Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Environ Health Res ; 31(8): 901-914, 2021 Dec.
Article in English | MEDLINE | ID: mdl-31829725

ABSTRACT

We examined whether exercising indoors vs. outdoors reduced the cardio-respiratory effects of outdoor air pollution. Adults ≥55 were randomly assigned to exercise indoors when the Air Quality Health Index was ≥5 and outdoors on other days (intervention group, n = 37), or outdoors everyday (control group, n = 35). Both groups completed cardio-respiratory measurements before and after exercise for up to 10 weeks. Data were analyzed using linear mixed effect regression models. In the control group, an interquartile range increase in fine particulate matter (PM2.5) was associated with increases of 1.4% in heart rate (standard error (SE) = 0.7%) and 5.6% (SE = 2.6%) in malondialdehyde, and decreases of 5.6% (SE = 2.5%) to 16.5% (SE = 7.5%) in heart rate variability measures. While the hypothesized benefit of indoor vs. outdoor exercise could not be demonstrated due to an insufficient number of intervention days (n = 2), the study provides evidence of short-term effects of air pollution in older adults. ISRCTN #26552763.


Subject(s)
Air Pollution/adverse effects , Environmental Exposure/adverse effects , Exercise/physiology , Aged , Aged, 80 and over , Air Pollutants/adverse effects , Air Pollutants/analysis , Air Pollution/analysis , Environmental Exposure/analysis , Female , Heart Rate , Humans , Male , Malondialdehyde/urine , Middle Aged , Oxidative Stress , Particulate Matter/adverse effects , Particulate Matter/analysis , Regression Analysis , Respiratory Function Tests
2.
J Cell Physiol ; 224(2): 501-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20432450

ABSTRACT

Type 1 diabetes is inhibited in diabetes-prone BioBreeding (BBdp) rats fed a low-antigen hydrolyzed casein (HC) diet. In cereal-fed BBdp rats, islet expansion is defective accompanied by a futile upregulation of islet neogenesis without increased islet mass, due to a subtle blockage in islet cell cycle. We hypothesized that islet growth is enhanced before insulitis in HC-fed young BBdp rats and that islet neogenesis could be stimulated by a trophic factor, islet neogenesis-associated protein (INGAP). beta-Cell homeostasis was analyzed using immunohistochemistry, morphometry, laser capture microdissection and RT-PCR in BBdp rats fed HC or cereal diets. beta-cell proliferation in small and medium islets, and the number and area fraction of medium and large islets were increased in HC-fed animals. In situ islet cell cycle analysis revealed an increased proportion of proliferating S + G2 cells in medium and large islets of 25-45 day HC-fed rats. Expression of the cell cycle inhibitor, p16(INK4a) correlated with islet size and the percentage of p16(INK4a+) beta-cells increased in HC-fed BBdp rats, likely reflecting an increase in large islet area fraction. In HC-fed rats, extra-islet insulin(+) clusters (EIC), insulin(+) duct cells, large islet area fraction, and beta-cell mass were increased. Neurogenin-3 and Pdx-1, markers of beta-cell progenitors, were increased in EIC of weanling HC-fed rats. Daily injection of INGAP (30-45 days) increased the number of small islets, total islets, and insulin(+) cells in small ducts. Thus, in BBdp rats fed a protective HC diet, beta-cell expansion is enhanced through increased beta-cell proliferation and stimulation of islet neogenesis.


Subject(s)
Aging/pathology , Cytoprotection , Diabetes Mellitus, Experimental/pathology , Diet , Feeding Behavior , Insulin-Secreting Cells/pathology , Aging/drug effects , Animals , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caseins/administration & dosage , Caseins/pharmacology , Cell Aggregation/drug effects , Cell Count , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cytokines/pharmacology , Cytoprotection/drug effects , Diabetes Mellitus, Experimental/metabolism , Disease Susceptibility/pathology , Homeodomain Proteins/metabolism , Homeostasis/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Organ Size/drug effects , Pancreatitis-Associated Proteins , Peptide Fragments/pharmacology , Rats , Trans-Activators/metabolism
3.
Toxicol In Vitro ; 23(5): 816-33, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19406224

ABSTRACT

Prior to its application for in vitro toxicological assays, thorough characterization of a cell line is essential. The present study uses global transcriptional profiling to characterize a lung epithelial cell line (FE1) derived from MutaMouse [White, P.A., Douglas, G.R., Gingerich, J., Parfett, C., Shwed, P., Seligy, V., Soper, L., Berndt, L., Bayley, J., Wagner, S., Pound, K., Blakey, D., 2003. Development and characterization of a stable epithelial cell line from Muta Mouse lung. Environmental and Molecular Mutagenesis 42, 166-184]. Results presented here demonstrate the origin of the FE1 lung cell line as epithelial, presenting both type I and type II alveolar phenotype. An assessment of toxicologically-relevant genes, including those involved in the response to stress and stimuli, DNA repair, cellular metabolism, and programmed cell death, revealed changes in expression of 22-27% of genes in one or more culture type (proliferating and static FE1 cultures, primary epithelial cultures) compared with whole lung isolates. Gene expression analysis at 4 and 24h following benzo(a)pyrene exposure revealed the induction of cyp1a1, cyp1a2, and cyp1b1 in FE1 cells and lung isolates. The use of DNA microarrays for gene expression profiling allows an improved understanding of global, coordinated cellular events arising in cells under different physiological conditions. Taken together, these data indicate that the FE1 cell line is derived from a cell type relevant to toxic responses in vivo, and shows some similarity in response to chemical insult as the original tissue.


Subject(s)
Benzo(a)pyrene/toxicity , Gene Expression Profiling/methods , Mutagens/toxicity , Oligonucleotide Array Sequence Analysis/methods , Animals , Cell Line , Enzyme Induction/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Transgenic , Mutagenesis/drug effects , Phenotype , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Time Factors
4.
Lab Invest ; 87(12): 1240-51, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17906659

ABSTRACT

We reported previously that young BioBreeding diabetes-prone (BBdp) rats display increased neogenic extra-islet insulin+ clusters (EICs, <4 insulin+ cells) without an increase in beta-cell mass. Therefore, we investigated the possibility that abnormal islet expansion occurs in BBdp rats before the appearance of islet inflammation. Islet expansion was analyzed in pancreata from 14 to 45 day BBdp and control (BioBreeding control, BBc) rats using immunohistochemistry, morphometry, laser capture microdissection and reverse transcriptase-PCR. mRNA expression for Neurogenin-3, a developmental marker of endocrine progenitors, was three-fold greater in EIC of weanling BBdp and BBc rats compared with islet cells. With increasing age (14-30 days), Neurogenin-3 expression decreased in EIC and increased in islets. In BBdp rats, EIC number and beta-cell proliferation within EIC was greater compared with BBc animals; apoptosis did not differ. The area of small and medium islets in BBdp rats was greater than BBc rats between 14 and 30 days, but this did not result in increased total islet area or beta-cell mass. In addition, the number and area of very large islets was low at 45 days. The frequency of proliferating beta-cells decreased with increasing islet size in BBdp but was constant in BBc rats. Cell cycle analysis of islets revealed more G1 cells and fewer G2 cells in BBdp rats. The ratio of cyclinD2/Cdkn1a, genes that respectively promote or inhibit cell cycle progression, was decreased in BBdp islets. These results suggest that despite increased islet neogenesis, the capacity for islet expansion in diabetes-prone rats is compromised possibly due to decreased proliferative capacity with increasing islet size associated with a partial block at the G1/S cell cycle boundary in islet cells.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/growth & development , Islets of Langerhans/pathology , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle , Cell Proliferation , Cell Size , Cyclin D2 , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/metabolism , Diabetes Mellitus, Type 1/metabolism , Homeodomain Proteins/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Nerve Tissue Proteins/metabolism , Rats , Trans-Activators/metabolism
5.
Diabetes ; 55(2): 452-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16443780

ABSTRACT

C57BL/6 (B6) mice develop glucose intolerance with age, whereas C3H/He (C3H) mice do not. In this study, we examined whether this differential glucose homeostasis was associated with differences of proteolytic activation of pancreatic prohormones. Radioimmunoassays showed comparable levels of fasting plasma insulin between the two strains but a significantly lower glucagon level in B6 mice. Pulse-chase analysis of glucagon biosynthesis in isolated pancreatic islets revealed that proglucagon was less efficiently processed in B6 mice. Because proprotein convertase (PC)2 and its 7B2 helper protein are required for this processing, we quantified islet mRNA levels by RT-PCR and protein levels by immunoblotting. The levels of proPC2 mRNA were similar between the two strains, but B6 protein extracts contained less of the mature PC2. In contrast, 7B2 mRNA and protein levels were both significantly lower in B6 pancreas. Sequencing of the 7B2 gene promoter and cDNA in the two strains revealed seven single nucleotide polymorphisms and one dinucleotide insertion/deletion in the cDNA as well as a single nucleotide polymorphism and two insertions/deletions in the promoter. Differential expression of 7B2 may contribute to the difference between B6 and C3H mice not only in glucagon production and secretion but also in glucose tolerance.


Subject(s)
Gene Expression Regulation , Neuroendocrine Secretory Protein 7B2/genetics , Neuroendocrine Secretory Protein 7B2/metabolism , Pancreas/metabolism , Polymorphism, Genetic/genetics , Aging , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Glucagon/blood , Glucose Intolerance/genetics , Humans , Islets of Langerhans/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Neuroendocrine Secretory Protein 7B2/chemistry , Promoter Regions, Genetic , Proprotein Convertase 2/metabolism , Protein Folding , Sequence Alignment
6.
Biochim Biophys Acta ; 1724(1-2): 23-36, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15882932

ABSTRACT

Glucose induces complex patterns of oscillations in intracellular Ca2+ concentration ([Ca2+]i), metabolism and secretion in islets of Langerhans including "slow" and "fast" pulses with period of 2-5 min and 10-20 s respectively. In an effort to elucidate the origin of slow oscillations, individual mouse islets were exposed to different fuels including glyceraldehyde, pyruvate, methyl pyruvate and alpha-ketoisocaproate (KIC), all of which bypass key steps of glycolytic metabolism, while monitoring [Ca2+]i, oxygen consumption and secretion. Glyceraldehyde gave rise to slow oscillations only when substimulatory glucose was also added to the media. Glucosamine, an inhibitor of glucokinase, blocked these slow oscillations. KIC, pyruvate, and methyl pyruvate did not give rise to slow oscillations alone or with glucose present. The addition of glucose to islets bathed in nutrient-rich cell culture media accelerated metabolism and initiated slow oscillations while glyceraldehyde did not. It is concluded that glucose has a special role in accelerating metabolism and generating slow oscillations in isolated islets of Langerhans from mice. Combined with previous observations of Ca2+ dependency for all oscillations in islets, we propose that interactions between Ca2+ influx and glycolysis are responsible for the slow oscillations. In contrast, fast oscillations can occur independent of glycolytic flux.


Subject(s)
Calcium/metabolism , Glucose/metabolism , Islets of Langerhans/metabolism , Animals , Glucose/pharmacology , Glyceraldehyde/metabolism , Glyceraldehyde/pharmacology , Glycolysis , Islets of Langerhans/drug effects , Keto Acids/metabolism , Keto Acids/pharmacology , Mice , Pyruvates/metabolism , Pyruvates/pharmacology , Pyruvic Acid/metabolism , Pyruvic Acid/pharmacology
7.
Diabetes ; 53(6): 1517-25, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161756

ABSTRACT

Mice with deletion of insulin receptor substrate (IRS)-1 (IRS-1 knockout [KO] mice) show mild insulin resistance and defective glucose-stimulated insulin secretion and reduced insulin synthesis. To further define the role of IRS-1 in islet function, we examined the insulin secretory defect in the knockouts using freshly isolated islets and primary beta-cells. IRS-1 KO beta-cells exhibited a significantly shorter increase in intracellular free Ca(2+) concentration ([Ca(2+)](i)) than controls when briefly stimulated with glucose or glyceraldehyde and when l-arginine was used to potentiate the stimulatory effect of glucose. These changes were paralleled by a lower number of exocytotic events in the KO beta-cells in response to the same secretagogues, indicating reduced insulin secretion. Furthermore, the normal oscillations in intracellular Ca(2+) and O(2) consumption after glucose stimulation were dampened in freshly isolated KO islets. Semiquantitative RT-PCR showed a dramatically reduced islet expression of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA)-2b and -3 in the mutants. These data provide evidence that IRS-1 modulation of insulin secretion is associated with Ca(2+) signaling and expression of SERCA-2b and -3 genes in pancreatic islets and provides a direct link between insulin resistance and defective insulin secretion.


Subject(s)
Calcium Signaling , Calcium-Transporting ATPases/metabolism , Islets of Langerhans/metabolism , Phosphoproteins/deficiency , Animals , Calcium/metabolism , Calcium-Transporting ATPases/genetics , Exocytosis/drug effects , Exocytosis/physiology , Gene Expression , Glucose/pharmacology , Glyceraldehyde/pharmacology , In Vitro Techniques , Insulin/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Insulin Secretion , Intracellular Membranes/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Male , Mice , Mice, Knockout , Oscillometry , Osmolar Concentration , Oxygen Consumption , Sarcoplasmic Reticulum Calcium-Transporting ATPases
8.
Endocrine ; 23(1): 77-84, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15034199

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is the most insulinogenic of the glucagon-like peptides secreted mainly by L cells in the small and large intestine in response to the ingestion of nutrients. It binds to a specific GLP-1 receptor (GLP-1R) on beta-cells and can increase islet neogenesis and beta-cell mass. It is not clear whether the transmission of information from the gut to islet beta-cells by messengers such as GLP-1 is different in individuals who develop autoimmune diabetes. In the present study the expression of bioactive GLP-1 protein in the gut and its receptor in the pancreas was examined in diabetes-prone BioBreeding (BBdp) rats in the period before overt diabetes and in age-matched control, non-diabetes-prone BB (BBc) rats. An N-terminal directed antibody specific for the bioactive forms of GLP-1 (GLP-1(7-37) and GLP-1(7-36amide)) was used to mea-sure GLP-1 by radioimmunoassay in proximal, median, and distal gut. Pancreas GLP-1R area fraction, GLP-1R gene expression, and insulin content were analyzed, as were plasma GLP-1, glucose, and insulin. The concentration of GLP-1 protein in the jejunum and ileum of BBdp rats was lower than in BBc rats. Although these animals maintained normal blood glucose, there was impaired pancreatic endocrine function, characterized by low baseline insulin concentration in plasma and pancreas. GLP-1R mRNA expression was threefold less in islets isolated from BBdp rats, and GLP-1R+ islet area fraction in pancreas sections was decreased. When injected iv with GLP-1, BBdp rats displayed lower second-phase insulin response (and insulin/glucose ratios) compared with BBc rats. Thus, young BBdp rats displayed decreased concentrations of bioactive GLP-1 in jejunum and ileum, reduced GLP-1R in islets, and lower second-phase insulin response to iv GLP-1 than controls. The decrease in insulinogenic and islet beta-cell mass-promoting signal from GLP-1 in BBdp rats may contribute to impaired glucoregulation and ineffective maintenance of normal islet mass that shifts islet homeostasis in favor of development of diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Gastrointestinal Tract/metabolism , Glucagon/metabolism , Insulin/metabolism , Pancreas/metabolism , Peptide Fragments/metabolism , Protein Precursors/metabolism , Receptors, Glucagon/metabolism , Animals , Blood Glucose/metabolism , Blotting, Northern , Female , Gene Expression , Glucagon/biosynthesis , Glucagon/genetics , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Immunohistochemistry , Insulin/blood , Male , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Protein Precursors/biosynthesis , Protein Precursors/genetics , Rats , Rats, Inbred BB , Receptors, Glucagon/biosynthesis , Receptors, Glucagon/genetics
9.
Analyst ; 128(8): 1013-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12964599

ABSTRACT

A microscale method for purines involved in intracellular signaling and energy metabolism, including ADP, ATP, cyclic-AMP, NADH and GTP, was developed. The analytes were separated on a fused-silica capillary liquid chromatography column (50 microm inner diameter by 25 cm long) packed with 7 microm reversed-phase particles and detected with a carbon fiber cylinder microelectrode at +1.50 V versus Ag/AgCl reference electrode. With an acetonitrile gradient, the separation was carried out within 15 min. With a 100 nl injection volume, the detection limits varied from 0.9 to 8 fmol depending upon the analyte. The low detection limits make the method suitable for analysis of small tissue samples. As a demonstration of the method, islets of Langerhans were analyzed for their adenosine-related messenger content.


Subject(s)
Microchemistry/methods , Purines/analysis , Animals , Chromatography, Liquid/methods , Culture Techniques , Islets of Langerhans/chemistry , Mice , Rats , Rats, Sprague-Dawley
10.
Biochem Biophys Res Commun ; 304(2): 371-7, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12711325

ABSTRACT

A glucose oxidase-based glucose microsensor (<10 micro m tip diameter) was used to measure the glucose concentration within single islets under static conditions and during step changes in glucose level. The sensors had response times of 4.1+/-0.5s (n=7) and sensitivities of 8.7+/-1.8 pA/mM (n=11). The sensors performed independent of oxygen up to 15 mM glucose as long as the oxygen level was > 70 mm Hg. Spatially resolved glucose measurements revealed a glucose gradient around and inside single islets. From measurement of the glucose gradient, a glucose consumption rate of 0.48+/-0.14 pmol/nL islet/min (n=6) and an intraislet glucose diffusion coefficient of 3.8 x 10(-7)cm(2)/s were determined. The measurement of the gradient demonstrates that not all cells within an islet in culture are exposed to the same glucose concentration. The sensor was also used to measure the time required for intraislet glucose concentration to reach steady state following a step increase in glucose concentration from 3 to 10 mM at the islet surface. At a depth of 70 micro m inside an islet, glucose reached steady state in 180+/-7s (n=7) for islets with a diameter of 180-220 micro m (smaller islets reach steady-state faster). In the presence of 10 mM mannoheptulose, an inhibitor of glucokinase, the equilibration time was reduced to 122+/-11s (n=6), indicating that glucose utilization by glycolysis limited the time required for glucose to diffuse into the islets. The long times to reach steady state and presence of glucose gradients are important in interpreting data from experiments involving islets in culture.


Subject(s)
Biosensing Techniques , Glucose/analysis , Glucose/metabolism , Islets of Langerhans/metabolism , Animals , Biological Transport , Cells, Cultured , Glucose Oxidase/metabolism , Islets of Langerhans/chemistry , Kinetics , Mice , Microelectrodes
11.
Diabetes ; 51 Suppl 1: S152-61, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11815475

ABSTRACT

Whereas the mechanisms underlying oscillatory insulin secretion remain unknown, several models have been advanced to explain if they involve generation of metabolic oscillations in beta-cells. Evidence, including measurements of oxygen consumption, glucose consumption, NADH, and ATP/ADP ratio, has accumulated to support the hypothesis that energy metabolism in beta-cells can oscillate. Where simultaneous measurements have been made, these oscillations are well correlated with oscillations in intracellular [Ca(2+)] and insulin secretion. Considerable evidence has been accumulated to suggest that entry of Ca(2+) into cells can modulate metabolism both positively and negatively. The main positive effect of Ca(2+) is an increase in oxygen consumption, believed to involve activation of mitochondrial dehydrogenases. Negative feedback by Ca(2+) includes decreases in glucose consumption and decreases in the mitochondrial membrane potential. Ca(2+) also provides negative feedback by increasing consumption of ATP. The negative feedback provided by Ca(2+) provides a mechanism for generating oscillations based on a model in which glucose stimulates a rise in ATP/ADP ratio that closes ATP-sensitive K(+) (K(ATP)) channels, thus depolarizing the cell membrane and allowing Ca(2+) entry through voltage-sensitive channels. Ca(2+) entry reduces the ATP/ADP ratio and allows reopening of the K(ATP) channel.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Periodicity , Animals , Humans , Insulin Secretion
SELECTION OF CITATIONS
SEARCH DETAIL