Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Circ Res ; 134(3): 252-265, 2024 02 02.
Article in English | MEDLINE | ID: mdl-38166470

ABSTRACT

BACKGROUND: Intracellular Ca2+ cycling determines myocardial contraction and relaxation in response to physiological demands. SERCA2a (sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2a) is responsible for the sequestration of cytosolic Ca2+ into intracellular stores during cardiac relaxation, and its activity is reversibly inhibited by PLN (phospholamban). However, the regulatory hierarchy of SERCA2a activity remains unclear. METHODS: Cardiomyocyte-specific ZBTB20 knockout mice were generated by crossing ZBTB20flox mice with Myh6-Cre mice. Echocardiography, blood pressure measurements, Langendorff perfusion, histological analysis and immunohistochemistry, quantitative reverse transcription-PCR, Western blot analysis, electrophysiological measurements, and chromatin immunoprecipitation assay were performed to clarify the phenotype and elucidate the molecular mechanisms. RESULTS: Specific ablation of ZBTB20 in cardiomyocyte led to a significant increase in basal myocardial contractile parameters both in vivo and in vitro, accompanied by an impairment in cardiac reserve and exercise capacity. Moreover, the cardiomyocytes lacking ZBTB20 showed an increase in sarcoplasmic reticular Ca2+ content and exhibited a remarkable enhancement in both SERCA2a activity and electrically stimulated contraction. Mechanistically, PLN expression was dramatically reduced in cardiomyocytes at the mRNA and protein levels by ZBTB20 deletion or silencing, and PLN overexpression could largely restore the basal contractility in ZBTB20-deficient cardiomyocytes. CONCLUSIONS: These data point to ZBTB20 as a fine-tuning modulator of PLN expression and SERCA2a activity, thereby offering new perspective on the regulation of basal contractility in the mammalian heart.


Subject(s)
Myocardium , Sarcoplasmic Reticulum , Animals , Mice , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Mammals , Mice, Knockout , Myocardial Contraction/physiology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
2.
Nat Commun ; 14(1): 7934, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38040719

ABSTRACT

Arginase, a manganese (Mn)-dependent enzyme, is indispensable for urea generation and ammonia disposal in the liver. The potential role of fructose in Mn and ammonia metabolism is undefined. Here we demonstrate that fructose overconsumption impairs hepatic Mn homeostasis and ammonia disposal in male mice. Fructose overexposure reduces liver Mn content as well as its activity of arginase and Mn-SOD, and impairs the clearance of blood ammonia under liver dysfunction. Mechanistically, fructose activates the Mn exporter Slc30a10 gene transcription in the liver in a ChREBP-dependent manner. Hepatic overexpression of Slc30a10 can mimic the effect of fructose on liver Mn content and ammonia disposal. Hepatocyte-specific deletion of Slc30a10 or ChREBP increases liver Mn contents and arginase activity, and abolishes their responsiveness to fructose. Collectively, our data establish a role of fructose in hepatic Mn and ammonia metabolism through ChREBP/Slc30a10 pathway, and postulate fructose dietary restriction for the prevention and treatment of hyperammonemia.


Subject(s)
Fructose , Manganese , Male , Mice , Animals , Manganese/toxicity , Manganese/metabolism , Fructose/metabolism , Ammonia/metabolism , Arginase/genetics , Arginase/metabolism , Liver/metabolism , Transcription Factors/metabolism , Homeostasis
3.
Proc Natl Acad Sci U S A ; 120(24): e2220867120, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37279265

ABSTRACT

The mammalian cochlear epithelium undergoes substantial remodeling and maturation before the onset of hearing. However, very little is known about the transcriptional network governing cochlear late-stage maturation and particularly the differentiation of its lateral nonsensory region. Here, we establish ZBTB20 as an essential transcription factor required for cochlear terminal differentiation and maturation and hearing. ZBTB20 is abundantly expressed in the developing and mature cochlear nonsensory epithelial cells, with transient expression in immature hair cells and spiral ganglion neurons. Otocyst-specific deletion of Zbtb20 causes profound deafness with reduced endolymph potential in mice. The subtypes of cochlear epithelial cells are normally generated, but their postnatal development is arrested in the absence of ZBTB20, as manifested by an immature appearance of the organ of Corti, malformation of tectorial membrane (TM), a flattened spiral prominence (SP), and a lack of identifiable Boettcher cells. Furthermore, these defects are related with a failure in the terminal differentiation of the nonsensory epithelium covering the outer border Claudius cells, outer sulcus root cells, and SP epithelial cells. Transcriptome analysis shows that ZBTB20 regulates genes encoding for TM proteins in the greater epithelial ridge, and those preferentially expressed in root cells and SP epithelium. Our results point to ZBTB20 as an essential regulator for postnatal cochlear maturation and particularly for the terminal differentiation of cochlear lateral nonsensory domain.


Subject(s)
Cochlea , Hair Cells, Auditory , Animals , Mice , Cochlea/metabolism , Hair Cells, Auditory/physiology , Hearing/physiology , Mammals , Spiral Ganglion , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Int J Obes (Lond) ; 46(5): 1068-1075, 2022 05.
Article in English | MEDLINE | ID: mdl-35152269

ABSTRACT

OBJECTIVES: Brown adipose tissue (BAT) plays a critical role in energy expenditure by uncoupling protein 1 (UCP1)-mediated thermogenesis and represents an important therapeutic target for metabolic diseases. Carbohydrate response element-binding protein (ChREBP) is a key transcription factor regulating de novo lipogenesis, and its activity is associated with UCP1 expression and thermogenesis in BAT. However, the exact physiological role of endogenous ChREBP in BAT thermogenesis remains unclear. METHODS: We used the Cre/LoxP system to generate ChREBP BAT-specific knockout mice, and examined their BAT thermogenesis under acute cold exposure and long-term cold acclimation. Gene expression was analyzed at the mRNA and protein levels, and lipogenesis was examined by 3H-H2O incorporation assay. RESULTS: The mice lacking ChREBP specifically in BAT displayed a significant decrease in the expression levels of lipogenic genes and the activity of de novo lipogenesis in BAT after cold exposure, with UCP1 expression decreased under thermoneutral conditions or after acute cold exposure but not chronic cold acclimation. Unexpectedly, BAT-specific ChREBP deletion did not significantly affect body temperature as well as local temperature or morphology of BAT after acute cold exposure or chronic cold acclimation. Of note, ChREBP deletion mildly aggravated glucose intolerance induced by a high-fat diet. CONCLUSIONS: Our work indicates that ChREBP regulates de novo lipogenesis in BAT and glucose tolerance, but is not required for non-shivering thermogenesis by BAT under acute or long-term cold exposure.


Subject(s)
Adipose Tissue, Brown , Lipogenesis , Adipose Tissue, Brown/metabolism , Animals , Cold Temperature , Energy Metabolism/physiology , Mice , Mice, Knockout , Thermogenesis/physiology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
5.
Hepatology ; 75(5): 1169-1180, 2022 05.
Article in English | MEDLINE | ID: mdl-34580885

ABSTRACT

BACKGROUND AND AIMS: Lipoprotein lipase (LPL) is responsible for the lipolytic processing of triglyceride-rich lipoproteins, the deficiency of which causes severe hypertriglyceridemia. Liver LPL expression is high in suckling rodents but relatively low at adulthood. However, the regulatory mechanism and functional significance of liver LPL expression are incompletely understood. We have established the zinc finger protein ZBTB20 as a critical factor for hepatic lipogenesis. Here, we evaluated the role of ZBTB20 in regulating liver Lpl gene transcription and plasma triglyceride metabolism. APPROACH AND RESULTS: Hepatocyte-specific inactivation of ZBTB20 in mice led to a remarkable increase in LPL expression at the mRNA and protein levels in adult liver, in which LPL protein was mainly localized onto sinusoidal epithelial cells and Kupffer cells. As a result, the LPL activity in postheparin plasma was substantially increased, and postprandial plasma triglyceride clearance was significantly enhanced, whereas plasma triglyceride levels were decreased. The dysregulated liver LPL expression and low plasma triglyceride levels in ZBTB20-deficient mice were normalized by inactivating hepatic LPL expression. ZBTB20 deficiency protected the mice against high-fat diet-induced hyperlipidemia without causing excessive triglyceride accumulation in the liver. Chromatin immunoprecipitation and gel-shift assay studies revealed that ZBTB20 binds to the LPL promoter in the liver. A luciferase reporter assay revealed that ZBTB20 inhibits the transcriptional activity of LPL promoter. The regulation of LPL expression by ZBTB20 is liver-specific under physiological conditions. CONCLUSIONS: Liver ZBTB20 serves as a key regulator of LPL expression and plasma triglyceride metabolism and could be a therapeutic target for hypertriglyceridemia.


Subject(s)
BTB-POZ Domain , Hypertriglyceridemia , Animals , Hepatocytes/metabolism , Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Liver/metabolism , Mice , Transcription Factors/metabolism , Transcription, Genetic , Triglycerides/metabolism , Zinc Fingers
6.
Chin Med J (Engl) ; 134(11): 1276-1285, 2021 May 18.
Article in English | MEDLINE | ID: mdl-34010200

ABSTRACT

ABSTRACT: Excessive consumption of fructose, the sweetest of all naturally occurring carbohydrates, has been linked to worldwide epidemics of metabolic diseases in humans, and it is considered an independent risk factor for cardiovascular diseases. We provide an overview about the features of fructose metabolism, as well as potential mechanisms by which excessive fructose intake is associated with the pathogenesis of metabolic diseases both in humans and rodents. To accomplish this aim, we focus on illuminating the cellular and molecular mechanisms of fructose metabolism as well as its signaling effects on metabolic and cardiovascular homeostasis in health and disease, highlighting the role of carbohydrate-responsive element-binding protein in regulating fructose metabolism.


Subject(s)
Fructose , Metabolic Diseases , Fructose/adverse effects , Homeostasis , Humans , Metabolic Diseases/etiology
7.
FASEB J ; 34(10): 13862-13876, 2020 10.
Article in English | MEDLINE | ID: mdl-32844471

ABSTRACT

The zinc-finger protein ZBTB20 regulates development and metabolism in multiple systems, and is essential for postnatal survival in mice. However, its potential role in the cardiovascular system remains undefined. Here, we demonstrate that ZBTB20 is critically involved in the regulation of cardiac contractility and blood pressure in mice. At the age of 16 days, the relatively healthy Zbtb20-null mice exhibited hypotension without obvious change of heart rate or other evidence for heart failure. Moreover, Zbtb20 deletion led to a marked reduction in heart size, left ventricular wall thickness, and cell size of cardiomyocytes, which was largely proportional to the decreased body growth. Notably, echocardiographic and hemodynamic analyses showed that cardiac contractility was greatly impaired in the absence of ZBTB20. Mechanistically, ZBTB20 deficiency decreased cardiac ATP contents, and compromised the enzyme activity of mitochondrial complex I in heart as well as L-type calcium current density in cardiomyocytes. Furthermore, the developmental activation of some mitochondrial function-related genes was significantly attenuated in Zbtb20-null myocardium, which included Hspb8, Ckmt2, Cox7a1, Tfrc, and Ogdhl. Put together, these results suggest that ZBTB20 plays a crucial role in the regulation of heart development, energy metabolism, and contractility.


Subject(s)
Heart Diseases/genetics , Hypotension/genetics , Myocardial Contraction , Transcription Factors/genetics , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling , Cells, Cultured , Creatine Kinase, Mitochondrial Form/genetics , Creatine Kinase, Mitochondrial Form/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Heart Diseases/metabolism , Heart Diseases/pathology , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hypotension/metabolism , Hypotension/pathology , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , Ventricular Function , Ventricular Remodeling
8.
Cell Death Dis ; 9(5): 462, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29700307

ABSTRACT

Liver has a unique regenerative capacity, however, its regulatory mechanism is not fully defined. We have established the zinc-finger protein ZBTB20 as a key transcriptional repressor for alpha-fetoprotein (AFP) gene in liver. As a marker of hepatic differentiation, AFP expression is closely associated with hepatocyte proliferation. Unexpectedly, here we showed that ZBTB20 acts as a positive regulator of hepatic replication and is required for efficient liver regeneration. The mice specifically lacking ZBTB20 in hepatocytes exhibited a remarkable defect in liver regeneration after partial hepatectomy, which was characterized by impaired hepatocyte proliferation along with delayed cyclin D1 induction and diminished AKT activation. Furthermore, we found that epithelial growth factor receptor (EGFR) expression was dramatically reduced in the liver in the absence of ZBTB20, thereby substantially attenuating the activation of EGFR signaling pathway in regenerating liver. Adenovirus-mediated EGFR overexpression in ZBTB20-deficient hepatocytes could largely restore AKT activation in response to EGFR ligands in vitro, as well as hepatocyte replication in liver regeneration. Furthermore, ZBTB20 overexpression could significantly restore hepatic EGFR expression and cell proliferation after hepatectomy in ZBTB20-deficient liver. Taken together, our data point to ZBTB20 as a critical regulator of EGFR expression and hepatocyte proliferation in mouse liver regeneration, and may serve as a potential therapeutic target in clinical settings of liver regeneration.


Subject(s)
Cell Proliferation , ErbB Receptors/metabolism , Gene Expression Regulation , Hepatocytes/metabolism , Liver Regeneration , Liver/metabolism , Transcription Factors/metabolism , Animals , ErbB Receptors/genetics , Hepatocytes/pathology , Liver/pathology , Male , Mice , Mice, Knockout , Signal Transduction , Transcription Factors/genetics
9.
J Vis Exp ; (129)2017 11 16.
Article in English | MEDLINE | ID: mdl-29286362

ABSTRACT

The pituitary gland or hypophysis is an important endocrine organ secreting hormones essential for homeostasis. It consists of two glands with separate embryonic origins and functions - the neurohypophysis and the adenohypophysis. The developing mouse pituitary gland is tiny and delicate with an elongated oval shape. A coronal section is preferred to display both the adenohypophysis and neurohypophysis in a single slice of the mouse pituitary. The goal of this protocol is to achieve proper pituitary coronal sections with well-preserved tissue architectures from developing mice. In this protocol, we describe in detail how to dissect and process pituitary glands properly from developing mice. First, mice are fixed by transcardial perfusion of formaldehyde prior to dissection. Then three different dissecting techniques are applied to obtain intact pituitary glands depending on the age of mice. For fetal mice aged embryonic days (E) 17.5 - 18.5 and neonates up to 4 days, the entire sella regions including the sphenoid bone, gland, and trigeminal nerves are dissected. For pups aged postnatal days (P) 5 - 14, the pituitary glands connected with trigeminal nerves are dissected as a whole. For mice over 3 weeks old, the pituitary glands are carefully dissected free from the surrounding tissues. We also display how to embed the pituitary glands in a proper orientation by using the surrounding tissues as landmarks to obtain satisfying coronal sections. These methods are useful in analyzing histological and developmental features of pituitary glands in developing mice.


Subject(s)
Dissection/methods , Pituitary Gland/embryology , Pituitary Gland/surgery , Animals , Embryo, Mammalian/surgery , Mice , Pituitary Gland/pathology
10.
Nat Commun ; 8: 14824, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28327662

ABSTRACT

Hepatic de novo lipogenesis (DNL) converts carbohydrates into triglycerides and is known to influence systemic lipid homoeostasis. Here, we demonstrate that the zinc finger protein Zbtb20 is required for DNL. Mice lacking Zbtb20 in the liver exhibit hypolipidemia and reduced levels of liver triglycerides, along with impaired hepatic lipogenesis. The expression of genes involved in glycolysis and DNL, including that of two ChREBP isoforms, is decreased in livers of knockout mice. Zbtb20 binds to and enhances the activity of the ChREBP-α promoter, suggesting that altered metabolic gene expression is mainly driven by ChREBP. In addition, ChREBP-ß overexpression largely restores hepatic expression of genes involved in glucose and lipid metabolism, and increases plasma and liver triglyceride levels in knockout mice. Finally, we show that Zbtb20 ablation protects from diet-induced liver steatosis and improves hepatic insulin resistance. We suggest ZBTB20 is an essential regulator of hepatic lipogenesis and may be a therapeutic target for the treatment of fatty liver disease.


Subject(s)
Lipogenesis , Liver/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Carbohydrates/chemistry , Cell Nucleus/metabolism , Dietary Carbohydrates , Fatty Liver/genetics , Fatty Liver/pathology , Gene Deletion , Gene Expression Regulation , Glucose/metabolism , Glycolysis , Homeostasis , Humans , Insulin Resistance , Lipogenesis/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Protein Transport , Transcription Factors/deficiency , Transcription, Genetic , Triglycerides/blood , Triglycerides/metabolism
11.
Sci Rep ; 6: 34936, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713535

ABSTRACT

The purpose of the current study was to investigate the effect of topical atropine on choroidal thickness using spectral-domain optical coherence tomography. A total of 30 healthy eyes from 30 children were analyzed in this study. A single drop of 1% atropine gel was administered twice daily for a week. Choroidal thickness (CT) was measured using SD-OCT, and changes in CT before and after administration of the eye drops were analyzed at the subfovea and at 1.0-mm intervals (up to 3.0 mm) from the fovea at superior, inferior, nasal, and temporal locations. Pre- and post-cycloplegic axial length (AL) was also measured using the IOLMaster. We observed that administration of 1% atropine gel led to a significant increase in the choroidal thickness under the fovea and at all intervals from the fovea. The greatest change in CT was observed in the inferior meridian, while the nasal meridian exhibited the least change. AL did not significantly differ before and after cycloplegia, and there was no significant correlation between the changes in AL and subfoveal CT. It was concluded that administration of 1% atropine gel can significantly increase CT in the eyes of young Chinese children, albeit with different magnitude at different locations.


Subject(s)
Atropine/administration & dosage , Choroid/anatomy & histology , Choroid/drug effects , Mydriatics/administration & dosage , Child , Child, Preschool , Choroid/physiology , Female , Fovea Centralis/anatomy & histology , Fovea Centralis/drug effects , Fovea Centralis/physiology , Humans , Male , Muscarinic Antagonists/administration & dosage , Myopia/pathology , Myopia/physiopathology , Myopia/prevention & control , Ophthalmic Solutions , Prospective Studies , Tomography, Optical Coherence/methods
12.
Nat Commun ; 7: 11121, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27079169

ABSTRACT

The anterior pituitary harbours five distinct hormone-producing cell types, and their cellular differentiation is a highly regulated and coordinated process. Here we show that ZBTB20 is essential for anterior pituitary development and lactotrope specification in mice. In anterior pituitary, ZBTB20 is highly expressed by all the mature endocrine cell types, and to some less extent by somatolactotropes, the precursors of prolactin (PRL)-producing lactotropes. Disruption of Zbtb20 leads to anterior pituitary hypoplasia, hypopituitary dwarfism and a complete loss of mature lactotropes. In ZBTB20-null mice, although lactotrope lineage commitment is normally initiated, somatolactotropes exhibit profound defects in lineage specification and expansion. Furthermore, endogenous ZBTB20 protein binds to Prl promoter, and its knockdown decreases PRL expression and secretion in a lactotrope cell line MMQ. In addition, ZBTB20 overexpression enhances the transcriptional activity of Prl promoter in vitro. In conclusion, our findings point to ZBTB20 as a critical regulator of anterior pituitary development and lactotrope specification.


Subject(s)
Cell Lineage/genetics , Lactotrophs/metabolism , Pituitary Gland, Anterior/metabolism , Transcription Factors/genetics , Animals , Blotting, Western , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Hypopituitarism/genetics , Hypopituitarism/metabolism , Hypothalamus/embryology , Hypothalamus/growth & development , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Lactotrophs/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pituitary Gland, Anterior/embryology , Pituitary Gland, Anterior/growth & development , Prolactin/genetics , Prolactin/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
13.
Sci Rep ; 6: 20438, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26830324

ABSTRACT

The recombination efficiency and cell specificity of Cre driver lines are critical for exploring pancreatic ß cell biology with the Cre/LoxP approach. Some commonly used Cre lines are based on the short Ins2 promoter fragment and show recombination activity in hypothalamic neurons; however, whether this stems from endogenous Ins2 promoter activity remains controversial. In this study, we generated Ins2-Cre knockin mice with a targeted insertion of IRES-Cre at the Ins2 locus and demonstrated with a cell lineage tracing study that the Ins2 gene is not transcriptionally active in the hypothalamus. The Ins2-Cre driver line displayed robust Cre expression and activity in pancreatic ß cells without significant alterations in insulin expression. In the brain, Cre activity was mainly restricted to the choroid plexus, without significant recombination detected in the hippocampus or hypothalamus by the LacZ or fluorescent tdTomato reporters. Furthermore, Ins2-Cre mice exhibited normal glucose tolerance and insulin secretion upon glucose stimulation in vivo. In conclusion, this Ins2-Cre driver line allowed high-fidelity detection of endogenous Ins2 promoter activity in vivo, and the negative activity in the hypothalamus demonstrated that this system is a promising alternative tool for studying ß cell biology.


Subject(s)
Gene Knock-In Techniques , Genetic Loci , Hypothalamus/cytology , Hypothalamus/metabolism , Insulin/genetics , Integrases/genetics , Integrases/metabolism , Neurons/metabolism , Animals , Enzyme Activation , Gene Order , Gene Targeting , Genetic Vectors/genetics , Glucose/metabolism , Homologous Recombination , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Transgenic , Phenotype
14.
Sci Rep ; 5: 11979, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26173901

ABSTRACT

Alpha-fetoprotein (AFP) represents a classical model system to study developmental gene regulation in mammalian cells. We previously reported that liver ZBTB20 is developmentally regulated and plays a central role in AFP postnatal repression. Here we show that ZBTB20 is a sequence-specific transcriptional repressor of AFP. By ELISA-based DNA-protein binding assay and conventional gel shift assay, we successfully identified a ZBTB20-binding site at -104/-86 of mouse AFP gene, flanked by two HNF1 sites and two C/EBP sites in the proximal promoter. Importantly, mutation of the core sequence in this site fully abolished its binding to ZBTB20 in vitro, as well as the repression of AFP promoter activity by ZBTB20. The unique ZBTB20 site was highly conserved in rat and human AFP genes, but absent in albumin genes. These help to explain the autonomous regulation of albumin and AFP genes in the liver after birth. Furthermore, we demonstrated that transcriptional repression of AFP gene by ZBTB20 was liver-specific. ZBTB20 was dispensable for AFP silencing in other tissues outside liver. Our data define a cognate ZBTB20 site in AFP promoter which mediates the postnatal repression of AFP gene in the liver.


Subject(s)
Transcription Factors/metabolism , alpha-Fetoproteins/metabolism , Animals , Base Sequence , Binding Sites , DNA/chemistry , DNA/metabolism , Electrophoretic Mobility Shift Assay , Genes, Reporter , Hep G2 Cells , Humans , Liver/metabolism , Mice , Mice, Knockout , Mutation , Promoter Regions, Genetic , Protein Binding , Rats , Real-Time Polymerase Chain Reaction , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , alpha-Fetoproteins/chemistry , alpha-Fetoproteins/genetics
15.
Development ; 142(2): 385-93, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25564625

ABSTRACT

The terminal differentiation of hypertrophic chondrocytes is a tightly regulated process that plays a pivotal role in endochondral ossification. As a negative regulator, Sox9 is essentially downregulated in terminally differentiated hypertrophic chondrocytes. However, the underlying mechanism of Sox9 silencing is undefined. Here we show that the zinc finger protein Zbtb20 regulates the terminal differentiation of hypertrophic chondrocytes by repressing Sox9. In the developing skeleton of the mouse, Zbtb20 protein is highly expressed by hypertrophic chondrocytes from late embryonic stages. To determine its physiological role in endochondral ossification, we have generated chondrocyte-specific Zbtb20 knockout mice and demonstrate that disruption of Zbtb20 in chondrocytes results in delayed endochondral ossification and postnatal growth retardation. Zbtb20 deficiency caused a delay in cartilage vascularization and an expansion of the hypertrophic zone owing to reduced expression of Vegfa in the hypertrophic zone. Interestingly, Sox9, a direct suppressor of Vegfa expression, was ectopically upregulated at both mRNA and protein levels in the late Zbtb20-deficient hypertrophic zone. Furthermore, knockdown of Sox9 greatly increased Vegfa expression in Zbtb20-deficient hypertrophic chondrocytes. Our findings point to Zbtb20 as a crucial regulator governing the terminal differentiation of hypertrophic chondrocytes at least partially through repression of Sox9.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/physiology , Osteogenesis/physiology , SOX9 Transcription Factor/antagonists & inhibitors , Transcription Factors/metabolism , Analysis of Variance , Animals , Chromatin Immunoprecipitation , Histological Techniques , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , RNA Interference , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Nat Commun ; 5: 4984, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25369838

ABSTRACT

In mammals, pain sensation is initiated by the detection of noxious stimuli through specialized transduction ion channels and receptors in nociceptive sensory neurons. Transient receptor potential (TRP) channels are the key sensory transducers that confer nociceptors distinct sensory modalities. However, the regulatory mechanisms about their expression are poorly defined. Here we show that the zinc-finger protein ZBTB20 regulates TRP channels expression in nociceptors. ZBTB20 is highly expressed in nociceptive sensory neurons of dorsal root ganglia. Disruption of ZBTB20 in nociceptors led to a marked decrease in the expression levels of TRPV1, TRPA1 and TRPM8 and the response of calcium flux and whole-cell currents evoked by their respective specific agonists. Phenotypically, the mice lacking ZBTB20 specifically in nociceptors showed a defect in nociception and pain sensation in response to thermal, mechanical and inflammatory stimulation. Our findings point to ZBTB20 as a critical regulator of nociception and pain sensation by modulating TRP channels expression in nociceptors.


Subject(s)
Nociception , Nociceptors/metabolism , TRPM Cation Channels/genetics , TRPV Cation Channels/genetics , Transcription Factors/metabolism , Transient Receptor Potential Channels/genetics , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , TRPA1 Cation Channel , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism , Transcription Factors/genetics , Transient Receptor Potential Channels/metabolism
17.
Diabetologia ; 56(9): 1999-2009, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23793716

ABSTRACT

AIMS/HYPOTHESIS: Adaptor protein, phosphotyrosine interaction, pleckstrin homology domain and leucine zipper containing 1 (APPL1) is an adapter protein that positively mediates adiponectin signalling. Deficiency of APPL1 in the target tissues of insulin induces insulin resistance. We therefore aimed, in the present study, to determine its role in regulating pancreatic beta cell function. METHODS: A hyperglycaemic clamp test was performed to determine insulin secretion in APPL1 knockout (KO) mice. Glucose- and adiponectin-induced insulin release was measured in islets from APPL1 KO mice or INS-1(832/13) cells with either APPL1 knockdown or overproduction. RT-PCR and western blotting were conducted to analyse gene expression and protein abundance. Oxygen consumption rate (OCR), ATP production and mitochondrial membrane potential were assayed to evaluate mitochondrial function. RESULTS: APPL1 is highly expressed in pancreatic islets, but its levels are decreased in mice fed a high-fat diet and db/db mice compared with controls. Deletion of the Appl1 gene leads to impairment of both the first and second phases of insulin secretion during hyperglycaemic clamp tests. In addition, glucose-stimulated insulin secretion (GSIS) is significantly decreased in islets from APPL1 KO mice. Conversely, overproduction of APPL1 leads to an increase in GSIS in beta cells. In addition, expression levels of several genes involved in insulin production, mitochondrial biogenesis and mitochondrial OCR, ATP production and mitochondrial membrane potential are reduced significantly in APPL1-knockdown beta cells. Moreover, suppression or overexproduction of APPL1 inhibits or stimulates adiponectin-potentiated GSIS in beta cells, respectively. CONCLUSIONS/INTERPRETATION: Our study demonstrates the roles of APPL1 in regulating GSIS and mitochondrial function in pancreatic beta cells, which implicates APPL1 as a therapeutic target in the treatment of type 2 diabetes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Glucose/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mitochondria/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Blotting, Western , Cell Line , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
J Physiol ; 590(19): 4917-32, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22777671

ABSTRACT

The mammalian hippocampus harbours neural circuitry that is crucial for associative learning and memory. The mechanisms that underlie the development and regulation of this complex circuitry are not fully understood. Our previous study established an essential role for the zinc finger protein Zbtb20 in the specification of CA1 field identity in the developing hippocampus. Here, we show that conditionally deleting Zbtb20 specifically in mature CA1 pyramidal neurons impaired hippocampus-dependent memory formation, without affecting hippocampal architecture or the survival, identity and basal excitatory synaptic activity of CA1 pyramidal neurons. We demonstrate that mature CA1-specific Zbtb20 knockout mice exhibited reductions in long-term potentiation (LTP) and NMDA receptor (NMDAR)-mediated excitatory post-synaptic currents. Furthermore, we show that activity-induced phosphorylation of ERK and CREB is impaired in the hippocampal CA1 of Zbtb20 mutant mice. Collectively, these results indicate that Zbtb20 in mature CA1 plays an important role in LTP and memory by regulating NMDAR activity, and activation of ERK and CREB.


Subject(s)
CA1 Region, Hippocampal/physiology , Memory/physiology , Pyramidal Cells/physiology , Transcription Factors/physiology , Animals , CA1 Region, Hippocampal/cytology , Cyclic AMP Response Element-Binding Protein/physiology , Excitatory Postsynaptic Potentials , Extracellular Signal-Regulated MAP Kinases/physiology , Hippocampus , Long-Term Potentiation/physiology , Mice , Mice, Knockout , Receptors, N-Methyl-D-Aspartate/physiology
19.
Gastroenterology ; 142(7): 1571-1580.e6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22374165

ABSTRACT

BACKGROUND & AIMS: Fructose-1,6-bisphosphatase (FBP)-1 is a gluconeogenic enzyme that regulates glucose metabolism and insulin secretion in ß cells, but little is known about how its transcription is controlled. The zinc finger protein ZBTB20 regulates glucose homeostasis, so we investigated its effects on expression of FBP-1. METHODS: We analyzed gene expression using real-time reverse-transcription polymerase chain reaction, immunoblotting, and immunohistochemistry. We generated mice with ß cell-specific disruption of Zbtb20 using Cre/LoxP technology. Expression of Zbtb20 in ß cells was reduced using small interfering RNAs, and promoter occupancy and transcriptional regulation were analyzed by chromatin immunoprecipitation and reporter assays. RESULTS: ZBTB20 was expressed at high levels by ß cells and other endocrine cells in islets of normal mice; expression levels were reduced in islets from diabetic db/db mice. Mice with ß cell-specific knockout of Zbtb20 had normal development of ß cells but had hyperglycemia, hypoinsulinemia, glucose intolerance, and impaired glucose-stimulated insulin secretion. Islets isolated from these mice had impaired glucose metabolism, adenosine triphosphate production, and insulin secretion after glucose stimulation in vitro, although insulin secretion returned to normal levels in the presence of KCl. ZBTB20 knockdown with small interfering RNAs impaired glucose-stimulated insulin secretion in the ß cell line MIN6. Expression of Fbp1 was up-regulated in ß cells with ZBTB20 knockout or knockdown; impairments to glucose-stimulated insulin secretion were restored by inhibition of FBPase activity. ZBTB20 was recruited to the Fbp1 promoter and repressed its transcription in ß cells. CONCLUSIONS: The transcription factor ZBTB20 regulates ß cell function and glucose homeostasis in mice. It might be a therapeutic target for type 2 diabetes mellitus.


Subject(s)
Fructose-Bisphosphatase/metabolism , Insulin-Secreting Cells/metabolism , Repressor Proteins/physiology , Zinc Fingers , Animals , Cell Line , Fructose-Bisphosphatase/genetics , Gene Expression , Gene Knockdown Techniques , Glucose/metabolism , Glucose Intolerance/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Insulin Secretion , Mice , Mice, Knockout , Mice, Transgenic , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
20.
BMC Cancer ; 11: 271, 2011 Jun 25.
Article in English | MEDLINE | ID: mdl-21702992

ABSTRACT

BACKGROUND: Our previous studies showed that ZBTB20, a new BTB/POZ-domain gene, could negatively regulate α feto-protein and other liver-specific genes, concerning such as bio-transformation, glucose metabolism and the regulation of the somatotropic hormonal axis. The aim of this study is to determine the potential clinical implications of ZBTB20 in hepatocellular carcinoma (HCC). METHODS: Quantitative real-time RT-PCR and Western blot analyses were used to detect expression levels of ZBTB20 in 50 paired HCC tumorous and nontumorous tissues and in 20 normal liver tissues. Moreover, expression of ZBTB20 was assessed by immunohistochemistry of paired tumor and peritumoral liver tissue from 102 patients who had undergone hepatectomy for histologically proven HCC. And its relationship with clinicopathological parameters and prognosis was investigated. RESULTS: Both messenger RNA and protein expression levels of ZBTB20 were elevated significantly in HCC tissues compared with the paired non-tumor tissues and normal liver tissues. Overexpressed ZBTB20 protein in HCC was significantly associated with vein invasion (P=0.016). Importantly, the recurrence or metastasis rates of HCCs with higher ZBTB20 expression were markedly greater than those of HCCs with lower expression (P=0.003, P=0.00015, respectively). Univariate and multivariate analyses revealed that ZBTB20 overexpression was an independent prognostic factor for HCC. The disease-free survival period and over-all survival period in patients with overexpressed ZBTB20 in HCC was significantly reduced. CONCLUSIONS: The expression of ZBTB20 is increased in HCC and associated with poor prognosis in patients with HCC, implicating ZBTB20 as a candidate prognostic marker in HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Transcription Factors/biosynthesis , Adult , Aged , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , China/epidemiology , Comorbidity , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Hepatitis B, Chronic/epidemiology , Hepatitis C, Chronic/epidemiology , Humans , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recurrence , Reverse Transcriptase Polymerase Chain Reaction , Sampling Studies , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL