Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38613153

ABSTRACT

BACKGROUND AND PURPOSE: Pancreatic islets are modulated by cross-talk among different cell types and paracrine signalling plays important roles in maintaining glucose homeostasis. Urocortin 3 (UCN3) secreted by pancreatic ß cells activates the CRF2 receptor (CRF2R) and downstream pathways mediated by different G protein or arrestin subtypes in δ cells to cause somatostatin (SST) secretion, and constitutes an important feedback circuit for glucose homeostasis. EXPERIMENTAL APPROACH: Here, we used Arrb1-/-, Arrb2-/-, Gsfl/fl and Gqfl/fl knockout mice, the G11-shRNA-GFPfl/fl lentivirus, as well as functional assays and pharmacological characterization to study how the coupling of Gs, G11 and ß-arrestin1 to CRF2R contributed to UCN3-induced SST secretion in pancreatic δ cells. KEY RESULTS: Our study showed that CRF2R coupled to a panel of G protein and arrestin subtypes in response to UCN3 engagement. While RyR3 phosphorylation by PKA at the S156, S2706 and S4697 sites may underlie the Gs-mediated UCN3- CRF2R axis for SST secretion, the interaction of SYT1 with ß-arrestin1 is also essential for efficient SST secretion downstream of CRF2R. The specific expression of the transcription factor Stat6 may contribute to G11 expression in pancreatic δ cells. Furthermore, we found that different UCN3 concentrations may have distinct effects on glucose homeostasis, and these effects may depend on different CRF2R downstream effectors. CONCLUSIONS AND IMPLICATIONS: Collectively, our results provide a landscape view of signalling mediated by different G protein or arrestin subtypes downstream of paracrine UCN3- CRF2R signalling in pancreatic ß-δ-cell circuits, which may facilitate the understanding of fine-tuned glucose homeostasis networks.

2.
Nat Chem Biol ; 20(4): 484-492, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37945893

ABSTRACT

GPR101 is an orphan G protein-coupled receptor actively participating in energy homeostasis. Here we report the cryo-electron microscopy structure of GPR101 constitutively coupled to Gs heterotrimer, which reveals unique features of GPR101, including the interaction of extracellular loop 2 within the 7TM bundle, a hydrophobic chain packing-mediated activation mechanism and the structural basis of disease-related mutants. Importantly, a side pocket is identified in GPR101 that facilitates in silico screening to identify four small-molecule agonists, including AA-14. The structure of AA-14-GPR101-Gs provides direct evidence of the AA-14 binding at the side pocket. Functionally, AA-14 partially restores the functions of GH/IGF-1 axis and exhibits several rejuvenating effects in wild-type mice, which are abrogated in Gpr101-deficient mice. In summary, we provide a structural basis for the constitutive activity of GPR101. The structure-facilitated identification of GPR101 agonists and functional analysis suggest that targeting this orphan receptor has rejuvenating potential.


Subject(s)
Receptors, G-Protein-Coupled , Mice , Animals , Cryoelectron Microscopy , Receptors, G-Protein-Coupled/metabolism , Ligands
3.
World J Gastrointest Surg ; 15(4): 634-642, 2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37206069

ABSTRACT

BACKGROUND: Hypersplenism and esophageal varices bleeding are the major complications of portal hypertension (PHT). In recent years, increasing attention has been given to spleen preservation operations. The mode and long-term effects of subtotal splenectomy and selective pericardial devascularization for PHT remain controversial. AIM: To investigate the clinical efficacy and safety of subtotal splenectomy combined with selective pericardial devascularization for the treatment of PHT. METHODS: This was a retrospective study of 15 patients with PHT who underwent subtotal splenectomy not preserving the splenic artery or vein combined with selective pericardial devascularization in the Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University from February 2011 to April 2022. Fifteen propensity score-matched patients with PHT who underwent total splenectomy at the same time served as the control group. The patients were followed for up to 11 years after surgery. We compared the postoperative platelet levels, perioperative splenic vein thrombosis, and serum immunoglobulin levels between the two groups. Abdominal enhanced computed tomography was used to evaluate the blood supply and function of the residual spleen. The operation time, intraoperative blood loss, evacuation time, and hospital stay were compared between the two groups. RESULTS: The postoperative platelet level of patients in the subtotal splenectomy group was significantly lower than that in the total splenectomy group (P < 0.05), and the postoperative portal system thrombosis rate in the subtotal splenectomy group was also much lower than that in the total splenectomy group. The levels of serum immunoglobulins (IgG, IgA, and IgM) showed no significant differences after surgery compared with before surgery in the subtotal splenectomy group (P > 0.05), but serum immunoglobulin IgG and IgM levels decreased dramatically after total splenectomy (P < 0.05). The operation time in the subtotal splenectomy group was longer than that in the total splenectomy group (P < 0.05), but there were no significant differences in the amount of intraoperative blood loss, evacuation time, or hospital stay between the two groups. CONCLUSION: Subtotal splenectomy not preserving the splenic artery or vein combined with selective pericardial devascularization is a safe and effective surgical treatment for patients with PHT, not only correcting hypersplenism but also preserving splenic function, especially immunological function.

4.
Cell Metab ; 34(2): 240-255.e10, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108512

ABSTRACT

Along with functionally intact insulin, diabetes-associated insulin peptides are secreted by ß cells. By screening the expression and functional characterization of olfactory receptors (ORs) in pancreatic islets, we identified Olfr109 as the receptor that detects insulin peptides. The engagement of one insulin peptide, insB:9-23, with Olfr109 diminished insulin secretion through Gi-cAMP signaling and promoted islet-resident macrophage proliferation through a ß cell-macrophage circuit and a ß-arrestin-1-mediated CCL2 pathway, as evidenced by ß-arrestin-1-/- mouse models. Systemic Olfr109 deficiency or deficiency induced by Pdx1-Cre+/-Olfr109fl/fl specifically alleviated intra-islet inflammatory responses and improved glucose homeostasis in Akita- and high-fat diet (HFD)-fed mice. We further determined the binding mode between insB:9-23 and Olfr109. A pepducin-based Olfr109 antagonist improved glucose homeostasis in diabetic and obese mouse models. Collectively, we found that pancreatic ß cells use Olfr109 to autonomously detect self-secreted insulin peptides, and this detection arrests insulin secretion and crosstalks with macrophages to increase intra-islet inflammation.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans , Animals , Blood Glucose/metabolism , Diet, High-Fat , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism
5.
Oncogene ; 38(6): 868-880, 2019 02.
Article in English | MEDLINE | ID: mdl-30177842

ABSTRACT

Perihilar cholangiocarcinoma (PHCCA) is the most common type of cholangiocarcinoma with low resection rate and high morbidity. The study of PHCCA biomarkers made progresses slowly compared with intrahepatic cholangiocarcinoma because of surgical complexity and low possibility of radical surgery, which resulted in the difficulty of specimen obtainment. To screen and identify new biomarkers in PHCCA, we constructed a retrospective cohort with 121 PHCCA patients and a prospective cohort consisting of 64 PHCCA patients, and screened the candidate biomarkers by immunohistochemistry and quantified PCR. In our study, expression of high mobility group box 1 (HMGB1) was demonstrated to be significantly associated with microvascular density (MVD) and unfavorable prognosis of PHCCA in both retrospective and prospective study. Moreover, HMGB1 concentrations in bile and serum of PHCCA patients and healthy controls were detected and compared. Postoperative serum HMGB1 and reflux cholangitis indicated recurrence and unfavorable prognosis of PHCCA. With experiments in vitro and in vivo, we demonstrated that intracellular HMGB1 could be released from PHCCA cells and induce invasion and angiogenesis with LPS stimulation. VEGFR2 expression in vessel endothelial cells was upregulated by the released HMGB1 from PHCCA, resulting in the ectopic angiogenesis. In conclusion, intracellular HMGB1 could be released from PHCCA cells and promote angiogenesis via elevating VEGFR2 in vessel endothelial cells. High expression of HMGB1 was associated with MVD and poor prognosis in clinical analyzation. Postoperative serum HMGB1 and cholangitis could predict high recurrence and unfavorable prognosis.


Subject(s)
Bile Duct Neoplasms , HMGB1 Protein/blood , Klatskin Tumor , Neoplasm Proteins/blood , Neovascularization, Pathologic , Bile Duct Neoplasms/blood , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/surgery , Disease-Free Survival , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Klatskin Tumor/blood , Klatskin Tumor/mortality , Klatskin Tumor/surgery , Male , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/mortality , Neovascularization, Pathologic/surgery , Prospective Studies , Survival Rate
6.
J Cell Mol Med ; 22(11): 5596-5606, 2018 11.
Article in English | MEDLINE | ID: mdl-30160357

ABSTRACT

Fibroblast growth factor receptor 2 (FGFR2) was demonstrated to correlate to the progression and prognosis of intrahepatic cholangiocarcinoma (ICC) by numerous evidences. However, as a well-recognized suppressor of FGFR2 signalling, the clinical significance of Sprouty (SPRY) family of ICC has not been investigated. In our study, the expressions of SPRY1-4 in 20 pairs of fresh tumour tissues were detected with qPCR, and in 108 cases of paraffin-embedded tissues with immunohistochemistry. The prognostic value of SPRY family in ICC was estimated with univariate analysis and multivariate analysis. As a result, SPRY2 was identified as an independent prognostic biomarker predicting favourable prognosis of ICC. High SPRY2 expression was correlated with good differentiation of ICC. With silencing SPRY2 expression, we demonstrated that SPRY2 could suppress FGFR2-induced ERK phosphorylation, migration, invasion and epithelial-mesenchymal transition (EMT) under FGF1 stimulation. By overexpressing SPRY2-wide type or SPRY2-Y55F, the tyrosine-55 of SPRY2 was demonstrated to be essential in suppressing ERK phosphorylation, tumour invasion and EMT of ICC cells. In conclusion, SPRY2 was correlated with favourable prognosis of ICC via suppressing FGFR2-induced ERK phosphorylation, invasion and EMT. The phosphorylation of SPRY2-Y55 was required in this tumour-suppressing function of SPRY2.


Subject(s)
Cholangiocarcinoma/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Prognosis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cholangiocarcinoma/epidemiology , Cholangiocarcinoma/pathology , Disease Progression , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Phosphorylation , Signal Transduction/genetics , Tissue Array Analysis
7.
Cell Death Dis ; 9(6): 682, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880854

ABSTRACT

The somatostatin (SST)-secreting cells were mainly distributed in the pancreatic islets, brain, stomach and intestine in mammals and have many physiological functions. In particular, the SST-secreting δ cell is the third most common cell type in the islets of Langerhans. Recent studies have suggested that dysregulation of paracrine interaction between the pancreatic δ cells and ß cells results in impaired glucose homeostasis and contributes to diabetes development. However, direct evidence of the functional importance of SST cells in glucose homeostasis control is still lacking. In the present study, we specifically ablated SST-secreting cells by crossing Sst-cre transgenic mice with R26 DTA mice (Sst Cre R26 DTA ). The Sst Cre R26 DTA mice exhibited neonatal death. The life spans of these mice with severe hypoglycemia were extended by glucose supplementation. Moreover, we observed that SST cells deficiency led to increased insulin content and excessive insulin release, which might contribute to the observed hypoglycemia. Unexpectedly, although SST is critical for the regulation of insulin content, factors other than SST that are produced by pancreatic δ cells via their endogenous corticotropin-releasing hormone receptor 2 (CRHR2) activity play the main roles in maintaining normal insulin release, as well as neonatal glucose homeostasis in the resting state. Taken together, our results identified that the SST cells in neonatal mouse played critical role in control of insulin release and normal islet function. Moreover, we provided direct in vivo evidence of the functional importance of the SST cells, which are essential for neonatal survival and the maintenance of glucose homeostasis.


Subject(s)
Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Somatostatin-Secreting Cells/metabolism , Animals , Animals, Newborn , Glucose/metabolism , Humans , Hypoglycemia/pathology , Insulin/biosynthesis , Insulin Secretion , Mice, Inbred C57BL , Models, Biological , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Somatostatin/metabolism
8.
J Clin Invest ; 127(7): 2631-2646, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28604389

ABSTRACT

Somatostatin secreted by pancreatic δ cells mediates important paracrine interactions in Langerhans islets, including maintenance of glucose metabolism through the control of reciprocal insulin and glucagon secretion. Disruption of this circuit contributes to the development of diabetes. However, the precise mechanisms that control somatostatin secretion from islets remain elusive. Here, we found that a super-complex comprising the cullin 4B-RING E3 ligase (CRL4B) and polycomb repressive complex 2 (PRC2) epigenetically regulates somatostatin secretion in islets. Constitutive ablation of CUL4B, the core component of the CRL4B-PRC2 complex, in δ cells impaired glucose tolerance and decreased insulin secretion through enhanced somatostatin release. Moreover, mechanistic studies showed that the CRL4B-PRC2 complex, under the control of the δ cell-specific transcription factor hematopoietically expressed homeobox (HHEX), determines the levels of intracellular calcium and cAMP through histone posttranslational modifications, thereby altering expression of the Cav1.2 calcium channel and adenylyl cyclase 6 (AC6) and modulating somatostatin secretion. In response to high glucose levels or urocortin 3 (UCN3) stimulation, increased expression of cullin 4B (CUL4B) and the PRC2 subunit histone-lysine N-methyltransferase EZH2 and reciprocal decreases in Cav1.2 and AC6 expression were found to regulate somatostatin secretion. Our results reveal an epigenetic regulatory mechanism of δ cell paracrine interactions in which CRL4B-PRC2 complexes, Cav1.2, and AC6 expression fine-tune somatostatin secretion and facilitate glucose homeostasis in pancreatic islets.


Subject(s)
Cullin Proteins/metabolism , Insulin/metabolism , Multienzyme Complexes/metabolism , Paracrine Communication , Somatostatin-Secreting Cells/metabolism , Somatostatin/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cullin Proteins/genetics , Cyclic AMP/metabolism , Epigenesis, Genetic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin Secretion , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Somatostatin/genetics , Somatostatin-Secreting Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Biol Psychiatry ; 81(8): 654-670, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27916196

ABSTRACT

BACKGROUND: Stress is a conserved physiological response in mammals. Whereas moderate stress strengthens memory to improve reactions to previously experienced difficult situations, too much stress is harmful. METHODS: We used specific ß-adrenergic agonists, as well as ß2-adrenergic receptor (ß2AR) and arrestin knockout models, to study the effects of adaptive ß2AR activation on cognitive function using Morris water maze and object recognition experiments. We used molecular and cell biological approaches to elucidate the signaling subnetworks. RESULTS: We observed that the duration of the adaptive ß2AR activation determines its consequences on learning and memory. Short-term formoterol treatment, for 3 to 5 days, improved cognitive function; however, prolonged ß2AR activation, for more than 6 days, produced harmful effects. We identified the activation of several signaling networks downstream of ß2AR, as well as an essential role for arrestin and lactate metabolism in promoting cognitive ability. Whereas Gs-protein kinase A-cyclic adenosine monophosphate response element binding protein signaling modulated monocarboxylate transporter 1 expression, ß-arrestin-1 controlled expression levels of monocarboxylate transporter 4 and lactate dehydrogenase A through the formation of a ß-arrestin-1/phospho-mitogen-activated protein kinase/hypoxia-inducible factor-1α ternary complex to upregulate lactate metabolism in astrocyte-derived U251 cells. Conversely, long-term treatment with formoterol led to the desensitization of ß2ARs, which was responsible for its decreased beneficial effects. CONCLUSIONS: Our results not only revealed that ß-arrestin-1 regulated lactate metabolism to contribute to ß2AR functions in improved memory formation, but also indicated that the appropriate management of one specific stress pathway, such as through the clinical drug formoterol, may exert beneficial effects on cognitive abilities.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Lactic Acid/metabolism , Learning/physiology , Memory/physiology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Stress, Psychological/metabolism , beta-Arrestin 1/metabolism , Adrenergic beta-2 Receptor Agonists/administration & dosage , Animals , Astrocytes/metabolism , Cell Line , Formoterol Fumarate/administration & dosage , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Learning/drug effects , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Receptors, Adrenergic, beta-2/genetics , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Transcriptome
10.
Br J Pharmacol ; 172(21): 5050-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26248680

ABSTRACT

BACKGROUND AND PURPOSE: Cholecystokinin (CCK) is secreted by intestinal I cells and regulates important metabolic functions. In pancreatic islets, CCK controls beta cell functions primarily through CCK1 receptors, but the signalling pathways downstream of these receptors in pancreatic beta cells are not well defined. EXPERIMENTAL APPROACH: Apoptosis in pancreatic beta cell apoptosis was evaluated using Hoechst-33342 staining, TUNEL assays and Annexin-V-FITC/PI staining. Insulin secretion and second messenger production were monitored using ELISAs. Protein and phospho-protein levels were determined by Western blotting. A glucose tolerance test was carried out to examine the functions of CCK-8s in streptozotocin-induced diabetic mice. KEY RESULTS: The sulfated carboxy-terminal octapeptide CCK26-33 amide (CCK-8s) activated CCK1 receptors and induced accumulation of both IP3 and cAMP. Whereas Gq -PLC-IP3 signalling was required for the CCK-8s-induced insulin secretion under low-glucose conditions, Gs -PKA/Epac signalling contributed more strongly to the CCK-8s-mediated insulin secretion in high-glucose conditions. CCK-8s also promoted formation of the CCK1 receptor/ß-arrestin-1 complex in pancreatic beta cells. Using ß-arrestin-1 knockout mice, we demonstrated that ß-arrestin-1 is a key mediator of both CCK-8s-mediated insulin secretion and of its the protective effect against apoptosis in pancreatic beta cells. The anti-apoptotic effects of ß-arrestin-1 occurred through cytoplasmic late-phase ERK activation, which activates the 90-kDa ribosomal S6 kinase-phospho-Bcl-2-family protein pathway. CONCLUSIONS AND IMPLICATIONS: Knowledge of different CCK1 receptor-activated downstream signalling pathways in the regulation of distinct functions of pancreatic beta cells could be used to identify biased CCK1 receptor ligands for the development of new anti-diabetic drugs.


Subject(s)
Cholecystokinin/physiology , Islets of Langerhans/metabolism , Receptors, Cholecystokinin/metabolism , Signal Transduction , Animals , Apoptosis/physiology , Arrestins/genetics , Cholecystokinin/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , MAP Kinase Signaling System , Mice , Mice, Knockout , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , beta-Arrestin 1 , beta-Arrestins
11.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 36(4): 426-31, 2014 Aug.
Article in Chinese | MEDLINE | ID: mdl-25176213

ABSTRACT

OBJECTIVE: To investigate the effect of ischemia/reperfusion (I/R) on tumor metastasis in a experimental mouse model of hematogenous metastasis after I/R and to quantify expression of vascular cell adhesion molecule-1 (VCAM-1) during I/R. METHODS: An experimental mouse model of metastasis after partial hepatic I/R was designed to determine the effects of I/R on tumor metastasis to liver. Tumor loads were valued 14 days after operation. In addition, the expressions of alanine transaminase (ALT), aspartate transaminase (AST), and VCAM-1 were detected. RESULTS: Two hours after hepatic reperfusion, ALT and AST levels in ischemia 45-minute group and ischemia 30-minute group were significantly higher than in the sham group (all P < 0.05). Also, the changes of ALT and AST were more obvious in the ischemia 45-minute group than in ischemia 30-minute group (all P < 0.05). In the sham group, both ALT and AST slightly and transiently increased. ALT and AST in the ischemia 45-minute group and ischemia 30-minute group at 8 hours were both significantly higher than those at 2 hours reperfusion (P<0.05). The tumor load (valued by hepatic replacement area) and the expression of VCAM-1 in ischemic lobe were significantly larger in the ischemia 45-minute group than in the ischemia 30-minute group and sham group (P = 0.013, P = 0.007). However, there was no statistical difference on tumor load between the right lobe of sham operated mice and the right lobe (nonischemic lobes) of mice subjected to I/R (P = 0.089). Mouse survivals were significantly longer in the sham group than in the ischemia 30-minute group (P = 0.041) but were not significantly different between the ischemia 45-minute group and ischemia 30-minute group (P = 0.055). VCAM-1 expression in ischemia 45-minute group was significantly higher than in ischemia 30-minute group and sham group(P = 0.003, P < 0.001), and it was positively correlated with the hepatic replacement area (r = 0.491, P = 0.045). CONCLUSION: Hepatic I/R promotes liver hematogenic metastasis of hepatocellular carcinoma in mice and at least in part, through the induction of VCAM-1 expression.


Subject(s)
Liver Neoplasms/pathology , Reperfusion Injury/complications , Vascular Cell Adhesion Molecule-1/physiology , Animals , Liver/blood supply , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis
12.
Cell Res ; 24(9): 1067-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25081058

ABSTRACT

The tyrosine phosphorylation barcode encoded in C-terminus of HER2 and its ubiquitination regulate diverse HER2 functions. PTPN18 was reported as a HER2 phosphatase; however, the exact mechanism by which it defines HER2 signaling is not fully understood. Here, we demonstrate that PTPN18 regulates HER2-mediated cellular functions through defining both its phosphorylation and ubiquitination barcodes. Enzymologic characterization and three crystal structures of PTPN18 in complex with HER2 phospho-peptides revealed the molecular basis for the recognition between PTPN18 and specific HER2 phosphorylation sites, which assumes two distinct conformations. Unique structural properties of PTPN18 contribute to the regulation of sub-cellular phosphorylation networks downstream of HER2, which are required for inhibition of HER2-mediated cell growth and migration. Whereas the catalytic domain of PTPN18 blocks lysosomal routing and delays the degradation of HER2 by dephosphorylation of HER2 on pY(1112), the PEST domain of PTPN18 promotes K48-linked HER2 ubiquitination and its rapid destruction via the proteasome pathway and an HER2 negative feedback loop. In agreement with the negative regulatory role of PTPN18 in HER2 signaling, the HER2/PTPN18 ratio was correlated with breast cancer stage. Taken together, our study presents a structural basis for selective HER2 dephosphorylation, a previously uncharacterized mechanism for HER2 degradation and a novel function for the PTPN18 PEST domain. The new regulatory role of the PEST domain in the ubiquitination pathway will broaden our understanding of the functions of other important PEST domain-containing phosphatases, such as LYP and PTPN12.


Subject(s)
Catalytic Domain , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Receptor, ErbB-2/metabolism , Ubiquitination , Amino Acid Motifs , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Crystallography, X-Ray , Female , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Lysine/metabolism , Lysosomes/metabolism , Models, Biological , Molecular Sequence Data , Neoplasm Staging , Peptides/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , Structure-Activity Relationship , Substrate Specificity , beta-Transducin Repeat-Containing Proteins/metabolism
13.
Diabetologia ; 57(9): 1899-910, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24947582

ABSTRACT

AIMS/HYPOTHESIS: Somatostatin secretion from islet delta cells plays an important role in regulating islet function and is tightly controlled by environmental changes. Activation of the adrenergic system promoted somatostatin secretion from islet delta cells; however, the role of the adrenergic system in regulating somatostatin content and transcription has not been defined. An imbalance between the somatostatin content and its secretion may cause dysfunctions in the islet delta cells. We have investigated the role of the adrenergic system in the modulation of somatostatin content and transcription in pancreatic delta cells and the detailed underlying mechanisms of this regulation. METHODS: The stress hormone adrenaline (epinephrine), specific adrenergic agonists or specific adrenergic antagonists were applied to islets from either wild-type or specific adrenergic receptor knockout mice and pancreatic delta cell lines to investigate their effects on somatostatin content and transcription. The GloSensor assay, quantitative real-time PCR, western blots and the dual luciferase assay were used to monitor the cAMP level, somatostatin expression, activations of kinases and transcriptional factors. Arrb1 knockout mice, specific Creb or Pax6 mutations and specific kinase inhibitors were used to dissect the signalling pathway. RESULTS: Adrenaline and isoprenaline increased somatostatin content and transcription through the activation of ß1-/ß2-adrenergic receptors (ß1-/ß2ARs). The somatostatin content in ß1AR(-/-) /ß2AR(-/-) (Adrb1/Adrb2 knockout) mice was 50% lower than in ß1AR(+/+)/ß2AR (+/+) mice. Two parallel signalling pathways, Gs-cAMP-protein kinase A (PKA)-cAMP response element binding protein (CREB) and ß-arrestin 1-extracellular signal-related kinase (ERK)-paired box protein 6 (PAX6), cooperatively regulated isoprenaline-induced somatostatin transcription. CONCLUSIONS/INTERPRETATION: A stress pathway increased somatostatin content and transcription through ß-adrenergic agonism. ß-Arrestin1, ERK and PAX6 are important pancreatic delta cell regulators in addition to cAMP, PKA and CREB. Dysfunction of ß-adrenergic agonism may impair pancreatic delta cell function.


Subject(s)
Arrestins/metabolism , Receptors, Adrenergic/metabolism , Somatostatin-Secreting Cells/metabolism , Somatostatin/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Mice , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , beta-Arrestin 1 , beta-Arrestins
14.
World J Gastroenterol ; 20(14): 4076-84, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24744599

ABSTRACT

AIM: To investigate the correlation between nerve growth factor-tropomyosin-receptor-kinase (NGF-TrkA) signaling pathway and prognosis in intrahepatic cholangiocarcinoma (IHCC). METHODS: NGF and TrkA expression in 83 samples of IHCC was assessed by immunohistochemistry. Correlations between NGF-TrkA expression and clinicopathological features were analyzed by χ² test. Moreover, we evaluated the association between NGF-TrkA and overall survival by univariate and multivariate analysis. With experiments in vitro, we investigated the crucial role of NGF-TrkA on proliferation and invasion of IHCC cells with recombinant NGF-ß stimulation. RESULTS: We found that NGF and TrkA expression was significantly related with differentiation (P = 0.024) and intraneural invasion (P = 0.003), respectively. Additionally, double higher expression of NGF and TrkA was identified as an independent prognostic factor in IHCC (P = 0.003). Moreover, we demonstrated that NGF-TrkA signaling pathway can promote IHCC proliferation and invasion. CONCLUSION: NGF-TrkA double higher expression is an independent prognostic factor in IHCC. NGF-TrkA pathway can promote IHCC progression, indicating that NGF-TrkA may become a potential drug target.


Subject(s)
Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/metabolism , Cholangiocarcinoma/metabolism , Gene Expression Regulation, Neoplastic , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Aged , Cell Line, Tumor , Cell Proliferation , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Signal Transduction , Treatment Outcome
15.
Biochem Biophys Res Commun ; 446(1): 54-60, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24565842

ABSTRACT

Fibroblast growth factor receptor 4 (FGFR4) is related to poor prognosis of several cancers, but the correlation between FGFR4 expression and cholangiocarcinoma (CCA) has not been well elucidated. We investigated the expression of FGFR4 in 83 intrahepatic cholangiocarcinomas (IHCCs), 75 perihilar cholangiocarcinomas (PHCCs) and 41 distal cholangiocarcinomas (DCCs) by immunohistochemistry (IHC), and subsequently evaluated association of FGFR4 with clinicopathologic parameters and survival rate. The rate of FGFR4 higher expression was 61.4% (51/83) in IHCCs, 53.3% (40/75) in PHCCs and 56.1% (23/41) in DCCs. FGFR4 expression was significantly related to poor prognosis of IHCC (P=0.002) and PHCC (P=0.019) with univariate analysis, and also identified as an independent prognostic factor in IHCC (P=0.045) and PHCC (P=0.049) with multivariate analysis. Additionally, with functional assays in vitro, we found FGFR4 can induce proliferation, invasion and epithelial-mesenchymal transition (EMT) of CCA cell lines with FGF19 stimulation. Moreover, FGFR4 inhibitor AP24354 can suppress proliferation, invasion and induce apoptosis of CCA cells. In conclusion, FGFR4 expression can be identified as a significant independent prognostic biomarker of IHCC and PHCC. FGFR4 played a pivotal role in proliferation, invasion and EMT of CCA. FGFR4 inhibitor can suppress proliferation, invasion and induce apoptosis of CCA, indicating that FGFR4 may act as a potential therapeutic target.


Subject(s)
Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic , Cholangiocarcinoma/metabolism , Liver Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Aged , Apoptosis/drug effects , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Disease Progression , Epithelial-Mesenchymal Transition , Female , Gene Knockdown Techniques , HEK293 Cells , Hep G2 Cells , Humans , Imidazoles/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Pyridazines/pharmacology , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...