Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cells ; 10(5)2021 04 21.
Article in English | MEDLINE | ID: mdl-33919154

ABSTRACT

Interleukin-18 receptor accessory protein (IL18RAP) is an indispensable subunit for the IL-18 receptor (IL-18R) complex's ability to mediate high-affinity IL-18 binding and signalling transduction. Interest in IL-18 in systemic lupus erythematosus (SLE) has been mostly focused on its role as a type 1 T helper cell-driving cytokine. The functional significance of IL18RAP in mediating the IL-18-driven response in myeloid cells in SLE remains largely unexplored. This study aimed to investigate the expression and function significance of IL18RAP in neutrophils of SLE patients. By qRT-PCR and Western blot analyses, elevated expressions of IL18RAP mRNA and protein were observed in neutrophils from SLE patients-particularly those with a history of nephritis. IL18RAP expression correlated negatively with complement 3 level and positively with disease activity, with higher expression in patients exhibiting renal and immunological manifestations. The increased IL18RAP expression in SLE neutrophils could be attributed to elevated type I interferon level in sera. Functionally, neutrophils from SLE patients showed higher IL-18-mediated enhancement in reactive oxygen species (ROS) generation, which showed positive correlation with IL18RAP expression and could be neutralized by anti-IL18RAP blocking antibodies. Taken together, our findings suggest that IL-18 could contribute to SLE pathogenesis through mediation of neutrophil dysfunction via the upregulation of IL18RAP expression.


Subject(s)
Interleukin-18 Receptor beta Subunit/immunology , Lupus Erythematosus, Systemic/immunology , Neutrophils/immunology , Reactive Oxygen Species/immunology , Adult , Case-Control Studies , Female , Humans , Interferon Type I/immunology , Interleukin-18/immunology , Male , Middle Aged , Neutrophils/cytology
2.
Transl Cancer Res ; 10(2): 656-668, 2021 Feb.
Article in English | MEDLINE | ID: mdl-35116399

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC), an aggressive breast cancer subtype, is associated with poor prognosis and high mortality rate. In the search for effective therapeutic options, preclinical studies have suggested using systemic oxygenation to inhibit tumor growth and metastasis in various cancer models, including TNBC, by weakening the hypoxia-A2A adenosine receptors (A2AR)-driven immunosuppression in the tumor microenvironment (TME). In our present study, a hemoglobin-based oxygen carrier (HBOC) "YQ23" was tested for its role in modulating the TME and tumor inhibition. METHODS: A syngeneic TNBC mouse model was established by inoculating 4T1 cells subcutaneously in BALB/c mice. Tumor (~100 mm3) bearing mice were treated either with saline or YQ23 (400 mg/kg) i.v. once weekly. To prove the immune-regulatory role of YQ23, CD4+ and CD8+ cells were depleted from a group of mice prior to treatment. Tumor growth was monitored for four weeks while xenografts were isolated at the end of the treatment for ex vivo immunohistological examination. RESULTS: YQ23 significantly inhibited the tumor growth, and this suppressive effect was abolished by depleting the host immune cells. Immunohistochemical staining of xenograft sections showed YQ23 reduced the level of hypoxia and adenosine producing ecto-enzyme CD73. Although there was no significant difference in the make up of the intra-tumoral immune populations, we observed a down-regulation of the immune checkpoint PD-1. In concordance with the weakened immunosuppression, the inflammatory cytokine interferon γ and cytolytic granzyme B were upregulated. CONCLUSIONS: YQ23 treatment may be a potential therapeutic strategy to modulate the TME in TNBC.

3.
Dev Cell ; 46(1): 112-125.e4, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29974860

ABSTRACT

Zebrafish is a powerful model for forward genetics. Reverse genetic approaches are limited by the time required to generate stable mutant lines. We describe a system for gene knockout that consistently produces null phenotypes in G0 zebrafish. Yolk injection of sets of four CRISPR/Cas9 ribonucleoprotein complexes redundantly targeting a single gene recapitulated germline-transmitted knockout phenotypes in >90% of G0 embryos for each of 8 test genes. Early embryonic (6 hpf) and stable adult phenotypes were produced. Simultaneous multi-gene knockout was feasible but associated with toxicity in some cases. To facilitate use, we generated a lookup table of four-guide sets for 21,386 zebrafish genes and validated several. Using this resource, we targeted 50 cardiomyocyte transcriptional regulators and uncovered a role of zbtb16a in cardiac development. This system provides a platform for rapid screening of genes of interest in development, physiology, and disease models in zebrafish.


Subject(s)
Gene Knockout Techniques/methods , Heart/embryology , Promyelocytic Leukemia Zinc Finger Protein/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Genetic Engineering/methods , Morpholinos/genetics , Myocytes, Cardiac/cytology , Transcription, Genetic/genetics , Zebrafish/embryology
4.
Cancer Lett ; 431: 201-212, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29859876

ABSTRACT

MicroRNAs (miRNAs) are endogenous small, non-coding RNAs that regulate genome expression at the post-transcriptional level. They are involved in a wide range of physiological processes including the maintenance of immune homeostasis and normal function. Accumulating evidence from animal studies show that alterations in pan or specific miRNA expression would break immunological tolerance, leading to autoimmunity. Differential miRNA expressions have also been documented in patients of many autoimmune disorders. In this review, we highlight the evidence that signifies the critical role of miRNAs in autoimmunity, specifically on their regulatory roles in the pathogenesis of several rheumatic diseases including systemic lupus erythematosus, rheumatoid arthritis and spondyloarthritis. The potential of miRNAs as biomarkers and therapeutic targets is also discussed. Manipulation of dysregulated miRNAs in vivo through miRNA delivery or inhibition offers promise for new therapeutic strategies in treating rheumatic diseases.


Subject(s)
MicroRNAs/metabolism , Rheumatic Diseases/immunology , Rheumatic Diseases/metabolism , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Autoimmunity , Biomarkers/metabolism , Cell Differentiation , Gene Expression Regulation , Homeostasis , Humans , Immune System , Immune Tolerance , Leukocytes/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/metabolism , T-Lymphocytes, Regulatory/metabolism
5.
J Cell Biol ; 217(3): 1097-1112, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29301867

ABSTRACT

Mechanisms that sense and regulate epithelial morphogenesis, integrity, and homeostasis are incompletely understood. Protease-activated receptor 2 (Par2), the Par2-activating membrane-tethered protease matriptase, and its inhibitor, hepatocyte activator inhibitor 1 (Hai1), are coexpressed in most epithelia and may make up a local signaling system that regulates epithelial behavior. We explored the role of Par2b in matriptase-dependent skin abnormalities in Hai1a-deficient zebrafish embryos. We show an unexpected role for Par2b in regulation of epithelial apical cell extrusion, roles in regulating proliferation that were opposite in distinct but adjacent epithelial monolayers, and roles in regulating cell-cell junctions, mobility, survival, and expression of genes involved in tissue remodeling and inflammation. The epidermal growth factor receptor Erbb2 and matrix metalloproteinases, the latter induced by Par2b, may contribute to some matriptase- and Par2b-dependent phenotypes and be permissive for others. Our results suggest that local protease-activated receptor signaling can coordinate cell behaviors known to contribute to epithelial morphogenesis and homeostasis.


Subject(s)
Cell Proliferation/physiology , Epithelial Cells/metabolism , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Epithelial Cells/cytology , Homeostasis/physiology , Morphogenesis/physiology , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Serine Endopeptidases/genetics , Zebrafish Proteins/genetics
6.
Dev Biol ; 418(1): 157-165, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27333774

ABSTRACT

Sphingosine 1-phosphate (S1P) is a bioactive lipid that acts via G protein-coupled receptors. The S1P receptor S1P1, encoded by S1pr1, is expressed in developing heart but its roles there remain largely unexplored. Analysis of S1pr1 LacZ knockin embryos revealed ß-galactosidase staining in cardiomyocytes in the septum and in the trabecular layer of hearts collected at 12.5 days post coitus (dpc) and weak staining in the inner aspect of the compact layer at 15.5 dpc and later. Nkx2-5-Cre- and Mlc2a-Cre-mediated conditional knockout of S1pr1 led to ventricular noncompaction and ventricular septal defects at 18.5 dpc and to perinatal lethality in the majority of mutants. Further analysis of Mlc2a-Cre conditional mutants revealed no gross phenotype at 12.5 dpc but absence of the normal increase in the number of cardiomyocytes and the thickness of the compact layer at 13.5 dpc and after. Consistent with relative lack of a compact layer, in situ hybridization at 13.5 dpc revealed expression of trabecular markers extending almost to the epicardium in mutants. Mutant hearts also showed decreased myofibril organization in the compact but not trabecular myocardium at 12.5 dpc. These results suggest that S1P signaling via S1P1 in cardiomyocytes plays a previously unknown and necessary role in heart development in mice.


Subject(s)
Heart/embryology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Animals , Cell Proliferation/genetics , Gene Knockout Techniques , Heart Septal Defects, Ventricular/genetics , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Myofibrils/genetics , Myofibrils/metabolism , Myosin Light Chains/genetics , Signal Transduction , Sphingosine-1-Phosphate Receptors
7.
Biochim Biophys Acta ; 1861(7): 594-605, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27090939

ABSTRACT

Non-alcoholic steatohepatitis (NASH), is the form of non-alcoholic fatty liver disease posing risk to progress into serious long term complications. Human and pre-clinical models implicate cellular cholesterol dysregulation playing important role in its development. Mouse model studies suggest synergism between dietary cholesterol and fat in contributing to NASH but the mechanisms remain poorly understood. Our laboratory previously reported the primary importance of hepatic endoplasmic reticulum cholesterol (ER-Chol) in regulating hepatic ER stress by comparing the responses of wild type, Ldlr-/-xLcat+/+ and Ldlr-/-xLcat-/- mice, to a 2% high cholesterol diet (HCD). Here we further investigated the roles of ER-Chol and ER stress in HFHS diet-induced NASH using the same strains. With HFHS diet feeding, both WT and Ldlr-/-xLcat+/+ accumulate ER-Chol in association with ER stress and inflammasome activation but the Ldlr-/-xLcat-/- mice are protected. By contrast, all three strains accumulate cholesterol crystal, in correlation with ER-Chol, albeit less so in Ldlr-/-xLcat-/- mice. By comparison, HCD feeding per se (i) is sufficient to promote steatosis and activate inflammasomes, and (ii) results in dramatic accumulation of cholesterol crystal which is linked to inflammasome activation in Ldlr-/-xLcat-/- mice, independent of ER-Chol. Our data suggest that both dietary fat and cholesterol each independently promote steatosis, cholesterol crystal accumulation and inflammasome activation through distinct but complementary pathways. In vitro studies using palmitate-induced hepatic steatosis in HepG2 cells confirm the key roles by cellular cholesterol in the induction of steatosis and inflammasome activations. These novel findings provide opportunities for exploring a cellular cholesterol-focused strategy for treatment of NASH.


Subject(s)
Cholesterol, Dietary/metabolism , Endoplasmic Reticulum Stress/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Receptors, LDL/genetics , Animals , Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Regulation , Hep G2 Cells , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lecithin Cholesterol Acyltransferase Deficiency/metabolism , Lipid Metabolism/genetics , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Oxidation-Reduction , Palmitic Acid/pharmacology , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Receptors, LDL/deficiency , Signal Transduction
8.
Cancer Chemother Pharmacol ; 76(6): 1199-207, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26553104

ABSTRACT

PURPOSE: Adjunct chemoradiation is offered to unresectable esophageal squamous cell carcinoma (ESCC) patients, while its use is limited in tumors with strong resistance. Oxygen carriers or anti-hypoxic drugs belong to an emerging class of regulators that can alleviate tumor hypoxia. METHODS: We investigate the potential use of a novel oxygen carrier YQ23 in sensitizing chemoresistant ESCC in a series of subcutaneous tumor xenograft models developed using ESCC cell lines with different strengths of chemosensitivities. RESULTS: Tumor xenografts were developed using SLMT-1 and HKESC-2 ESCC cell lines with different strengths of resistance to two chemotherapeutic drugs, 5-fluorouracil and cisplatin. More resistant SLMT-1 xenografts responded better to YQ23 treatment than HKESC-2, as reflected by the induced tumor oxygen level. YQ23 sensitized SLMT-1 xenografts toward 5-fluorouracil via its effect on reducing the level of a hypoxic marker HIF-1α. Furthermore, a derangement of tumor microvessel density and integrity was demonstrated with a concurrent decrease in the level of a tumor mesenchymal marker vimentin. Similar to the 5-fluorouracil sensitizing effect, YQ23 also enhanced the response of SLMT-1 xenografts toward cisplatin by reducing the tumor size and the number of animals with invasive tumors. Chemosensitive HKESC-2 xenografts were irresponsive to combined YQ23 and cisplatin treatment. CONCLUSIONS: In all, YQ23 functions selectively on chemoresistant ESCC xenografts, which implicates its potential use as a chemosensitizing agent for ESCC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/drug therapy , Hemoglobins/pharmacology , Xenograft Model Antitumor Assays , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Synergism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Fluorouracil/pharmacology , Humans , Male , Mice, Nude , Oxygen/metabolism , Tumor Burden/drug effects
9.
PLoS One ; 8(7): e68843, 2013.
Article in English | MEDLINE | ID: mdl-23894351

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the major malignancies worldwide and is associated with poor prognosis due to the high incidences of metastasis and tumor recurrence. Our previous study showed that overexpression of p21-activated protein kinase 1 (PAK1) is frequently observed in HCC and is associated with a more aggressive tumor behavior, suggesting that PAK1 is a potential therapeutic target in HCC. In the current study, an allosteric small molecule PAK1 inhibitor, IPA-3, was evaluated for the potential in suppressing hepatocarcinogenesis. Consistent with other reports, inhibition of PAK1 activity was observed in several human HCC cell lines treated with various dosages of IPA-3. Using cell proliferation, colony formation and BrdU incorporation assays, we demonstrated that IPA-3 treatment significantly inhibited the growth of HCC cells. The mechanisms through which IPA-3 treatment suppresses HCC cell growth are enhancement of apoptosis and blockage of activation of NF-κB. Furthermore, our data suggested that IPA-3 not only inhibits the HCC cell growth, but also suppresses the metastatic potential of HCC cells. Nude mouse xenograft assay demonstrated that IPA-3 treatment significantly reduced the tumor growth rate and decreased tumor volume, indicating that IPA-3 can suppress the in vivo tumor growth of HCC cells. Taken together, our demonstration of the potential preclinical efficacy of IPA-3 in HCC provides the rationale for cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Disulfides/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , NF-kappa B/metabolism , Naphthols/pharmacology , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Male , Protein Transport , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
PLoS One ; 7(6): e39913, 2012.
Article in English | MEDLINE | ID: mdl-22761926

ABSTRACT

Osmoregulation via maintenance of water and salt homeostasis is a vital process. In the brain, a functional secretin (SCT) and secretin receptor (SCTR) axis has recently been shown to mediate central actions of angiotensin II (ANGII), including initiation of water intake and stimulation of vasopressin (VP) expression and release. In this report, we provide evidence that estrogen-related receptor α (ERRα, NR3B1), a transcription factor mainly involved in metabolism, acts as an upstream activator of the SCT gene. In vitro studies using mouse hypothalamic cell line N-42 show that ERRα upregulates SCT promoter and gene expression. More importantly, knockdown of endogenous ERRα abolishes SCT promoter activation in response to hypertonic and ANGII stimulations. In mouse brain, ERRα coexpresses with SCT in various osmoregulatory brain regions, including the lamina terminalis and the paraventricular nucleus of the hypothalamus, and its expression is induced by hyperosmotic and ANGII treatments. Based on our data, we propose that both the upregulation of ERRα and/or the increased binding of ERRα to the mouse SCT promoter are two possible mechanisms for the elevated SCT expression upon hyperosmolality and central ANGII stimulation.


Subject(s)
Angiotensin II/pharmacology , Estrogen Receptor alpha/physiology , Secretin/metabolism , Up-Regulation/physiology , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers , Electrophoretic Mobility Shift Assay , Humans , Immunohistochemistry , Injections, Intraventricular , Mice , Polymerase Chain Reaction , Rats
11.
FASEB J ; 24(12): 5024-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20739612

ABSTRACT

Fluid balance is critical to life and hence is tightly controlled in the body. Angiotensin II (ANGII), one of the most important components of this regulatory system, is recognized as a dipsogenic hormone that stimulates vasopressin (VP) expression and release. However, detailed mechanisms regarding how ANGII brings about these changes are not fully understood. In the present study, we show initially that the osmoregulatory functions of secretin (SCT) in the brain are similar to those of ANGII in mice and, more important, we discovered the role of SCT as the link between ANGII and its downstream effects. This was substantiated by the use of two knockout mice, SCTR(-/-) and SCT(-/-), in which we show the absence of an intact SCT/secretin receptor (SCTR) axis resulted in an abolishment or much reduced ANGII osmoregulatory functions. By immunohistochemical staining and in situ hybridization, the proteins and transcripts of SCT and its receptor are found in the paraventricular nucleus (PVN) and lamina terminalis. We propose that SCT produced in the circumventricular organs is transported and released in the PVN to stimulate vasopressin expression and release. In summary, our findings identify SCT and SCTR as novel elements of the ANGII osmoregulatory pathway in maintaining fluid balance in the body.


Subject(s)
Angiotensin II/pharmacology , Secretin/metabolism , Secretin/pharmacology , Animals , Drinking/drug effects , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Secretin/genetics , Vasopressins/metabolism , Water-Electrolyte Balance/drug effects
12.
Hepatology ; 52(1): 204-14, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20578263

ABSTRACT

UNLABELLED: During bile duct ligation (BDL), the growth of large cholangiocytes is regulated by the cyclic adenosine monophosphate (cAMP)/extracellular signal-regulated kinase 1/2 (ERK1/2) pathway and is closely associated with increased secretin receptor (SR) expression. Although it has been suggested that SR modulates cholangiocyte growth, direct evidence for secretin-dependent proliferation is lacking. SR wild-type (WT) (SR(+/+)) or SR knockout (SR(-/-)) mice underwent sham surgery or BDL for 3 or 7 days. We evaluated SR expression, cholangiocyte proliferation, and apoptosis in liver sections and proliferating cell nuclear antigen (PCNA) protein expression and ERK1/2 phosphorylation in purified large cholangiocytes from WT and SR(-/-) BDL mice. Normal WT mice were treated with secretin (2.5 nmoles/kg/day by way of osmotic minipumps for 1 week), and biliary mass was evaluated. Small and large cholangiocytes were used to evaluate the in vitro effect of secretin (100 nM) on proliferation, protein kinase A (PKA) activity, and ERK1/2 phosphorylation. SR expression was also stably knocked down by short hairpin RNA, and basal and secretin-stimulated cAMP levels (a functional index of biliary growth) and proliferation were determined. SR was expressed by large cholangiocytes. Knockout of SR significantly decreased large cholangiocyte growth induced by BDL, which was associated with enhanced apoptosis. PCNA expression and ERK1/2 phosphorylation were decreased in large cholangiocytes from SR(-/-) BDL compared with WT BDL mice. In vivo administration of secretin to normal WT mice increased ductal mass. In vitro, secretin increased proliferation, PKA activity, and ERK1/2 phosphorylation of large cholangiocytes that was blocked by PKA and mitogen-activated protein kinase kinase inhibitors. Stable knockdown of SR expression reduced basal cholangiocyte proliferation. SR is an important trophic regulator sustaining biliary growth. CONCLUSION: The current study provides strong support for the potential use of secretin as a therapy for ductopenic liver diseases.


Subject(s)
Bile Ducts/pathology , Cholestasis, Extrahepatic/complications , Liver Diseases/etiology , Liver/pathology , Receptors, G-Protein-Coupled/physiology , Receptors, Gastrointestinal Hormone/physiology , Animals , Apoptosis , Bile Ducts/drug effects , Cell Proliferation , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Gene Knockout Techniques , Hyperplasia/genetics , Hyperplasia/pathology , Liver/drug effects , Liver Diseases/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Organ Size , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone/genetics , Secretin/pharmacology
13.
Int J Cancer ; 127(1): 43-54, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-19904746

ABSTRACT

Secretin plays a key role in the regulation of normal cholangiocyte physiology via secretin receptor (SCTR). SCTR expression is upregulated during extrahepatic cholestasis induced by bile duct ligation and closely associated with cholangiocyte proliferative responses. Although well studied in normal cholangiocytes, the role of secretin and the expression of SCTR in the regulation of cholangiocarcinoma proliferation are unknown. In vitro, secretin (10(-7) M) displayed differential effects on normal cholangiocyte [H-69 and human intrahepatic biliary epithelial cell line (HIBEpiC)] and cholangiocarcinoma (Mz-ChA-1, HuH-28, TFK-1, SG231, CCLP1 and HuCC-T1) cell lines as such secretin is mitogenic for normal cholangiocytes and antiproliferative for cholangiocarcinoma. As expected in normal cholangiocytes (HIBEpiC), secretin increased intracellular cyclic adenosine monophosphate (cAMP) levels. However, the effect of secretin on intracellular cAMP levels was suppressed in Mz-ChA-1 cells. Secretin-stimulated intracellular cAMP levels in Mz-ChA-1 were restored by pretreatment with pertussis toxin, suggesting that the receptor coupled to Galpha(i) rather than Galpha(s). SCTR expression was found to be downregulated in 4 of the 6 cholangiocarcinoma cell lines evaluated and in human cholangiocarcinoma biopsy samples. In vivo, secretin significantly inhibited the tumor size and more than doubled tumor latency, which was associated with a decrease in proliferating cell nuclear antigen and an increase in cleaved-caspase 3 expression levels. Our results demonstrate that secretin and/or the modulation of SCTR expression might have potential as a therapeutic tool in the treatment of cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Cyclic AMP/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Secretin/physiology , Signal Transduction/physiology , Cell Division/physiology , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Humans , Polymerase Chain Reaction
14.
Article in English | MEDLINE | ID: mdl-19372104

ABSTRACT

Small heterodimer partner (SHP) is an orphan nuclear receptor in which gene expression can be upregulated by bile acids. It regulates its target genes by repressing the transcriptional activities of other nuclear receptors including NeuroD, which has been shown to regulate secretin gene expression. Here, we evaluated the regulation on duodenal secretin gene expression by SHP and selected bile acids, cholic acid (CA) and chenodeoxycholic acid (CDCA). In vitro treatment of CDCA or fexaramine elevated the SHP transcript level and occupancy on secretin promoter. The increase in the SHP level, induced by bile acid treatment or overexpression, reduced secretin gene expression, whereas this gene inhibitory effect was reversed by silencing of endogenous SHP. In in vivo studies, double-immunofluorescence staining demonstrated the coexpression of secretin and SHP in mouse duodenum. Feeding mice with 1% CA-enriched rodent chow resulted in upregulation of SHP and a concomitant decrease in secretin transcript and protein levels in duodenum compared with the control group fed with normal chow. A diet enriched with 5% cholestyramine led to a decrease in SHP level and a corresponding increase in secretin expression. Overall, this study showed that bile acids via SHP inhibit duodenal secretin gene expression. Because secretin is a key hormone that stimulates bile flow in cholangiocytes, this pathway thus provides a novel means to modulate secretin-stimulated choleresis in response to intraduodenal bile acids.


Subject(s)
Chenodeoxycholic Acid/metabolism , Cholic Acid/metabolism , Duodenum/metabolism , Enteroendocrine Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Secretin/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzene Derivatives/pharmacology , Binding Sites , Cell Line, Tumor , Chenodeoxycholic Acid/administration & dosage , Cholestyramine Resin/administration & dosage , Diet , Down-Regulation , Duodenum/drug effects , Enteroendocrine Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Secretin/genetics , Transcription, Genetic , Transfection
15.
J Mol Neurosci ; 36(1-3): 125-31, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18566919

ABSTRACT

Secretin is a peptide hormone playing multiple functions in the brain-gut axis. In this report, we investigated, by promoter analysis, the potential function of the variable of tandem repeats (VNTR), located at the 5' upstream region of the human secretin gene, and we demonstrated for the first time that this VNTR could downregulate transcription of the human secretin gene in a promoter-specific manner. The efficiency of VNTR in silencing the promoter was found to be directly related the number of repetitive units residing within. We also showed the deoxyribonucleic acid sequence as well as the length polymorphism of the VNTR of 76 Chinese individuals. These results collectively suggest that VNTR could potentially be a functional regulator to control the expression of the human secretin gene in different individuals.


Subject(s)
5' Flanking Region/genetics , Gene Expression Regulation , Minisatellite Repeats , Secretin/genetics , Animals , Base Sequence , Down-Regulation , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Secretin/metabolism , Sequence Alignment
16.
Int Rev Cytol ; 265: 159-90, 2008.
Article in English | MEDLINE | ID: mdl-18275888

ABSTRACT

The discovery of secretin initiated the field of endocrinology. Over the past century, multiple gastrointestinal functions of secretin have been extensively studied, and it was discovered that the principal function of this peptide in the gastrointestinal system is to facilitate digestion and to provide protection. In view of the late identification of secretin and the secretin receptor in various tissues, including the central nervous system, the pleiotropic functions of secretin have more recently been an area of intense focus. Secretin is a classical hormone, and recent studies clearly showed secretin's involvement in neural and neuroendocrine pathways, although the neuroactivity and neural regulation of its release are yet to be elucidated. This chapter reviews our current understanding of the pleiotropic actions of secretin with a special focus on the hormonal and neural interdependent pathways that mediate these actions.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Secretin/physiology , Humans , Signal Transduction
17.
BMC Physiol ; 7: 6, 2007 Jul 19.
Article in English | MEDLINE | ID: mdl-17640386

ABSTRACT

BACKGROUND: Prolactin promotes proliferation of several cells. Prolactin receptor exists as two isoforms: long and short, which activate different transduction pathways including the Ca2+-dependent PKC-signaling. No information exists on the role of prolactin in the regulation of the growth of female cholangiocytes. The rationale for using cholangiocytes from female rats is based on the fact that women are preferentially affected by specific cholangiopathies including primary biliary cirrhosis. We propose to evaluate the role and mechanisms of action by which prolactin regulates the growth of female cholangiocytes. RESULTS: Normal cholangiocytes express both isoforms (long and short) of prolactin receptors, whose expression increased following BDL. The administration of prolactin to normal female rats increased cholangiocyte proliferation. In purified normal female cholangiocytes, prolactin stimulated cholangiocyte proliferation, which was associated with increased [Ca2+]i levels and PKCbeta-I phosphorylation but decreased PKCalpha phosphorylation. Administration of an anti-prolactin antibody to BDL female rats decreased cholangiocyte proliferation. Normal female cholangiocytes express and secrete prolactin, which was increased in BDL rats. The data show that prolactin stimulates normal cholangiocyte growth by an autocrine mechanism involving phosphorylation of PKCbeta-I and dephosphorylation of PKCalpha. CONCLUSION: We suggest that in female rats: (i) prolactin has a trophic effect on the growth of normal cholangiocytes by phosphorylation of PKCbeta-I and dephosphorylation of PKCalpha; and (iii) cholangiocytes express and secrete prolactin, which by an autocrine mechanism participate in regulation of cholangiocyte proliferation. Prolactin may be an important therapeutic approach for the management of cholangiopathies affecting female patients.


Subject(s)
Bile Ducts/cytology , Bile Ducts/enzymology , Calcium Signaling/physiology , Cell Proliferation , Prolactin/metabolism , Protein Kinase C/metabolism , Animals , Bile Ducts/drug effects , Calcium Signaling/drug effects , Cell Proliferation/drug effects , Female , Isoenzymes/metabolism , Male , Phosphorylation , Prolactin/pharmacology , Protein Kinase C beta , Protein Kinase C-alpha/metabolism , Rats , Rats, Inbred F344 , Receptors, Prolactin/biosynthesis
18.
Ann N Y Acad Sci ; 1070: 371-5, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16888194

ABSTRACT

Previous studies have demonstrated the transcriptional repressive property of the atypical nuclear receptor, small heterodimer partner (SHP), on NeuroD. NeuroD is a basic helix-loop-helix transcription factor that has also been shown to be important in modulating secretin gene expression. The present study revealed the activation of the human secretin core promotor by overexpressing NeuroD, and the localization of SHP and secretin-producing cells in mouse duodenal epithelium by immunohistochemical stainings. These results indicated that SHP and secretin are potentially co-expressed and lead us to propose a novel regulatory pathway, in which SHP represses NeuroD's positive regulatory activity on secretin gene.


Subject(s)
Duodenum/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Secretin/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Humans , Immunohistochemistry , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/genetics , Secretin/genetics
19.
Regul Pept ; 137(1-2): 95-104, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16930743

ABSTRACT

Secretin, a 27-amino acid gastrointestinal peptide, was initially discovered based on its activities in stimulating pancreatic juice. In the past 20 years, secretin was demonstrated to exhibit pleiotropic functions in many different tissues and more importantly, its role as a neuropeptide was substantiated. To carry out its activities in the central nervous system and in peripheral organs, secretin interacts specifically with one known receptor. Secretin receptor, a member of guanine nucleotide-binding protein (G protein)-coupled receptor (GPCR) in the secretin/VIP/glucagon subfamily, possesses the characteristics of GPCR with seven conserved transmembrane domains, a relatively large amino-terminal extracellular domain and an intracellular carboxyl terminus. The structural features and signal transduction pathways of the secretin receptor in various tissues are reviewed in this article.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Signal Transduction , Humans , Ligands
SELECTION OF CITATIONS
SEARCH DETAIL