Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Am J Physiol Endocrinol Metab ; 326(3): E308-E325, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38265288

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 [2-3H] and [3-3H]-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.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Animals , Male , Rats , Blood Glucose/metabolism , Cyclic AMP , Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Glucose/metabolism , Hyperglycemia/metabolism , Insulin/metabolism , Rats, Zucker
2.
Int J Mol Sci ; 24(8)2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37108594

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.


Subject(s)
Acute Kidney Injury , Mercuric Chloride , Rats , Male , Guinea Pigs , Animals , Mercuric Chloride/toxicity , Kidney/metabolism , Kidney Function Tests , Acute Kidney Injury/metabolism , Liver/metabolism
3.
Mol Ther Methods Clin Dev ; 28: 355-365, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36879848

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.

4.
Toxicol Appl Pharmacol ; 430: 115713, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34492290

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.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Gene Expression Profiling , Liver/metabolism , Metabolome , Metabolomics , Thioacetamide , Transcriptome , 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
5.
Int J Mol Sci ; 21(21)2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33158035

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.


Subject(s)
Amino Acids/metabolism , Chemical and Drug Induced Liver Injury/diagnosis , Hazardous Substances/pharmacology , Lipid Metabolism/drug effects , Metabolome/drug effects , 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
6.
Toxicology ; 441: 152493, 2020 08.
Article in English | MEDLINE | ID: mdl-32479839

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.


Subject(s)
Chemical and Drug Induced Liver Injury/blood , 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
7.
Toxicology ; 442: 152530, 2020 09.
Article in English | MEDLINE | ID: mdl-32599119

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 (HgCl2)-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.


Subject(s)
Kidney Diseases/chemically induced , Kidney Diseases/pathology , Mercuric Chloride/toxicity , Toxicogenetics/methods , 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
8.
J Biol Chem ; 295(15): 4858-4869, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32122971

ABSTRACT

The interplay between the transforming growth factor ß (TGF-ß) signaling proteins, SMAD family member 2 (SMAD2) and 3 (SMAD3), and the TGF-ß-inhibiting SMAD, SMAD7, seems to play a vital role in proper pancreatic endocrine development and also in normal ß-cell function in adult pancreatic islets. Here, we generated conditional SMAD7 knockout mice by crossing insulin1Cre mice with SMAD7fx/fx mice. We also created a ß cell-specific SMAD7-overexpressing mouse line by crossing insulin1Dre mice with HPRT-SMAD7/RosaGFP mice. We analyzed ß-cell function in adult islets when SMAD7 was either absent or overexpressed in ß cells. Loss of SMAD7 in ß 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 ß cells. Our results show that both the absence and overexpression of SMAD7 affect TGF-ß signaling and modulates ß-cell proliferation but does not appear to alter ß-cell function. Reversible SMAD7 overexpression may represent an attractive therapeutic option to enhance ß-cell proliferation without negative effects on ß-cell function.


Subject(s)
Cell Proliferation , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Insulin/physiology , Smad7 Protein/physiology , Transforming Growth Factor beta/metabolism , Animals , Female , Glucose/pharmacology , Male , Mice , Mice, Knockout , Signal Transduction , Sweetening Agents/pharmacology , Transforming Growth Factor beta/genetics
9.
J Endocr Soc ; 3(11): 1979-1992, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31620668

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 ß-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 ß-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.

10.
Development ; 146(13)2019 06 27.
Article in English | MEDLINE | ID: mdl-31160417

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 ß-cells is termed ß-cell heterogeneity, which was recently associated with diabetic phenotypes. Nevertheless, the presence of a developmentally derived ß-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 ß-cells had fewer Ki-67+ proliferating ß-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 ß-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 ß-cell heterogeneity.


Subject(s)
Cell Lineage , Cell Tracking/methods , Insulin-Secreting Cells/cytology , Pancreas/cytology , Stem Cells/cytology , Animals , Cell Differentiation/genetics , Cell Lineage/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
11.
Cell Stem Cell ; 22(1): 78-90.e4, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29304344

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.


Subject(s)
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 , 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
12.
Theranostics ; 8(21): 5960-5971, 2018.
Article in English | MEDLINE | ID: mdl-30613274

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/ß-cell function and the occurrence of pancreatitis in these mice were assessed via ELISA, qPCR, and immunohistochemical analyses. Results: The LTßR/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 ß-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 ß-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.


Subject(s)
Death , Glucagon-Secreting Cells/pathology , Growth Disorders/physiopathology , Hypoglycemia/physiopathology , Insulin-Secreting Cells/pathology , Pancreatitis/physiopathology , Protein Serine-Threonine Kinases/metabolism , 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
13.
Diabetologia ; 60(12): 2399-2408, 2017 12.
Article in English | MEDLINE | ID: mdl-28884202

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 Gcg CreERT2 mouse line was crossed to the Rosa26 tdTomato (R26 tdTomato ) Cre reporter mouse line in order to evaluate the tissue specificity, efficiency and tamoxifen dependency of Gcg CreERT2 -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 Gcg CreERT2/w heterozygous mice. RESULTS: Tamoxifen-treated Gcg CreERT2/w ;R26 tdTomato/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 Gcg CreERT2/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 Gcg CreERT2 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.


Subject(s)
Glucagon-Secreting Cells/metabolism , Proglucagon/metabolism , Animals , Female , Glucagon/blood , Glucagon-Like Peptide 1/blood , Immunohistochemistry , Male , Mice , Mice, Transgenic , Proglucagon/genetics , Tamoxifen/pharmacology
14.
Diabetes ; 66(10): 2646-2658, 2017 10.
Article in English | MEDLINE | ID: mdl-28775125

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 ß-cell dedifferentiation, followed by a time-dependent decrease in functional ß-cell mass-all specifically in the ligated tail portion of the pancreas (PDL-tail). High levels of transforming growth factor ß1 (TGFß1) 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 ß-cell mass was then found to result from TGFß1-triggered epithelial-mesenchymal transition (EMT) by ß-cells, rather than resulting directly from ß-cell apoptosis. Mechanistically, TGFß1-treated ß-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 ß-cells ameliorated ß-cell EMT and ß-cell loss and prevented the onset of diabetes in mice undergoing PDL. Together, our data suggest that chronic pancreatitis may trigger TGFß1-mediated ß-cell EMT to lead to CPRD, which could substantially be prevented by sustained expression of FoxO1 in ß-cells.


Subject(s)
Pancreatitis, Chronic/metabolism , STAT3 Transcription Factor/metabolism , Smad3 Protein/metabolism , 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
15.
J Biol Chem ; 292(8): 3456-3465, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28057752

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 ß cell dysfunction with accelerated glucose intolerance, followed by overt diabetes. The accelerated ß cell dysfunction and loss of proliferation capacity, two features of ß 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 ß cells that marked the onset of diabetes. Moreover, forced FoxO1 expression in ß cells prevented ß cell dysfunction and loss in this model. Thus, we present a model of accelerated ß cell aging that may be useful for studying the mechanisms underlying ß cell failure in diabetes. Moreover, we provide evidence highlighting a critical role of FoxO1 in maintaining ß cell identity in the context of SMAD7 failure.


Subject(s)
Diabetes Mellitus/metabolism , Forkhead Box Protein O1/metabolism , Insulin-Secreting Cells/pathology , Smad7 Protein/metabolism , 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
16.
J Biol Chem ; 291(43): 22630-22637, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27587395

ABSTRACT

A thorough understanding of the signaling pathways involved in the regulation of ß cell proliferation is an important initial step in restoring ß 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 ß cell proliferation. Specific deletion of EGFR in the ß cells of adult mice impaired ß cell proliferation at baseline and after 50% PPx, suggesting that the EGFR signaling pathway plays an essential role in adult ß cell proliferation. Further analyses showed that ß cell-specific depletion of EGFR resulted in impaired expression of cyclin D1 and impaired suppression of p27 after PPx, both of which enhance ß cell proliferation. These data highlight the importance of EGFR signaling and its downstream signaling cascade in postnatal ß cell growth.


Subject(s)
Cell Proliferation/physiology , ErbB Receptors/metabolism , Insulin-Secreting Cells/metabolism , Signal Transduction/physiology , Animals , Cyclin D1/genetics , Cyclin D1/metabolism , ErbB Receptors/genetics , Mice , Mice, Transgenic
17.
Cell Mol Life Sci ; 73(19): 3623-37, 2016 10.
Article in English | MEDLINE | ID: mdl-27105622

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.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Animals , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Organ Size , Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
18.
Endocrinology ; 157(4): 1348-56, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26872091

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 TGFß receptor signaling in ß-cells has been shown to increase ß-cell proliferation in mice, but has not been rigorously examined in humans. Here, treatment of human islets with a TGFß receptor I inhibitor, SB-431542 (SB), significantly improved C-peptide secretion by ß-cells, and significantly increased ß-cell number by increasing ß-cell proliferation. In addition, SB increased cell-cycle activators and decreased cell-cycle suppressors in human ß-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 ß-cell proliferation in the graft, compared with controls. Thus, our data suggest that transient suppression of TGFß receptor signaling may improve the outcome of human islet transplantation, seemingly through increasing ß-cell number and function.


Subject(s)
Islets of Langerhans Transplantation/methods , Islets of Langerhans/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , 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
19.
Sci Rep ; 6: 21127, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26884345

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.


Subject(s)
Acinar Cells/cytology , Cell Separation/methods , Pancreas/cytology , Pancreatitis/pathology , Peanut Agglutinin , Acinar Cells/metabolism , Animals , Flow Cytometry/methods , Mice , Pancreas/pathology , Peanut Agglutinin/metabolism
20.
Endocrinology ; 157(1): 166-75, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26505114

ABSTRACT

A key question in diabetes research is whether new ß-cells can be derived from endogenous, nonendocrine cells. The potential for pancreatic ductal cells to convert into ß-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-ß 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 ß-cells in normal young developing mice as well as in adult TGF-ß 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 ß-cells in the postnatal pancreas but also importantly implicates TGF-ß signaling in this process.


Subject(s)
Cell Transdifferentiation , Insulin-Secreting Cells/cytology , Insulin/biosynthesis , Islets of Langerhans/cytology , Pancreatic Ducts/cytology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , 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
SELECTION OF CITATIONS
SEARCH DETAIL
...