Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Morphol ; 279(6): 724-746, 2018 06.
Article in English | MEDLINE | ID: mdl-29464754

ABSTRACT

We analyzed the development of the pancreatic ducts in grass snake Natrix natrix L. embryos with special focus on the three-dimensional (3D)-structure of the duct network, ultrastructural differentiation of ducts with attention to cell types and lumen formation. Our results indicated that the system of ducts in the embryonic pancreas of the grass snake can be divided into extralobular, intralobular, and intercalated ducts, similarly as in other vertebrate species. However, the pattern of branching was different from that in other vertebrates, which was related to the specific topography of the snake's internal organs. The process of duct remodeling in Natrix embryos began when the duct walls started to change from multilayered to single-layered and ended together with tube formation. It began in the dorsal pancreatic bud and proceeded toward the caudal direction. The lumen of pancreatic ducts differentiated by cavitation because a population of centrally located cells was cleared through cell death resembling anoikis. During embryonic development in the pancreatic duct walls of the grass snake four types of cells were present, that is, principal, endocrine, goblet, and basal cells, which is different from other vertebrate species. The principal cells were electron-dense, contained indented nuclei with abundant heterochromatin, microvilli and cilia, and were connected by interdigitations of lateral membranes and junctional complexes. The endocrine cells were electron-translucent and some of them included endocrine granules. The goblet cells were filled with large granules with nonhomogeneous, moderately electron-dense material. The basal cells were small, electron-dense, and did not reach the duct lumen.


Subject(s)
Colubridae/embryology , Embryonic Development , Pancreas, Exocrine/embryology , Animals , Pancreas, Exocrine/ultrastructure
2.
PLoS One ; 12(6): e0179398, 2017.
Article in English | MEDLINE | ID: mdl-28617859

ABSTRACT

To search for clues suggesting that beta cells may generate by transdifferentiation in humans, we assessed the presence of cells double positive for exocrine (amylase, carboxypeptidase A) and endocrine (insulin) markers in the pancreas of non-diabetic individuals (ND) and patients with type 2 diabetes (T2D). Samples from twelve ND and twelve matched T2D multiorgan donors were studied by electron microscopy, including amylase and insulin immunogold labeling; carboxypeptidase A immunofluorescence light microscopy assessment was also performed. In the pancreas from four T2D donors, cells containing both zymogen-like and insulin-like granules were observed, scattered in the exocrine compartment. Nature of granules was confirmed by immunogold labeling for amylase and insulin. Double positive cells ranged from 0.82 to 1.74 per mm2, corresponding to 0.26±0.045% of the counted exocrine cells. Intriguingly, cells of the innate immune systems (mast cells and/or macrophages) were adjacent to 33.3±13.6% of these hybrid cells. No cells showing co-localization of amylase and insulin were found in ND samples by electron microscopy. Similarly, cells containing both carboxypeptidase A and insulin were more frequently observed in the diabetic pancreata. These results demonstrate more abundant presence of cells containing both acinar markers and insulin in the pancreas of T2D subjects, which suggests possible conversion from one cellular type to the other and specific association with the diseased condition.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Pancreas, Exocrine/metabolism , Aged , Biomarkers/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Humans , Male , Middle Aged , Pancreas, Exocrine/ultrastructure
3.
Adv Exp Med Biol ; 913: 163-176, 2016.
Article in English | MEDLINE | ID: mdl-27796887

ABSTRACT

Stroma is viewed as the supportive framework of a predominant epithelial organ, comprising mostly of connective tissue, blood vessels and nerves. Since the discovery of telocytes one decade ago (Popescu and Faussone-Pellegrini J Cell Mol Med 2010;14(4):729-40), their presence was proven in several exocrine gland stromata, including major and minor salivary glands, mammary glands as well as exocrine pancreas.Telocytes have been found in a close connection with acinar and ductal structures but also with their stromal neighbours - nerves, blood vessels or other connective elements, either cells or collagen fibres.The approaches used to reveal the telocytes' location were immunohistochemistry and electron microscopy.


Subject(s)
Mammary Glands, Human/ultrastructure , Pancreas, Exocrine/ultrastructure , Salivary Glands/ultrastructure , Telocytes/ultrastructure , Acinar Cells/metabolism , Acinar Cells/ultrastructure , Animals , Blood Vessels/metabolism , Blood Vessels/ultrastructure , Connective Tissue/metabolism , Connective Tissue/ultrastructure , Humans , Immunohistochemistry , Mammary Glands, Human/metabolism , Microscopy, Electron , Pancreas, Exocrine/metabolism , Rats , Salivary Glands/metabolism , Telocytes/metabolism
4.
Hum Pathol ; 54: 127-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27063474

ABSTRACT

Microscopic pale-staining acinar nodules were characterized in native pancreas in the 1980s under a variety of names but have been infrequently reported since. We retrospectively studied the frequency and characteristics of pale acinar nodules in allograft pancreas biopsies, as compared to a sampling of native pancreas specimens at our center. Pale acinar nodules were present in 13% (9/69) of allograft biopsies from 22% (7/32) of transplant patients, and 23% (5/22) of native pancreas surgical specimens, although more nodules per pancreas area were present in allograft needle biopsies. Acinar nodules had size of 100 to 700 µm, were periodic acid-Schiff pale, were synaptophysin negative, stained more weakly with keratin CAM 5.2 compared to surrounding parenchyma, and had a low proliferative rate. Ultrastructural evaluation revealed paucity of zymogen granules with dilated cistern-like structures. In our experience, pale acinar nodules have similar features in allograft and native pancreas specimens, yet remain of uncertain etiology and significance.


Subject(s)
Acinar Cells/ultrastructure , Pancreas Transplantation/adverse effects , Pancreas, Exocrine/ultrastructure , Acinar Cells/chemistry , Acinar Cells/transplantation , Allografts , Biomarkers/analysis , Biopsy, Needle , Humans , Immunohistochemistry , Keratins/analysis , Ki-67 Antigen/analysis , Microscopy, Electron, Transmission , Pancreas, Exocrine/chemistry , Pancreas, Exocrine/surgery , Retrospective Studies , Synaptophysin/analysis , Treatment Outcome
5.
PLoS One ; 11(2): e0148458, 2016.
Article in English | MEDLINE | ID: mdl-26895040

ABSTRACT

The aim of the present study was to investigate the pancreatic exocrine function in a canine model and to analyze the changes in organelles of pancreatic acinar cells during the early stage of acute pancreatitis (AP). AP was induced by retrograde injection of 5% sodium taurocholate (0.5 ml/kg) into the main pancreatic duct of dogs. The induction of AP resulted in serum hyperamylasemia and a marked reduction of amylase activity in the pancreatic fluid (PF). The pancreatic exocrine function was markedly decreased in subjects with AP compared with the control group. After the induction of AP, histological examination showed acinar cell edema, cytoplasmic vacuolization, fibroblasts infiltration, and inflammatory cell infiltration in the interstitium. Electron micrographs after the induction of AP revealed that most of the rough endoplasmic reticulum (RER) were dilated and that some of the ribosomes were no longer located on the RER. The mitochondria were swollen, with shortened and broken cristae. The present study demonstrated, in a canine model, a reduced volume of PF secretion with decreased enzyme secretion during the early stage of AP. Injury of mitochondria and dilatation and degranulation of RER may be responsible for the reduced exocrine function in AP. Furthermore, the present model and results may be useful for researching novel therapeutic measures in AP.


Subject(s)
Organelles/metabolism , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Pancreatitis/metabolism , Pancreatitis/pathology , Acute Disease , Amylases/biosynthesis , Amylases/blood , Animals , Bicarbonates/metabolism , Disease Models, Animal , Dogs , Extracellular Fluid/metabolism , Lipase/biosynthesis , Lipase/blood , Organelles/pathology , Organelles/ultrastructure , Pancreas, Exocrine/ultrastructure , Pancreatitis/blood
6.
Biochem Biophys Res Commun ; 471(4): 610-5, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26845357

ABSTRACT

Small G protein Rab27B is expressed in various secretory cell types and plays a role in mediating secretion. In pancreatic acinar cells, Rab27B was found to be expressed on the zymogen granule membrane and by overexpression to regulate the secretion of zymogen granules. However, the effect of Rab27B deletion on the physiology of pancreatic acinar cells is unknown. In the current study, we utilized the Rab27B KO mouse model to better understand the role of Rab27B in the secretion of pancreatic acinar cells. Our data show that Rab27B deficiency had no obvious effects on the expression of major digestive enzymes and other closely related proteins, e.g. similar small G proteins, such as Rab3D and Rab27A, and putative downstream effectors. The overall morphology of acinar cells was not changed in the knockout pancreas. However, the size of zymogen granules was decreased in KO acinar cells, suggesting a role of Rab27B in regulating the maturation of secretory granules. The secretion of digestive enzymes was moderately decreased in KO acini, compared with the WT control. These data indicate that Rab27B is involved at a different steps of zymogen granule maturation and secretion, which is distinct from that of Rab3D.


Subject(s)
Pancreas, Exocrine/enzymology , Pancreatic alpha-Amylases/metabolism , Secretory Vesicles/enzymology , rab GTP-Binding Proteins/physiology , Acinar Cells , Animals , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas, Exocrine/ultrastructure , Secretory Vesicles/ultrastructure , rab GTP-Binding Proteins/biosynthesis , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins , rab3 GTP-Binding Proteins/biosynthesis
7.
Fiziol Zh (1994) ; 62(6): 88-94, 2016.
Article in English, Ukrainian | MEDLINE | ID: mdl-29762976

ABSTRACT

Morphological changes of the pancreas of young rats after exposure of exogenous melatonin in the spring and autumn periods was investigated. Exogenous melatonin (Unipharm Inc., USA) was administered to experimental group of animals daily'at a dose 5 mg/kg. The duration of the experiment was 28 days. It was revealed that the exocrine part of the.pancreas responds differently to the effects of melatonin at different times of the year. Thus, after administration of melatonin in the spring increase of the size of acinus, the height of the epithelium (by 7 %), area exocrinocytes (by 58 %), of their nucleus (by 20 %) and cytoplasm (69 %), the amount of nucleoli in cells (18 %), reduction the amount of connective tissue elements. Melatonin introduction in the autumn decrease in the size of acinus, height and area of exocrinocytes, growth the number of exocrinocytes in the acinus, nucleoli and width layers interlobular connective tissue in the gland. This may indicate that melatonin increases in the spring of the synthetic activity of the exocrine pancreas, whereas in the autumn (for the majority of the morphometric parameters) - somewhat reduces its functional state. The administration of melatonin in the spring (mostly) and in the autumn periods increased the functional activity of the endocrine pancreas. This is indicated by growth in the size of Langerhans islets, increasing the number and density of the (autumn) endocrinocytes.


Subject(s)
Antioxidants/pharmacology , Islets of Langerhans/drug effects , Melatonin/pharmacology , Pancreas, Exocrine/drug effects , Animals , Antioxidants/administration & dosage , Islets of Langerhans/physiology , Islets of Langerhans/ultrastructure , Male , Melatonin/administration & dosage , Pancreas, Exocrine/physiology , Pancreas, Exocrine/ultrastructure , Rats, Wistar , Seasons
8.
Cell Calcium ; 58(3): 237-45, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26100948

ABSTRACT

Intracellular Ca(2+) release is mostly mediated by inositol trisphosphate, but intracellular cyclic-ADP-ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP) are important messengers in many systems. Whereas cADPR generally activates type 2 ryanodine receptors (RyR2s), the NAADP-activated Ca(2+) release mechanism is less clear. Using knockouts and antibodies against RyRs and Two-Pore Channels (TPCs), we have compared their relative importance for NAADP-induced Ca(2+) release from two-photon permeabilized pancreatic acinar cells. In these cells, cholecystokinin-elicited Ca(2+) release is mediated by NAADP. TPC2-KO reduced NAADP-induced Ca(2+) release by 64%, but the combination of TPC2-KO and an antibody against TPC1, significantly reduced Ca(2+) release by 86% (64% vs. 86%, p<0.0002). In RyR3-KO, NAADP-evoked Ca(2+) release reduced by ∼50% but, when combined with antibodies against RyR1, responses were 90% inhibited. Antibodies against RyR2 had practically no effect on NAADP-evoked Ca(2+) release, but reduced release in response to cADPR by 55%. Antibodies to RyR1 inhibited NAADP-induced Ca(2+) liberation by 81%, but only reduced cADPR responses by 30%. We conclude that full NAADP-mediated Ca(2+) release requires both TPCs and RyRs. The sequence of relative importance for NAADP-elicited Ca(2+) release from the all stores is RyR1>TPC2>RyR3>TPC1>>RyR2. However, when assessing NAADP-induced Ca(2+) release solely from the acidic stores (granules/endosomes/lysosomes), antibodies against TPC2 and TPC1 virtually abolished the Ca(2+) liberation as did antibodies against RyR1 and RyR3. Our results indicate that the primary, but very small, NAADP-elicited Ca(2+) release via TPCs from endosomes/lysosomes triggers the detectable Ca(2+)-induced Ca(2+) release via RyR1 and RyR3 occurring from the granules and the ER.


Subject(s)
Calcium Channels/physiology , Calcium Signaling , Calcium/metabolism , NADP/analogs & derivatives , Pancreas, Exocrine/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Acinar Cells/metabolism , Acinar Cells/ultrastructure , Animals , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Mice , Mice, Knockout , NADP/metabolism , Pancreas, Exocrine/ultrastructure
9.
Proc Natl Acad Sci U S A ; 111(5): E582-91, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24453213

ABSTRACT

Suppressor/Enhancer of Lin-12-like (Sel1L) is an adaptor protein for the E3 ligase hydroxymethylglutaryl reductase degradation protein 1 (Hrd1) involved in endoplasmic reticulum-associated degradation (ERAD). Sel1L's physiological importance in mammalian ERAD, however, remains to be established. Here, using the inducible Sel1L knockout mouse and cell models, we show that Sel1L is indispensable for Hrd1 stability, ER homeostasis, and survival. Acute loss of Sel1L leads to premature death in adult mice within 3 wk with profound pancreatic atrophy. Contrary to current belief, our data show that mammalian Sel1L is required for Hrd1 stability and ERAD function both in vitro and in vivo. Sel1L deficiency disturbs ER homeostasis, activates ER stress, attenuates translation, and promotes cell death. Serendipitously, using a biochemical approach coupled with mass spectrometry, we found that Sel1L deficiency causes the aggregation of both small and large ribosomal subunits. Thus, Sel1L is an indispensable component of the mammalian Hrd1 ERAD complex and ER homeostasis, which is essential for protein translation, pancreatic function, and cellular and organismal survival.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Homeostasis , Mammals/metabolism , Proteins/metabolism , Animals , Atrophy , Cell Culture Techniques , Cell Death , Cell Proliferation , Cell Survival , Endoplasmic Reticulum/ultrastructure , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Pancreas, Exocrine/abnormalities , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Pancreas, Exocrine/ultrastructure , Polyribosomes/metabolism , Protein Biosynthesis , Protein Stability , Secretory Vesicles/metabolism , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response
10.
Micron ; 47: 50-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416165

ABSTRACT

It is well established that the status of the endoplasmic reticulum (ER) and mitochondria, and the interactions between them, is critical to numerous cellular functions including apoptosis. Mitochondrial dynamics is greatly influenced by cell stress, and recent studies implicate ER in mitochondrial fission. Although a number of proteins have been identified to participate in ER-induced mitochondrial fission, the molecular mechanism of the process is little understood. In the current study, we confirm the involvement of ER in mitochondrial fission and hypothesize the involvement of water channels or aquaporins (AQP) in the process. Previous studies demonstrate the presence of AQP both in the ER and mitochondrial membranes. Mitochondrial swelling has been observed following mitochondrial calcium overload, and studies report that chelation of cytosolic calcium induces extensive mitochondrial division at ER contact sites. Based on this information, the involvement of ER in mitochondrial division, possibly via water channels, is hypothesized. Utilizing a multi-faceted imaging approach consisting of atomic force microscopy on aldehyde-fixed and semi-dry cells, transmission electron microscopy, and immunofluorescence microscopy on live cells, the physical interactions between the two organelles are demonstrated. Mitochondrial fission following ER stress was abrogated with exposure of cells to the AQP inhibitor mercuric chloride, suggesting the involvement of AQP(s) especially AQP8 and AQP9 known to be present in the mitochondrial membrane, in mitochondrial fission.


Subject(s)
Aquaporins/metabolism , Endoplasmic Reticulum/physiology , Mitochondrial Dynamics/physiology , Pancreas, Exocrine , Animals , Aquaporins/pharmacology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Humans , Male , Mice , Microscopy, Electron , Microscopy, Fluorescence , Mitochondrial Dynamics/drug effects , Mitochondrial Membranes/metabolism , Pancreas, Exocrine/cytology , Pancreas, Exocrine/ultrastructure , Pancreatitis/chemically induced , Rats , Rats, Wistar
11.
Gastroenterology ; 143(3): 832-843.e7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22710192

ABSTRACT

BACKGROUND & AIMS: During development of alcoholic pancreatitis, oxidative (acetaldehyde) and nonoxidative metabolites (ethyl palmitate, ethyl oleate), rather than ethanol itself, mediate toxic injury. Exposure of pancreatic acini to ethanol blocks cholecystokinin (CCK)-8-stimulated apical exocytosis and redirects exocytosis to the basolateral plasma membrane, causing interstitial pancreatitis. We examined how each ethanol metabolite contributes to these changes in exocytosis. METHODS: Rat pancreatic acini were incubated with concentrations of ethanol associated with alcoholic pancreatitis (20-50 mmol/L) or ethanol metabolites (1-3 mmol/L) and then stimulated with CCK-8. We performed single zymogen granule (ZG) exocytosis assays, Ca(2+) imaging studies, ultrastructural analyses (with electron microscopy), and confocal microscopy to assess the actin cytoskeleton and track the movement of vesicle-associated membrane protein (VAMP)-8-containing ZGs. Coimmunoprecipitation assays were used to identify complexes that contain the distinct combinations of Munc18 and the soluble N-ethylmaleimide sensitive factor attachment protein receptor proteins, which mediate apical (ZG-apical plasma membrane) and basolateral exocytosis and fusion between ZGs (ZG-ZG). RESULTS: The ethanol metabolites acetaldehyde, ethyl palmitate, and ethyl oleate reduced CCK-8-stimulated apical exocytosis and formation of apical exocytotic complexes (between Munc18b and Syntaxin-2, synaptosomal-associated protein of 23 kilodaltons [SNAP23], and VAMP2) in rat pancreatic acini. Acetaldehyde and ethyl oleate redirected CCK-8-stimulated exocytosis to the basal and lateral plasma membranes and translocation of VAMP8-containing ZGs toward the basolateral plasma membrane. This process was mediated primarily via formation of basolateral exocytotic complexes (between Munc18c and Syntaxin-4, SNAP23, and VAMP8). Exposure of the acini to acetaldehyde and ethyl oleate followed by CCK-8 stimulation mildly perturbed the actin cytoskeleton and Ca(2+) signaling; exposure to ethyl palmitate severely affected Ca(2+) signaling. Acetaldehyde, like ethanol, promoted fusion between ZGs by the formation of ZG-ZG exocytotic complexes (between Munc18b and Syntaxin-3, SNAP23, and VAMP8), whereas ethyl palmitate and ethyl oleate reduced ZG-ZG fusion and formation of these complexes. CONCLUSIONS: The ethanol metabolites acetaldehyde, ethyl palmitate, and ethyl oleate perturb exocytosis processes in cultured rat pancreatic acini (apical blockade, basolateral exocytosis, and fusion between ZGs). Acetaldehyde and, to a lesser degree, ethyl oleate produce many of the same pathologic effects of ethanol on CCK-8-stimulated exocytosis in pancreatic acini.


Subject(s)
Amylases/metabolism , Ethanol/toxicity , Exocytosis/drug effects , Pancreas, Exocrine/drug effects , Pancreatitis, Alcoholic/etiology , Secretory Vesicles/drug effects , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Actin Cytoskeleton/metabolism , Animals , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Ethanol/metabolism , Immunoprecipitation , Male , Membrane Fusion/drug effects , Microscopy, Confocal , Microscopy, Electron, Transmission , Munc18 Proteins/metabolism , Oleic Acids/metabolism , Oleic Acids/toxicity , Palmitic Acids/metabolism , Palmitic Acids/toxicity , Pancreas, Exocrine/enzymology , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/ultrastructure , Pancreatitis, Alcoholic/enzymology , Pancreatitis, Alcoholic/pathology , Qa-SNARE Proteins/metabolism , Rats , Rats, Sprague-Dawley , Secretory Vesicles/enzymology , Secretory Vesicles/metabolism , Sincalide/pharmacology , Time Factors , Tissue Culture Techniques , Vesicle-Associated Membrane Protein 2/metabolism , Vesicular Transport Proteins/metabolism
12.
Pancreas ; 41(6): 949-56, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22318257

ABSTRACT

OBJECTIVES: Pancreatic interstitial cells are located among acini, ducts, nerves, and blood vessels. They are essential for pancreas development, physiology, and for oncogenic microenvironment. We identified cells with characteristic ultrastructural features of telocytes in pancreatic interstitium. Telocytes were initially described as interstitial Cajal-like cells, but it gradually became clear that they were a distinct novel cell type not directly related to canonical interstitial Cajal cells. METHODS: Serial ultrathin sections of human pancreatic tissue were studied by transmission electron microscopy. Computer analysis software was used to obtain 2-dimensional compositions from serial micrographs and to perform morphometry. RESULTS: Pancreatic telocytes appear as small-body cells with prolongations called telopodes. The ultrastructural features of telopodes are the following: (a) number: 1 to 3; (b) length: tens of micrometers; (c) moniliform aspect: with podoms (thicker portions) and podomers (thin segments, with a mean width of 60 nm, undetectable by light microscopy); (d) dichotomous branching forming a network; (e) establish homocellular and heterocellular junctions; (f) release of microvesicles/multivesicular bodies. Telopodes pass close to blood vessels, nerves, and pancreatic acinar cells and ducts. CONCLUSIONS: Telocytes are present as distinct interstitial cells in the exocrine pancreatic stroma. They act as important players in intercellular signaling via stromal synapses and shed vesicle transfer.


Subject(s)
Pancreas, Exocrine/ultrastructure , Stromal Cells/ultrastructure , Cell Size , Cell Surface Extensions/ultrastructure , Humans , Image Processing, Computer-Assisted , Intercellular Junctions/ultrastructure , Microscopy, Electron, Transmission , Multivesicular Bodies/ultrastructure
13.
Gastroenterology ; 142(4): 999-1009.e6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22178591

ABSTRACT

BACKGROUND & AIMS: The exocrine portion of the pancreas functions in digestion and preserves pancreatic homeostasis. Learning how this tissue forms during embryogenesis could improve our understanding of human pancreatic diseases. Expression of the homeobox gene Prox1 in the exocrine pancreas changes throughout development in mice. We investigated the role of Prox1 in development of the exocrine pancreas in mice. METHODS: Mice with pancreas-specific deletion of Prox1 (Prox1(ΔPanc)) were generated and their pancreatic tissues were analyzed using immunohistochemistry, transmission electron microscopy, histologic techniques, quantitative real-time polymerase chain reaction, immunoblotting, and morphometric analysis. RESULTS: Loss of Prox1 from the pancreas led to multiple exocrine alterations, most notably premature acinar cell differentiation, increased ductal cell proliferation, altered duct morphogenesis, and imbalanced expression of claudin proteins. Prox1(ΔPanc) mice also had some minor alterations in islet cells, but beta-cell development was not affected. The exocrine congenital defects of Prox1(ΔPanc) pancreata appeared to initiate a gradual process of deterioration that resulted in extensive loss of acinar cells, lipomatosis, and damage to ductal tissue in adult mice. CONCLUSIONS: Pancreas-specific deletion of Prox1 causes premature differentiation of acinar cells and poor elongation of epithelial branches; these defects indicate that Prox1 controls the expansion of tip progenitors in the early developing pancreas. During later stages of embryogenesis, Prox1 appears to regulate duct cell proliferation and morphogenesis. These findings identify Prox1 as an important regulator of pancreatic exocrine development.


Subject(s)
Embryonic Stem Cells/metabolism , Pancreas, Exocrine/metabolism , Tumor Suppressor Proteins/deficiency , Age Factors , Aging , Animals , Blotting, Western , Cell Differentiation , Cell Proliferation , Claudins/metabolism , Embryonic Stem Cells/ultrastructure , Gene Expression Regulation, Developmental , Genotype , Gestational Age , Homeodomain Proteins/genetics , Homeostasis , Immunohistochemistry , Islets of Langerhans/embryology , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Morphogenesis , Pancreas, Exocrine/embryology , Pancreas, Exocrine/ultrastructure , Pancreatic Ducts/embryology , Pancreatic Ducts/metabolism , Phenotype , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
14.
Pancreas ; 40(5): 715-22, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602737

ABSTRACT

OBJECTIVE: We sought to evaluate the effects of telmisartan, sitagliptin, or their combination on pancreatic ultrastructural alterations in high-fat-fed C57BL/6 mice. METHODS: Three-month-old C57BL/6 mice were fed with standard chow (SC, 10% lipids) or high-fat diet (HF, 60% lipids) during 10 weeks to induce obesity and its comorbidities. After this period, treatment began (lasted 6 weeks), and the HF group was divided into 4 subgroups: untreated HF, HF plus telmisartan (5 mg/kg per day), HF plus sitagliptin (1.1 g/kg per day), and HF plus telmisartan plus sitagliptin. Drugs were mixed with diet. Biochemical analyses, radioimmunoassay, immunofluorescence, stereology, and transmission electron microscopy were performed to assess pancreatic remodeling. RESULTS: Overweight, hyperinsulinemia, hyperglycemia, and dyslipidemia were found in the HF group, but these outcomes were controlled by the different treatments. Untreated HF animals also showed alterations concerning distribution of α/ß cell followed by large and numerous lipid droplets within pancreas. Telmisartan and sitagliptin as monotherapy alleviated these findings, and a complete reversal of pancreatic steatosis was observed after treating with the combination of the 2 drugs. CONCLUSIONS: AT1 receptor blockade, partial peroxisome proliferator-activated receptor gamma activation, and extended incretin action emerge as feasible strategies to control pancreatic steatosis and avoid progression of pancreatic diseases due to lipotoxicity.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Benzimidazoles/administration & dosage , Benzoates/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Obesity/drug therapy , Obesity/pathology , Pancreas/drug effects , Pancreas/ultrastructure , Pyrazines/administration & dosage , Triazoles/administration & dosage , Animals , Blood Glucose/metabolism , Dietary Fats/administration & dosage , Drug Therapy, Combination , Insulin/blood , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Pancreas/metabolism , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/ultrastructure , Sitagliptin Phosphate , Telmisartan
15.
J Vet Med Sci ; 73(2): 161-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20847540

ABSTRACT

Obstructive jaundice causes multiple malfunctions in various organs including the pancreas. To establish how such malfunctions occur, we experimentally induced obstructive jaundice through bile duct ligation (BDL) using rats, measured serum bilirubin, amylase and insulin levels, and examined histological, immunohistochemical and cytological changes in the pancreas at 3 days, 1 week, and 4 weeks after the BDL. Morphometrical analysis was also conducted. Serum amylase levels steeply increased at 3 days, and then decreased at 1 and 4 weeks after the BDL to lower than the control level. In contrast, the number of zymogen granules decreased at 3 days after the BDL, then increased and eventually surpassed the control group at 4 weeks after the BDL. On the other hand, serum insulin levels dramatically decreased at 3 days after the BDL but recovered to a level close to that of the control group at 1 week after the BDL. At 4 weeks after the BDL, however, the serum insulin levels again showed a marked decline. Slight decrease in insulin immunoreactivity and number of insulin granules were observed at 4 weeks after the BDL. Cholecystokinin receptors (CCK-R) were expressed in both acinar and islet cells; their immunoreactivity significantly decreased in the acinar cells at 4 weeks after the BDL. Our results suggest that CCK may play a role in regulating changes in the pancreas after obstructive jaundice.


Subject(s)
Islets of Langerhans/pathology , Jaundice, Obstructive/pathology , Pancreas, Exocrine/pathology , Amylases/blood , Animals , Bilirubin/blood , Immunohistochemistry , Insulin/blood , Islets of Langerhans/enzymology , Islets of Langerhans/ultrastructure , Jaundice, Obstructive/blood , Male , Microscopy, Electron, Transmission , Pancreas, Exocrine/enzymology , Pancreas, Exocrine/ultrastructure , Rats , Rats, Wistar , Receptors, Cholecystokinin/biosynthesis
16.
J Proteome Res ; 9(10): 4927-39, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20707389

ABSTRACT

Zymogen granules (ZG) are specialized storage organelles in the exocrine pancreas that allow the sorting, packaging, and regulated apical secretion of digestive enzymes. As there is a critical need for further understanding of the key processes in regulated secretion to develop new therapeutic options in medicine, we applied a suborganellar proteomics approach to identify peripheral membrane-associated ZG proteins. We focused on the analysis of a "basic" group (pH range 6.2-11) with about 46 spots among which 44 were identified by tandem mass spectrometry. These spots corresponded to 16 unique proteins, including rat mast cell chymase (RMCP-1) and peptidyl-prolyl cis-trans isomerase B (PpiB; cyclophilin B), an ER-resident protein. To confirm that these proteins were specific to zymogen granules and not contaminants of the preparation, we conducted a series of validation experiments. Immunoblotting of ZG subfractions revealed that chymase and PpiB behaved like bona fide peripheral membrane proteins. Their expression in rat pancreas was regulated by feeding behavior. Ultrastructural and immunofluorescence studies confirmed their ZG localization. Furthermore, a chymase-YFP fusion protein was properly targeted to ZG in pancreatic AR42J cells. Interestingly, for both proteins, proteoglycan-binding properties have been reported. The importance of our findings for sorting and packaging during ZG formation is discussed.


Subject(s)
Membrane Proteins/metabolism , Pancreas, Exocrine/metabolism , Proteomics/methods , Secretory Vesicles/metabolism , Animals , Cell Line, Tumor , Chymases/genetics , Chymases/metabolism , Cyclophilins/genetics , Cyclophilins/metabolism , Electrophoresis, Gel, Two-Dimensional , Gene Expression , Hydrogen-Ion Concentration , Immunoblotting , Membrane Proteins/genetics , Microscopy, Fluorescence , Microscopy, Immunoelectron , Pancreas, Exocrine/ultrastructure , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
17.
Toxicol Pathol ; 38(4): 658-65, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20448086

ABSTRACT

This was a study of the microscopic, ultrastructural, immunohistochemical, and enzyme cytochemical features of giant eosinophilic granules encountered in pancreatic acinar cells of alloxan-induced diabetic rats. Seven male F344 rats with diabetes induced by a single i.v. dose of alloxan were sacrificed after twenty-five weeks of treatment. Histologically, the pancreatic acini were diffusely atrophied, and the islets showed marked atrophy or had disappeared, and giant eosinophilic granules and small vacuoles were observed in almost all acinar cells. The eosinophilic granules showed negative reactions for periodic acid-Schiff (PAS) and acid phosphatase, as well as fat stains such as Nile blue, Oil red O, and Sudan III. Ultrastructurally, the giant eosinophilic granules were huge structures surrounded by a double membrane containing many irregular cristae. A large amount of small lipid droplets was also apparent in the basal area of the acinar cells. Immunohistochemical analysis of prohibitin, a kind of protein located in the mitochondrial inner membrane, was partially positive in the marginal area of some giant eosinophilic granules, but negative for the central area. The enzyme activity for succinic dehydrogenase (SDH), one of the mitochondrial enzymes, showed a localizing pattern similar to that of prohibitin. These findings confirmed that the giant eosinophilic granules in the exocrine pancreas of alloxan-induced diabetic rats were giant mitochondria.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Mitochondria/pathology , Pancreas, Exocrine/pathology , Alloxan , Animals , Blood Glucose/metabolism , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Eosinophils/metabolism , Glycosuria/blood , Glycosuria/pathology , Histocytochemistry , Male , Microscopy, Electron , Mitochondria/metabolism , Pancreas, Exocrine/ultrastructure , Prohibitins , Rats , Rats, Inbred F344 , Repressor Proteins/metabolism , Vacuoles/metabolism , Vacuoles/ultrastructure
18.
Am J Physiol Gastrointest Liver Physiol ; 298(4): G518-24, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20110462

ABSTRACT

Endogenous trypsin inhibitors are synthesized, stored, and secreted by pancreatic acinar cells. It is believed that they play a protective role in the pancreas by inhibiting trypsin within the cell should trypsinogen become prematurely activated. Rodent trypsin inhibitors are highly homologous to human serine protease inhibitor Kazal-type 1 (SPINK1). The mouse has one pancreatic trypsin inhibitor known as SPINK3, and the rat has two trypsin inhibitors commonly known as pancreatic secretory trypsin inhibitors I and II (PSTI-I and -II). Rat PSTI-I is a 61-amino acid protein that shares 65% sequence identity with mouse SPINK3. It was recently demonstrated that mice with genetic deletion of the Spink3 gene (Spink3(-/-)) do not survive beyond 15 days and lack normal pancreata because of pancreatic autophagy. We have shown that targeted transgenic expression of the rat Psti1 gene to acinar cells in mice [TgN(Psti1)] protects mice against caerulein-induced pancreatitis. To determine whether the autophagic phenotype and lethality in Spink3(-/-) mice were due to lack of pancreatic trypsin inhibitor, we conducted breeding studies with Spink3(+/-) heterozygous mice and TgN(Psti1) mice. We observed that, whereas Spink3(+/+), Spink3(+/-), and Spink3(-/-)/TgN(Psti1) mice had similar survival rates, no Spink3(-/-) mice survived longer than 1 wk. The level of expression of SPINK3 protein in acini was reduced in heterozygote mice compared with wild-type mice. Furthermore, endogenous trypsin inhibitor capacity was reduced in the pancreas of heterozygote mice compared with wild-type or knockout mice rescued with the rat Psti1 gene. Surprisingly, the lesser amount of SPINK3 present in the pancreata of heterozygote mice did not predispose animals to increased susceptibility to caerulein-induced acute pancreatitis. We propose that a threshold level of expression is sufficient to protect against pancreatitis.


Subject(s)
Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Pancreas/pathology , Pancreatitis/genetics , Prostatic Secretory Proteins/genetics , Transgenes/genetics , Amino Acid Sequence , Amylases/blood , Animals , Body Size/genetics , Ceruletide/pharmacology , Female , Glycoproteins/metabolism , Heterozygote , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Organ Size/genetics , Pancreas/drug effects , Pancreas/metabolism , Pancreas, Exocrine/pathology , Pancreas, Exocrine/ultrastructure , Pancreatitis/chemically induced , Pancreatitis/pathology , Prostatic Secretory Proteins/metabolism , Rats , Sequence Homology, Amino Acid , Survival Rate , Trypsin/metabolism , Trypsin Inhibitor, Kazal Pancreatic
19.
Cell Biol Int ; 34(1): 3-12, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20017733

ABSTRACT

In contrast with the observation in electron micrographs of partially empty vesicles in cells following secretion, it has been believed since the 1950s that during cell secretion, secretory vesicles completely merge at the cell plasma membrane, resulting in the diffusion of intravesicular contents to the cell exterior and the compensatory retrieval of the excess membrane by endocytosis. In the interim, a large body of work has been published arguing both for and against the complete merger of secretory vesicle membrane at the cell plasma membrane during secretion. The only definitive determination of the mechanism of cell secretion remained in its direct observation at nanometre resolution in live cells. In the past decade, this finally became a reality through the power and scope of the atomic force microscope, which has made it possible to resolve a major conundrum in cell biology. This paradigm shift in our understanding of cell secretion is briefly outlined here.


Subject(s)
Secretory Vesicles/physiology , Cell Membrane/physiology , Cell Membrane/ultrastructure , Membrane Fusion , Microscopy, Atomic Force , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/ultrastructure , Protein Transport , SNARE Proteins/metabolism , Secretory Vesicles/ultrastructure
20.
J Comp Pathol ; 141(1): 78-83, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19410259

ABSTRACT

Three Inland Bearded Dragons (Pogona vitticeps) from two breeding groups were humanely destroyed following a period of anorexia. Two of the animals were 8-months old and related and one animal was approximately 2-weeks old. Necropsy examination revealed poor bodily condition but no other gross abnormalities. Microscopically there was non-suppurative hepatitis and interstitial nephritis. Multiple large, amphophilic, intranuclear inclusion bodies were present within hepatocytes and epithelial cells of the bile ducts, renal tubules, small and large intestinal mucosa, pancreatic acini and oral mucous membranes. Transmission electron microscopy (TEM) demonstrated that the inclusions comprised viral particles with morphology consistent with an adenovirus. A fragment of the adenoviral polymerase gene was amplified, sequenced and compared with other reptilian adenoviral sequences.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/ultrastructure , DNA, Viral/ultrastructure , Inclusion Bodies/ultrastructure , Lizards/virology , Virion/ultrastructure , Adenoviridae Infections/pathology , Animals , Bile Ducts/pathology , Bile Ducts/ultrastructure , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Hepatitis, Animal/pathology , Hepatocytes/pathology , Hepatocytes/ultrastructure , Inclusion Bodies/pathology , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Intestine, Large/pathology , Intestine, Large/ultrastructure , Intestine, Small/pathology , Intestine, Small/ultrastructure , Kidney Tubules/pathology , Kidney Tubules/ultrastructure , Liver/pathology , Liver/ultrastructure , Mouth/pathology , Mouth/ultrastructure , Mouth Mucosa/pathology , Mouth Mucosa/ultrastructure , Pancreas, Exocrine/pathology , Pancreas, Exocrine/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL