Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Methods Enzymol ; 645: 231-242, 2020.
Article in English | MEDLINE | ID: mdl-33565974

ABSTRACT

Extracellular vesicles (EVs) are mediators of intercellular communication in diverse cellular functions. Visualizing EVs in vivo is important to elucidate the biogenesis of EVs, and various approaches have been developed for in vivo EV tracking. The ubiquitously expressed tetraspanin CD63 is liberally incorporated into EVs. Thus, fluorescently tagged CD63 has been used in many studies to label EVs. In the present study, we presented isolation and transfer assays for EVs from two transgenic rats expressing CD63-GFP in their body fluids or brains.


Subject(s)
Body Fluids , Extracellular Vesicles , Animals , Cell Communication , Rats , Rats, Transgenic
2.
Dis Model Mech ; 11(1)2018 01 30.
Article in English | MEDLINE | ID: mdl-29208635

ABSTRACT

Extracellular vesicles (EVs) can modulate microenvironments by transferring biomolecules, including RNAs and proteins derived from releasing cells, to target cells. To understand the molecular mechanisms maintaining the neural stem cell (NSC) niche through EVs, a new transgenic (Tg) rat strain that can release human CD63-GFP-expressing EVs from the NSCs was established. Human CD63-GFP expression was controlled under the rat Sox2 promoter (Sox2/human CD63-GFP), and it was expressed in undifferentiated fetal brains. GFP signals were specifically observed in in vitro cultured NSCs obtained from embryonic brains of the Tg rats. We also demonstrated that embryonic NSC (eNSC)-derived EVs were labelled by human CD63-GFP. Furthermore, when we examined the transfer of EVs, eNSC-derived EVs were found to be incorporated into astrocytes and eNSCs, thus implying an EV-mediated communication between different cell types around NSCs. This new Sox2/human CD63-GFP Tg rat strain should provide resources to analyse the cell-to-cell communication via EVs in NSC microenvironments.


Subject(s)
Extracellular Vesicles/metabolism , Green Fluorescent Proteins/metabolism , Neural Stem Cells/metabolism , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics , Tetraspanin 30/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Brain/growth & development , Brain/metabolism , Cell Differentiation , Coculture Techniques , Embryo, Mammalian/metabolism , Humans , Models, Animal , Rats, Transgenic , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/metabolism
3.
Sci Rep ; 6: 31172, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27539050

ABSTRACT

Extracellular vesicles (EVs) play an important role in the transfer of biomolecules between cells. To elucidate the intercellular transfer fate of EVs in vivo, we generated a new transgenic (Tg) rat model using green fluorescent protein (GFP)-tagged human CD63. CD63 protein is highly enriched on EV membranes via trafficking into late endosomes and is often used as an EV marker. The new Tg rat line in which human CD63-GFP is under control of the CAG promoter exhibited high expression of GFP in various body tissues. Exogenous human CD63-GFP was detected on EVs isolated from three body fluids of the Tg rats: blood serum, breast milk and amniotic fluid. In vitro culture allowed transfer of serum-derived CD63-GFP EVs into recipient rat embryonic fibroblasts, where the EVs localized in endocytic organelles. These results suggested that this Tg rat model should provide significant information for understanding the intercellular transfer and/or mother-child transfer of EVs in vivo.


Subject(s)
Body Fluids/metabolism , Extracellular Vesicles/metabolism , Amniotic Fluid/metabolism , Animals , Biological Transport, Active , Cells, Cultured , Endosomes/metabolism , Female , Fibroblasts/metabolism , Humans , Maternal-Fetal Exchange , Milk/metabolism , Models, Animal , Pregnancy , Rats , Rats, Transgenic , Rats, Wistar , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetraspanin 30/blood , Tetraspanin 30/genetics , Tetraspanin 30/metabolism , Tissue Distribution
4.
Neurochem Int ; 97: 26-33, 2016 07.
Article in English | MEDLINE | ID: mdl-27131735

ABSTRACT

MicroRNAs (miRs) play important roles in neuronal differentiation, maturation, and synaptic function in the central nervous system. They have also been suggested to be implicated in the pathogenesis of neurodegenerative and psychiatric diseases. Although miR-132 is one of the well-studied brain-specific miRs, which regulates synaptic structure and function in the postnatal brain, its function in the prenatal brain is still unclear. Here, we investigated miR-132 function during differentiation of rat embryonic neural stem cells (eNSCs). We found that miR-132 expression significantly increased during the fetal rat brain development and neural differentiation of eNSCs in vitro. Furthermore, miR-132 expression was increased during differentiation through MAPK/ERK1/2 pathway. Inhibition of ERK1/2 activation resulted in increased levels of synaptic proteins including PSD-95, GluR1 and synapsin I. Silencing of miR-132 also increased PSD-95 and GluR1. Considering that miR-132 increases synaptic proteins in differentiated cortical neurons, our result shows a novel function of miR-132 as a negative regulator for synaptic maturation in the neuronal differentiation of eNSCs.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , MicroRNAs/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neural Stem Cells/metabolism , Neurogenesis/physiology , Animals , Cells, Cultured , Gene Expression , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Rats , Rats, Wistar , Synapses/genetics , Synapses/metabolism
5.
Comput Math Methods Med ; 2016: 9713280, 2016.
Article in English | MEDLINE | ID: mdl-28096896

ABSTRACT

Lung motion due to respiration causes image degradation in medical imaging, especially in nuclear medicine which requires long acquisition times. We have developed a method for image correction between the respiratory-gated (RG) PET images in different respiration phases or breath-hold (BH) PET images in an inconsistent respiration phase. In the method, the RG or BH-PET images in different respiration phases are deformed under two criteria: similarity of the image intensity distribution and smoothness of the estimated motion vector field (MVF). However, only these criteria may cause unnatural motion estimation of lung. In this paper, assuming the use of a PET-CT scanner, we add another criterion that is the similarity for the motion direction estimated from inhalation and exhalation CT images. The proposed method was first applied to a numerical phantom XCAT with tumors and then applied to BH-PET image data for seven patients. The resultant tumor contrasts and the estimated motion vector fields were compared with those obtained by our previous method. Through those experiments we confirmed that the proposed method can provide an improved and more stable image quality for both RG and BH-PET images.


Subject(s)
Lung Neoplasms/diagnostic imaging , Lung/diagnostic imaging , Positron-Emission Tomography/methods , Tomography, X-Ray Computed/methods , Artifacts , Breath Holding , Computer Simulation , Contrast Media/chemistry , Exhalation , Fluorodeoxyglucose F18/chemistry , Humans , Image Processing, Computer-Assisted/methods , Lung/physiopathology , Lung Neoplasms/physiopathology , Motion , Phantoms, Imaging , Respiration
6.
Autophagy ; 9(3): 403-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23291500

ABSTRACT

Regulated degradation of cellular components by lysosomes is essential to maintain biological homeostasis. In mammals, three forms of autophagy, macroautophagy, microautophagy and chaperone-mediated autophagy (CMA), have been identified. Here, we showed a novel type of autophagy, in which RNA is taken up directly into lysosomes for degradation. This pathway, which we term "RNautophagy," is ATP-dependent, and unlike CMA, is independent of HSPA8/Hsc70. LAMP2C, a lysosomal membrane protein, serves as a receptor for this pathway. The cytosolic tail of LAMP2C specifically binds to almost all total RNA derived from mouse brain. The cytosolic sequence of LAMP2C and its affinity for RNA are evolutionarily conserved from nematodes to humans. Our findings shed light on the mechanisms underlying RNA homeostasis in higher eukaryotes.


Subject(s)
Autophagy , Lysosomes/metabolism , RNA/metabolism , Animals , Brain/embryology , Brain/metabolism , Conserved Sequence , HeLa Cells , Humans , Lysosomal-Associated Membrane Protein 2/metabolism , Mice , Mice, Knockout , Sequence Homology, Amino Acid
7.
Circ Res ; 107(1): 30-4, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20570919

ABSTRACT

RATIONALE: The (pro)renin receptor [(P)RR], encoded in ATP6AP2, plays a key role in the activation of local renin-angiotensin system (RAS). A truncated form of (P)RR, termed M8.9, was also found to be associated with the vacuolar H(+)-ATPase (V-ATPase), implicating a non-RAS-related function of ATP6AP2. OBJECTIVE: We investigated the role of (P)RR/ATP6AP2 in murine cardiomyocytes. METHODS AND RESULTS: Cardiomyocyte-specific ablation of Atp6ap2 resulted in lethal heart failure; the cardiomyocytes contained RAB7- and lysosomal-associated membrane protein 2 (LAMP2)-positive multivesicular vacuoles, especially in the perinuclear regions. The myofibrils and mitochondria remained at the cell periphery. Cardiomyocyte death was accompanied by numerous autophagic vacuoles that contained undigested cellular constituents, as a result of impaired autophagic degradation. Notably, ablation of Atp6ap2 selectively suppressed expression of the V(O) subunits of V-ATPase, resulting in deacidification of the intracellular vesicles. Furthermore, the inhibition of intracellular acidification by treatment with bafilomycin A1 or chloroquine reproduced the phenotype observed for the (P)RR/ATP6AP2-deficient cardiomyocytes. CONCLUSIONS: Genetic ablation of Atp6ap2 created a loss-of-function model for V-ATPase. The gene product of ATP6AP2 is considered to act as in 2 ways: (1) as (P)RR, exerting a RAS-related function; and (2) as the V-ATPase-associated protein, exerting a non-RAS-related function that is essential for cell survival.


Subject(s)
Myocytes, Cardiac/enzymology , Protein Precursors/physiology , Receptors, Cell Surface/physiology , Renin/physiology , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cell Survival/genetics , Heart Failure/enzymology , Heart Failure/mortality , Heart Failure/pathology , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Protein Precursors/genetics , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Renin/genetics , Vacuolar Proton-Translocating ATPases/deficiency , Vacuolar Proton-Translocating ATPases/physiology , Prorenin Receptor
8.
Biochem Biophys Res Commun ; 390(3): 377-81, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19732742

ABSTRACT

Triglyceride ingestion releases gut peptides from enteroendocrine cells located in the intestinal epithelia and provides feedback regulations of gastrointestinal function. The precise mechanisms sensing lipids in the intestinal wall, however, are not well characterized. In the current study, we investigated the release of gut peptides following oral triglyceride loading in mice deficient for monoacylglycerol acyltransferase 2 (MGAT2KO) and diacylglycerol acyltransferase 1 (DGAT1KO), enzymes that sequentially re-synthesize triglyceride to secrete as chylomicron at the small intestine. In wild-type (Wt) mice, oral triglyceride loading resulted in hypertriglycemia. In addition, plasma glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) were significantly increased 30 min after triglyceride loading, before decaying in 2h. In MGAT2KO and DGAT1KO mice, oral triglyceride loading did not result in hypertriglycemia and the increase in GIP was significantly suppressed in both KO mouse strains. In contrast, the increases in plasma GLP-1 and PYY in both KO mouse strains were comparable to Wt mice 30 min after triglyceride loading, however, they remained elevated in DGAT1KO mice even 2h after triglyceride loading. In parallel to the changes in GLP-1 and PYY, gastric emptying was delayed after oral triglyceride loading in MGAT2KO mice comparably to Wt type mice and was further delayed in DGAT1KO mice. STC-1 and GLUTag, GLP-1-producing intestinal endocrine L-cell lines, displayed a significant level of DGAT1 activity but not MGAT activity. These findings suggest that synthesis and/or secretion of triglyceride-rich lipoproteins play an important role in the release of GIP. Moreover, DGAT1 may directly regulate the release of GLP-1 and PYY in L-cells.


Subject(s)
Acyltransferases/physiology , Diacylglycerol O-Acyltransferase/physiology , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Intestinal Mucosa/metabolism , Triglycerides/metabolism , Acyltransferases/genetics , Animals , Diacylglycerol O-Acyltransferase/genetics , Eating , Lipoproteins/biosynthesis , Mice , Mice, Knockout , Triglycerides/administration & dosage
9.
Cancer Sci ; 100(8): 1544-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19459854

ABSTRACT

The advances in preclinical cancer models, including orthotopic implantation models or genetically engineered mouse models of cancer, enable pursuing the molecular mechanism of cancer disease that might mimic genetic and biological processes in humans. Lung cancer is the major cause of cancer deaths; therefore, the treatment and prevention of lung cancer are expected to be improved by a better understanding of the complex mechanism of disease. In this study, we have examined the quantification of two distinct mouse lung cancer models by utilizing imaging modalities for monitoring tumor progression and drug efficacy evaluation. The utility of microcomputed tomography (micro-CT) for real-time/non-invasive monitoring of lung cancer progression has been confirmed by combining bioluminescent imaging and histopathological analyses. Further, we have developed a more clinically relevant lung cancer model by utilizing K-ras(LSL-G12D)/p53(LSL-R270H) mutant mice. Using micro-CT imaging, we monitored the development and progression of solitary lung tumor in K-ras(LSL-G12D)/p53(LSL-R270H) mutant mouse, and further demonstrated tumor growth inhibition by anticancer drug treatment. These results clearly indicate that imaging-guided evaluation of more clinically relevant tumor models would improve the process of new drug discovery and increase the probability of success in subsequent clinical studies.


Subject(s)
Carcinoma, Lewis Lung/diagnostic imaging , Carcinoma, Lewis Lung/pathology , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , X-Ray Microtomography/methods , Adenoviridae/genetics , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Lewis Lung/drug therapy , Cell Line, Tumor , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Models, Animal , Disease Progression , Drug Therapy, Combination , Genes, ras/genetics , Lentivirus/genetics , Luciferases/metabolism , Luminescent Measurements , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Random Allocation , Time Factors , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays , Gemcitabine
10.
Genesis ; 47(6): 414-22, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19391115

ABSTRACT

C57BL/6 (B6)-derived embryonic stem (ES) cells are not widely used to generate knockout mice despite the advantage of a well-defined genetic background because of poor developmental potential. We newly established serum- and feeder-free B6 ES cells with full developmental potential by using leukemia inhibitory factor (LIF) and 6-bromoindirubin-3'-oxime (BIO), a glycogen synthase kinase-3 (GSK3) inhibitor. BIO treatment significantly increased the expression levels of 364 genes including pluripotency markers such as Nanog and Klf family. Unexpectedly, by aggregating or microinjecting those ES cells to each eight-cell-stage diploid embryo, we stably generated germline-competent ES-derived mice. Furthermore, founder mice completely derived from female XO, heterozygous, or homozygous mutant B6 ES cells were directly available for intercross breeding and phenotypic analysis. We hereby propose that serum- and feeder-free B6 ES cells stimulated with LIF plus GSK3 inhibitor are valuable for generating mouse models on B6 background.


Subject(s)
Embryonic Stem Cells/drug effects , Indoles/pharmacology , Oximes/pharmacology , Totipotent Stem Cells/drug effects , Animals , Cell Culture Techniques , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Leukemia Inhibitory Factor/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Inbred Strains , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Totipotent Stem Cells/cytology , Totipotent Stem Cells/metabolism
11.
PLoS One ; 3(7): e2800, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18665239

ABSTRACT

The rat is a reference animal model for physiological studies and for the analysis of multigenic human diseases such as hypertension, diabetes, neurological disorders, and cancer. The rats have long been used in extensive chemical carcinogenesis studies. Thus, the rat embryonic stem (rES) cell is an important resource for the study of disease models. Attempts to derive ES cells from various mammals, including the rat, have not succeeded. Here we have established two independent rES cells from Wister rat blastocysts that have undifferentiated characters such as Nanog and Oct3/4 genes expression and they have stage-specific embryonic antigen (SSEA) -1, -3, -4, and TRA-1-81 expression. The cells were successfully cultured in an undifferentiated state and can be possible over 18 passages with maintaining more than 40% of normal karyotype. Their pluripotent potential was confirmed by the differentiation into derivatives of the endoderm, mesoderm, and ectoderm. Most importantly, the rES cells are capable of producing chimera rats. Therefore, we established pluripotent rES cell lines that are widely used to produce genetically modified experimental rats for study of human diseases.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , Cell Line , Chimera , Disease Models, Animal , Female , Green Fluorescent Proteins/metabolism , Karyotyping , Male , Models, Biological , Rats , Rats, Wistar
12.
Arch Biochem Biophys ; 466(2): 283-9, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17880910

ABSTRACT

To investigate the effect of elevation of liver glycogen synthase (GYS2) activity on glucose and glycogen metabolism, we performed adenoviral overexpression of the mutant GYS2 with six serine-to-alanine substitutions in rat primary hepatocytes. Cell-free assays demonstrated that the serine-to-alanine substitutions caused constitutive activity and electrophoretic mobility shift. In rat primary hepatocytes, overexpression of the mutant GYS2 significantly reduced glucose production by 40% and dramatically induced glycogen synthesis via the indirect pathway rather than the direct pathway. Thus, we conclude that elevation of glycogen synthase activity has an inhibitory effect on glucose production in hepatocytes by shunting gluconeogenic precursors into glycogen. In addition, although intracellular compartmentation of glucose-6-phosphate (G6P) remains unclear in hepatocytes, our results imply that there are at least two G6P pools via gluconeogenesis and due to glucose phosphorylation, and that G6P via gluconeogenesis is preferentially used for glycogen synthesis in hepatocytes.


Subject(s)
Alanine/genetics , Glycogen Synthase/biosynthesis , Hepatocytes/metabolism , Serine/genetics , Amino Acid Substitution , Animals , Cells, Cultured , Glucose/biosynthesis , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Glycogen Synthase/genetics , Male , Rats , Rats, Wistar
13.
FEBS J ; 274(18): 4766-77, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17697114

ABSTRACT

Hepatocytes of the periportal and perivenous zones of the liver lobule show marked differences in the contents and activities of many enzymes and other proteins. Previous studies from our and other groups have pointed towards an important role of beta-catenin-dependent signaling in the regulation of expression of genes encoding proteins with preferential perivenous localization, whereas, in contrast, signaling through Ras-dependent pathway(s) may induce a 'periportal' phenotype. We have now conducted a series of experiments to further investigate this hypothesis. In transgenic mice with scattered expression of an activated Ha-ras (Ha-ras(G12V)) mutant in liver, expression of the perivenous markers glutamine synthetase and two cytochrome P450 isoforms was completely abolished in those hepatocytes demonstrating constitutively activated extracellular signal-regulated kinase activity, even though they were located directly adjacent to central veins. Similarly, incubation of primary hepatocytes or hepatoma cells with increasing amounts of serum caused a concentration-dependent attenuation of expression of perivenous marker mRNAs, whereas the expression of periportal markers was increased. The inhibitory effect of high amounts of serum on the expression of perivenous markers was also observed if their expression was stimulated by activation of beta-catenin signaling, and comparable inhibitory effects were seen in cells stably transfected with a T-cell factor/lymphoid-enhancing factor-driven luciferase reporter. Epidermal growth factor could partly mimic serum effects in hepatoma cells, and its effect could be blocked by an inhibitor of extracellular signal-regulated kinase activity. These data suggest that activation of the Ras/mitogen-activated protein kinase (extracellular signal-regulated kinase) pathway favors periportal gene expression while simultaneously antagonizing a perivenous phenotype of hepatocytes.


Subject(s)
Biomarkers , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Oncogene Protein p21(ras)/metabolism , Serum/chemistry , Signal Transduction , beta Catenin/metabolism , Animals , Axin Protein , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytochrome P-450 Enzyme System/genetics , Cytoskeletal Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutamate-Ammonia Ligase/genetics , Hepatocytes/drug effects , Mice , Mice, Transgenic , Oncogene Protein p21(ras)/genetics , TCF Transcription Factors/metabolism , Transcription, Genetic/drug effects
14.
Mol Cell Biol ; 27(5): 1881-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17210641

ABSTRACT

Acetyl coenzyme A (acetyl-CoA) carboxylase (ACC) catalyzes carboxylation of acetyl-CoA to form malonyl-CoA. In mammals, two isozymes exist with distinct physiological roles: cytosolic ACC1 participates in de novo lipogenesis (DNL), and mitochondrial ACC2 is involved in negative regulation of mitochondrial beta-oxidation. Since systemic ACC1 null mice were embryonic lethal, to clarify the physiological role of ACC1 in hepatic DNL, we generated the liver-specific ACC1 null mouse by crossbreeding of an Acc1(lox(ex46)) mouse, in which exon 46 of Acc1 was flanked by two loxP sequences and the liver-specific Cre transgenic mouse. In liver-specific ACC1 null mice, neither hepatic Acc1 mRNA nor protein was detected. However, to compensate for ACC1 function, hepatic ACC2 protein and activity were induced 1.4 and 2.2 times, respectively. Surprisingly, hepatic DNL and malonyl-CoA were maintained at the same physiological levels as in wild-type mice. Furthermore, hepatic DNL was completely inhibited by an ACC1/2 dual inhibitor, 5-tetradecyloxyl-2-furancarboxylic acid. These results strongly demonstrate that malonyl-CoA from ACC2 can access fatty acid synthase and become the substrate for the DNL pathway under the unphysiological circumstances that result with ACC1 disruption. Therefore, there does not appear to be strict compartmentalization of malonyl-CoA from either of the ACC isozymes in the liver.


Subject(s)
Acetyl-CoA Carboxylase/deficiency , Acetyl-CoA Carboxylase/genetics , Lipogenesis , Liver/metabolism , Animals , Enzyme Inhibitors/pharmacology , Liver/enzymology , Malonyl Coenzyme A/analysis , Malonyl Coenzyme A/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Polymerase Chain Reaction
15.
J Endocrinol ; 191(1): 197-205, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17065403

ABSTRACT

G protein-coupled bile acid receptor 1 (Gpbar1/M-Bar) is a novel G protein-coupled receptor for bile acid. Tissue distribution and cell-type specificity of Gpbar1 mRNA suggest a potential role for the receptor in the endocrine system; however, the precise physiological role of Gpbar1 still remains to be elucidated. To investigate the role of Gpbar1 in vivo, the Gpbar1 gene was disrupted in mice. In homozygous mice, total bile acid pool size was significantly decreased by 21-25% compared with that of the wild-type mice, suggesting that Gpbar1 contributes to bile acid homeostasis. In order to assess the impact of Gpbar1 deficiency in bile acid homeostasis more precisely, Gpbar1 homozygous mice were fed a high-fat diet for 2 months. As a result, female Gpbar1 homozygous mice showed significant fat accumulation with body weight gain compared with that of the wild-type mice. These findings were also observed in heterozygous mice to the same extent. Although the precise mechanism for fat accumulation in female Gpbar1 homozygous mice remains to be addressed, these data indicate that Gpbar1 is a potential new player in energy homeostasis. Thus, Gpbar1-deficient mice are useful in elucidating new physiological roles for Gpbar1.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Animals , Base Sequence , Bile/metabolism , Bile Acids and Salts/analysis , Bile Acids and Salts/metabolism , Blotting, Northern/methods , Body Composition , Cholesterol/blood , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Feces/chemistry , Female , Gallbladder/metabolism , Homeostasis , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Molecular Sequence Data , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Triglycerides/blood
16.
Mol Cell Biol ; 25(9): 3658-69, 2005 May.
Article in English | MEDLINE | ID: mdl-15831471

ABSTRACT

Appropriate expression of the selenocysteine tRNA (tRNA(Sec)) gene is necessary for the production of an entire family of selenoprotein enzymes. This study investigates the consequence of disrupting an upstream enhancer region of the mouse tRNA(Sec) gene (Trsp) known as the distal sequence element (DSE) by use of a conditional repair gene targeting strategy, in which a 3.2-kb insertion was introduced into the promoter of the gene. In the absence of DSE activity, homozygous mice failed to develop in utero beyond embryonic day 7.5 and had severely decreased levels of selenoprotein transcript. Cre-mediated removal of the selection cassette recovered DSE regulation of Trsp, restoring wild-type levels of tRNA(Sec) expression and allowing the generation of viable rescued mice. Further analysis of targeted heterozygous adult mice revealed that the enhancer activity of the DSE is tissue dependent since, in contrast to liver, heart does not require the DSE for normal expression of Trsp. Similarly, in mouse cell lines we showed that the DSE functions as a cell-line-specific inducible element of tRNA(Sec). Together, our data demonstrate that the DSE is a tissue-dependent regulatory element of tRNA(Sec) expression and that its activity is vital for sufficient tRNA(Sec) production during mouse embryogenesis.


Subject(s)
Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental , Mice/embryology , RNA, Transfer, Amino Acid-Specific/genetics , Animals , Antioxidants/metabolism , Base Sequence , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Embryonic Development/genetics , Embryonic Development/physiology , Enhancer Elements, Genetic/genetics , Gene Targeting , Genes, Lethal/genetics , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Liver/metabolism , Membrane Proteins , Mice/genetics , Molecular Sequence Data , Myocardium/metabolism , Proteins/genetics , Proteins/metabolism , RNA, Transfer, Amino Acid-Specific/analysis , Selenoproteins , Tissue Distribution , Up-Regulation , Glutathione Peroxidase GPX1
17.
Nat Genet ; 35(4): 323-30, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625550

ABSTRACT

The mammalian homeobox transcription factor CDX2 has key roles in intestinal development and differentiation. Heterozygous Cdx2 mice develop one or two benign hamartomas in the proximal colon, whereas heterozygous Apc(Delta716) mice develop numerous adenomatous polyps, mostly in the small intestine. Here we show that the colonic polyp number is about six times higher in Apc+/Delta716 Cdx2+/- compound mutant mice. Levels of both APC and CDX2 were significantly lower in the distal colon, which caused high anaphase bridge index (ABI) associated with a higher frequency of loss of heterozygosity (LOH) at Apc. In cultured rat intestinal epithelial and human colon cancer cell lines, suppression of CDX2 by antisense RNA caused marked increases in ABI and chromosomal aberrations. This was mediated by stimulation of the mTOR pathway, causing translational deregulation and G1-S acceleration, associated with low levels of p27 and activation of cyclin E-Cdk2. We obtained similar results in the colonic mucosa of Apc+/Delta716) Cdx2+/- compound mutant mice. Forced activation of mTOR through upstream regulator Akt also increased ABI in colon cancer cells. High ABI in all cell lines was suppressed by mTOR inhibitors LY294002 and rapamycin. These results suggest that reduced expression of CDX2 is important in colon tumorigenesis through mTOR-mediated chromosomal instability.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Chromosomal Instability/genetics , Homeodomain Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Anaphase , Animals , Bromodeoxyuridine , CDX2 Transcription Factor , Cell Survival , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Polyps/genetics , Colonic Polyps/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , G1 Phase , Heterozygote , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , In Situ Nick-End Labeling , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Antisense/pharmacology , Rats , S Phase , TOR Serine-Threonine Kinases , Trans-Activators
18.
J Cell Sci ; 116(Pt 22): 4629-38, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14576356

ABSTRACT

Keratin transgenic mouse models and the association of human keratin mutations with liver disease highlight the importance of keratins in protecting the liver from environmental insults, but little is known regarding keratins and their function in the gallbladder. We characterized keratin expression pattern and filament organization in normal and keratin polypeptide-8 (K8)-null, K18-null and K19-null gallbladders, and examined susceptibility to liver and gallbladder injury induced by a high-fat lithogenic diet (LD) in K8-null mice. The major keratins of normal mouse gallbladder are K8>K19>K18 which become markedly depleted in K8-null mice with minor K18/K19 remnants and limited K7 over-expression. Compensatory K18/K20 protein and RNA overexpression occur in K19-null but not in K18-null gallbladders, probably because of the higher levels of K19 than K18 in normal gallbladder. LD challenge causes more severe liver injury in K8-null than wild-type mice without altering keratin protein levels. In contrast, wild-type and K8-null gallbladders are equally susceptible to LD-induced injury and stone formation, but wild-type gallbladders do overexpress keratins upon LD challenge. LD-induced injury triggers keratin hyperphosphorylation in wild-type livers and gallbladders. Hence, mouse gallbladder K8/K18/K19 expression is induced in response to cholelithiasis injury. A high-fat LD increases the susceptibility of K8-null mice to liver but not gallbladder injury, which suggests that keratin mutations may increase the risk of liver damage in patients with steatohepatitis. Differences between K8-null mouse gallbladder and hepatocyte susceptibility to injury may be related to their minimal versus absent keratin expression, respectively.


Subject(s)
Diet , Gallbladder/metabolism , Keratins/metabolism , Liver/metabolism , Animals , Disease Susceptibility/pathology , Fluorescent Antibody Technique, Indirect , Gallbladder/pathology , Keratin-8 , Keratins/genetics , Liver/pathology , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
19.
Dev Biol ; 253(2): 230-46, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12645927

ABSTRACT

Tumor suppressor Apc (adenomatous polyposis coli) is implicated in the Wnt signaling pathway that is involved in the early embryonic development and tumorigenesis in vertebrates. While the heterozygous null mutant mice develop intestinal polyps, the homozygous embryos die before gastrulation. To investigate the role of Apc in later embryonic development, we constructed a novel hypomorphic Apc allele whose expression was attenuated by approximately 80%. In the hypomorphic Apc homozygous ES cells, reduction in Apc expression caused beta-catenin accumulation and Wnt signaling activation. The homozygous mutant mouse embryos survived 3 days longer than the null mutant embryos. Interestingly, they showed anterior truncation, partial axis duplication, and defective ventral morphogenesis. To determine the tissues where Apc functions for anterior and ventral morphogenesis, we constructed chimeric embryos whose epiblast was derived predominantly from the Apc hypomorphic homozygous cells but the visceral endoderm was from the wild type. Although these chimeric embryos still showed some anterior defects, their ventral morphogenesis was rescued. In addition, marker studies indicated that the axial mesendoderm was also defective in the homozygous embryos. Our results provide genetic evidence that expression of Apc at the normal level is essential for both anterior and ventral development, in the epiblast derivatives and visceral endoderm.


Subject(s)
Embryonic and Fetal Development/genetics , Genes, APC , Alleles , Animals , Base Sequence , Body Patterning/genetics , Chimera , DNA/genetics , Gastrula/metabolism , Heterozygote , Homozygote , In Situ Hybridization , Lac Operon , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
20.
Biochem Biophys Res Commun ; 298(5): 714-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12419312

ABSTRACT

Bile acids play an essential role in the solubilization and absorption of dietary fat and lipid-soluble vitamins. Bile acids also modulate the transcription of various genes for enzymes and transport proteins for their own and cholesterol homeostasis through binding to nuclear receptors. Here we report a novel category of bile acid receptor, a membrane-type G protein-coupled receptor (GPCR), BG37. Bile acids induced rapid and dose-dependent elevation of intracellular cAMP levels in BG37-expressing cells, but not in mock-transfected cells, independently of nuclear receptor expression. The rank order of potency of various bile acids for BG37-expressing cells was different from that for the nuclear receptor-mediated response. These observations demonstrate the presence of two independent signaling pathways for bile acids; membrane-type GPCR for rapid signaling and nuclear receptors for delayed signaling. Expression of BG37 was detected in various specific tissues, suggesting its physiological role, although it remains to be further characterized.


Subject(s)
Bile Acids and Salts/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Base Sequence , Bile Acids and Salts/pharmacology , Caco-2 Cells , Cell Line , Cyclic AMP/biosynthesis , DNA/genetics , GTP-Binding Proteins/metabolism , Gene Expression , Humans , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Homology, Amino Acid , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...