a physical barrier to signaling may exist between organs in the embryo. Here we describe a previously unrecognized role for the embryonic coelomic epithelium in providing a physical barrier to contain morphogenic signaling in the developing mouse pancreas. This layer of cells appears to function both to contain key factors required for pancreatic epithelial differentiation, and to prevent fusion of adjacent organs during critical developmental windows. During early foregut development, this barrier appears to play a role in preventing splenic anlage-derived activin signaling from inducing intestinalization of the pancreas-specified epithelium., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
45. Pancreatic duct cells as a source of VEGF in mice.
- Author
-
Xiao X, Prasadan K, Guo P, El-Gohary Y, Fischbach S, Wiersch J, Gaffar I, Shiota C, and Gittes GK
- Subjects
- Animals, Epithelial Cells cytology, Gene Knockdown Techniques, Human Umbilical Vein Endothelial Cells, Humans, Islets of Langerhans cytology, Islets of Langerhans Transplantation, Mice, Neovascularization, Physiologic, RNA, Small Interfering metabolism, SOX9 Transcription Factor genetics, Vascular Endothelial Growth Factor A genetics, Insulin-Secreting Cells metabolism, Pancreatic Ducts metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Aims/hypothesis: Vascular endothelial growth factor (VEGF) is essential for proper pancreatic development, islet vascularisation and insulin secretion. In the adult pancreas, VEGF is thought to be predominantly secreted by beta cells. Although human duct cells have previously been shown to secrete VEGF at angiogenic levels in culture, an analysis of the kinetics of VEGF synthesis and secretion, as well as elucidation of an in vivo role for this ductal VEGF in affecting islet function and physiology, has been lacking., Methods: We analysed purified duct cells independently prepared by flow cytometry, surgical isolation or laser-capture microdissection. We infected duct cells in vivo with Vegf (also known as Vegfa) short hairpin RNA (shRNA) in an intrapancreatic ductal infusion system and examined the effect of VEGF knockdown in duct cells in vitro and in vivo., Results: Pancreatic duct cells express high levels of Vegf mRNA. Compared with beta cells, duct cells had a much higher ratio of secreted to intracellular VEGF. As a bioassay, formation of tubular structures by human umbilical vein endothelial cells was essentially undetectable when cultured alone and was substantially increased when co-cultured with pancreatic duct cells but significantly reduced when co-cultured with duct cells pretreated with Vegf shRNA. Compared with islets transplanted alone, improved vascularisation and function was detected in the islets co-transplanted with duct cells but not in islets co-transplanted with duct cells pretreated with Vegf shRNA., Conclusions/interpretation: Human islet preparations for transplantation typically contain some contaminating duct cells and our findings suggest that the presence of duct cells in the islet preparation may improve transplantation outcomes.
- Published
- 2014
- Full Text
- View/download PDF
46. Toll-like receptor 4-mediated endoplasmic reticulum stress in intestinal crypts induces necrotizing enterocolitis.
- Author
-
Afrazi A, Branca MF, Sodhi CP, Good M, Yamaguchi Y, Egan CE, Lu P, Jia H, Shaffiey S, Lin J, Ma C, Vincent G, Prindle T Jr, Weyandt S, Neal MD, Ozolek JA, Wiersch J, Tschurtschenthaler M, Shiota C, Gittes GK, Billiar TR, Mollen K, Kaser A, Blumberg R, and Hackam DJ
- Subjects
- Activating Transcription Factor 6 genetics, Activating Transcription Factor 6 metabolism, Animals, Apoptosis genetics, Enterocolitis, Necrotizing genetics, Enterocolitis, Necrotizing pathology, HEK293 Cells, Humans, Intestinal Mucosa pathology, Mice, Mice, Knockout, Stem Cells pathology, Toll-Like Receptor 4 genetics, Transcription Factor CHOP genetics, Transcription Factor CHOP metabolism, eIF-2 Kinase genetics, eIF-2 Kinase metabolism, Endoplasmic Reticulum Stress, Enterocolitis, Necrotizing metabolism, Intestinal Mucosa metabolism, Stem Cells metabolism, Toll-Like Receptor 4 metabolism
- Abstract
The cellular cues that regulate the apoptosis of intestinal stem cells (ISCs) remain incompletely understood, yet may play a role in diseases characterized by ISC loss including necrotizing enterocolitis (NEC). Toll-like receptor-4 (TLR4) was recently found to be expressed on ISCs, where its activation leads to ISC apoptosis through mechanisms that remain incompletely explained. We now hypothesize that TLR4 induces endoplasmic reticulum (ER) stress within ISCs, leading to their apoptosis in NEC pathogenesis, and that high ER stress within the premature intestine predisposes to NEC development. Using transgenic mice and cultured enteroids, we now demonstrate that TLR4 induces ER stress within Lgr5 (leucine-rich repeat-containing G-protein-coupled receptor 5)-positive ISCs, resulting in crypt apoptosis. TLR4 signaling within crypts was required, because crypt ER stress and apoptosis occurred in TLR4(ΔIEC-OVER) mice expressing TLR4 only within intestinal crypts and epithelium, but not TLR4(ΔIEC) mice lacking intestinal TLR4. TLR4-mediated ER stress and apoptosis of ISCs required PERK (protein kinase-related PKR-like ER kinase), CHOP (C/EBP homologous protein), and MyD88 (myeloid differentiation primary response gene 88), but not ATF6 (activating transcription factor 6) or XBP1 (X-box-binding protein 1). Human and mouse NEC showed high crypt ER stress and apoptosis, whereas genetic inhibition of PERK or CHOP attenuated ER stress, crypt apoptosis, and NEC severity. Strikingly, using intragastric delivery into fetal mouse intestine, prevention of ER stress reduced TLR4-mediated ISC apoptosis and mucosal disruption. These findings identify a novel link between TLR4-induced ER stress and ISC apoptosis in NEC pathogenesis and suggest that increased ER stress within the premature bowel predisposes to NEC development.
- Published
- 2014
- Full Text
- View/download PDF
47. M2 macrophages promote beta-cell proliferation by up-regulation of SMAD7.
- Author
-
Xiao X, Gaffar I, Guo P, Wiersch J, Fischbach S, Peirish L, Song Z, El-Gohary Y, Prasadan K, Shiota C, and Gittes GK
- Subjects
- Animals, Cell Movement, Cell Nucleus metabolism, Cell Proliferation, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Disease Models, Animal, ErbB Receptors metabolism, Inflammation metabolism, Inflammation pathology, Ligation, Mice, Mice, Inbred C57BL, Models, Biological, Signal Transduction, Transforming Growth Factor beta metabolism, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells pathology, Macrophages metabolism, Smad7 Protein metabolism, Up-Regulation
- Abstract
Determination of signaling pathways that regulate beta-cell replication is critical for beta-cell therapy. Here, we show that blocking pancreatic macrophage infiltration after pancreatic duct ligation (PDL) completely inhibits beta-cell proliferation. The TGFβ superfamily signaling inhibitor SMAD7 was significantly up-regulated in beta cells after PDL. Beta cells failed to proliferate in response to PDL in beta-cell-specific SMAD7 mutant mice. Forced expression of SMAD7 in beta cells by itself was sufficient to promote beta-cell proliferation in vivo. M2, rather than M1 macrophages, seem to be the inducers of SMAD7-mediated beta-cell proliferation. M2 macrophages not only release TGFβ1 to directly induce up-regulation of SMAD7 in beta cells but also release EGF to activate EGF receptor signaling that inhibits TGFβ1-activated SMAD2 nuclear translocation, resulting in TGFβ signaling inhibition. SMAD7 promotes beta-cell proliferation by increasing CyclinD1 and CyclinD2, and by inducing nuclear exclusion of p27. Our study thus reveals a molecular pathway to potentially increase beta-cell mass through enhanced SMAD7 activity induced by extracellular stimuli.
- Published
- 2014
- Full Text
- View/download PDF
48. A smad signaling network regulates islet cell proliferation.
- Author
-
El-Gohary Y, Tulachan S, Wiersch J, Guo P, Welsh C, Prasadan K, Paredes J, Shiota C, Xiao X, Wada Y, Diaz M, and Gittes G
- Subjects
- Animals, Cell Proliferation, Insulin-Secreting Cells cytology, Mice, Mice, Transgenic, Phosphorylation, Receptors, Transforming Growth Factor beta metabolism, Smad Proteins genetics, Transforming Growth Factor beta metabolism, Cell Dedifferentiation physiology, Insulin-Secreting Cells metabolism, Signal Transduction physiology, Smad Proteins metabolism
- Abstract
Pancreatic β-cell loss and dysfunction are critical components of all types of diabetes. Human and rodent β-cells are able to proliferate, and this proliferation is an important defense against the evolution and progression of diabetes. Transforming growth factor-β (TGF-β) signaling has been shown to affect β-cell development, proliferation, and function, but β-cell proliferation is thought to be the only source of new β-cells in the adult. Recently, β-cell dedifferentiation has been shown to be an important contributory mechanism to β-cell failure. In this study, we tie together these two pathways by showing that a network of intracellular TGF-β regulators, smads 7, 2, and 3, control β-cell proliferation after β-cell loss, and specifically, smad7 is necessary for that β-cell proliferation. Importantly, this smad7-mediated proliferation appears to entail passing through a transient, nonpathologic dedifferentiation of β-cells to a pancreatic polypeptide-fold hormone-positive state. TGF-β receptor II appears to be a receptor important for controlling the status of the smad network in β-cells. These studies should help our understanding of properly regulated β-cell replication.
- Published
- 2014
- Full Text
- View/download PDF
49. Specific transduction and labeling of pancreatic ducts by targeted recombinant viral infusion into mouse pancreatic ducts.
- Author
-
Guo P, Xiao X, El-Gohary Y, Criscimanna A, Prasadan K, Rymer C, Shiota C, Wiersch J, Gaffar I, Esni F, and Gittes GK
- Subjects
- Animals, Cell Lineage, Genetic Vectors, Green Fluorescent Proteins genetics, HEK293 Cells, Humans, Infusion Pumps, Mice, Pancreas cytology, Pancreas metabolism, Pancreatic Ducts cytology, Promoter Regions, Genetic, Recombinant Proteins genetics, Regeneration, SOX9 Transcription Factor genetics, Transduction, Genetic instrumentation, Dependovirus genetics, Pancreatic Ducts metabolism, Transduction, Genetic methods
- Abstract
Specific labeling of pancreatic ducts has proven to be quite difficult. Such labeling has been highly sought after because of the power it would confer to studies of pancreatic ductal carcinogenesis, as well as studies of the source of new insulin-producing β-cells. Cre-loxp recombination could, in theory, lineage-tag pancreatic ducts, but results have been conflicting, mainly due to low labeling efficiencies. Here, we achieved a high pancreatic duct labeling efficiency using a recombinant adeno-associated virus (rAAV) with a duct-specific sox9 promoter infused into the mouse common biliary/pancreatic duct. We saw rapid, diffuse duct-specific labeling, with 50 and 89% labeling in the pancreatic tail and head region, respectively. This highly specific labeling of ducts should greatly enhance our ability to study the role of pancreatic ducts in numerous aspects of pancreatic growth, development and function.
- Published
- 2013
- Full Text
- View/download PDF
50. α-Cells are dispensable in postnatal morphogenesis and maturation of mouse pancreatic islets.
- Author
-
Shiota C, Prasadan K, Guo P, El-Gohary Y, Wiersch J, Xiao X, Esni F, and Gittes GK
- Subjects
- Ablation Techniques, Animals, Animals, Newborn, Biomarkers metabolism, Exons, Female, Glucagon chemistry, Glucagon genetics, Glucose Transporter Type 2 metabolism, Hypertrophy, Hypoglycemia etiology, Insulin-Secreting Cells cytology, Insulin-Secreting Cells metabolism, Islets of Langerhans cytology, Islets of Langerhans surgery, Luminescent Proteins chemistry, Luminescent Proteins metabolism, Male, Mice, Mice, Transgenic, Pancreas pathology, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Promoter Regions, Genetic, Recombinant Fusion Proteins metabolism, Urocortins metabolism, Glucagon metabolism, Glucagon-Secreting Cells physiology, Islets of Langerhans growth & development
- Abstract
Glucagon-producing α-cells are the second-most abundant cell type in the islet. Whereas α-cells make up less than 20% of the cells in a mature mouse islet, they occupy a much larger proportion of the pancreatic endocrine cell population during the early postnatal period, the time when morphological and functional maturation occurs to form adult islets. To determine whether α-cells have a role in postnatal islet development, a diphtheria toxin-mediated α-cell ablation mouse model was established. Rapid and persistent depletion of α-cells was achieved by daily injection of the toxin for 2 wk starting at postnatal day 1 (P1). Total pancreatic glucagon content in the α-cell-ablated mice was undetectable at P14 and still less than 0.3% of that of the control mice at 4 mo of age. Histological analyses revealed that formation of spherical islets occurred normally, and the islet size distribution was not changed despite the near-total lack of α-cells. Furthermore, there were no differences in expression of β-cell maturation marker proteins, including urocortin 3 and glucose transporter 2, in the α-cell-ablated islets at P14. Mice lacking α-cells grew normally and appeared healthy. Both glucose and insulin tolerance tests demonstrated that the α-cell-ablated mice had normal glucose homeostasis. These results indicate that α-cells do not play a critical role in postnatal islet morphogenesis or functional maturation of β-cells.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.