101
|
Yang Y, Chan L. Gene Therapy for Diabetes. TRANSLATING GENE THERAPY TO THE CLINIC 2015:115-128. [DOI: 10.1016/b978-0-12-800563-7.00008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
102
|
Sommer CA, Mostoslavsky G. RNA-Seq analysis of enteroendocrine cells reveals a role for FABP5 in the control of GIP secretion. Mol Endocrinol 2014; 28:1855-65. [PMID: 25268051 DOI: 10.1210/me.2014-1194] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In response to fat intake, enteroendocrine K cells release the hormone glucose-dependent insulinotropic polypeptide (GIP). GIP acts on adipocytes to increase lipid uptake and enhance adipokine secretion, promoting weight gain and insulin resistance. Modulation of intestinal GIP release could therefore represent a therapeutic strategy for the treatment and prevention of obesity and diabetes. However, the prospects of using drugs to effectively target specific enteroendocrine cell types have been tempered by the realization that these cells share similar transcriptional programs and frequently employ common mechanisms of hormone secretion. To gain novel insights into the regulation of GIP release, we generated knock-in mice expressing green fluorescent protein (GFP) under the control of the endogenous GIP promoter that enable the isolation of a purified population of small intestine K cells. Using RNA sequencing, we comprehensively characterized the transcriptomes of GIP(GFP) cells as well as the entire enteroendocrine lineage derived from Neurogenin3-expressing progenitors. Among the genes differentially expressed in GIP(GFP) cells, we identified and validated fatty acid-binding protein 5 (FABP5) as a highly expressed marker of GIP-producing cells that is absent in other enteroendocrine cell types. FABP5 promotes intracellular transport and inactivation of endocannabinoids, including anandamide, which inhibits GIP release. Remarkably, we found that circulating levels of GIP were significantly decreased in FABP5-deficient mice in the fasting state and in response to acute, oral fat diet administration. Our findings highlight the power of RNA sequencing to uncover molecular signatures of specific enteroendocrine cell types that can potentially be exploited for therapeutic purposes in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Cesar A Sommer
- Section of Gastroenterology, Department of Medicine, and Center for Regenerative Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | | |
Collapse
|
103
|
Bell GD, Reddy S, Sun X, Yang Y, Krissansen GW. Distribution of insulin mRNA transcripts within the human body. Biochem Biophys Res Commun 2014; 451:425-430. [PMID: 25110147 DOI: 10.1016/j.bbrc.2014.07.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 07/31/2014] [Indexed: 11/27/2022]
Abstract
Here we sought evidence for the existence of insulin mRNA-producing cells outside the human pancreas. Commercially available complementary DNA (cDNA) arrays prepared from 72 different types of adult human tissues were screened by PCR for transcripts encoding insulin, and other classic pancreatic hormones. Insulin mRNA transcripts were detected by standard PCR in the pancreas, stomach, pylorus region of the stomach, and the duodenum; and additionally by nested PCR in the jejunum, ileum and cecum, but not in other body tissues including the brain and colon. Most of these tissues also variably expressed mRNA transcripts for amylase α2B, amylin, glucagon, somatostatin, and pancreatic polypeptide. In summary, using sensitive PCR methods we have provided evidence for the presence of rare insulin mRNA-expressing cells within the stomach, small intestine, and cecum. Their role at these sites may be to support classical enteroendocrine cells as sentinels to sense and monitor gastric contents passing into and through the bowel.
Collapse
Affiliation(s)
- Glenn D Bell
- Department of Molecular Medicine & Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1005, New Zealand
| | - Shiva Reddy
- Department of Molecular Medicine & Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1005, New Zealand
| | - Xueying Sun
- Department of Molecular Medicine & Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1005, New Zealand
| | - Yi Yang
- Department of Molecular Medicine & Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1005, New Zealand
| | - Geoffrey W Krissansen
- Department of Molecular Medicine & Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1005, New Zealand.
| |
Collapse
|
104
|
Walker EM, Thompson CA, Battle MA. GATA4 and GATA6 regulate intestinal epithelial cytodifferentiation during development. Dev Biol 2014; 392:283-94. [PMID: 24929016 PMCID: PMC4149467 DOI: 10.1016/j.ydbio.2014.05.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/06/2014] [Accepted: 05/21/2014] [Indexed: 11/18/2022]
Abstract
The intestinal epithelium performs vital roles in organ function by absorbing nutrients and providing a protective barrier. The zinc-finger containing transcription factors GATA4 and GATA6 regulate enterocyte gene expression and control regional epithelial cell identity in the adult intestinal epithelium. Although GATA4 and GATA6 are expressed in the developing intestine, loss of either factor alone during the period of epithelial morphogenesis and cytodifferentiation fails to disrupt these processes. Therefore, we tested the hypothesis that GATA4 and GATA6 function redundantly to control these aspects of intestinal development. We used Villin-Cre, which deletes specifically in the intestinal epithelium during the period of villus development and epithelial cytodifferentiation, to generate Gata4Gata6 double conditional knockout embryos. Mice lacking GATA4 and GATA6 in the intestinal epithelium died within 24h of birth. At E18.5, intestinal villus architecture and epithelial cell populations were altered. Enterocytes were lost, and goblet cells were increased. Proliferation was also increased in GATA4-GATA6 deficient intestinal epithelium. Although villus morphology appeared normal at E16.5, the first time at which both Gata4 and Gata6 were efficiently reduced, changes in expression of markers of enterocytes, goblet cells, and proliferative cells were detected. Moreover, goblet cell number was increased at E16.5. Expression of the Notch ligand Dll1 and the Notch target Olfm4 were reduced in mutant tissue indicating decreased Notch signaling. Finally, we found that GATA4 occupies chromatin near the Dll1 transcription start site suggesting direct regulation of Dll1 by GATA4. We demonstrate that GATA4 and GATA6 play an essential role in maintaining proper intestinal epithelial structure and in regulating intestinal epithelial cytodifferentiation. Our data highlight a novel role for GATA factors in fine tuning Notch signaling during intestinal epithelial development to repress goblet cell differentiation.
Collapse
Affiliation(s)
- Emily M Walker
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | - Cayla A Thompson
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | - Michele A Battle
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.
| |
Collapse
|
105
|
Zissimopoulos A, Vradelis S, Konialis M, Chadolias D, Bampali A, Constantinidis T, Efremidou E, Kouklakis G. Chromogranin A as a biomarker of disease activity and biologic therapy in inflammatory bowel disease: a prospective observational study. Scand J Gastroenterol 2014; 49:942-9. [PMID: 24897131 DOI: 10.3109/00365521.2014.920910] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To access the correlation of Chromogranin A (CgA) with inflammatory bowel disease (IBD) activity and responsiveness to medical therapy. MATERIAL AND METHODS A prospective observational study was conducted in 56 patients with moderate ulcerative colitis (UC) or Crohn's disease (CD) (UC, n = 29, CD, n = 27), 17 patients with irritable bowel syndrome and predominant diarrhea (IBS-D) and 40 healthy volunteers. IBD patients were treated by biologics (infliximab or adalimumab) or conventional agents (aminosalicylates, thiopurines or methotrexate and steroids) and were classified according to their treatment in two groups. Serum CgA was measured at baseline and 4-week posttreatment period. RESULTS Serum CgA was significantly higher in IBD patients than in those with IBS-D or healthy volunteers (p < 0.01). Furthermore, serum CgA was markedly increased in CD patients than in UC patients (p < 0.01). CgA value was significantly reduced in 'biologic' group (24 IBD patients, UC, n = 15, CD, n = 9) at 4-week posttreatment period (p < 0.01), while 18/24 (72%) patients were already in remission during that time. In contrast, CgA value was significantly increased in the 'conventional' treatment group (32 IBD patients, UC, n = 14, CD, n = 18) between the two visits (p < 0.01), although 22/32 (69%) patients were in remission during the 4-week posttreatment period. CONCLUSION CgA appears to be a reliable marker of disease activity in IBD patients and especially in those who received biologic therapy. IBS-D patients presented normal CgA values.
Collapse
Affiliation(s)
- Athanasios Zissimopoulos
- Deartment of Nuclear Medicine, Democritus University of Thrace , Alexandroupolis, 68100 , Greece
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Mojibian M, Lam AWY, Fujita Y, Asadi A, Grassl GA, Dickie P, Tan R, Cheung AT, Kieffer TJ. Insulin-producing intestinal K cells protect nonobese diabetic mice from autoimmune diabetes. Gastroenterology 2014; 147:162-171.e6. [PMID: 24662331 DOI: 10.1053/j.gastro.2014.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 03/12/2014] [Accepted: 03/18/2014] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Type 1 diabetes is caused by an aberrant response against pancreatic β cells. Intestinal K cells are glucose-responsive endocrine cells that might be engineered to secrete insulin. We generated diabetes-prone non-obese diabetic (NOD) mice that express insulin, via a transgene, in K cells. We assessed the effects on immunogenicity and diabetes development. METHODS Diabetes incidence and glucose homeostasis were assessed in NOD mice that expressed mouse preproinsulin II from a transgene in K cells and nontransgenic NOD mice (controls); pancreas and duodenum tissues were collected and analyzed by histology. We evaluated T cell responses to insulin, levels of circulating autoantibodies against insulin, and the percentage of circulating antigen-specific T cells. Inflammation of mesenteric and pancreatic lymph node cells was also evaluated. RESULTS The transgenic mice tended to have lower blood levels of glucose than control mice, associated with increased plasma levels of immunoreactive insulin and proinsulin. Fewer transgenic mice developed diabetes than controls. In analyses of pancreas and intestine tissues from the transgenic mice, insulin-producing K cells were not affected by the immune response and the mice had reduced destruction of endogenous β cells. Fewer transgenic mice were positive for insulin autoantibodies compared with controls. Cells isolated from mesenteric lymph nodes of the transgenic mice had significantly lower rates of proliferation and T cells from transgenic mice tended to secrete lower levels of inflammatory cytokines than from controls. At 15 weeks, transgenic mice had fewer peripheral CD8(+) T cells specific for NRP-V7 than control mice. CONCLUSIONS NOD mice with intestinal K cells engineered to express insulin have reduced blood levels of glucose, are less likely to develop diabetes, and have reduced immunity against pancreatic β cells compared with control NOD mice. This approach might be developed to treat patients with type 1 diabetes.
Collapse
Affiliation(s)
- Majid Mojibian
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ada W Y Lam
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yukihiro Fujita
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ali Asadi
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Guntram A Grassl
- Institute for Experimental Medicine, Christian Albrechts University Kiel, Kiel, Germany
| | - Peter Dickie
- University of Alberta, Edmonton, Alberta, Canada
| | - Rusung Tan
- Child and Family Research Institute, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Pathology, Sidra Medical and Research Center, Doha, Qatar
| | | | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
107
|
Bouchi R, Foo KS, Hua H, Tsuchiya K, Ohmura Y, Sandoval PR, Ratner LE, Egli D, Leibel RL, Accili D. FOXO1 inhibition yields functional insulin-producing cells in human gut organoid cultures. Nat Commun 2014; 5:4242. [PMID: 24979718 PMCID: PMC4083475 DOI: 10.1038/ncomms5242] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 05/28/2014] [Indexed: 01/12/2023] Open
Abstract
Generation of surrogate sources of insulin-producing β-cells remains a goal of diabetes therapy. While most efforts have been directed at differentiating embryonic or induced pluripotent stem (iPS) cells into β-like-cells through endodermal progenitors, we have shown that gut endocrine progenitor cells of mice can be differentiated into glucose-responsive, insulin-producing cells by ablation of transcription factor Foxo1. Here we show that FOXO1 is present in human gut endocrine progenitor and serotonin-producing cells. Using gut organoids derived from human iPS cells, we show that FOXO1 inhibition using a dominant-negative mutant or lentivirus-encoded shRNA promotes generation of insulin-positive cells that express all markers of mature pancreatic β-cells, release C-peptide in response to secretagogues, and survive in vivo following transplantation into mice. The findings raise the possibility of using gut-targeted FOXO1 inhibition or gut organoids as a source of insulin-producing cells to treat human diabetes.
Collapse
Affiliation(s)
- Ryotaro Bouchi
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | - Kylie S Foo
- 1] New York Stem Cell Foundation Research Institute, New York, New York 10032, USA [2] Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | - Haiqing Hua
- 1] New York Stem Cell Foundation Research Institute, New York, New York 10032, USA [2] Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | - Yoshiaki Ohmura
- Department of Surgery, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | - P Rodrigo Sandoval
- Department of Surgery, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | - Lloyd E Ratner
- Department of Surgery, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | - Dieter Egli
- 1] New York Stem Cell Foundation Research Institute, New York, New York 10032, USA [2] Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | - Rudolph L Leibel
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | - Domenico Accili
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| |
Collapse
|
108
|
CtBP and associated LSD1 are required for transcriptional activation by NeuroD1 in gastrointestinal endocrine cells. Mol Cell Biol 2014; 34:2308-17. [PMID: 24732800 DOI: 10.1128/mcb.01600-13] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Gene expression programs required for differentiation depend on both DNA-bound transcription factors and surrounding histone modifications. Expression of the basic helix-loop-helix (bHLH) protein NeuroD1 is restricted to endocrine cells in the gastrointestinal (GI) tract, where it is important for endocrine differentiation. RREB1 (RAS-responsive element binding protein 1), identified as a component of the CtBP corepressor complex, binds to nearby DNA elements to associate with NeuroD and potentiate transcription of a NeuroD1 target gene. Transcriptional activation by RREB1 depends on recruitment of CtBP with its associated proteins, including LSD1, through its PXDLS motifs. The mechanism of transcriptional activation by CtBP has not been previously characterized. Here we found that activation was dependent on the histone H3 lysine 9 (H3K9) demethylase activity of LSD1, which removes repressive methyl marks from dimethylated H3K9 (H3K9Me2), to facilitate subsequent H3K9 acetylation by the NeuroD1-associated histone acetyltransferase, P300/CBP-associated factor (PCAF). The secretin, β-glucokinase, insulin I, and insulin II genes, four known direct targets of NeuroD1 in intestinal and pancreatic endocrine cells, all show similar promoter occupancy by CtBP-associated proteins and PCAF, with acetylation of H3K9. This work may indicate a mechanism for selective regulation of transcription by CtBP and LSD1 involving their association with specific transcription factors and cofactors to drive tissue-specific transcription.
Collapse
|
109
|
Maruccio L, D'Angelo L, de Girolamo P, Lucini C, Castaldo L. GDNF and GFRα co-receptor family in the developing feline gut. Ann Anat 2014; 196:296-302. [PMID: 24834895 DOI: 10.1016/j.aanat.2014.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 01/19/2023]
Abstract
Glial cell-line derived neurotrophic factor (GDNF) and the GFRα co-receptors play a role in the developing enteric nervous system. The co-receptors elicit their action by binding receptor tyrosine kinase RET. This immunohistochemical study reports the presence of GDNF and its specific co-receptor GFRα1 in the cat gastrointestinal apparatus during development, from stage 9 to 22. At stage 9 and 11, immunoreactivity (IR) to GDNF was observed in the cells of mesenchyme of the anterior gut. From stage 14 to 22, GDNF IR was detected in nervous plexuses; moreover, GDNF and GFRα1 IR appeared localized in gastrointestinal endocrine cells. The presence of GDNF in the enteric nervous system and in the endocrine cells suggests an involvement of this neurotrophic factor in the gastrointestinal development. Moreover, the presence of the co-receptor GFRα1 in endocrine cells and its absence in the enteric nervous system seems to indicate a different mode of transduction of GDNF signal. GFRα2 and GFRα3 co-receptors were not detected.
Collapse
Affiliation(s)
- L Maruccio
- Department of Veterinary Medicine and Animal Productions, University of Naples, Federico II, Naples, Italy.
| | - L D'Angelo
- Department of Veterinary Medicine and Animal Productions, University of Naples, Federico II, Naples, Italy
| | - P de Girolamo
- Department of Veterinary Medicine and Animal Productions, University of Naples, Federico II, Naples, Italy
| | - C Lucini
- Department of Veterinary Medicine and Animal Productions, University of Naples, Federico II, Naples, Italy
| | - L Castaldo
- Department of Veterinary Medicine and Animal Productions, University of Naples, Federico II, Naples, Italy
| |
Collapse
|
110
|
Nagatake T, Fujita H, Minato N, Hamazaki Y. Enteroendocrine cells are specifically marked by cell surface expression of claudin-4 in mouse small intestine. PLoS One 2014; 9:e90638. [PMID: 24603700 PMCID: PMC3948345 DOI: 10.1371/journal.pone.0090638] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/05/2014] [Indexed: 01/24/2023] Open
Abstract
Enteroendocrine cells are solitary epithelial cells scattered throughout the gastrointestinal tract and produce various types of hormones, constituting one of the largest endocrine systems in the body. The study of these rare epithelial cells has been hampered by the difficulty in isolating them because of the lack of specific cell surface markers. Here, we report that enteroendocrine cells selectively express a tight junction membrane protein, claudin-4 (Cld4), and are efficiently isolated with the use of an antibody specific for the Cld4 extracellular domain and flow cytometry. Sorted Cld4+ epithelial cells in the small intestine exclusively expressed a chromogranin A gene (Chga) and other enteroendocrine cell–related genes (Ffar1, Ffar4, Gpr119), and the population was divided into two subpopulations based on the activity of binding to Ulex europaeus agglutinin-1 (UEA-1). A Cld4+UEA-1− cell population almost exclusively expressed glucose-dependent insulinotropic polypeptide gene (Gip), thus representing K cells, whereas a Cld4+UEA-1+ cell population expressed other gut hormone genes, including glucagon-like peptide 1 (Gcg), pancreatic polypeptide–like peptide with N-terminal tyrosine amide (Pyy), cholecystokinin (Cck), secretin (Sct), and tryptophan hydroxylase 1 (Tph1). In addition, we found that orally administered luminal antigens were taken up by the solitary Cld4+ cells in the small intestinal villi, raising the possibility that enteroendocrine cells might also play a role in initiation of mucosal immunity. Our results provide a useful tool for the cellular and functional characterization of enteroendocrine cells.
Collapse
Affiliation(s)
- Takahiro Nagatake
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Harumi Fujita
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nagahiro Minato
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoko Hamazaki
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
111
|
Hansen CF, Vassiliadis E, Vrang N, Sangild PT, Cummings BP, Havel P, Jelsing J. The effect of ileal interposition surgery on enteroendocrine cell numbers in the UC Davis type 2 diabetes mellitus rat. ACTA ACUST UNITED AC 2014; 189:31-9. [PMID: 24512816 DOI: 10.1016/j.regpep.2014.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 01/09/2014] [Accepted: 01/31/2014] [Indexed: 02/07/2023]
Abstract
AIM To investigate the short-term effect of ileal interposition (IT) surgery on gut morphology and enteroendocrine cell numbers in the pre-diabetic UC Davis type 2 diabetes mellitus (UCD-T2DM) rat. STUDY DESIGN Two-month old male UCD-T2DM rats underwent either sham (n=5) or IT (n=5) surgery. Intestines were collected 1.5months after surgery. The jejunum, ileum and colon regions were processed for histochemical and immunohistochemical labeling and stereological analyses of changes in gut morphometry and number of enteroendocrine cells. RESULTS Stereological analysis showed that intestinal volume, luminal surface area and the number of all chromogranin A-positive enteroendocrine cells were markedly increased in the IT rats compared with sham-operated animals. Subanalyses of the glucagon-like peptide 2, cholecystokinin, serotonin cells and the neurotensin immunoreactive sub-pool of enteroendocrine cells in the IT region revealed an increase in numbers across phenotypes. However, the density of the different cell types varied. CONCLUSION IT surgery in the UCD-T2DM rat leads to rapid alterations in gut morphometry and an increase in the number of enteroendocrine cells. This effect may potentially explain why IT surgery delays the onset of type 2 diabetes in the UCD-T2DM rat.
Collapse
Affiliation(s)
- Carl Frederik Hansen
- Gubra, Hørsholm, Denmark; Department of Human Nutrition, University of Copenhagen, Frederiksberg, Denmark
| | | | | | - Per T Sangild
- Department of Human Nutrition, University of Copenhagen, Frederiksberg, Denmark
| | - Bethany P Cummings
- Department of Biomedical Sciences, School of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Peter Havel
- Department of Molecular Biosciences, University of California, Davis, CA, USA
| | | |
Collapse
|
112
|
González-Abuín N, Martínez-Micaelo N, Blay M, Ardévol A, Pinent M. Grape-seed procyanidins prevent the cafeteria-diet-induced decrease of glucagon-like peptide-1 production. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:1066-1072. [PMID: 24410268 DOI: 10.1021/jf405239p] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Grape-seed procyanidin extract (GSPE) has been reported to improve insulin resistance in cafeteria rats. Because glucagon-like peptide-1 (GLP-1) is involved in glucose homeostasis, the preventive effects of GSPE on GLP-1 production, secretion, and elimination were evaluated in a model of diet-induced insulin resistance. Rats were fed a cafeteria diet for 12 weeks, and 25 mg of GSPE/kg of body weight was administered concomitantly. Vehicle-treated cafeteria-fed rats and chow-fed rats were used as controls. The cafeteria diet decreased active GLP-1 plasma levels, which is attributed to a decreased intestinal GLP-1 production, linked to reduced colonic enteroendocrine cell populations. Such effects were prevented by GSPE. In the same context, GSPE avoided the decrease on intestinal dipeptidyl-peptidase 4 (DPP4) activity and modulated the gene expression of GLP-1 and its receptor in the hypothalamus. In conclusion, the preventive treatment with GSPE abrogates the effects of the cafeteria diet on intestinal GLP-1 production and DPP4 activity.
Collapse
Affiliation(s)
- Noemi González-Abuín
- Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili , 43007 Tarragona, Spain
| | | | | | | | | |
Collapse
|
113
|
Petersen N, Reimann F, Bartfeld S, Farin HF, Ringnalda FC, Vries RGJ, van den Brink S, Clevers H, Gribble FM, de Koning EJP. Generation of L cells in mouse and human small intestine organoids. Diabetes 2014; 63:410-20. [PMID: 24130334 PMCID: PMC4306716 DOI: 10.2337/db13-0991] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Upon a nutrient challenge, L cells produce glucagon-like peptide 1 (GLP-1), a powerful stimulant of insulin release. Strategies to augment endogenous GLP-1 production include promoting L-cell differentiation and increasing L-cell number. Here we present a novel in vitro platform to generate functional L cells from three-dimensional cultures of mouse and human intestinal crypts. We show that short-chain fatty acids selectively increase the number of L cells, resulting in an elevation of GLP-1 release. This is accompanied by the upregulation of transcription factors associated with the endocrine lineage of intestinal stem cell development. Thus, our platform allows us to study and modulate the development of L cells in mouse and human crypts as a potential basis for novel therapeutic strategies in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Natalia Petersen
- Hubrecht Institute for Development Biology and Stem Cell Research, Utrecht, Netherlands
| | - Frank Reimann
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, UK
| | - Sina Bartfeld
- Hubrecht Institute for Development Biology and Stem Cell Research, Utrecht, Netherlands
| | - Henner F. Farin
- Hubrecht Institute for Development Biology and Stem Cell Research, Utrecht, Netherlands
| | - Femke C. Ringnalda
- Hubrecht Institute for Development Biology and Stem Cell Research, Utrecht, Netherlands
| | - Robert G. J. Vries
- Hubrecht Institute for Development Biology and Stem Cell Research, Utrecht, Netherlands
| | | | - Hans Clevers
- Hubrecht Institute for Development Biology and Stem Cell Research, Utrecht, Netherlands
- Utrecht University Medical Center, Utrecht, Netherlands
| | - Fiona M. Gribble
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, Addenbrooke’s Hospital, Cambridge, UK
| | - Eelco J. P. de Koning
- Hubrecht Institute for Development Biology and Stem Cell Research, Utrecht, Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Department of Endocrinology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
114
|
Impact of high-fat feeding on basic helix-loop-helix transcription factors controlling enteroendocrine cell differentiation. Int J Obes (Lond) 2014; 38:1440-8. [PMID: 24480860 DOI: 10.1038/ijo.2014.20] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/03/2013] [Accepted: 01/17/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES Gut hormones secreted by enteroendocrine cells (EECs) play a major role in energy regulation. Differentiation of EEC is controlled by the expression of basic helix-loop-helix (bHLH) transcription factors. High-fat (HF) feeding alters gut hormone levels; however, the impact of HF feeding on bHLH transcription factors in mediating EEC differentiation and subsequent gut hormone secretion and expression is not known. METHODS Outbred Sprague-Dawley rats were maintained on chow or HF diet for 12 weeks. Gene and protein expression of intestinal bHLH transcription factors, combined with immunofluorescence studies, were analyzed for both groups in the small intestine and colon. Gut permeability, intestinal lipid and carbohydrate transporters as well as circulating levels and intestinal protein expression of gut peptides were determined. RESULTS We showed that HF feeding resulted in hyperphagia and increased adiposity. HF-fed animals exhibited decreased expression of bHLH transcription factors controlling EEC differentiation (MATH1, NGN3, NEUROD1) and increased expression of bHLH factors modulating enterocyte expression. Furthermore, HF-fed animals had decreased number of total EECs and L-cells. This was accompanied by increased gut permeability and expression of lipid and carbohydrate transporters, and a decrease in circulating and intestinal gut hormone levels. CONCLUSIONS Taken together, our results demonstrate that HF feeding caused decreased secretory lineage (that is, EECs) differentiation through downregulation of bHLH transcription factors, resulting in reduced EEC number and gut hormone levels. Thus, impaired EEC differentiation pathways by HF feeding may promote hyperphagia and subsequent obesity.
Collapse
|
115
|
El-Salhy M, Gundersen D, Gilja OH, Hatlebakk JG, Hausken T. Is irritable bowel syndrome an organic disorder? World J Gastroenterol 2014; 20:384-400. [PMID: 24574708 PMCID: PMC3923014 DOI: 10.3748/wjg.v20.i2.384] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 11/05/2013] [Accepted: 11/13/2013] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder that is generally considered to be functional because there appears to be no associated anatomical defect. Stress and psychological factors are thought to play an important role in IBS. The gut neuroendocrine system (NES), which regulates all functions of the gastrointestinal tract, consists of endocrine cells that are scattered among the epithelial cells of the mucosa, and the enteric nervous system. Although it is capable of operating independently from the central nervous system (CNS), the gut NES is connected to and modulated by the CNS. This review presents evidence for the presence of an anatomical defect in IBS patients, namely in the gastrointestinal endocrine cells. These cells have specialized microvilli that project into the lumen and function as sensors for the luminal content and respond to luminal stimuli by releasing hormones into the lamina propria, which starts a chain reaction that progresses throughout the entire NES. The changes in the gastrointestinal endocrine cells observed in IBS patients are highly consistent with the other abnormalities reported in IBS patients, such as visceral hypersensitivity, dysmotility, and abnormal secretion.
Collapse
|
116
|
Kuwahara A. Contributions of colonic short-chain Fatty Acid receptors in energy homeostasis. Front Endocrinol (Lausanne) 2014; 5:144. [PMID: 25228897 PMCID: PMC4150999 DOI: 10.3389/fendo.2014.00144] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/19/2014] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal (GI) tract is separated from the body's internal environment by a single layer of epithelial cells, through which nutrients must pass for their absorption into the bloodstream. Besides food and drink, the GI lumen is also exposed to bioactive chemicals and bacterial products including short-chain fatty acids (SCFAs). Therefore, the GI tract has to monitor the composition of its contents continuously to discriminate between necessary and unnecessary compounds. Recent molecular identification of epithelial membrane receptor proteins has revealed the sensory roles of intestinal epithelial cells in the gut chemosensory system. Malfunctioning of these receptors may be responsible for a variety of metabolic dysfunctions associated with obesity and related disorders. Recent studies suggest that SCFAs produced by microbiota fermentation act as signaling molecules and influence the host's metabolism; uncovering the sensory mechanisms of such bacterial metabolites would help us understand the interactions between the host and microbiota in host energy homeostasis. In this review, the contribution of colonic SCFA receptors in energy metabolism and our recent findings concerning the possible link between SCFA receptors and host energy homeostasis are discussed.
Collapse
Affiliation(s)
- Atsukazu Kuwahara
- Laboratory of Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
- *Correspondence: Atsukazu Kuwahara, Laboratory of Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan e-mail:
| |
Collapse
|
117
|
Abstract
The colon serves as the habitat for trillions of microbes, which it must maintain, regulate, and sequester. This is managed by what is termed the mucosal barrier. The mucosal barrier separates the gut flora from the host tissues; regulates the absorption of water, electrolytes, minerals, and vitamins; and facilitates host-flora interactions. Colonic homeostasis depends on a complex interaction between the microflora and the mucosal epithelium, immune system, vasculature, stroma, and nervous system. Disruptions in the colonic microenvironment such as changes in microbial composition, epithelial cell function/proliferation/differentiation, mucus production/makeup, immune function, diet, motility, or blood flow may have substantial local and systemic consequences. Understanding the complex activities of the colon in health and disease is important in drug development, as xenobiotics can impact all segments of the colon. Direct and indirect effects of pharmaceuticals on intestinal function can produce adverse findings in laboratory animals and humans and can negatively impact drug development. This review will discuss normal colon homeostasis with examples, where applicable, of xenobiotics that disrupt normal function.
Collapse
Affiliation(s)
- Rani S Sellers
- 1Albert Einstein College of Medicine, Bronx, New York, USA
| | | |
Collapse
|
118
|
González-Abuín N, Martínez-Micaelo N, Blay M, Green BD, Pinent M, Ardévol A. Grape-seed procyanidins modulate cellular membrane potential and nutrient-induced GLP-1 secretion in STC-1 cells. Am J Physiol Cell Physiol 2013; 306:C485-92. [PMID: 24371039 DOI: 10.1152/ajpcell.00355.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Grape-seed procyanidin extracts (GSPE) modulate glucose homeostasis, and it was suggested that GSPE may achieve this by enhancing the secretion of incretin hormones such as glucagon-like peptide-1 (GLP-1). Therefore, the aim of the present study is to examine in detail the effects of GSPE on intestinal endocrine cells (STC-1). GSPE was found to modulate plasma membrane potential in enteroendocrine cells, inducing depolarization at low concentrations (0.05 mg/l) and hyperpolarization at high concentrations (50 mg/l), and surprisingly this was also accompanied by suppressed GLP-1 secretion. Furthermore, how GSPE affects STC-1 cells under nutrient-stimulated conditions (i.e., glucose, linoleic acid, and l-proline) was also explored, and we found that the higher GSPE concentration was effective in limiting membrane depolarization and reducing GLP-1 secretion. Next, it was also examined whether GSPE affected mitochondrial membrane potential, and it was found that this too is altered by GSPE; however, this does not appear to explain the observed effects on plasma membrane potential and GLP-1 secretion. In conclusion, our results show that grape-seed procyanidins modulate cellular membrane potential and nutrient-induced enteroendocrine hormone secretion in STC-1 cells.
Collapse
Affiliation(s)
- Noemi González-Abuín
- Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Tarragona, Spain; and
| | | | | | | | | | | |
Collapse
|
119
|
Quantification of endocrine cells and ultrastructural study of insulin granules in the large intestine of opossum Didelphis aurita (Wied-Neuwied, 1826). Tissue Cell 2013; 46:70-7. [PMID: 24359801 DOI: 10.1016/j.tice.2013.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 11/14/2013] [Accepted: 11/14/2013] [Indexed: 11/22/2022]
Abstract
This study aimed to investigate the distribution of argyrophil, argentaffin, and insulin-immunoreactive endocrine cells in the large intestine of opossums (Didelphis aurita) and to describe the ultrastructure of the secretory granules of insulin-immunoreactive endocrine cells. Fragments of the large intestine of 10 male specimens of D. aurita were collected, processed, and subjected to staining, immunohistochemistry, and transmission electron microscopy. The argyrophil, the argentaffin, and the insulin-immunoreactive endocrine cells were sparsely distributed in the intestinal glands of the mucous layer, among other cell types of the epithelium in all regions studied. Proportionally, the argyrophil, the argentaffin, and the insulin-immunoreactive endocrine cells represented 62.75%, 36.26%, and 0.99% of the total determined endocrine cells of the large intestine, respectively. Quantitatively, there was no difference between the argyrophil and the argentaffin endocrine cells, whereas insulin-immunoreactive endocrine cells were less numerous. The insulin-immunoreactive endocrine cells were elongated or pyramidal, with rounded nuclei of irregularly contoured, and large amounts of secretory granules distributed throughout the cytoplasm. The granules have different sizes and electron densities and are classified as immature and mature, with the mature granules in predominant form in the overall granular population. In general, the granule is shown with an external electron-lucent halo and electron-dense core. The ultrastructure pattern in the granules of the insulin-immunoreactive endocrine cells was similar to that of the B cells of pancreatic islets in rats.
Collapse
|
120
|
Lee E, Ryu GR, Moon SD, Ko SH, Ahn YB, Song KH. Reprogramming of enteroendocrine K cells to pancreatic β-cells through the combined expression of Nkx6.1 and Neurogenin3, and reaggregation in suspension culture. Biochem Biophys Res Commun 2013; 443:1021-7. [PMID: 24365150 DOI: 10.1016/j.bbrc.2013.12.093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 12/17/2013] [Indexed: 10/25/2022]
Abstract
Recent studies have demonstrated that adult cells such as pancreatic exocrine cells can be converted to pancreatic β-cells in a process called cell reprogramming. Enteroendocrine cells and β-cells share similar pathways of differentiation during embryonic development. Notably, enteroendocrine K cells express many of the key proteins found in β-cells. Thus, K cells could be reprogrammed to β-cells under certain conditions. However, there is no clear evidence on whether these cells convert to β-cells. K cells were selected from STC-1 cells, an enteroendocrine cell line expressing multiple hormones. K cells were found to express many genes of transcription factors crucial for islet development and differentiation except for Nkx6.1 and Neurogenin3. A K cell clone stably expressing Nkx6.1 (Nkx6.1(+)-K cells) was established. Induction of Neurogenin3 expression in Nkx6.1(+)-K cells, by either treatment with a γ-secretase inhibitor or infection with a recombinant adenovirus expressing Neurogenin3, led to a significant increase in Insulin1 mRNA expression. After infection with the adenovirus expressing Neurogenin3 and reaggregation in suspension culture, about 50% of Nkx6.1(+)-K cells expressed insulin as determined by immunostaining. The intracellular insulin content was increased markedly. Electron microscopy revealed the presence of insulin granules. However, glucose-stimulated insulin secretion was defective, and there was no glucose lowering effect after transplantation of these cells in diabetic mice. In conclusion, we demonstrated that K cells could be reprogrammed partially to β-cells through the combined expression of Nkx6.1 and Neurogenin3, and reaggregation in suspension culture.
Collapse
Affiliation(s)
- Esder Lee
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Gyeong Ryul Ryu
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Dae Moon
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Hyun Ko
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yu-Bae Ahn
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ki-Ho Song
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
121
|
|
122
|
Rasouli M, Abbasi S, Sarsaifi K, Hani H, Ahmad Z, Omar AR. The L-cell isolation from heterogonous population of intestinal cell line using antibiotic selection method. Appl Biochem Biotechnol 2013; 172:394-404. [PMID: 24081707 DOI: 10.1007/s12010-013-0514-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 09/15/2013] [Indexed: 11/30/2022]
Abstract
Enteroendocrine cells are the largest population of hormone-producing cells in the body and play important roles in many aspects of body functions. The enteroendocrine cell population is divided into different subpopulations that secrete different hormones and peptides. Characterization of each subpopulation is particularly useful for analyzing the cellular mechanisms responsible for specific cell types. Therefore, the necessity of a pure cell line for a specific study purpose was the important motivation for the separation of cell lines for each subpopulation of enteroendocrine cells. The present research introduces a method for the isolation of L-cells, one of the important subpopulations of enteroendocrine cells. The antibiotic selection method was conducted in order to isolate the L-cells from a heterogonous population of intestinal cell line. In this method, a neomycin resistance gene (as selected marker) was expressed under the control of a specific promoter of L-cells. After transfection of manipulated plasmid, only the cells which determine the specific promoter and express neomycin resistance protein would be able to survive under Geneticin antibiotic treatment condition. In order to confirm that the isolated cells were L-cells, reverse transcriptase polymerase chain reaction (PCR) and quantitative PCR assays were performed. Based on the results, the isolated cells were pure L-cells that could be able to express specific mRNA of L-cells efficiently. This technique provides a unique method for the isolation and purification of any cell line. The purified isolated L-cells by this method can be used for future studies and for analyzing cellular mechanisms that involve L-cells' functions.
Collapse
Affiliation(s)
- Mina Rasouli
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, 43400, Serdang, Selangor, Malaysia,
| | | | | | | | | | | |
Collapse
|
123
|
Abstract
The author aims to review the established medical treatment options of neuroendocrine tumours, which have expanded greatly in recent years and present the most important aspects to be considered in planning patients' management. Medical treatment is usually considered in advanced stages of these tumours, as well as in cases of hormone overproduction. Somatostatin analogues have been known to be effective in alleviating hormone excess syndromes, especially carcinoid syndrome for the past 25 years. There is a convincing evidence that the somatostatin analogue octreotide is useful as an antitumor agent, at least in well-differentiated small intestinal neuroendocrine tumours and probably also in those of pancreatic origin. Interferons may be also used and the indications for their use may be almost the same. Optimal patient selection is mandatory for the use of cytotoxic chemotherapy. Streptozotocin- and, recently, temozolomide-based chemotherapies should be considered in progressive phases of well differentiated (G1/G2) pancreatic neuroendocrine tumours. A cisplatin-etoposide combination is the first choice for the treatment of G3 neuroendocrine carcinomas of any origin. Recently, the mammalian target of rapamycin inhibitor everolimus and the combined tyrosine kinase inhibitor sunitinib were registered for the treatment of G1/G2 pancreatic neuroendocrine tumours. The most recent drug treatment recommendations and therapeutic algorithms to improve systemic therapy in patients with neuroendocrine tumours are summarized and novel drug candidates with particular potential for future management of these tumours are outlined.
Collapse
Affiliation(s)
- Miklós Tóth
- Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| |
Collapse
|
124
|
Sentani K, Uraoka N, Oue N, Yasui W. Alpha-fetoprotein-producing clear cell carcinoma of the gallbladder with neuroendocrine differentiation. Med Mol Morphol 2013; 47:54-6. [PMID: 23860909 DOI: 10.1007/s00795-013-0049-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 06/25/2013] [Indexed: 11/28/2022]
Abstract
An uncommon case of alpha-fetoprotein (AFP) producing clear cell carcinoma of the gallbladder with neuroendocrine differentiation in a 78-year-old Japanese woman, who complained of epigastralgia, is reported. Macroscopically, the nodular infiltrative type of tumor, measuring approximately 4.5 × 3.5 cm in size, was located in the fundus of the gallbladder. Histologically, the tumor was composed of clear cell carcinoma with AFP production, non-clear cell adenocarcinoma with neuroendocrine differentiation, and poorly or undifferentiated carcinoma with extensive ulceration. Carcinoma in situ was found in the surrounding gallbladder epithelium. Her postoperative laboratory tests showed a decrease in AFP levels to normal. The clinical and pathologic significance of AFP production or neuroendocrine differentiation in the gallbladder carcinomas have thus far remained completely obscure. However, we should recognize the entity of this tumor because the accurate diagnosis of primary clear cell carcinoma of the gallbladder may have important therapeutic implications.
Collapse
Affiliation(s)
- Kazuhiro Sentani
- Department of Molecular Pathology, Hiroshima University Institute of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | | | | | | |
Collapse
|
125
|
Dos Santos DCM, Cupertino MDC, Novaes RD, Soares ÍADC, Fonseca CC, da Matta SLP, Sartori SSR. Morphologic characterization and distribution of endocrine cells in the large intestine of the opossum Didelphis aurita (Wied-Neuwied, 1826). Tissue Cell 2013; 45:338-49. [PMID: 23810437 DOI: 10.1016/j.tice.2013.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 05/11/2013] [Accepted: 05/27/2013] [Indexed: 01/13/2023]
Abstract
This study was designed to investigate the morphology and distribution of argyrophil, argentaffins, and insulin-immunoreactive endocrine cells in the large intestine of the opossum Didelphis aurita. Fragments of the large intestine of 10 male specimens of the opossum D. aurita were collected, processed, and submitted for histochemistry, immunohistochemistry, and scanning electron microscopy. The tunics of the large intestine of D. aurita presented morphological characteristics that have already been described for eutherian mammals. The morphometric data showed that the inner circular layer of all portions and regions analyzed is thicker relative to the longitudinal layer, and these layers in the rectum are thicker compared to the cecum and ascending colon. The majority of mucus-secreting cells have acid and neutral mucins, suggesting that the production of mucus is mixed. The number of these cells increases in the region of the cecum toward the rectum. Important findings include the occurrence of argyrophil, argentaffins, and insulin-immunoreactive endocrine cells in all segments of the large intestine of the opossum (D. aurita). To the best of our knowledge, this is the first report about the presence of insulin-immunoreactive endocrine cells in the large intestine of the opossum (D. aurita).
Collapse
|
126
|
Hansen CF, Bueter M, Theis N, Lutz T, Paulsen S, Dalbøge LS, Vrang N, Jelsing J. Hypertrophy dependent doubling of L-cells in Roux-en-Y gastric bypass operated rats. PLoS One 2013; 8:e65696. [PMID: 23776529 PMCID: PMC3679162 DOI: 10.1371/journal.pone.0065696] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 04/29/2013] [Indexed: 12/31/2022] Open
Abstract
Background and Aims Roux-en-Y gastric bypass (RYGB) leads to a rapid remission of type 2 diabetes mellitus (T2DM), but the underlying mode of action remains incompletely understood. L-cell derived gut hormones such as glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) are thought to play a central role in the anti-diabetic effects of RYGB; therefore, an improved understanding of intestinal endocrine L-cell adaptability is considered pivotal. Methods The full rostrocaudal extension of the gut was analyzed in rats after RYGB and in sham-operated controls ad libitum fed or food restricted to match the body weight of RYGB rats. Total number of L-cells, as well as regional numbers, densities and mucosa volumes were quantified using stereological methods. Preproglucagon and PYY mRNA transcripts were quantified by qPCR to reflect the total and relative hormone production capacity of the L-cells. Results RYGB surgery induced hypertrophy of the gut mucosa in the food exposed regions of the small intestine coupled with a doubling in the total number of L-cells. No changes in L-cell density were observed in any region regardless of surgery or food restriction. The total gene expression capacity of the entire gut revealed a near 200% increase in both PYY and preproglucagon mRNA levels in RYGB rats associated with both increased L-cell number as well as region-specific increased transcription per cell. Conclusions Collectively, these findings indicate that RYGB in rats is associated with gut hypertrophy, an increase in L-cell number, but not density, and increased PYY and preproglucagon gene expression. This could explain the enhanced gut hormone dynamics seen after RYGB.
Collapse
Affiliation(s)
- Carl Frederik Hansen
- Department of Histology, Gubra, Hørsholm, Denmark
- Department of Human Nutrition, University of Copenhagen, Frederiksberg, Denmark
| | - Marco Bueter
- Department of Visceral and Transplant Surgery, University Hospital Zurich, Zurich, Switzerland
- Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Nadine Theis
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Thomas Lutz
- Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | | | - Niels Vrang
- Department of Histology, Gubra, Hørsholm, Denmark
| | - Jacob Jelsing
- Department of Histology, Gubra, Hørsholm, Denmark
- * E-mail:
| |
Collapse
|
127
|
Li B, Zhou X, Wu J, Zhou H. From gut changes to type 2 diabetes remission after gastric bypass surgeries. Front Med 2013; 7:191-200. [PMID: 23553469 DOI: 10.1007/s11684-013-0258-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/14/2013] [Indexed: 12/18/2022]
Abstract
Increasing evidence suggests that the gut may influence the host's metabolism and ultimately change the outcomes of type 2 diabetes mellitus (T2DM). We review the evidence on the relationship between the gut and T2DM remission after gastric bypass surgery, and discuss the potential mechanisms underlying the above relationship: gut anatomical rearrangement, microbial composition changes, altered gut cells, and gut hormone modulation. However, the exact changes and their relative importance in the metabolic improvements after gastric bypass surgery remain to be further clarified. Elucidating the precise metabolic mechanisms of T2DM resolution after bypass surgery will help to reveal the molecular mechanisms of pathogenesis, and facilitate the development of novel diagnoses and preventative interventions for this common disease.
Collapse
Affiliation(s)
- Bing Li
- Key Laboratory of Systems Biology, SIBS-Novo Nordisk Translational Research Centre for PreDiabetes, Shanghai Institutes for Biological Sciences, CAS, Shanghai, China
| | | | | | | |
Collapse
|
128
|
Takashima S, Gold D, Hartenstein V. Stem cells and lineages of the intestine: a developmental and evolutionary perspective. Dev Genes Evol 2013; 223:85-102. [PMID: 23179635 PMCID: PMC3873164 DOI: 10.1007/s00427-012-0422-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 10/12/2012] [Indexed: 12/19/2022]
Abstract
The intestine consists of epithelial cells that secrete digestive enzymes and mucus (gland cells), absorb food particles (enterocytes), and produce hormones (endocrine cells). Intestinal cells are rapidly turned over and need to be replaced. In cnidarians, mitosis of differentiated intestinal cells accounts for much of the replacement; in addition, migratory, multipotent stem cells (interstitial cells) contribute to the production of intestinal cells. In other phyla, intestinal cell replacement is solely the function of stem cells entering the gut from the outside (such as in case of the neoblasts of platyhelminths) or intestinal stem cells located within the midgut epithelium (as in both vertebrates or arthropods). We will attempt in the following to review important aspects of midgut stem cells in different animal groups: where are they located, what types of lineages do they produce, and how do they develop. We will start out with a comparative survey of midgut cell types found across the animal kingdom; then briefly look at the specification of these cells during embryonic development; and finally focus on the stem cells that regenerate midgut cells during adult life. In a number of model systems, including mouse, zebrafish and Drosophila, the molecular pathways controlling intestinal stem cells proliferation and the specification of intestinal cell types are under intensive investigation. We will highlight findings of the recent literature, focusing on aspects that are shared between the different models and that point at evolutionary ancient mechanisms of intestinal cell formation.
Collapse
Affiliation(s)
- Shigeo Takashima
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
129
|
Roach G, Heath Wallace R, Cameron A, Emrah Ozel R, Hongay CF, Baral R, Andreescu S, Wallace KN. Loss of ascl1a prevents secretory cell differentiation within the zebrafish intestinal epithelium resulting in a loss of distal intestinal motility. Dev Biol 2013; 376:171-86. [PMID: 23353550 DOI: 10.1016/j.ydbio.2013.01.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 01/05/2013] [Accepted: 01/08/2013] [Indexed: 02/07/2023]
Abstract
The vertebrate intestinal epithelium is renewed continuously from stem cells at the base of the crypt in mammals or base of the fold in fish over the life of the organism. As stem cells divide, newly formed epithelial cells make an initial choice between a secretory or enterocyte fate. This choice has previously been demonstrated to involve Notch signaling as well as Atonal and Her transcription factors in both embryogenesis and adults. Here, we demonstrate that in contrast to the atoh1 in mammals, ascl1a is responsible for formation of secretory cells in zebrafish. ascl1a-/- embryos lack all intestinal epithelial secretory cells and instead differentiate into enterocytes. ascl1a-/- embryos also fail to induce intestinal epithelial expression of deltaD suggesting that ascl1a plays a role in initiation of Notch signaling. Inhibition of Notch signaling increases the number of ascl1a and deltaD expressing intestinal epithelial cells as well as the number of developing secretory cells during two specific time periods: between 30 and 34hpf and again between 64 and 74hpf. Loss of enteroendocrine products results in loss of anterograde motility in ascl1a-/- embryos. 5HT produced by enterochromaffin cells is critical in motility and secretion within the intestine. We find that addition of exogenous 5HT to ascl1a-/- embryos at near physiological levels (measured by differential pulse voltammetry) induce anterograde motility at similar levels to wild type velocity, distance, and frequency. Removal or doubling the concentration of 5HT in WT embryos does not significantly affect anterograde motility, suggesting that the loss of additional enteroendocrine products in ascl1a-/- embryos also contributes to intestinal motility. Thus, zebrafish intestinal epithelial cells appear to have a common secretory progenitor from which all subtypes form. Loss of enteroendocrine cells reveals the critical need for enteroendocrine products in maintenance of normal intestinal motility.
Collapse
Affiliation(s)
- Gillian Roach
- Department of Biology, Clarkson University, 8 Clarkson Ave., Potsdam, NY 13699, USA
| | | | | | | | | | | | | | | |
Collapse
|
130
|
Flasse LC, Stern DG, Pirson JL, Manfroid I, Peers B, Voz ML. The bHLH transcription factor Ascl1a is essential for the specification of the intestinal secretory cells and mediates Notch signaling in the zebrafish intestine. Dev Biol 2013; 376:187-97. [PMID: 23352790 DOI: 10.1016/j.ydbio.2013.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 01/09/2013] [Accepted: 01/11/2013] [Indexed: 11/24/2022]
Abstract
Notch signaling has a fundamental role in stem cell maintenance and in cell fate choice in the intestine of different species. Canonically, Notch signaling represses the expression of transcription factors of the achaete-scute like (ASCL) or atonal related protein (ARP) families. Identifying the ARP/ASCL genes expressed in the gastrointestinal tract is essential to build the regulatory cascade controlling the differentiation of gastrointestinal progenitors into the different intestinal cell types. The expression of the ARP/ASCL factors was analyzed in zebrafish to identify, among all the ARP/ASCL factors found in the zebrafish genome, those expressed in the gastrointestinal tract. ascl1a was found to be the earliest factor detected in the intestine. Loss-of-function analyses using the pia/ascl1a mutant, revealed that ascl1a is crucial for the differentiation of all secretory cells. Furthermore, we identify a battery of transcription factors expressed during secretory cell differentiation and downstream of ascl1a. Finally, we show that the repression of secretory cell fate by Notch signaling is mediated by the inhibition of ascl1a expression. In conclusion, this work identifies Ascl1a as a key regulator of the secretory cell lineage in the zebrafish intestine, playing the same role as Atoh1 in the mouse intestine. This highlights the diversity in the ARP/ASCL family members acting as cell fate determinants downstream from Notch signaling.
Collapse
Affiliation(s)
- Lydie C Flasse
- Unit of Molecular Biology and Genetic Engineering, Giga-Research, University of Liège, 1 avenue de l'Hôpital B34, B-4000 Sart-Tilman (Liège), Belgium
| | | | | | | | | | | |
Collapse
|
131
|
Abstract
Current therapies for the treatment of type 1 diabetes include daily administration of exogenous insulin and, less frequently, whole-pancreas or islet transplantation. Insulin injections often result in inaccurate insulin doses, exposing the patient to hypo- and/or hyperglycemic episodes that lead to long-term complications. Islet transplantation is also limited by lack of high-quality islet donors, early graft failure, and chronic post-transplant immunosuppressive treatment. These barriers could be circumvented by designing a safe and efficient strategy to restore insulin production within the patient's body. Porcine islets have been considered as a possible alternative source of transplantable insulin-producing cells to replace human cadaveric islets. More recently, embryonic or induced pluripotent stem cells have also been examined for their ability to differentiate in vitro into pancreatic endocrine cells. Alternatively, it may be feasible to generate new β-cells by ectopic expression of key transcription factors in endogenous non-β-cells. Finally, engineering surrogate β-cells by in vivo delivery of the insulin gene to specific tissues is also being studied as a possible therapy for type 1 diabetes. In the present review, we discuss these different approaches to restore insulin production.
Collapse
Affiliation(s)
- Eva Tudurí
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
132
|
Egerod KL, Engelstoft MS, Grunddal KV, Nøhr MK, Secher A, Sakata I, Pedersen J, Windeløv JA, Füchtbauer EM, Olsen J, Sundler F, Christensen JP, Wierup N, Olsen JV, Holst JJ, Zigman JM, Poulsen SS, Schwartz TW. A major lineage of enteroendocrine cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatostatin. Endocrinology 2012; 153:5782-95. [PMID: 23064014 PMCID: PMC7958714 DOI: 10.1210/en.2012-1595] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
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.
Collapse
Affiliation(s)
- Kristoffer L Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, 2200 Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Thanasupawat T, Hammje K, Adham I, Ghia JE, Del Bigio MR, Krcek J, Hoang-Vu C, Klonisch T, Hombach-Klonisch S. INSL5 is a novel marker for human enteroendocrine cells of the large intestine and neuroendocrine tumours. Oncol Rep 2012; 29:149-54. [PMID: 23128569 DOI: 10.3892/or.2012.2119] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 09/19/2012] [Indexed: 11/05/2022] Open
Abstract
We report for the first time the distribution of human INSL5 and its cognate leucine rich G-protein coupled receptor RXFP4 in the large intestine and in neuroendocrine/carcinoid tissues. Immunoreactive INSL5 was uniquely expressed by enteroendocrine cells (EECs) located within the colonic mucosa, whereas colonocytes were immunopositive for RXFP4. INSL5+ and RXFP4+ cells were also detected in human neuroendocrine/carcinoid tissues. We employed a recently described Insl5 knockout mouse model and 2 mouse models of induced colitis to address the relevance of Insl5 in EEC development and in acute inflammation of the colon. We identified INSL5 as a specific marker for synaptophysin+ EECs in the mucosa of the normal human and mouse colon. Insl5 was not essential for the development of mouse synaptophysin+ EECs. The mouse models of chemically induced colitis (dextran sulfate sodium and dinitrobenzene-sulfonic acid) failed to show changes in the numbers of Insl5+ EECs at inflammatory sites during the acute phase of colitis. In conclusion, we showed that INSL5 is a novel marker of colorectal EECs and provide first evidence for the presence of a potentially autocrine/paracrine INSL5-RXFP4 signaling system in the normal human and mouse colon and in rare human neuroendocrine tumours.
Collapse
Affiliation(s)
- Thatchawan Thanasupawat
- Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Muniz LR, Knosp C, Yeretssian G. Intestinal antimicrobial peptides during homeostasis, infection, and disease. Front Immunol 2012; 3:310. [PMID: 23087688 PMCID: PMC3466489 DOI: 10.3389/fimmu.2012.00310] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/17/2012] [Indexed: 12/25/2022] Open
Abstract
Antimicrobial peptides (AMPs), including defensins and cathelicidins, constitute an arsenal of innate regulators of paramount importance in the gut. The intestinal epithelium is exposed to myriad of enteric pathogens and these endogenous peptides are essential to fend off microbes and protect against infections. It is becoming increasingly evident that AMPs shape the composition of the commensal microbiota and help maintain intestinal homeostasis. They contribute to innate immunity, hence playing important functions in health and disease. AMP expression is tightly controlled by the engagement of pattern recognition receptors (PRRs) and their impairment is linked to abnormal host responses to infection and inflammatory bowel diseases (IBD). In this review, we provide an overview of the mucosal immune barriers and the intricate crosstalk between the host and the microbiota during homeostasis. We focus on the AMPs and pay particular attention to how PRRs promote their secretion in the intestine. Furthermore, we discuss their production and main functions in three different scenarios, at steady state, throughout infection with enteric pathogens and IBD.
Collapse
Affiliation(s)
- Luciana R Muniz
- Department of Medicine, Immunology Institute, Mount Sinai School of Medicine New York, NY, USA
| | | | | |
Collapse
|
135
|
Affiliation(s)
- Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8.
| |
Collapse
|
136
|
Grigoryan M, Kedees MH, Charron MJ, Guz Y, Teitelman G. Regulation of mouse intestinal L cell progenitors proliferation by the glucagon family of peptides. Endocrinology 2012; 153:3076-88. [PMID: 22569789 PMCID: PMC3380309 DOI: 10.1210/en.2012-1120] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Glucagon like peptide-1 (GLP-1) and GLP-2 are hormones secreted by intestinal L cells that stimulate glucose-dependent insulin secretion and regulate intestinal growth, respectively. Mice with deletion of the glucagon receptor (Gcgr) have high levels of circulating GLP-1 and GLP-2. We sought to determine whether the increased level of the glucagon-like peptides is due to L cell hyperplasia. We found, first, that high levels of the glucagon-like peptides increase L cell number but does not affect the number of other intestinal epithelial cell types. Second, a large proportion of ileal L cells of Gcgr(-/-) mice coexpressed glucose-dependent insulinotropic peptide (GIP). Cells coexpressing GIP and GLP-1 are termed LK cells. Third, the augmentation in L cell number was due to a higher rate of proliferation of L cell progenitors rather than to the entrance of mature L cells into the cell cycle. Fourth, a high concentration of the glucagon-like peptides in the circulation augmented the mRNA levels of transcription factors expressed by late but not early enteroendocrine progenitors. Fifth, the administration of exendin 9-39, a GLP-1 receptor antagonist, resulted in a decrease in the rate of L cell precursor proliferation. Finally, we determined that L cells do not express the GLP-1 receptor, suggesting that the effect of GLP-1 is mediated by paracrine and/or neuronal signals. Our results suggest that GLP-1 plays an important role in the regulation of L cell number.
Collapse
Affiliation(s)
- Marine Grigoryan
- Department of Cell Biology, State University of New York-Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, New York 11203, USA
| | | | | | | | | |
Collapse
|
137
|
Takashima S, Hartenstein V. Genetic control of intestinal stem cell specification and development: a comparative view. Stem Cell Rev Rep 2012; 8:597-608. [PMID: 22529012 PMCID: PMC3950647 DOI: 10.1007/s12015-012-9351-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cells of the adult vertebrate intestine (ISCs) are responsible for the continuous replacement of intestinal cells, but also serve as site of origin of intestinal neoplasms. The interaction between multiple signaling pathways, including Wnt/Wg, Shh/Hh, BMP, and Notch, orchestrate mitosis, motility, and differentiation of ISCs. Many fundamental questions of how these pathways carry out their function remain unanswered. One approach to gain more insight is to look at the development of stem cells, to analyze the "programming" process which these cells undergo as they emerge from the large populations of embryonic progenitors. This review intends to summarize pertinent data on vertebrate intestinal stem cell biology, to then take a closer look at recent studies of intestinal stem cell development in Drosophila. Here, stem cell pools and their niche environment consist of relatively small numbers of cells, and questions concerning the pattern of cell division, niche-stem cell contacts, or differentiation can be addressed at the single cell level. Likewise, it is possible to analyze the emergence of stem cells during development more easily than in vertebrate systems: where in the embryo do stem cells arise, what structures in their environment do they interact with, and what signaling pathways are active sequentially as a result of these interactions. Given the high degree of conservation among genetic mechanisms controlling stem cell behavior in all animals, findings in Drosophila will provide answers that inform research in the vertebrate stem cell field.
Collapse
Affiliation(s)
- Shigeo Takashima
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | |
Collapse
|
138
|
Goto Y, Kiyono H. Epithelial barrier: an interface for the cross-communication between gut flora and immune system. Immunol Rev 2012; 245:147-63. [PMID: 22168418 DOI: 10.1111/j.1600-065x.2011.01078.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Large numbers of environmental antigens, including commensal bacteria and food-derived antigens, constitutively interact with the epithelial layer of the gastrointestinal (GI) tract. Commensal bacteria peacefully cohabit with the host GI tract and exert multiple beneficial or destructive effects on their host. Intestinal epithelial cells (IECs) constitute the first physical and immunological protective wall against invasive pathogens and a cohabitation niche for commensal bacteria. As the physiological homeostasis of IECs is maintained by multiple biological processes such as apoptosis, autophagy, and the handling of endoplasmic reticulum stress, the aberrant kinetics of these biological events, which have genetic and environmental causes, leads to the development of host intestinal pathogenesis such as inflammatory bowel disease. In addition, IECs recognize and interact with commensal bacteria and give instructions to mucosal immune cells to initiate an immunological balance between active and quiescent conditions, eventually establishing intestinal homeostasis. The mucosal immune system regulates the homeostasis of gut microbiota by producing immunological molecules such as secretory immunoglobulin A, the production of which is mediated by IECs. IECs therefore play a central role in the creation and maintenance of a physiologically and immunologically stable intestinal environment.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
139
|
Nasoohi S, Hemmati AA, Moradi F, Ahmadiani A. The γ-secretase blocker DAPT impairs recovery from lipopolysaccharide-induced inflammation in rat brain. Neuroscience 2012; 210:99-109. [PMID: 22445932 DOI: 10.1016/j.neuroscience.2012.02.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/28/2012] [Accepted: 02/29/2012] [Indexed: 11/15/2022]
Abstract
γ-Secretase is an important contributing enzyme in Alzheimer's disease and is therefore an important therapeutic target. However, the impact of γ-secretase inhibition is not well studied in acute neuroinflammation induced by systemic infection. In this study the influence of γ-secretase on the expression of some proinflammatory markers was assessed in the acute phase as well as the subsiding phase of neuroinflammation. Cerebral γ-secretase cleavage activity was measured by a fluorometric assay after lipopolysaccharide (LPS) intraperitoneal administration. Time profiles of TNF-α and COX-II expression were then determined to detect the time points relevant to the maximal inflammatory responses and the subsequent recovery phase. γ-Secretase activity coincident with TNF-α protein expression returned to its basal level till 8-12 h after systemic challenge with low dose LPS while COX-II over expression lasted for 48-72 h later. Pharmacological inhibition of γ-secretase with local or systemic administration of DAPT (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester) was performed to indicate the results on the developmental and sinking phases of inflammatory responses in 6 and 72 h post LPS respectively. Our results demonstrate that both local and systemic modulation of γ-secretase hyper-activity with DAPT increase the duration of TNF-α, COX-II, and NFκB induction. We consistently found mild augmented apoptosis in animals treated with DAPT as determined by measuring cleaved caspase-3 expression and by TUNEL assay 72 h following LPS injection. These results suggest that γ-secretase modulation interferes with certain immune regulatory pathways which may restrict some inflammatory transcription factors such as NFκB.
Collapse
Affiliation(s)
- S Nasoohi
- Department of Pharmacology and Toxicology, School of Pharmacy and Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan, PO Box 6287, Ahvaz, Iran
| | | | | | | |
Collapse
|
140
|
Generation of functional insulin-producing cells in the gut by Foxo1 ablation. Nat Genet 2012; 44:406-12, S1. [PMID: 22406641 PMCID: PMC3315609 DOI: 10.1038/ng.2215] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 02/07/2012] [Indexed: 12/31/2022]
Abstract
Restoration of regulated insulin secretion is the ultimate goal of therapy for type 1 diabetes. Here, we show that, unexpectedly, somatic ablation of Foxo1 in Neurog3(+) enteroendocrine progenitor cells gives rise to gut insulin-positive (Ins(+)) cells that express markers of mature β cells and secrete bioactive insulin as well as C-peptide in response to glucose and sulfonylureas. Lineage tracing experiments showed that gut Ins(+) cells arise cell autonomously from Foxo1-deficient cells. Inducible Foxo1 ablation in adult mice also resulted in the generation of gut Ins(+) cells. Following ablation by the β-cell toxin streptozotocin, gut Ins(+) cells regenerate and produce insulin, reversing hyperglycemia in mice. The data indicate that Neurog3(+) enteroendocrine progenitors require active Foxo1 to prevent differentiation into Ins(+) cells. Foxo1 ablation in gut epithelium may provide an approach to restore insulin production in type 1 diabetes.
Collapse
|
141
|
Wijtten PJA, Langhout DJ, Verstegen MWA. Small intestine development in chicks after hatch and in pigs around the time of weaning and its relation with nutrition: A review. ACTA AGR SCAND A-AN 2012. [DOI: 10.1080/09064702.2012.676061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
142
|
Moran-Ramos S, Tovar AR, Torres N. Diet: friend or foe of enteroendocrine cells--how it interacts with enteroendocrine cells. Adv Nutr 2012; 3:8-20. [PMID: 22332097 PMCID: PMC3262619 DOI: 10.3945/an.111.000976] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gut hormones play a key role in the regulation of food intake, energy expenditure, glucose homeostasis, lipid metabolism, and a wide range of metabolic functions in response to food ingestion. These hormones are altered in metabolic diseases, such as obesity and type 2 diabetes, and are thus proposed to be possible targets for the prevention or treatment of these diseases. It is clear that food composition, macronutrients, and other non-nutrient components as well as the physical properties of food not only modulate the secretion of gut peptides but also modulate transcription and enteroendocrine cell differentiation, which ultimately modifies gut hormone response. The specific mechanisms or sensing machinery that respond to the different components of the diet have been studied for many years; however, over the last few years, new molecular genetic techniques have led to important advances, thereby allowing a deeper understanding of these mechanisms. This review addresses the current knowledge regarding enteroendocrine cells and how diet interacts with this machinery to stimulate and regulate the secretion of gut peptides. The potential for diet interventions as a promising strategy for modulating gut hormone responses to food ingestion and, ultimately, preventing or treating metabolic diseases is being emphasized considering that these diseases are currently a public health burden.
Collapse
Affiliation(s)
- Sofia Moran-Ramos
- Departamento de Fisiología de la Nutrición Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán,” México, México City, México; and,Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
| | - Armando R. Tovar
- Departamento de Fisiología de la Nutrición Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán,” México, México City, México; and
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán,” México, México City, México; and,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
143
|
Grande E, Capdevila J, Barriuso J, Antón-Aparicio L, Castellano D. Gastroenteropancreatic neuroendocrine tumor cancer stem cells: do they exist? Cancer Metastasis Rev 2011; 31:47-53. [DOI: 10.1007/s10555-011-9328-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
144
|
Lahar N, Lei NY, Wang J, Jabaji Z, Tung SC, Joshi V, Lewis M, Stelzner M, Martín MG, Dunn JCY. Intestinal subepithelial myofibroblasts support in vitro and in vivo growth of human small intestinal epithelium. PLoS One 2011; 6:e26898. [PMID: 22125602 PMCID: PMC3219641 DOI: 10.1371/journal.pone.0026898] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 10/06/2011] [Indexed: 01/17/2023] Open
Abstract
The intestinal crypt-niche interaction is thought to be essential to the function, maintenance, and proliferation of progenitor stem cells found at the bases of intestinal crypts. These stem cells are constantly renewing the intestinal epithelium by sending differentiated cells from the base of the crypts of Lieberkühn to the villus tips where they slough off into the intestinal lumen. The intestinal niche consists of various cell types, extracellular matrix, and growth factors and surrounds the intestinal progenitor cells. There have recently been advances in the understanding of the interactions that regulate the behavior of the intestinal epithelium and there is great interest in methods for isolating and expanding viable intestinal epithelium. However, there is no method to maintain primary human small intestinal epithelium in culture over a prolonged period of time. Similarly no method has been published that describes isolation and support of human intestinal epithelium in an in vivo model. We describe a technique to isolate and maintain human small intestinal epithelium in vitro from surgical specimens. We also describe a novel method to maintain human intestinal epithelium subcutaneously in a mouse model for a prolonged period of time. Our methods require various growth factors and the intimate interaction between intestinal sub-epithelial myofibroblasts (ISEMFs) and the intestinal epithelial cells to support the epithelial in vitro and in vivo growth. Absence of these myofibroblasts precluded successful maintenance of epithelial cell formation and proliferation beyond just a few days, even in the presence of supportive growth factors. We believe that the methods described here can be used to explore the molecular basis of human intestinal stem cell support, maintenance, and growth.
Collapse
Affiliation(s)
- Nicholas Lahar
- Division of Gastroenterology and Nutrition, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Nan Ye Lei
- Division of Gastroenterology and Nutrition, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- Division of Gastroenterology and Nutrition, Department of Bioengineering, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Jiafang Wang
- Division of Gastroenterology and Nutrition, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Ziyad Jabaji
- Division of Gastroenterology and Nutrition, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Stephaine C. Tung
- Division of Gastroenterology and Nutrition, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- Division of Gastroenterology and Nutrition, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Vaidehi Joshi
- Division of Gastroenterology and Nutrition, Department of Bioengineering, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Michael Lewis
- Department of Pathology, Veterans Affairs Greater Los Angeles Healthcare System, University of California, Los Angeles, California, United States of America
| | - Matthias Stelzner
- Division of Gastroenterology and Nutrition, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Martín G. Martín
- Division of Gastroenterology and Nutrition, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - James C. Y. Dunn
- Division of Gastroenterology and Nutrition, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- Division of Gastroenterology and Nutrition, Department of Bioengineering, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
145
|
Endocrine differentiation of rat enterocytes in long-term three-dimensional co-culture with intestinal myofibroblasts. In Vitro Cell Dev Biol Anim 2011; 47:707-15. [DOI: 10.1007/s11626-011-9458-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 09/15/2011] [Indexed: 11/26/2022]
|
146
|
Abstract
Multiple approaches have been investigated with the ultimate goal of providing insulin independence to patients with either type 1 or type 2 diabetes. Approaches to produce insulin-secreting cells in culture, convert non-β-cells into functional β-cells or engineer autologous cells to express and secrete insulin in a meal-responsive manner have all been described. This research has been facilitated by significant improvements in both viral and non-viral gene delivery approaches that have enabled new experimental strategies. Many studies have examined possible avenues to confer islet cytoprotection against immune rejection, inflammation and apoptosis by genetic manipulation of islet cells prior to islet transplantation. Here we review several reports based on the reprogramming of pancreas and gut endocrine cells to treat diabetes.
Collapse
Affiliation(s)
- E Tudurí
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
147
|
Belacortu Y, Paricio N. Drosophila as a model of wound healing and tissue regeneration in vertebrates. Dev Dyn 2011; 240:2379-404. [PMID: 21953647 DOI: 10.1002/dvdy.22753] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2011] [Indexed: 11/11/2022] Open
Abstract
Understanding the molecular basis of wound healing and regeneration in vertebrates is one of the main challenges in biology and medicine. This understanding will lead to medical advances allowing accelerated tissue repair after wounding, rebuilding new tissues/organs and restoring homeostasis. Drosophila has emerged as a valuable model for studying these processes because the genetic networks and cytoskeletal machinery involved in epithelial movements occurring during embryonic dorsal closure, larval imaginal disc fusion/regeneration, and epithelial repair are similar to those acting during wound healing and regeneration in vertebrates. Recent studies have also focused on the use of Drosophila adult stem cells to maintain tissue homeostasis. Here, we review how Drosophila has contributed to our understanding of these processes, primarily through live-imaging and genetic tools that are impractical in mammals. Furthermore, we highlight future research areas where this insect may provide novel insights and potential therapeutic strategies for wound healing and regeneration.
Collapse
Affiliation(s)
- Yaiza Belacortu
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, Burjasot, Spain
| | | |
Collapse
|
148
|
Abstract
Neuroendocrine neoplasms arise in almost every organ of the body and are variably defined according to the site of origin. This Review focuses on neuroendocrine neoplasms of the digestive tract and pancreas. The 2010 WHO classification of tumors of the digestive system introduces grading and staging tools for neuroendocrine neoplasms. A carcinoid is now defined as a grade 1 or 2 neuroendocrine tumor and grade 3, small-cell or large-cell carcinomas are defined as neuroendocrine carcinoma. Epidemiological data show a worldwide increase in the prevalence and incidence of gastroentero-pancreatic neuroendocrine tumors in the past few decades, which is probably due to improved methods of detection of these tumors. The current diagnostic procedures and treatment options for neuroendocrine neoplasms are defined and summarized in the Review, although evidence-based data are lacking. Surgery remains the treatment mainstay and somatostatin analogues the basis for both diagnosis and therapy as the only 'theranostic' tool. Emerging compounds including chemotherapeutic agents, small molecules and biological therapies may provide new hope for patients.
Collapse
Affiliation(s)
- Guido Rindi
- Institute of Pathology, Università Cattolica del Sacro Cuore-Policlinico A. Gemelli, Largo A. Gemelli 8, I-00168 Rome, Italy.
| | | |
Collapse
|
149
|
Hermann G, Konukiewitz B, Schmitt A, Perren A, Klöppel G. Hormonally defined pancreatic and duodenal neuroendocrine tumors differ in their transcription factor signatures: expression of ISL1, PDX1, NGN3, and CDX2. Virchows Arch 2011; 459:147-54. [PMID: 21739268 DOI: 10.1007/s00428-011-1118-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/15/2011] [Accepted: 04/20/2011] [Indexed: 12/26/2022]
Abstract
We recently identified the transcription factor (TF) islet 1 gene product (ISL1) as a marker for well-differentiated pancreatic neuroendocrine tumors (P-NETs). In order to better understand the expression of the four TFs, ISL1, pancreatico-duodenal homeobox 1 gene product (PDX1), neurogenin 3 gene product (NGN3), and CDX-2 homeobox gene product (CDX2), that mainly govern the development and differentiation of the pancreas and duodenum, we studied their expression in hormonally defined P-NETs and duodenal (D-) NETs. Thirty-six P-NETs and 14 D-NETs were immunostained with antibodies against the four pancreatic hormones, gastrin, serotonin, calcitonin, ISL1, PDX1, NGN3, and CDX2. The TF expression pattern of each case was correlated with the tumor's hormonal profile. Insulin-positive NETs expressed only ISL1 (10/10) and PDX1 (9/10). Glucagon-positive tumors expressed ISL1 (7/7) and were almost negative for the other TFs. Gastrin-positive NETs, whether of duodenal or pancreatic origin, frequently expressed PDX1 (17/18), ISL1 (14/18), and NGN3 (14/18). CDX2 was mainly found in the gastrin-positive P-NETs (5/8) and rarely in the D-NETs (1/10). Somatostatin-positive NETs, whether duodenal or pancreatic in origin, expressed ISL1 (9/9), PDX1 (3/9), and NGN3 (3/9). The remaining tumors showed labeling for ISL1 in addition to NGN3. There was no association between a particular TF pattern and NET features such as grade, size, location, presence of metastases, and functional activity. We conclude from our data that there is a correlation between TF expression patterns and certain hormonally defined P-NET and D-NET types, suggesting that most of the tumor types originate from embryologically determined precursor cells. The observed TF signatures do not allow us to distinguish P-NETs from D-NETs.
Collapse
Affiliation(s)
- Gratiana Hermann
- Department of Pathology, Assaf Harofeh Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
150
|
Gupta D, Harvey SAK, Kaminski N, Swamynathan SK. Mouse conjunctival forniceal gene expression during postnatal development and its regulation by Kruppel-like factor 4. Invest Ophthalmol Vis Sci 2011; 52:4951-62. [PMID: 21398290 DOI: 10.1167/iovs.10-7068] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
PURPOSE To identify the changes in postnatal mouse conjunctival forniceal gene expression and their regulation by Klf4 during the eye-opening stage when the goblet cells first appear. METHODS Laser microdissection (LMD) was used to collect conjunctival forniceal cells from postnatal (PN) day 9, PN14 and PN20 wild-type (WT), and PN14 Klf4-conditional null (Klf4CN) mice, in which goblet cells are absent, developing, present, and missing, respectively. Microarrays were used to compare gene expression among these groups. Expression of selected genes was validated by quantitative RT-PCR, and spatiotemporal expression was assessed by in situ hybridization. RESULTS This study identified 668, 251, 1160, and 139 transcripts that were increased and 492, 377, 1419, and 57 transcripts that were decreased between PN9 and PN14, PN14 and PN20, PN9 and PN20, and PN14 WT and Klf4CN conjunctiva, respectively. Transcripts encoding transcription factors Spdef, FoxA1, and FoxA3 that regulate goblet cell development in other mucosal epithelia, and epithelium-specific Ets (ESE) transcription factor family members were increased during conjunctival development. Components of pathways related to the mesenchymal-epithelial transition, glycoprotein biosynthesis, mucosal immunity, signaling, and endocytic and neural regulation were increased during conjunctival development. Conjunctival Klf4 target genes differed significantly from the previously identified corneal Klf4 target genes, implying tissue-dependent regulatory targets for Klf4. CONCLUSIONS The changes in gene expression accompanying mouse conjunctival development were identified, and the role of Klf4 in this process was determined. This study provides new probes for examining conjunctival development and function and reveals that the gene regulatory network necessary for goblet cell development is conserved across different mucosal epithelia.
Collapse
Affiliation(s)
- Divya Gupta
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Eye and Ear Institute, 203 Lothrop Street, Pittsburgh PA 15213, USA
| | | | | | | |
Collapse
|