Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Pathophysiol. Nov 15, 2015; 6(4): 131-139
Published online Nov 15, 2015. doi: 10.4291/wjgp.v6.i4.131
Elusive liver factor that causes pancreatic α cell hyperplasia: A review of literature
Run Yu, Carcinoid and Neuroendocrine Tumor Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
Yun Zheng, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
Matthew B Lucas, Princeton University, Princeton, NJ 08544, United States
Yun-Guang Tong, Department of Medicine, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA 90048, United States
Yun-Guang Tong, Department of Pathology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
Author contributions: All authors contributed to this paper.
Supported by National Cancer Institute of the National Institutes of Health, No. R00CA138914 (YT); and by National Natural Science Foundation, No. 81372216 (YT).
Conflict-of-interest statement: The authors have no conflicts of interest to disclose. Dr. Yunguang Tong is supported by the National Cancer Institute of the National Institutes of Health under award number R00CA138914 (YT) and National Natural Science Foundation under grant number 81372216 (YT). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Run Yu, MD, PhD, Carcinoid and Neuroendocrine Tumor Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, B-131, Los Angeles, CA 90048, United States. run.yu@cshs.org
Telephone: +1-310-4234774 Fax: +1-310-4230440
Received: April 1, 2015
Peer-review started: April 2, 2015
First decision: June 18, 2015
Revised: July 3, 2015
Accepted: July 24, 2015
Article in press: July 27, 2015
Published online: November 15, 2015

Abstract

Tumors and cancers of the gastrointestinal tract and pancreas are commonly derived from precursor lesions so that understanding the physiological, cellular, and molecular mechanisms underlying the pathogenesis of precursor lesions is critical for the prevention and treatment of those neoplasms. Pancreatic neuroendocrine tumors (PNETs) can also be derived from precursor lesions. Pancreatic α cell hyperplasia (ACH), a specific and overwhelming increase in the number of α cells, is a precursor lesion leading to PNET pathogenesis. One of the 3 subtypes of ACH, reactive ACH is caused by glucagon signaling disruption and invariably evolves into PNETs. In this article, the existing work on the mechanisms underlying reactive ACH pathogenesis is reviewed. It is clear that the liver secretes a humoral factor regulating α cell numbers but the identity of the liver factor remains elusive. Potential approaches to identify the liver factor are discussed.

Key Words: Pancreatic α cell hyperplasia, Humoral factor, Pancreatic neuroendocrine tumors, Digestive system hormone, Liver

Core tip: Tumors and cancers of the gastrointestinal tract and pancreas are commonly derived from precursor lesions. One of the precursor lesions, reactive pancreatic α cell hyperplasia is caused by glucagon signaling disruption and invariably evolves into pancreatic neuroendocrine tumors. In this article, the existing work on the mechanisms underlying the novel precursor lesion is reviewed. It is clear that the liver secretes a humoral factor regulating pancreatic α cell numbers but the identity of the liver factor remains elusive. Potential approaches to identify the liver factor are discussed.



INTRODUCTION

Tumors and cancers of the gastrointestinal tract and pancreas are commonly derived from precursor lesions[1-3]. For example, colon cancer is derived from polypoid or non-polypoid pre-neoplastic lesions in the colon, and pancreatic ductal carcinoma from pancreatic intraepithelial neoplasia. Neuroendocrine tumors in the gastrointestinal tract and pancreas (GEP-NETs) are relatively rare and indolent tumors with variable biological behaviors[4-6]. GEP-NETs can also be derived from precursor lesions[7-9]. In atrophic gastritis, hypergastrinemia drives enterochromaffin-like cell hyperplasia, which in turn can give rise to gastric carcinoids[7,10]. In ulcerative colitis, microscopic neuroendocrine tumors can arise after long disease duration, probably in response to inflammation[7,11]. Recently, precursor lesions giving rise to pancreatic neuroendocrine tumors (PNETs) have drawn much attention and become more understood. It is well known now that diffuse precursor lesions including endocrine cell hyperplasia, dysplasia, and microadenomas are present in the pancreata of patients with familial tumor syndromes such as multiple endocrine neoplasia syndrome type 1 (MEN1) and von Hippel-Lindau disease, and of animal models of PNETs[12-16]. In the pancreata of patients with MEN1 and mice with heterozygous MEN1 inactivation, the hyperplastic endocrine cells are polyclonal and multi-hormonal and contain the normal menin allele, while microadenomas have to first lose the normal menin allele[17,18]. In contrast, uni-hormonal pancreatic endocrine cell hyperplasia such as pancreatic α cell hyperplasia (ACH) and pancreatic polypeptide cell hyperplasia has only been recognized in the last several years[8,9,19]. Although pancreatic polypeptide cell hyperplasia may be a physiological variation of normal pancreatic polypeptide cell distribution, ACH is clearly a pathologic precursor lesion leading to PNET pathogenesis[19].

In this article, we will summarize how the discovery of a novel hereditary tumor syndrome, Mahvash disease, has stimulated interest in the pathogenesis of ACH, and discuss the possible identify of an elusive liver factor that may cause the ACH.

The data we review are based on work in our own laboratories and PubMed and major endocrine conferences search using key words pancreatic α cell hyperplasia, glucagon receptor mutation, glucagon receptor antagonism, and hyperglucagonemia.

PANCREATIC ACH

ACH is defined as an overwhelming and specific increase of pancreatic α cell numbers[8,19]. Based on etiology and glucagon levels, 3 types of ACH are observed. Reactive ACH is caused in humans by inactivating glucagon receptor mutations and is associated with marked hyperglucagonemia. Because the glucagon receptor is inactive, the severe hyperglucagonemia in reactive ACH does not result in glucagonoma syndrome. Non-functional ACH has an unknown cause and is associated with normal glucagon levels. Functional ACH also has an unknown cause but is associated with hyperglucagonemia that results in glucagonoma syndrome.

Reactive ACH is most extensively studied due to the novel Mahvash disease and the existence of multiple animal models. We first described the Mahvash disease which is hyperglucagonemia, ACH, and PNETs but without glucagonoma syndrome, caused by an inactivating glucagon receptor mutation[20,21]. Later, we and others have confirmed the Mahvash disease (Tang L and Yu R, unpublished results)[19,22]. Currently, 8 inactivating glucagon receptor mutations are known.

We further established that the glucagon receptor knockout (Gcgr-/-) mice are a murine model of Mahvash disease[23-25]. The Gcgr-/- mice exhibit ACH throughout their lifespan. Dysplastic islets consisted of mostly α cells are evident from 5-7 mo on and glucagonomas are detected from 10-12 mo to death. Hyperplasia is also observed in the exocrine compartment but dysplasia, carcinoma in situ, or frank exocrine carcinoma is not found. Large PNETs contribute at least partially to the premature demise of the Gcgr-/- mice. Three other murine models also mimic the Mahvash disease in some aspects. The prohormone convertase 2 knockout (PC-/-) mice cannot make mature glucagon; they exhibit ACH and eventually develop PNETs[26,27]. The preproglucagon knockout (Gcg-/-) mice cannot make any proglucagon- derived peptide hormones, including mature glucagon; they also exhibit ACH and eventually develop PNETs[28,29]. The liver-specific Gsα knockout mice cannot transduce the glucagon signaling in hepatocytes; they exhibit hyperglucagonemia and ACH, and eventually develop PNETs as well[30,31].

Thus both in humans and in mice, reactive ACH ensues whenever glucagon signaling is disrupted and evolves into PNETs eventually. Reactive ACH thus is clearly a precursor lesion leading to PNET pathogenesis.

PATHOGENESIS OF REACTIVE ACH

The pathogenesis of reactive ACH in Gcgr-/- mice is studied in detail. As increased pancreatic endocrine cell numbers can be due to proliferation, neogenesis, or reduced apoptosis, they are each examined at 6-7 mo[23]. α cell proliferation measured by proliferating cell nuclear antigen or Ki-67 labeling is very low and not significantly different in WT and Gcgr-/- mice. α cell neogenesis measured by counting singlet and doublet α cells and exocrine ducts harboring glucagon-positive cells is much higher in Gcgr-/- than in WT mice. α cell apoptosis measured by TUNEL labeling is very low in both Gcgr-/- and WT mice and not significantly different. While upregulated α cell neogenesis is also seen in the PC-/- mice throughout their lifespan, higher α cell proliferation is found at 3 mo[27].

The hyperplastic α cells in the Gcgr-/- mice exhibit abnormal differentiation. A few of these cells are positively labeled with both glucagon and insulin, and some express pancreatic and duodenal homeobox 1, a β cell marker[23]. Most α cells express embryonic α cell markers such as GLUT2[32]. Abnormal α cell differentiation is also seen in humans with mutated Gcgr and in the PC-/- mice as they both express glucagon-like peptide 1, which is normally not expressed in the α cells[21,27].

REACTIVE ACH AND THE LIVER

As reactive ACH universally occurs after glucagon signaling inhibition (see above), it is logical to hypothesize that glucagon signaling negatively feeds back on α cell number regulation and loss of the negative feedback causes the ACH[33]. A number of lines of evidence point to the liver as the organ which sends inhibitory signals to the α cells during normal glucagon signaling and a stimulatory signal to them when glucagon signaling is disrupted. First, liver is the natural target organ of glucagon signaling. Second, liver-specific Gsα deletion in mice recapitulates ACH pathogenesis[30]. Third, liver-specific glucagon receptor deletion in mice results in a phenotype very similar to that of mice with global glucagon receptor deletion[34]. Fourth, glucagon receptor re-expression in the liver of Gcgr-/- mice reduces glucagon levels by almost 99%[35]. Therefore, if the liver does not respond to glucagon but all other organs do, reactive ACH ensues; conversely, if the liver does respond to glucagon but all other organs do not, reactive ACH likely reverses. In other words, the liver is likely necessary and sufficient to be the organ regulating the number of α cells in response to glucagon signaling.

THE ELUSIVE LIVER FACTOR

The liver communicates with the pancreas via neuronal and humoral signals. It has been shown that the liver can regulate insulin secretion and pancreatic β cell proliferation through neuronal signals[36-38]. In a similar manner, the liver may regulate glucagon secretion and pancreatic α cell proliferation through neuronal connection, but there has not been any direct experimental evidence supporting or disputing that. In contrast, there is strong evidence that the liver regulates glucagon secretion and pancreatic α cell proliferation through a humoral factor as shown by islet transplantation experiments[34]. Transplanted wildtype islets in Gcgr-/- recipient mice exhibit higher α/β cell ratio and increased α cell proliferation, compared with those in wildtype recipient animals. Conversely, transplanted Gcgr-/- islets in wildtype recipient mice exhibited reduced α-cell proliferation compared with those in Gcgr-/- recipient animals.

The nature and identity of the liver factor that causes reactive ACH have been sought after. As the liver gene expression must be different between the wildtype and the Gcgr-/- mice, systems approaches such as DNA microarray studies are done to efficiently provide systemic and novel insights into the nature of the liver factor. We compared gene expression profile of 4 WT and 4 Gcgr-/- mouse livers at 2.5 mo (2 females and 2 males in each group) by Affymetrix GeneChip Mouse Gene 1.0 ST Array. The microarray data were analyzed using Genespring 11 (Tables 1 and 2). A total of 125 genes were significantly differentially expressed (> 2 fold change and P < 0.05). Since ACH occurs regardless of sex, we eliminated 47 genes with differential expression only limited in one sex, leaving 35 genes upregulated and 43 genes downregulated in both female and male Gcgr-/- mouse liver. The differential expression of some of the 78 genes was validated by realtime polymerase chain reaction (Figure 1). We reason that potential candidate genes should encode secretory proteins. Of the genes overexpressed in Gcgr-/- liver, Igfbp1, Defb1, Serpina7, Inhba, Cxcl13, Il1b, and Cxcl9 are secretory proteins and may stimulate α-cell differentiation and proliferation. Defb1 is particularly interesting as it is very significantly overexpressed in the Gcgr-/- liver (Table 1). Defensins are a group of cysteine-rich antimicrobial peptides that function to help defend against microbial infections[39]. They are mostly secreted by leukocytes and epithelial cells and their anti-microbial mechanisms are multiple. There are a few families of defensins according to their structures in mice and humans. Originally, defensin β1 (DB1, encoded by Defb1) is found to be expressed in the lung and urogenital epithelials cells[40-42]. Later, DB1 is also found in the liver, especially in the biliary epithelial cells under obstructive jaundice[40,43,44]. DB1 released in circulation may result in ACH. Serpina7 encodes thyroxine-binding globulin (TBG) which binds thyroxine and increases total thyroxine levels[45]. As the index patient with Mahvash disease has normal thyroid functions, suggesting normal TBG levels, it is unlikely that TBG is the liver factor that causes ACH. For Cxcl13, Il1b, and Cxcl9, please see below. Literature review does not give us any clues on which underexpressed genes in Gcgr-/- mouse liver might encode a secretory protein that acts as inhibitor of α-cell differentiation and proliferation.

Table 1 Genes significantly overexpressed in the Gcgr-/- mouse liver.
Gene symbolmRNA descriptionGO biological process termFold increase
Cdkn1aCyclin-dependent kinase inhibitor 1A (P21), transcript variant 1Response to DNA damage stimulus/cell cycle/cell cycle arrest/negative regulation of cell proliferation5.6
Igfbp1Insulin-like growth factor binding protein 1Regulation of cell growth5.5
Defb1Defensin beta 1Defense response/response to bacterium/defense response to bacterium/innate immune response5.2
Gpr64G protein-coupled receptor 64, transcript variant 1Signal transduction/cell surface receptor linked signaling pathway/G-protein coupled receptor protein signaling pathway/neuropeptide signaling pathway5.1
Serpina7Serine (or cysteine) peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 7Post-embryonic development/response to vitamin A/response to drug4.9
Cpt1bCarnitine palmitoyltransferase 1b, muscle, nuclear gene encoding mitochondrial proteinLipid metabolic process/fatty acid metabolic process/transport/long-chain fatty acid transport/long-chain fatty acid transport4.6
Chac1ChaC, cation transport regulator-like 1 (E. coli)Apoptosis/response to unfolded protein/biological_process4.3
Npas2Neuronal PAS domain protein 2Transcription/regulation of transcription, DNA-dependent/signal transduction/circadian sleep/wake cycle/regulation of transcription/locomotor rhythm/positive regulation of transcription from RNA polymerase II promoter/rhythmic process3.9
Slc34a2Solute carrier family 34 (sodium phosphate), member 2In utero embryonic development/transport/ion transport/sodium ion transport/phosphate transport/phosphate transport/phosphate transport3.6
Fabp5Fatty acid binding protein 5, epidermalGlucose metabolic process/lipid metabolic process/phosphatidylcholine biosynthetic process/transport/glucose transport3.5
BC023105cDNA sequence BC023105Unknown3.1
Cav1Caveolin 1, caveolae proteinMAPKKK cascade/inactivation of MAPK activity/vasculogenesis/response to hypoxia/negative regulation of endothelial cell proliferation/negative regulation of cytokine-mediated signaling pathway/triglyceride metabolic process/calcium ion transport/cellular calcium ion homeostasis/cellular calcium ion homeostasis/endocytosis/regulation of smooth muscle contraction2.8
LplLipoprotein lipaseLipid metabolic process/positive regulation of macrophage derived foam cell differentiation/lipid catabolic process/triglyceride biosynthetic process/triglyceride catabolic process2.7
Il22ra1Interleukin 22 receptor, alpha 1Blood coagulation2.6
AcacaAcetyl-Coenzyme A carboxylase alphaTissue homeostasis/acetyl-CoA metabolic process/lipid metabolic process/fatty acid biosynthetic process/metabolic process/lipid biosynthetic process/response to organic cyclic substance/multicellular organismal protein metabolic process2.6
InhbaInhibin beta-AMesoderm formation/hemopoietic progenitor cell differentiation/growth/positive regulation of transcription from RNA polymerase II promoter/mesodermal cell differentiation/negative regulation of hair follicle development2.6
Gadd45bGrowth arrest and DNA-damage-inducible 45 betaActivation of MAPKK activity/negative regulation of protein kinase activity/apoptosis/multicellular organismal development/cell differentiation/regulation of cell cycle2.4
Tgtp1T-cell specific GTPase 1Immune response/response to virus2.4
Rassf4Ras association (RalGDS/AF-6) domain family member 4Cell cycle/signal transduction2.4
Cxcl13Chemokine (C-X-C motif) ligand 13Chemotaxis/inflammatory response/immune response/lymph node development2.4
Il1bInterleukin 1 betaAngiogenesis/fever/inflammatory response/immune response/elevation of cytosolic calcium ion concentration/aging2.4
Tgtp1T-cell specific GTPase 1Immune response/response to virus2.3
AsnsAsparagine synthetaseAsparagine biosynthetic process/glutamine metabolic process/metabolic process/cellular amino acid biosynthetic process2.3
Socs2Suppressor of cytokine signaling 2, transcript variant 1Lactation/regulation of growth/regulation of multicellular organism growth/negative regulation of multicellular organism growth/negative regulation of multicellular organism growth/positive regulation of neuron differentiation/negative regulation of JAK-STAT cascade/mammary gland alveolus development2.3
Meig1Meiosis expressed gene 1Meiosis2.3
Cxcl9Chemokine (C-X-C motif) ligand 9Inflammatory response/immune response2.2
Vtcn1V-set domain containing T cell activation inhibitor 1Negative regulation of T cell activation2.2
H2-Ab1Histocompatibility 2, class II antigen A, beta 1Antigen processing and presentation of peptide or polysaccharide antigen via MHC class II/immune response/antigen processing and presentation/antigen processing and presentation of exogenous peptide antigen via MHC class II2.2
Spon2Spondin 2, extracellular matrix proteinCell adhesion/innate immune response2.2
Rgs16Regulator of G-protein signaling 16G-protein coupled receptor protein signaling pathway/negative regulation of signal transduction2.1
Il2rgInterleukin 2 receptor, gamma chainRegulation of gene expression/positive regulation of CD4-positive, CD25-positive, alpha-beta regulatory T cell differentiation/positive regulation of T cell differentiation in the thymus/positive regulation of B cell differentiation2.1
Wdr67WD repeat domain 67, transcript variant 1Regulation of Rab GTPase activity2.1
Prss8Protease, serine, 8 (prostasin)Hair follicle development/proteolysis2.1
Bach2BTB and CNC homology 2Transcription/regulation of transcription, DNA-dependent/regulation of transcription2.0
Serpina12Serine (or cysteine) peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 12Unknown2.0
Figure 1
Figure 1 Realtime polymerase chain reaction of several genes differentially expressed in the Gcgr-/- mice. See text for details.
Table 2 Genes significantly underexpressed in the Gcgr-/- mouse liver.
Gene symbolmRNA descriptionGO biological process termFold decrease
Mmd2Monocyte to macrophage differentiation-associated 2Cytolysis9.7
NnmtNicotinamide N-methyltransferaseUnknown6.2
GcgrGlucagon receptorExocytosis/signal transduction/cell surface receptor linked signaling pathway/G-protein coupled receptor protein signaling pathway/G-protein signaling, coupled to cAMP nucleotide second messenger/activation of adenylate cyclase activity by G-protein signaling pathway5.3
Mfsd2aMajor facilitator superfamily domain containing 2ATransport/transmembrane transport4.2
OatOrnithine aminotransferase, nuclear gene encoding mitochondrial proteinUnknown4.1
Slc10a2Solute carrier family 10, member 2Transport/ion transport/sodium ion transport/organic anion transport/bile acid and bile salt transport3.9
A1bgAlpha-1-B glycoproteinUnknown3.5
Gm129Gene model 129 (NCBI)Unknown3.3
SdsSerine dehydrataseGluconeogenesis/cellular amino acid metabolic process/metabolic process3.1
Pck1Phosphoenolpyruvate carboxykinase 1, cytosolicGluconeogenesis/gluconeogenesis/oxaloacetate metabolic process/lipid metabolic process/glycerol biosynthetic process from pyruvate3.0
Lrtm1Leucine-rich repeats and transmembrane domains 1Unknown3.0
Ntrk2Neurotrophic tyrosine kinase, receptor, type 2, transcript variant 1Vasculogenesis/protein amino acid phosphorylation/transmembrane receptor protein tyrosine kinase signaling pathway/multicellular organismal development/nervous system development/feeding behavior/glutamate secretion/regulation of metabolic process/cell differentiation/brain-derived neurotrophic factor receptor signaling pathway/mechanoreceptor differentiation3.0
Gls2Glutaminase 2 (liver, mitochondrial), nuclear gene encoding mitochondrial proteinGutamine metabolic process3.0
Susd4Sushi domain containing 4Unknown2.9
Slc16a5Solute carrier family 16 (monocarboxylic acid transporters), member 5Unknown2.9
Ccrn4lCCR4 carbon catabolite repression 4-like (S. cerevisiae)Rhythmic process2.9
LhppPhospholysine phosphohistidine inorganic pyrophosphate phosphataseMetabolic process2.7
NebNebulinRegulation of actin filament length/sarcomere organization2.6
Got1Glutamate oxaloacetate transaminase 1, solubleOxaloacetate metabolic process/glycerol biosynthetic process/cellular amino acid metabolic process/aspartate metabolic process/aspartate biosynthetic process/biosynthetic process/glutamate catabolic process to aspartate/glutamate catabolic process to 2-oxoglutarate/dicarboxylic acid metabolic process/fatty acid homeostasis2.6
Sult5a1Sulfotransferase family 5A, member 1Unknown2.6
Hapln1Hyaluronan and proteoglycan link protein 1Cell adhesion2.5
Mt2Metallothionein 2Cellular zinc ion homeostasis/nitric oxide mediated signal transduction/detoxification of copper ion2.5
Mt1Metallothionein 1Cellular metal ion homeostasis/cellular zinc ion homeostasis/nitric oxide mediated signal transduction/detoxification of copper ion2.4
Slc3a1Solute carrier family 3, member 1Amino acid transport2.4
TrdnTriadinCellular calcium ion homeostasis/regulation of release of sequestered calcium ion into cytosol by sarcoplasmic reticulum/negative regulation of calcium ion transport via store-operated calcium channel activity2.4
Bhlhe41Basic helix-loop-helix family, member e41Negative regulation of transcription from RNA polymerase II promoter/transcription/regulation of transcription, DNA-dependent/circadian rhythm/entrainment of circadian clock/regulation of transcription2.3
Usp2Ubiquitin specific peptidase 2, transcript variant 3Ubiquitin-dependent protein catabolic process2.3
Derl3Der1-like domain family, member 3Unknown2.3
Mrap2Melanocortin 2 receptor accessory protein 2, transcript variant 1Unknown2.2
Ncam2Neural cell adhesion molecule 2, transcript variant 1Cell adhesion2.2
S1pr5Sphingosine-1-phosphate receptor 5Signal transduction/G-protein coupled receptor protein signaling pathway2.2
1810046K07RikRIKEN cDNA 1810046K07 geneUnknown2.2
Nrg4Neuregulin 4Unknown2.2
Gas2Growth arrest specific 2Apoptosis/cell cycle/cell cycle arrest/regulation of cell shape2.2
Ttc39bTetratricopeptide repeat domain 39BUnknown2.2
Cyp17a1Cytochrome P450, family 17, subfamily a, polypeptide 1Steroid biosynthetic process/glucocorticoid biosynthetic process/oxidation reduction2.1
Creld2cysteine-rich with EGF-like domains 2Unknown2.1
Upp2Uridine phosphorylase 2Nucleoside metabolic process/nucleotide catabolic process2.1
ArAndrogen receptorIn utero embryonic development/transcription/regulation of transcription, DNA-dependent/regulation of transcription from RNA polymerase II promoter/male gonad development/cellular process/regulation of gene expression/male somatic sex determination/androgen receptor signaling pathway/androgen receptor signaling pathway/positive regulation of estrogen receptor signaling pathway/positive regulation of MAPKKK cascade/positive regulation of insulin-like growth factor receptor signaling pathway2.1
Gm10419Lung RCB-0558 LLC cDNA, RIKEN full-length enriched library, clone:G730014J15 product: Hypothetical proteinUnknown2.1
Sdf2l1Stromal cell-derived factor 2-like 1Unknown2.0
TrdnTriadinCellular calcium ion homeostasis/regulation of release of sequestered calcium ion into cytosol by sarcoplasmic reticulum/negative regulation of calcium ion transport via store-operated calcium channel activity2.0
Antxr2Anthrax toxin receptor 2Unknown2.0
Figure 2
Figure 2 Schematic drawing of regulation of pancreatic α cell number by a humoral liver factor. The numbers indicate specific ways to disrupt glucagon signaling. (1) Glucagon deletion; (2) PC2 deletion; (3) Glucagon receptor (Gcgr) global deletion; (4) Gcgr liver-specific deletion; (5) Gcgr inactivating mutation; (6) Gcgr antisense RNA; (7) Gcgr antagonists; (8) Gcgr antibodies; and (9) Gsa liver-specific deletion. See text for details.

Pathway analysis suggested that WT and Gcgr-/- liver exhibit different metabolic profiles. As expected, genes involved in gluconeogenesis, glycogen synthesis, and glycogenolysis were downregulated in the Gcgr-/- liver, compared with those in WT (Table 3). Interestingly, genes involved in inflammation and cell proliferation were upregulated in the Gcgr-/- liver. The protein products of genes regulating cell proliferation unlikely diffuse out of the liver thus are improbable signals for regulating α cell mass. In contrast, the protein products of genes regulating inflammation are mostly cytokines which are secreted into the circulation and can reach the α cells, such as Cxcl13, Il1b, and Cxcl9. Interestingly, interleukin-6 (IL6), a cytokine secreted by T cells and macrophages (but not by the liver), upregulates α-cell mass but circulating IL6 levels are normal in the Gcgr-/- mice[34,46]. Alternatively, multiple liver-elaborated cytokines may act synergistically to cause ACH.

Table 3 Pathway analysis of differentially expressed genes in the Gcgr-/- mouse liver.
Glucose homeostasisInflammationCell proliferationMetabolism
Underexpressed in Gcgr-/- mouse liverNnmt, Got1, Sds, Pck1Gas2Slc10a2, Ntrk2, Gls2, Lhpp, Sult5a1, Mt2, Mt1, Slc3a1, Trdn, Usp2, Cyp17a1, Upp2
Overexpressed in Gcgr-/- mouse liverIl1b, Cxcl13, Tgtp1, Cxcl9, Defb1, Vtcn1, H2-Ab1, Spon2, Il2rgCdkn1a, Igfbp1, Chac1, Cav1, Inhba, Gadd45b, Rassf4, Socs2, Meig1Serpina7, Cpt1b, Slc34a2, Fabp5, Lpl, Acaca, Asns

The liver may indirectly regulate α cell differentiation and proliferation by metabolic signals. Not surprisingly, the metabolic profile of Gcgr-/- mice and wildtype counterparts are vastly different, as shown by polyomic metabolic profiling[47]. Similar to our results, genes involved in gluconeogenesis and amino acid catabolism are downregulated. Furthermore, genes involved in fatty acid oxidation processes are also downregulated and genes involved in glycolysis, fatty acid synthesis, and cholesterol synthesis are upregulated. More pertinent to the potential mechanisms for ACH pathogenesis are the dramatic changes in the levels of metabolites[47]. As reported before[21,23], glucose levels are decreased by 1.4-fold. Consistent with decreased gluconeogenesis in the Gcgr-/- mice, amino acids and amino acid derivatives levels are significantly elevated. The most upregulated amino acids are threonine (9.6-fold), serine (8.7-fold), and asparagine (8.1-fold). Amino acid derivatives levels are also higher in the Gcgr-/- mice, the highest being 2-aminodipic acid and ornithine (both 5.4-fold). Levels of certain nucleotides and their derivatives are elevated, e.g., pyridoxine levels are 3.6-fold elevated. Levels of some vitamins are different; those of dihydrofolic acid are 5.3-fold elevated. Glycerol and glycerol derivatives levels are about 2-fold lower. Intriguingly, the levels of cholic acid and glycocholic acid, two bile acids, are markedly and unexpectedly elevated (244- and 154-fold, respectively). There have been only a few studies addressing glucagon signaling and bile acids. In the rats, glucagon increases cholic acid levels[48]; in cultured cells, one bile acid, chenodeoxycholic acid, desensitizes the glucagon receptor[49]. Bile acids, however, are recognized recently as metabolic regulators[50]. Wildtype mice fed with cholic acid exhibit markedly elevated bile acid levels but their pancreas weight and glucagon levels are not changed[34]. Interestingly, α cell mass is somewhat increased (approximately 80%) by cholic acid feeding. Thus a metabolic signal that causes ACH has not been identified yet.

FUTURE DIRECTIONS

The elusive, yet-to-be identified liver factor that causes ACH fulfills the definition of a novel digestive system hormone (Figure 2). The liver factor is produced by the liver and released into the circulation; it then acts remotely on the pancreas to result in ACH. The liver factor could be more than one molecule but we use singular form here for conciseness. To identify this liver factor, the process of discovering leptin may offer some insights. When the first obese mouse models were described, it was not clear why they are obese. A circulating factor was hypothesized[51]. In the obese mouse models, the factor may either stimulate appetite and be overproduced or inhibit appetite and be underproduced. The circulating factor hypothesis was tested by parabiosis which joins the circulation of two mice of various lean and obese phenotypes. Eventually it was found that the ob/ob obese mice lack an inhibitor of appetite (leptin) and the db/db obese mice lack the receptor for the inhibitor (leptin receptor). Analogously, the Gcgr-/- mice liver could produce a stimulator or lack an inhibitor of α cell mass. Whether the liver factor is a stimulator or an inhibitor in nature is critical in guiding the search for the identity of the liver factor that causes ACH. As parabiosis is technically challenging, it probably should be used as a last resort. Alternative approaches such as primary pancreatic islet culture[52,53] may be used first to resolve the stimulator/inhibitor question and later used as a high-throughput model for identifying the factor. For example, if a 10:1 mixture of wildtype and Gcgr-/- serum stimulates α cell proliferation of the wildtype islets, then it is likely that the Gcgr-/- mice have a stimulator of α cell mass. Another potential systems approach is to compare the liver gene expression and metabolic profile of multiple animal models of reactive ACH. As all the models develop reactive ACH, any differentially expressed genes unlikely encode the liver factor and metabolites of various levels unlikely cause ACH, thus greatly narrowing down the list of candidate genes or metabolites. It is also important to point out that other subtypes of ACH exist and not all ACH is associated with glucagon receptor mutation[22,54].

CONCLUSION

Pancreatic ACH is a precursor lesion that gives rise to PNETs. Reactive ACH is associated with hyperglucagonemia and invariably evolves into PNETs in both humans and animal models. The glucagon receptor knockout (Gcgr-/-) mice are one of the murine model of reactive ACH and current research has shown that the liver produces a factor that regulates pancreatic α cell mass. Liver gene expression arrays and metabolic profiling suggest a number of potential candidates for the novel liver hormone but none of them so far tested has been confirmed. As understanding the physiological, cellular, and molecular mechanisms underlying reactive ACH pathogenesis is important to the prevention and treatment of PNETs, the search for the elusive liver factor is worthwhile but may require a substantial effort to find it.

Footnotes

P- Reviewer: Barreto S, Welsch T, Zielinski J S- Editor: Ji FF L- Editor: A E- Editor: Liu SQ

References
1.  Geboes K, Ectors N, Geboes KP. Pathology of early lower GI cancer. Best Pract Res Clin Gastroenterol. 2005;19:963-978.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Vieth M, Stolte M. Pathology of early upper GI cancers. Best Pract Res Clin Gastroenterol. 2005;19:857-869.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Scarlett CJ, Salisbury EL, Biankin AV, Kench J. Precursor lesions in pancreatic cancer: morphological and molecular pathology. Pathology. 2011;43:183-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
4.  Klöppel G. Classification and pathology of gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 2011;18 Suppl 1:S1-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 232]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
5.  Díez M, Teulé A, Salazar R. Gastroenteropancreatic neuroendocrine tumors: diagnosis and treatment. Ann Gastroenterol. 2013;26:29-36.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ro C, Chai W, Yu VE, Yu R. Pancreatic neuroendocrine tumors: biology, diagnosis,and treatment. Chin J Cancer. 2013;32:312-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 99]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
7.  Klöppel G, Anlauf M, Perren A. Endocrine precursor lesions of gastroenteropancreatic neuroendocrine tumors. Endocr Pathol. 2007;18:150-155.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Ouyang D, Dhall D, Yu R. Pathologic pancreatic endocrine cell hyperplasia. World J Gastroenterol. 2011;17:137-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 37]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
9.  Klöppel G, Anlauf M, Perren A, Sipos B. Hyperplasia to neoplasia sequence of duodenal and pancreatic neuroendocrine diseases and pseudohyperplasia of the PP-cells in the pancreas. Endocr Pathol. 2014;25:181-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
10.  Bordi C, Yu JY, Baggi MT, Davoli C, Pilato FP, Baruzzi G, Gardini G, Zamboni G, Franzin G, Papotti M. Gastric carcinoids and their precursor lesions. A histologic and immunohistochemical study of 23 cases. Cancer. 1991;67:663-672.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Stewart CJ, Matsumoto T, Jo Y, Mibu R, Hirahashi M, Yao T, Iida M. Multifocal microcarcinoid tumours in ulcerative colitis. J Clin Pathol. 2005;58:111-1112; author reply 1112.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Thompson NW, Lloyd RV, Nishiyama RH, Vinik AI, Strodel WE, Allo MD, Eckhauser FE, Talpos G, Mervak T. MEN I pancreas: a histological and immunohistochemical study. World J Surg. 1984;8:561-574.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Lubensky IA, Pack S, Ault D, Vortmeyer AO, Libutti SK, Choyke PL, Walther MM, Linehan WM, Zhuang Z. Multiple neuroendocrine tumors of the pancreas in von Hippel-Lindau disease patients: histopathological and molecular genetic analysis. Am J Pathol. 1998;153:223-231.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Jensen RT, Berna MJ, Bingham DB, Norton JA. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies. Cancer. 2008;113:1807-1843.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Vinik AI, Gonzales MR. New and emerging syndromes due to neuroendocrine tumors. Endocrinol Metab Clin North Am. 2011;40:19-63, vii.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
16.  Babu V, Paul N, Yu R. Animal models and cell lines of pancreatic neuroendocrine tumors. Pancreas. 2013;42:912-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
17.  Anlauf M, Perren A, Henopp T, Rudolf T, Garbrecht N, Schmitt A, Raffel A, Gimm O, Weihe E, Knoefel WT. Allelic deletion of the MEN1 gene in duodenal gastrin and somatostatin cell neoplasms and their precursor lesions. Gut. 2007;56:637-644.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Crabtree JS, Scacheri PC, Ward JM, Garrett-Beal L, Emmert-Buck MR, Edgemon KA, Lorang D, Libutti SK, Chandrasekharappa SC, Marx SJ. A mouse model of multiple endocrine neoplasia, type 1, develops multiple endocrine tumors. Proc Natl Acad Sci USA. 2001;98:1118-1123.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Yu R. Pancreatic α-cell hyperplasia: facts and myths. J Clin Endocrinol Metab. 2014;99:748-756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
20.  Yu R, Nissen NN, Dhall D, Heaney AP. Nesidioblastosis and hyperplasia of alpha cells, microglucagonoma, and nonfunctioning islet cell tumor of the pancreas: review of the literature. Pancreas. 2008;36:428-431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
21.  Zhou C, Dhall D, Nissen NN, Chen CR, Yu R. Homozygous P86S mutation of the human glucagon receptor is associated with hyperglucagonemia, alpha cell hyperplasia, and islet cell tumor. Pancreas. 2009;38:941-946.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 104]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
22.  Sipos B, Sperveslage J, Anlauf M, Hoffmeister M, Henopp T, Buch S, Hampe J, Weber A, Hammel P, Couvelard A. Glucagon cell hyperplasia and neoplasia with and without glucagon receptor mutations. J Clin Endocrinol Metab. 2015;100:E783-E788.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Yu R, Dhall D, Nissen NN, Zhou C, Ren SG. Pancreatic neuroendocrine tumors in glucagon receptor-deficient mice. PLoS One. 2011;6:e23397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
24.  Yu R, Ren SG, Mirocha J. Glucagon receptor is required for long-term survival: a natural history study of the Mahvash disease in a murine model. Endocrinol Nutr. 2012;59:523-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
25.  Yu R, Nissen NN, Dhall D. Exocrine pancreas hyperplasia without dysplasia in glucagon receptor knockout mice. Pancreas. 2014;43:143-145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
26.  Furuta M, Yano H, Zhou A, Rouillé Y, Holst JJ, Carroll R, Ravazzola M, Orci L, Furuta H, Steiner DF. Defective prohormone processing and altered pancreatic islet morphology in mice lacking active SPC2. Proc Natl Acad Sci USA. 1997;94:6646-6651.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Jones HB, Reens J, Brocklehurst SR, Betts CJ, Bickerton S, Bigley AL, Jenkins RP, Whalley NM, Morgan D, Smith DM. Islets of Langerhans from prohormone convertase-2 knockout mice show α-cell hyperplasia and tumorigenesis with elevated α-cell neogenesis. Int J Exp Pathol. 2014;95:29-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
28.  Hayashi Y, Yamamoto M, Mizoguchi H, Watanabe C, Ito R, Yamamoto S, Sun XY, Murata Y. Mice deficient for glucagon gene-derived peptides display normoglycemia and hyperplasia of islet {alpha}-cells but not of intestinal L-cells. Mol Endocrinol. 2009;23:1990-1999.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 83]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
29.  Hayashi Y, Takano Y, Kasai K, Imai T, Murata Y.  Pancreatic Neuroendocrine Tumors in Mice Deficient in Glucagon Gene. 2014;.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Chen M, Gavrilova O, Zhao WQ, Nguyen A, Lorenzo J, Shen L, Nackers L, Pack S, Jou W, Weinstein LS. Increased glucose tolerance and reduced adiposity in the absence of fasting hypoglycemia in mice with liver-specific Gs alpha deficiency. J Clin Invest. 2005;115:3217-3227.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Weinstein LS Role of the Gnas Gene in Metabolic Regulation. 2012;.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Vuguin PM, Kedees MH, Cui L, Guz Y, Gelling RW, Nejathaim M, Charron MJ, Teitelman G. Ablation of the glucagon receptor gene increases fetal lethality and produces alterations in islet development and maturation. Endocrinology. 2006;147:3995-4006.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Lucas MB, Yu VE, Yu R. Mahvash disease: pancreatic neuroendocrine tumor syndrome caused by inactivating glucagon receptor mutation. J Mol Genet Med. 2013;7:84.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Longuet C, Robledo AM, Dean ED, Dai C, Ali S, McGuinness I, de Chavez V, Vuguin PM, Charron MJ, Powers AC. Liver-specific disruption of the murine glucagon receptor produces α-cell hyperplasia: evidence for a circulating α-cell growth factor. Diabetes. 2013;62:1196-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 138]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
35.  Lee Y, Berglund ED, Wang MY, Fu X, Yu X, Charron MJ, Burgess SC, Unger RH. Metabolic manifestations of insulin deficiency do not occur without glucagon action. Proc Natl Acad Sci USA. 2012;109:14972-14976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 118]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
36.  Imai J, Katagiri H, Yamada T, Ishigaki Y, Suzuki T, Kudo H, Uno K, Hasegawa Y, Gao J, Kaneko K. Regulation of pancreatic beta cell mass by neuronal signals from the liver. Science. 2008;322:1250-1254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 170]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
37.  Imai J, Oka Y, Katagiri H. Identification of a novel mechanism regulating β-cell mass: neuronal relay from the liver to pancreatic β-cells. Islets. 2009;1:75-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
38.  Demir IE, Schäfer KH, Tieftrunk E, Friess H, Ceyhan GO. Neural plasticity in the gastrointestinal tract: chronic inflammation, neurotrophic signals, and hypersensitivity. Acta Neuropathol. 2013;125:491-509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
39.  Lehrer RI. Primate defensins. Nat Rev Microbiol. 2004;2:727-738.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Bals R, Goldman MJ, Wilson JM. Mouse beta-defensin 1 is a salt-sensitive antimicrobial peptide present in epithelia of the lung and urogenital tract. Infect Immun. 1998;66:1225-1232.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  McCray PB, Bentley L. Human airway epithelia express a beta-defensin. Am J Respir Cell Mol Biol. 1997;16:343-349.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Valore EV, Park CH, Quayle AJ, Wiles KR, McCray PB, Ganz T. Human beta-defensin-1: an antimicrobial peptide of urogenital tissues. J Clin Invest. 1998;101:1633-1642.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Huang YH, Wang PW, Tiao MM, Chou MH, Du YY, Huang CC, Chuang JH. Glucocorticoid modulates high-mobility group box 1 expression and Toll-like receptor activation in obstructive jaundice. J Surg Res. 2011;170:e47-e55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
44.  Harada K, Ohba K, Ozaki S, Isse K, Hirayama T, Wada A, Nakanuma Y. Peptide antibiotic human beta-defensin-1 and -2 contribute to antimicrobial defense of the intrahepatic biliary tree. Hepatology. 2004;40:925-932.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Schussler GC. The thyroxine-binding proteins. Thyroid. 2000;10:141-149.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Ellingsgaard H, Ehses JA, Hammar EB, Van Lommel L, Quintens R, Martens G, Kerr-Conte J, Pattou F, Berney T, Pipeleers D. Interleukin-6 regulates pancreatic alpha-cell mass expansion. Proc Natl Acad Sci USA. 2008;105:13163-13168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 200]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
47.  Yang J, MacDougall ML, McDowell MT, Xi L, Wei R, Zavadoski WJ, Molloy MP, Baker JD, Kuhn M, Cabrera O. Polyomic profiling reveals significant hepatic metabolic alterations in glucagon-receptor (GCGR) knockout mice: implications on anti-glucagon therapies for diabetes. BMC Genomics. 2011;12:281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 67]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
48.  Guettet C, Mathe D, Riottot M, Lutton C. Effects of chronic glucagon administration on cholesterol and bile acid metabolism. Biochim Biophys Acta. 1988;963:215-223.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Ikegami T, Krilov L, Meng J, Patel B, Chapin-Kennedy K, Bouscarel B. Decreased glucagon responsiveness by bile acids: a role for protein kinase Calpha and glucagon receptor phosphorylation. Endocrinology. 2006;147:5294-5302.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. Bile acids as regulatory molecules. J Lipid Res. 2009;50:1509-1520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 484]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
51.  Coleman DL. A historical perspective on leptin. Nat Med. 2010;16:1097-1099.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 78]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
52.  Andersson A. Tissue culture of isolated pancreatic islets. Acta Endocrinol Suppl (Copenh). 1976;205:283-294.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Andersson A, Eriksson U, Ostenson CG. Glucagon production by cultured pancreatic islets: effects of different culture conditions and media. In Vitro. 1981;17:378-384.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Al-Sarireh B, Haidermota M, Verbeke C, Rees DA, Yu R, Griffiths AP. Glucagon cell adenomatosis without glucagon receptor mutation. Pancreas. 2013;42:360-362.  [PubMed]  [DOI]  [Cited in This Article: ]