Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Ups J Med Sci ; 120(4): 299-304, 2015.
Article in English | MEDLINE | ID: mdl-26095011

ABSTRACT

BACKGROUND: A 57-year old man with low-back pain was found to have a 3 × 3 × 3 cm presacral neuroendocrine tumour (NET) with widespread metastases, mainly to the skeleton. His neoplastic disease responded well to peptide receptor radionuclide therapy (PRRT) with the radiotagged somatostatin agonist (177)Lu-DOTATATE. During almost 10 years he was fit for a normal life. He succumbed to an intraspinal dissemination. PROCEDURES: A resection of the rectum, with a non-radical excision of the adjacent NET, was made. In addition to computerized tomography (CT), receptor positron emission tomography (PET) with (68)Ga-labelled somatostatin analogues was used. OBSERVATIONS: The NET showed the growth pattern and immunoprofile of a G2 carcinoid. A majority cell population displayed immunoreactivity to ghrelin, exceptionally with co-immunoreactivity to motilin. Somatostatin receptor scintigraphy and (68)Ga-DOTATATE PET-CT demonstrated uptake in the metastatic lesions. High serum concentrations of total (desacyl-)ghrelin were found with fluctuations reflecting the severity of the symptoms. In contrast, the concentrations of active (acyl-)ghrelin were consistently low, as were those of chromogranin A (CgA). CONCLUSIONS: Neoplastically transformed ghrelin cells can release large amounts of desacyl-ghrelin, evoking an array of non-specific clinical symptoms. Despite an early dissemination to the skeleton, a ghrelinoma can be compatible with longevity after adequate radiotherapy.


Subject(s)
Carcinoma, Neuroendocrine/secondary , Ghrelin/metabolism , Neoplasms, Multiple Primary/diagnosis , Rectal Neoplasms/pathology , Spinal Neoplasms/secondary , Biopsy, Needle , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/surgery , Disease Progression , Fatal Outcome , Humans , Immunohistochemistry , Low Back Pain/diagnosis , Low Back Pain/etiology , Magnetic Resonance Imaging/methods , Male , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Positron-Emission Tomography/methods , Rectal Neoplasms/surgery , Sacrococcygeal Region , Spinal Neoplasms/physiopathology
2.
J Mol Endocrinol ; 52(1): R35-49, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24049065

ABSTRACT

The islets of Langerhans are key regulators of glucose homeostasis and have been known as a structure for almost one and a half centuries. During the twentieth century several different cell types were described in the islets of different species and at different developmental stages. Six cell types with identified hormonal product have been described so far by the use of histochemical staining methods, transmission electron microscopy, and immunohistochemistry. Thus, glucagon-producing α-cells, insulin-producing ß-cells, somatostatin-producing δ-cells, pancreatic polypeptide-producing PP-cells, serotonin-producing enterochromaffin-cells, and gastrin-producing G-cells have all been found in the mammalian pancreas at least at some developmental stage. Species differences are at hand and age-related differences are also to be considered. Eleven years ago a novel cell type, the ghrelin cell, was discovered in the human islets. Subsequent studies have shown the presence of islet ghrelin cells in several animals, including mouse, rat, gerbils, and fish. The developmental regulation of ghrelin cells in the islets of mice has gained a lot of interest and several studies have added important pieces to the puzzle of molecular mechanisms and the genetic regulation that lead to differentiation into mature ghrelin cells. A body of evidence has shown that ghrelin is an insulinostatic hormone, and the potential for blockade of ghrelin signalling as a therapeutic avenue for type 2 diabetes is intriguing. Furthermore, ghrelin-expressing pancreatic tumours have been reported and ghrelin needs to be taken into account when diagnosing pancreatic tumours. In this review article, we summarise the knowledge about islet ghrelin cells obtained so far.


Subject(s)
Ghrelin/metabolism , Islets of Langerhans/physiology , Animals , Gene Expression , Ghrelin/genetics , Homeostasis , Humans , Islets of Langerhans/cytology
3.
Endocrinology ; 153(12): 5782-95, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23064014

ABSTRACT

Enteroendocrine cells such as duodenal cholecystokinin (CCK cells) are generally thought to be confined to certain segments of the gastrointestinal (GI) tract and to store and release peptides derived from only a single peptide precursor. In the current study, however, transgenic mice expressing enhanced green fluorescent protein (eGFP) under the control of the CCK promoter demonstrated a distribution pattern of CCK-eGFP positive cells that extended throughout the intestine. Quantitative PCR and liquid chromatography-mass spectrometry proteomic analyses of isolated, FACS-purified CCK-eGFP-positive cells demonstrated expression of not only CCK but also glucagon-like peptide 1 (GLP-1), gastric inhibitory peptide (GIP), peptide YY (PYY), neurotensin, and secretin, but not somatostatin. Immunohistochemistry confirmed this expression pattern. The broad coexpression phenomenon was observed both in crypts and villi as demonstrated by immunohistochemistry and FACS analysis of separated cell populations. Single-cell quantitative PCR indicated that approximately half of the duodenal CCK-eGFP cells express one peptide precursor in addition to CCK, whereas an additional smaller fraction expresses two peptide precursors in addition to CCK. The coexpression pattern was further confirmed through a cell ablation study based on expression of the human diphtheria toxin receptor under the control of the proglucagon promoter, in which activation of the receptor resulted in a marked reduction not only in GLP-1 cells, but also PYY, neurotensin, GIP, CCK, and secretin cells, whereas somatostatin cells were spared. Key elements of the coexpression pattern were confirmed by immunohistochemical double staining in human small intestine. It is concluded that a lineage of mature enteroendocrine cells have the ability to coexpress members of a group of functionally related peptides: CCK, secretin, GIP, GLP-1, PYY, and neurotensin, suggesting a potential therapeutic target for the treatment and prevention of diabetes and obesity.


Subject(s)
Cholecystokinin/biosynthesis , Enteroendocrine Cells/cytology , Gastric Inhibitory Polypeptide/biosynthesis , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Neurotensin/biosynthesis , Peptide YY/metabolism , Animals , Cell Lineage , Cell Separation , Diabetes Mellitus/prevention & control , Enteroendocrine Cells/metabolism , Flow Cytometry , Ghrelin/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry/methods , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Obesity/prevention & control , Promoter Regions, Genetic
4.
Int J Endocrinol ; 2011: 401258, 2011.
Article in English | MEDLINE | ID: mdl-22164158

ABSTRACT

Mice deficient in the zinc-sensor GPR39, which has been demonstrated to protect cells against endoplasmatic stress and cell death in vitro, display moderate glucose intolerance and impaired glucose-induced insulin secretion. Here, we use the Tet-On system under the control of the proinsulin promoter to selectively overexpress GPR39 in the ß cells in a double transgenic mouse strain and challenge them with multiple low doses of streptozotocin, which in the wild-type littermates leads to a gradual increase in nonfasting glucose levels and glucose intolerance observed during both food intake and OGTT. Although the overexpression of the constitutively active GPR39 receptor in animals not treated with streptozotocin appeared by itself to impair the glucose tolerance slightly and to decrease the ß-cell mass, it nevertheless totally protected against the gradual hyperglycemia in the steptozotocin-treated animals. It is concluded that GPR39 functions in a ß-cell protective manner and it is suggested that it is involved in some of the beneficial, ß-cell protective effects observed for Zn(++) and that GPR39 may be a target for antidiabetic drug intervention.

5.
Neuroendocrinology ; 94(3): 228-36, 2011.
Article in English | MEDLINE | ID: mdl-21832817

ABSTRACT

BACKGROUND/AIMS: Cocaine- and amphetamine-regulated transcript (CART) is an anorexigenic regulatory peptide highly expressed in the brain's appetite control centers, but also in peripheral neurons and in endocrine cells in the adrenal medulla, thyroid, pancreatic islets, and in the gastrointestinal tract. Plasma levels of CART were recently shown to be elevated in patients with neuroendocrine tumors (NETs), but the cellular sources of CART in NETs have remained unknown. The aim of the study was to establish whether CART is expressed in various types of NETs and, if so, to examine the frequency, distribution and phenotype of CART-expressing cells. METHODS: Tumor specimens from 133 NETs originating in the stomach, ileum, rectum, pancreas and thyroid were examined with immunohistochemistry and in situ hybridization. The expression of CART was quantified and the CART-expressing cells were phenotyped by double staining for established markers and hormones. RESULTS: CART-expressing tumor cells were found in the majority of the examined NETs. The expression pattern of CART was highly heterogeneous not only between tumors, but also within individual tumors. In 14% of the NETs, CART was found in a major population of the tumor cells. CONCLUSION: CART is produced in the majority of NETs, regardless of tumor origin. This likely explains the elevated levels of circulating CART in certain NETs patients, as recently described. CART could therefore prove to be a useful tool in the diagnostics of NETs not only in blood samples, but also in histopathological specimens.


Subject(s)
Gastrointestinal Neoplasms/genetics , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/genetics , Thyroid Neoplasms/genetics , Carcinoid Tumor/genetics , Carcinoid Tumor/metabolism , Carcinoid Tumor/pathology , Carcinoma, Neuroendocrine , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Neoplasm Grading , Nerve Tissue Proteins/metabolism , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
6.
PLoS One ; 6(4): e18213, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21483673

ABSTRACT

BACKGROUND: Voltage-dependent K(+) channels (Kv) mediate repolarisation of ß-cell action potentials, and thereby abrogate insulin secretion. The role of the Kv1.1 K(+) channel in this process is however unclear. We tested for presence of Kv1.1 in different species and tested for a functional role of Kv1.1 by assessing pancreatic islet function in BALB/cByJ (wild-type) and megencephaly (mceph/mceph) mice, the latter having a deletion in the Kv1.1 gene. METHODOLOGY/PRINCIPAL FINDINGS: Kv1.1 expression was detected in islets from wild-type mice, SD rats and humans, and expression of truncated Kv1.1 was detected in mceph/mceph islets. Full-length Kv1.1 protein was present in islets from wild-type mice, but, as expected, not in those from mceph/mceph mice. Kv1.1 expression was localized to the ß-cell population and also to α- and δ-cells, with evidence of over-expression of truncated Kv1.1 in mceph/mceph islets. Blood glucose, insulin content, and islet morphology were normal in mceph/mceph mice, but glucose-induced insulin release from batch-incubated islets was (moderately) higher than that from wild-type islets. Reciprocal blocking of Kv1.1 by dendrotoxin-K increased insulin secretion from wild-type but not mceph/mceph islets. Glucose-induced action potential duration, as well as firing frequency, was increased in mceph/mceph mouse ß-cells. This duration effect on action potential in ß-cells from mceph/mceph mice was mimicked by dendrotoxin-K in ß-cells from wild-type mice. Observations concerning the effects of both the mceph mutation, and of dendrotoxin-K, on glucose-induced insulin release were confirmed in pancreatic islets from Kv1.1 null mice. CONCLUSION/SIGNIFICANCE: Kv1.1 channels are expressed in the ß-cells of several species, and these channels can influence glucose-stimulated insulin release.


Subject(s)
Insulin-Secreting Cells/metabolism , Kv1.1 Potassium Channel/metabolism , Action Potentials , Animals , Blood Glucose/metabolism , Body Weight , Data Collection , Female , Fura-2/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Kv1.1 Potassium Channel/deficiency , Kv1.1 Potassium Channel/genetics , Male , Mice , Molecular Imaging , Perfusion , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Deletion , Shab Potassium Channels/metabolism , Species Specificity
7.
Br J Pharmacol ; 162(5): 1156-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21091642

ABSTRACT

BACKGROUND AND PURPOSE: Caveolin-1-deficiency is associated with substantial urogenital alterations. Here, a mechanical, histological and biochemical characterization of female detrusors from wild-type and caveolin-1-deficient (KO) mice was made to increase the understanding of detrusor changes caused by lack of caveolae. EXPERIMENTAL APPROACH: Length-tension relationships were generated, and we recorded responses to electrical field stimulation, the muscarinic receptor agonist carbachol and the purinoceptor agonist ATP. Tyrosine nitration and the contents of caveolin-1, cavin-1, muscarinic M3 receptors, phospholipase C(ß1), muscle-specific kinase (MuSK) and L-type Ca(2+) channels were determined by immunoblotting. Innervation was assessed by immunohistochemistry. KEY RESULTS: Bladder to body weight ratio was not changed, nor was there any change in the optimum circumference for force development. Depolarization- and ATP-induced stress was reduced, as was carbachol-induced stress between 0.1 and 3 µM, but the supramaximal relative (% K(+)) response to carbachol was increased, as was M3 expression. The scopolamine-sensitive component of the electrical field stimulation response was impaired, and yet bladder nerves contained little caveolin-1. The density of cholinergic nerves was unchanged, whereas CART- and CGRP-positive nerves were reduced. Immunoblotting revealed loss of MuSK. CONCLUSIONS AND IMPLICATIONS: Ablation of caveolae in the female detrusor leads to generalized impairment of contractility, ruling out prostate hypertrophy as a contributing factor. Cholinergic neuroeffector transmission is impaired without conspicuous changes in the density of cholinergic nerves or morphology of their terminals, but correlating with reduced expression of MuSK.


Subject(s)
Caveolin 1/deficiency , Muscle, Smooth/innervation , Muscle, Smooth/physiopathology , Adenosine Triphosphate/pharmacology , Animals , Base Sequence , Biomechanical Phenomena , Carbachol/pharmacology , Caveolae/pathology , Caveolae/physiology , Caveolin 1/genetics , Caveolin 1/physiology , DNA Primers/genetics , Diuresis , Electric Stimulation , Female , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Muscle Contraction/drug effects , Muscle, Smooth/pathology , Organ Size , Prostatic Hyperplasia/complications , Synaptic Transmission , Urinary Bladder/innervation , Urinary Bladder/pathology , Urinary Bladder/physiopathology , Urinary Bladder Neck Obstruction/etiology , Urinary Bladder Neck Obstruction/pathology , Urinary Bladder Neck Obstruction/physiopathology
8.
Neuroendocrinology ; 91(4): 318-23, 2010.
Article in English | MEDLINE | ID: mdl-20460879

ABSTRACT

BACKGROUND/AIMS: There is an established association between the multiple endocrine neoplasia type 1 (MEN 1) syndrome and foregut carcinoids. Some registry studies also indicate that offspring to carcinoid patients run an increased risk of developing a carcinoid tumor themselves. However, there are only scattered reports of gastrointestinal carcinoids in two generations. The aim of this study was to describe the clinical characteristics as well as the histopathological, immunohistochemical (IHC) and genetic data of metastasizing ileal carcinoids in three consecutive first-degree relatives. METHODS: The histopathological and IHC analyses were performed on newly cut sections of the tumor specimens and included growth pattern, proliferation index (Ki-67) as well as expression of established neuroendocrine markers and recently introduced cocaine-amphetamine-regulated transcript (CART). The genetic analyses were focused on establishing whether a connection with the MEN 1 syndrome existed in this family, by means of mutation screening using polymerase chain reaction, multiple ligation-dependent probe amplification, and genotyping using fluorescent-labeled microsatellite markers. RESULTS: Histopathology and IHC revealed that the tumors were virtually identical, with only minor differences in proliferation index and expression of CART. Genetic analyses indicated that the inheritance of the small bowel carcinoids in the family was not linked to the MEN1 gene. CONCLUSION: Metastasizing small bowel carcinoids have been found in first-degree relatives in three consecutive generations. All three tumors were very similar when characterized by histopathology and IHC. Based on clinical findings and genetic analyses, it seems unlikely, although not completely excluded, that inheritance was linked to the MEN 1 syndrome.


Subject(s)
Carcinoid Tumor/genetics , Carcinoid Tumor/pathology , Genetic Predisposition to Disease , Intestinal Neoplasms/genetics , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoid Tumor/metabolism , DNA Mutational Analysis , Female , Humans , Immunohistochemistry , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Male , Middle Aged , Mutation , Neoplasm Metastasis , Nerve Tissue Proteins/biosynthesis , Pedigree , Polymerase Chain Reaction , Proto-Oncogene Proteins/genetics
9.
Endocrinology ; 150(7): 3049-57, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19299455

ABSTRACT

Lipids have been shown to play a dual role in pancreatic beta-cells: a lipid-derived signal appears to be necessary for glucose-stimulated insulin secretion, whereas lipid accumulation causes impaired insulin secretion and apoptosis. The ability of the protein perilipin to regulate lipolysis prompted an investigation of the presence of perilipin in the islets of Langerhans. In this study evidence is presented for perilipin expression in rat, mouse, and human islets of Langerhans as well as the rat clonal beta-cell line INS-1. In rat and mouse islets, perilipin was verified to be present in beta-cells. To examine whether the development of lipotoxicity could be prevented by manipulating the conditions for lipid storage in the beta-cell, INS-1 cells with adenoviral-mediated overexpression of perilipin were exposed to lipotoxic conditions for 72 h. In cells exposed to palmitate, perilipin overexpression caused increased accumulation of triacylglycerols and decreased lipolysis compared with control cells. Whereas glucose-stimulated insulin secretion was retained after palmitate exposure in cells overexpressing perilipin, it was completely abolished in control beta-cells. Thus, overexpression of perilipin appears to confer protection against the development of beta-cell dysfunction after prolonged exposure to palmitate by promoting lipid storage and limiting lipolysis.


Subject(s)
Insulin-Secreting Cells/metabolism , Phosphoproteins/metabolism , Adult , Aged , Animals , Carrier Proteins , Cell Line , Female , Humans , Lipolysis/drug effects , Male , Mice , Middle Aged , Palmitates/pharmacology , Perilipin-1 , Rats , Rats, Wistar
10.
Endocrinology ; 150(6): 2577-85, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19213833

ABSTRACT

G protein-coupled receptor (GPR)-39 is a seven-transmembrane receptor expressed mainly in endocrine and metabolic tissues that acts as a Zn(++) sensor signaling mainly through the G(q) and G(12/13) pathways. The expression of GPR39 is regulated by hepatocyte nuclear factor (HNF)-1alpha and HNF-4alpha, and in the present study, we addressed the importance of GPR39 for glucose homeostasis and pancreatic islets function. The expression and localization of GPR39 were characterized in the endocrine pancreas and pancreatic cell lines. Gpr39(-/-) mice were studied in vivo, especially in respect of glucose tolerance and insulin sensitivity, and in vitro in respect of islet architecture, gene expression, and insulin secretion. Gpr39 was down-regulated on differentiation of the pluripotent pancreatic cell line AR42J cells toward the exocrine phenotype but was along with Pdx-1 strongly up-regulated on differentiation toward the endocrine phenotype. Immunohistochemistry demonstrated that GRP39 is localized selectively in the insulin-storing cells of the pancreatic islets as well as in the duct cells of the exocrine pancreas. Gpr39(-/-) mice displayed normal insulin sensitivity but moderately impaired glucose tolerance both during oral and iv glucose tolerance tests, and Gpr39(-/-) mice had decreased plasma insulin response to oral glucose. Islet architecture was normal in the Gpr39 null mice, but expression of Pdx-1 and Hnf-1alpha was reduced. Isolated, perifused islets from Gpr39 null mice secreted less insulin in response to glucose stimulation than islets from wild-type littermates. It is concluded that GPR39 is involved in the control of endocrine pancreatic function, and it is suggested that this receptor could be a novel potential target for the treatment of diabetes.


Subject(s)
Glucose/metabolism , Homeostasis/physiology , Islets of Langerhans/physiopathology , Receptors, G-Protein-Coupled/genetics , Animals , Cells, Cultured , Disease Models, Animal , Female , Glucose/pharmacology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Homeodomain Proteins/metabolism , Insulin/blood , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Zinc/metabolism
11.
Nat Genet ; 41(1): 82-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19060908

ABSTRACT

Genome-wide association studies have shown that variation in MTNR1B (melatonin receptor 1B) is associated with insulin and glucose concentrations. Here we show that the risk genotype of this SNP predicts future type 2 diabetes (T2D) in two large prospective studies. Specifically, the risk genotype was associated with impairment of early insulin response to both oral and intravenous glucose and with faster deterioration of insulin secretion over time. We also show that the MTNR1B mRNA is expressed in human islets, and immunocytochemistry confirms that it is primarily localized in beta cells in islets. Nondiabetic individuals carrying the risk allele and individuals with T2D showed increased expression of the receptor in islets. Insulin release from clonal beta cells in response to glucose was inhibited in the presence of melatonin. These data suggest that the circulating hormone melatonin, which is predominantly released from the pineal gland in the brain, is involved in the pathogenesis of T2D. Given the increased expression of MTNR1B in individuals at risk of T2D, the pathogenic effects are likely exerted via a direct inhibitory effect on beta cells. In view of these results, blocking the melatonin ligand-receptor system could be a therapeutic avenue in T2D.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Insulin/metabolism , Polymorphism, Single Nucleotide/genetics , Receptor, Melatonin, MT2/genetics , Receptors, Melatonin/genetics , Aged , Animals , Cohort Studies , Female , Gene Expression Regulation , Humans , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Middle Aged , Protein Transport , Rats , Receptor, Melatonin, MT2/metabolism , Receptors, Melatonin/metabolism
12.
J Mol Histol ; 39(6): 635-42, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18998217

ABSTRACT

Rosiglitazone, peroxisome proliferator-activated receptor-gamma agonist, is an insulin sensitizing agent in peripheral tissues. This study investigated islet hormones and hormone-like peptides expression patterns in rosiglitazone treated streptozotocin (STZ)-diabetic rats by using immunohistochemistry and in situ hybridization methods. Animals were divided into four groups. I. Group: Intact control rats. II. Group: Rosiglitazone-treated controls. III. Group: STZ-diabetic rats. IV. Group: Rosiglitazone-treated diabetic animals. Rosiglitazone was given for 7 days at a dose of 20 mg/kg body weight. In the STZ-diabetic group, there were significant differences in islet hormones and hormone like peptides cell numbers compared to rosiglitazone control group and intact control group. There were significant differences in cocaine- and amphetamine-regulated transcript (CART) and pancreatic polypeptide (PP) cell numbers between rosiglitazone control group and rosiglitazone + STZ-diabetic group. We detected a significant decrease in glucagon mRNA signals in rosiglitazone-treated control group compared to intact controls. We found a statistically significant difference in islet amyloid polypeptide (IAPP) mRNA signals between the STZ-diabetic group and the rosiglitazone + STZ-diabetic group. Besides, we also demonstrated co-localization of peptides by using double and triple histochemistry. In conclusion, our results show that short-term rosiglitazone treatment had a preservative effect to some extent on the expression of islet hormones and hormone-like peptides to maintain the islet function.


Subject(s)
Hormones/metabolism , Hypoglycemic Agents/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , PPAR gamma/agonists , Peptides/metabolism , Thiazolidinediones/pharmacology , Animals , Diabetes Mellitus, Experimental , Immunohistochemistry , In Situ Hybridization , Islets of Langerhans/cytology , Male , Rats , Rats, Wistar , Rosiglitazone
13.
Pancreas ; 37(3): 288-94, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18815551

ABSTRACT

UNLABELLED: To develop radioimmunoassays (RIAs) for rat trypsinogens 1 and 2 and to investigate the effect of nicotine exposure on concentration and production of pancreatic zymogens in the rat. METHODS: Male Sprague-Dawley rats were supplied with either normal or nicotine-containing (0.77 mM) water for 28 days and were then killed. Rabbit antibodies for the activation peptides of trypsinogens 1 and 2 were obtained for use in the RIAs. Concentrations of the both trypsinogens in pancreatic extracts were measured by the RIAs after activation by enterokinase. DNA and amylase were measured using commercial kits. mRNA for trypsinogens 1 and 2, procolipase, and cholecystokinin receptor was measured by in situ hybridization. RESULTS: The specificity of the RIA for the trypsinogen 1 activation peptide was satisfactory. The RIA for the trypsinogen 2 activation peptide showed a limited cross-reaction toward the synthetic trypsinogen 1 activation peptide, but the importance of this cross-reaction was moderate when investigated in samples of activated trypsinogens. Weight gain was reduced in nicotine-treated animals. Concentrations of amylase, trypsinogen 1, trypsinogen 2, and the ratio of trypsinogen 2 to 1 were all increased in pancreatic extracts of nicotine-fed animals. Total DNA and mRNA for the trypsinogens, procolipase, and cholecystokinin receptor were not affected by nicotine exposure. CONCLUSIONS: The combination of increased proenzyme concentrations and unaffected mRNA levels suggests that nicotine impairs secretion rather than production of pancreatic zymogens.


Subject(s)
Amylases/metabolism , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Pancreas/drug effects , Trypsinogen/metabolism , Animals , Colipases/metabolism , Enteropeptidase/metabolism , Enzyme Precursors/metabolism , In Situ Hybridization , Male , Oligopeptides/metabolism , Pancreas/enzymology , Pancreas/metabolism , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/metabolism , Signal Processing, Computer-Assisted , Time Factors , Trypsinogen/genetics , Up-Regulation
14.
Biochem Biophys Res Commun ; 376(3): 558-62, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18804448

ABSTRACT

The observations that hormone-sensitive lipase (HSL) is located in close association to insulin granules in beta-cells and that cholesterol ester hydrolase activity is completely blunted in islets of HSL null mice made us hypothesize that the role of HSL in beta-cells is to provide cholesterol for the exocytosis of insulin. To test this hypothesis, wild type (wt) and HSL null islets were depleted of plasma membrane cholesterol using methyl-beta-cyclodextrin (mbetacd). A significant reduction in insulin secretion from HSL null islets was observed whereas wt islets were unaffected. Using synaptosomal protein of 25 kDa (SNAP-25) as indicator of cholesterol-rich microdomains, confocal microscopy was used to show that HSL null beta-cells treated with mbetacd contained fewer clusters than wt beta-cells. These results indicate that HSL plays an important role in insulin secretion by providing free cholesterol for the formation and maintenance of cholesterol-rich patches for docking of SNARE-proteins to the plasma membrane.


Subject(s)
Cholesterol/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Sterol Esterase/physiology , Synaptosomal-Associated Protein 25/metabolism , Animals , Cationic Amino Acid Transporter 1/biosynthesis , Cell Membrane/metabolism , Exocytosis/drug effects , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Mice , Mice, Knockout , Sterol Esterase/genetics , Synaptosomal-Associated Protein 25/analysis , beta-Cyclodextrins/pharmacology
15.
PLoS One ; 3(3): e1793, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18335062

ABSTRACT

BACKGROUND: Hormone-sensitive lipase (HSL) is expressed predominantly in adipose tissue, where it plays an important role in catecholamine-stimulated hydrolysis of stored tri- and diglycerides, thus mobilizing fatty acids. HSL exhibits broad substrate specificity and besides acylglycerides it hydrolyzes cholesteryl esters, retinyl esters and lipoidal esters. Despite its role in fatty acid mobilization, HSL null mice have been shown to be resistant to diet-induced obesity. METHODOLOGY/PRINCIPAL FINDINGS: Following a high-fat diet (HFD) regimen, energy expenditure, measured using indirect calorimetry, was increased in HSL null mice. White adipose tissue of HSL null mice was characterized by reduced mass and reduced protein expression of PPARgamma, a key transcription factor in adipogenesis, and stearoyl-CoA desaturase 1, the expression of which is known to be positively correlated to the differentiation state of the adipocyte. The protein expression of uncoupling protein-1 (UCP-1), the highly specific marker of brown adipocytes, was increased 7-fold in white adipose tissue of HSL null mice compared to wildtype littermates. Transmission electron microscopy revealed an increase in the size of mitochondria of white adipocytes of HSL null mice. The mRNA expression of pRb and RIP140 was decreased in isolated white adipocytes, while the expression of UCP-1 and CPT1 was increased in HSL null mice compared to wildtype littermates. Basal oxygen consumption was increased almost 3-fold in white adipose tissue of HSL null mice and was accompanied by increased uncoupling activity. CONCLUSIONS: These data suggest that HSL is involved in the determination of white versus brown adipocytes during adipocyte differentiation The exact mechanism(s) underlying this novel role of HSL remains to be elucidated, but it seems clear that HSL is required to sustain normal expression levels of pRb and RIP140, which both promote differentiation into the white, rather than the brown, adipocyte lineage.


Subject(s)
Adipocytes/cytology , Adipose Tissue, Brown/cytology , Sterol Esterase/physiology , Animals , Cell Differentiation , Diet , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Obesity/genetics , Sterol Esterase/genetics
16.
Histochem Cell Biol ; 129(5): 623-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18259770

ABSTRACT

Neurodegenerative disorders are often associated with metabolic alterations. This has received little attention, but might be clinically important because it can contribute to symptoms and influence the course of the disease. Patients with Huntington's disease (HD) exhibit increased incidence of diabetes mellitus (DM). This is replicated in mouse models of HD, e.g., the R6/2 mouse, in which DM is primarily caused by a deficiency of beta-cells with impaired insulin secretion. Pancreatic tissue from HD patients has previously not been studied and, thus, the pathogenesis of DM in HD is unclear. To address this issue, we examined pancreatic tissue sections from HD patients at different disease stages. We found that the pattern of insulin immunostaining, levels of insulin transcripts and islet beta-cell area were similar in HD patients and controls. Further, there was no sign of amyloid deposition in islets from HD patients. Thus, our data show that pancreatic islets in HD patients appear histologically normal. Functional studies of HD patients with respect to insulin secretion and islet function are required to elucidate the pathogenesis of DM in HD. This may lead to a better understanding of HD and provide novel therapeutic targets for symptomatic treatment in HD.


Subject(s)
Huntington Disease/pathology , Insulin-Secreting Cells/pathology , Insulin/metabolism , Islets of Langerhans/pathology , Adult , Aged , Aged, 80 and over , Congo Red/chemistry , Female , Gene Expression , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Immunochemistry , In Situ Hybridization , Insulin/genetics , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Middle Aged , Peptide Hormones/genetics , Peptide Hormones/metabolism , Transcription, Genetic
17.
BMC Neurosci ; 8: 51, 2007 Jul 11.
Article in English | MEDLINE | ID: mdl-17625001

ABSTRACT

BACKGROUND: The peptide CART is widely expressed in central and peripheral neurons, as well as in endocrine cells. Known peripheral sites of expression include the gastrointestinal (GI) tract, the pancreas, and the adrenal glands. In rodent pancreas CART is expressed both in islet endocrine cells and in nerve fibers, some of which innervate the islets. Recent data show that CART is a regulator of islet hormone secretion, and that CART null mutant mice have islet dysfunction. CART also effects GI motility, mainly via central routes. In addition, CART participates in the regulation of the hypothalamus-pituitary-adrenal-axis. We investigated CART expression in porcine pancreas, GI-tract, adrenal glands, and thyroid gland using immunocytochemistry. RESULTS: CART immunoreactive (IR) nerve cell bodies and fibers were numerous in pancreatic and enteric ganglia. The majority of these were also VIP IR. The finding of intrinsic CART containing neurons indicates that pancreatic and GI CART IR nerve fibers have an intrinsic origin. No CART IR endocrine cells were detected in the pancreas or in the GI tract. The adrenal medulla harboured numerous CART IR endocrine cells, most of which were adrenaline producing. In addition CART IR fibers were frequently seen in the adrenal cortex and capsule. The capsule also contained CART IR nerve cell bodies. The majority of the adrenal CART IR neuronal elements were also VIP IR. CART IR was also seen in a substantial proportion of the C-cells in the thyroid gland. The majority of these cells were also somatostatin IR, and/or 5-HT IR, and/or VIP IR. CONCLUSION: CART is a major neuropeptide in intrinsic neurons of the porcine GI-tract and pancreas, a major constituent of adrenaline producing adrenomedullary cells, and a novel peptide of the thyroid C-cells. CART is suggested to be a regulatory peptide in the porcine pancreas, GI-tract, adrenal gland and thyroid.


Subject(s)
Ganglia, Autonomic/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurosecretory Systems/physiology , Sus scrofa/metabolism , Viscera/innervation , Adrenal Glands/cytology , Adrenal Glands/innervation , Adrenal Glands/physiology , Adrenal Medulla/cytology , Adrenal Medulla/innervation , Adrenal Medulla/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Female , Ganglia, Autonomic/cytology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiology , Immunohistochemistry , Male , Neurons/cytology , Pancreas/cytology , Pancreas/innervation , Pancreas/physiology , Serotonin/metabolism , Somatostatin/metabolism , Sus scrofa/anatomy & histology , Thyroid Gland/cytology , Thyroid Gland/innervation , Thyroid Gland/physiology , Vasoactive Intestinal Peptide/metabolism , Viscera/cytology , Viscera/physiology
18.
J Clin Endocrinol Metab ; 92(9): 3573-81, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17595255

ABSTRACT

CONTEXT: Ghrelin is a novel hormone produced mainly in the gastric body. Hitherto, mapping studies of ghrelin cells covering the entire gastrointestinal (GI) tract in humans have been lacking. Furthermore, the phenotype of extragastric ghrelin cells is not known. OBJECTIVE: The objective of the study was to perform a detailed mapping with specimens from all parts of the GI tract, and colocalization studies to phenotype ghrelin cells along the tract. In addition, mapping of ghrelin cells was performed in porcine GI tract, and the plasma profiles of ghrelin and motilin in blood from the porcine intestine were measured. DESIGN: Biopsies from patients were obtained during gastroscopy or surgery. Ghrelin cell density and phenotyping was assessed with immunocytochemistry, in situ hybridization, and immunogold electron microscopy. Plasma ghrelin and motilin levels were measured in pigs, fitted with cannulas in the mesenteric vein. RESULTS: The upper small intestine is unexpectedly rich in ghrelin cells, and these cells contribute to circulating ghrelin. Ghrelin and motilin are coproduced in the same cells in the duodenum and jejunum of both species, and ghrelin and motilin are stored in all secretory granules of such cells in humans, indicating cosecretion. The plasma profiles of ghrelin and motilin in pig were parallel, and a correlation between ghrelin and motilin (r(2) = 0.22; P < 0.001) was evident in intestinal blood. CONCLUSIONS: The upper small intestine is an important source of ghrelin. The likely cosecretion of intestinal ghrelin and motilin suggests concerted actions of the two hormones. These data may have implications for understanding gut motility and clinical implications for dysmotility and bariatric surgery.


Subject(s)
Enteroendocrine Cells/metabolism , Intestine, Small/metabolism , Motilin/metabolism , Peptide Hormones/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Female , Gastric Mucosa/metabolism , Gastrointestinal Motility/physiology , Ghrelin , Humans , Intestine, Small/cytology , Male , Middle Aged , Swine
19.
Regul Pept ; 143(1-3): 97-103, 2007 Oct 04.
Article in English | MEDLINE | ID: mdl-17482289

ABSTRACT

Inhibition of dipeptidyl peptidase-4 (DPP-4) is currently explored as a novel therapy of type 2 diabetes. The strategy has been shown to improve glycemia in most, but not all, rodent forms of glucose intolerance. In this study, we explored the effects of DPP-4 inhibition in mice with beta-cell overexpression of human islet amyloid polypeptide (IAPP). We therefore administered the orally active and highly selective DPP-4 inhibitor, vildagliptin (3 micromol/mouse daily) to female mice with beta-cell overexpression of human IAPP. Controls were given plain water, and a series of untreated wildtype mice was also included. After five weeks, an intravenous glucose tolerance test showed improved glucose disposal and a markedly enhanced insulin response in mice treated with vildagliptin. After eight weeks, a gastric tolerance test showed that vildagliptin improved glucose tolerance and markedly (approximately ten-fold) augmented the insulin response in association with augmented (approximately five-fold) levels of intact glucagon-like peptide-1 (GLP-1). Furthermore, after nine weeks, islets were isolated. Islets from vildagliptin-treated mice showed augmented glucose-stimulated insulin response and a normalization of the islet insulin content, which was reduced by approximately 50% in transgenic controls versus wildtype animals. Double immunostaining of pancreatic islets for insulin and glucagon revealed that transgenic islets displayed severely disturbed intra-islet topography with frequently observed centrally located alpha-cells. Treatment with vildagliptin restored the islet topography. We therefore conclude that DPP-4 inhibition improves islet function and islet topography in mice with beta-cell specific transgenic overexpression of human IAPP.


Subject(s)
Amyloid/metabolism , Dipeptidyl-Peptidase IV Inhibitors , Glucagon-Like Peptide 1/pharmacology , Insulin/pharmacology , Islets of Langerhans/drug effects , Adamantane/analogs & derivatives , Adamantane/pharmacology , Amyloid/genetics , Animals , Dipeptidyl Peptidase 4/metabolism , Female , Glucose/metabolism , Glucose Tolerance Test , Humans , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Mice, Transgenic , Nitriles/pharmacology , Pyrrolidines/pharmacology , Vildagliptin
20.
Eur J Neurosci ; 25(1): 213-23, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17241282

ABSTRACT

The system that regulates insulin secretion from beta-cells in the islet of Langerhans has a capsaicin-sensitive inhibitory component. As calcitonin gene-related peptide (CGRP)-expressing primary sensory fibers innervate the islets, and a major proportion of the CGRP-containing primary sensory neurons is sensitive to capsaicin, the islet-innervating sensory fibers may represent the capsaicin-sensitive inhibitory component. Here, we examined the expression of the capsaicin receptor, vanilloid type 1 transient receptor potential receptor (TRPV1) in CGRP-expressing fibers in the pancreatic islets, and the effect of selective elimination of capsaicin-sensitive primary afferents on the decline of glucose homeostasis and insulin secretion in Zucker diabetic fatty (ZDF) rats, which are used to study various aspects of human type 2 diabetes mellitus. We found that CGRP-expressing fibers in the pancreatic islets also express TRPV1. Furthermore, we also found that systemic capsaicin application before the development of hyperglycemia prevents the increase of fasting, non-fasting, and mean 24-h plasma glucose levels, and the deterioration of glucose tolerance assessed on the fifth week following the injection. These effects were accompanied by enhanced insulin secretion and a virtually complete loss of CGRP- and TRPV1-coexpressing islet-innervating fibers. These data indicate that CGRP-containing fibers in the islets are capsaicin sensitive, and that elimination of these fibers contributes to the prevention of the deterioration of glucose homeostasis through increased insulin secretion in ZDF rats. Based on these data we propose that the activity of islet-innervating capsaicin-sensitive fibers may have a role in the development of reduced insulin secretion in human type 2 diabetes mellitus.


Subject(s)
Capsaicin/pharmacology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Insulin/deficiency , Islets of Langerhans/cytology , Nerve Fibers/drug effects , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Calcitonin Gene-Related Peptide/metabolism , Disease Models, Animal , Glucose Tolerance Test/methods , Glycated Hemoglobin/metabolism , Immunohistochemistry/methods , Insulin/blood , Male , Nerve Fibers/physiology , Rats , Rats, Zucker , TRPV Cation Channels/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...