Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 17(4)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38675383

ABSTRACT

To overcome the limitations of current nano/micro-scale drug delivery systems, an Escherichia coli (E. coli)-based drug delivery system could be a potential alternative, and an effective tumor-targeting delivery system can be developed by attempting to perform chemical binding to the primary amine group of a cell membrane protein. In addition, positron emission tomography (PET) is a representative non-invasive imaging technology and is actively used in the field of drug delivery along with radioisotopes capable of long-term tracking, such as zirconium-89 (89Zr). The membrane proteins were labeled with 89Zr using chelate (DFO), and not only was the long-term biodistribution in tumors and major organs evaluated in the body, but the labeling stability of 89Zr conjugated to the membrane proteins was also evaluated through continuous tracking. E. coli accumulated at high levels in the tumor within 5 min (initial time) after tail intravenous injection, and when observed after 6 days, 89Zr-DFO on the surface of E. coli was found to be stable for a long period of time in the body. In this study, we demonstrated the long-term biodistribution and tumor-targeting effect of an E. coli-based drug delivery system and verified the in vivo stability of radioisotopes labeled on the surface of E. coli.

2.
Biomedicines ; 9(7)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34356855

ABSTRACT

The spliced form of X-box binding protein 1 (XBP1s) is an active transcription factor that plays a vital role in the unfolded protein response (UPR). Under endoplasmic reticulum (ER) stress, unspliced Xbp1 mRNA is cleaved by the activated stress sensor IRE1α and converted to the mature form encoding spliced XBP1 (XBP1s). Translated XBP1s migrates to the nucleus and regulates the transcriptional programs of UPR target genes encoding ER molecular chaperones, folding enzymes, and ER-associated protein degradation (ERAD) components to decrease ER stress. Moreover, studies have shown that XBP1s regulates the transcription of diverse genes that are involved in lipid and glucose metabolism and immune responses. Therefore, XBP1s has been considered an important therapeutic target in studying various diseases, including cancer, diabetes, and autoimmune and inflammatory diseases. XBP1s is involved in several unique mechanisms to regulate the transcription of different target genes by interacting with other proteins to modulate their activity. Although recent studies discovered numerous target genes of XBP1s via genome-wide analyses, how XBP1s regulates their transcription remains unclear. This review discusses the roles of XBP1s in target genes transcriptional regulation. More in-depth knowledge of XBP1s target genes and transcriptional regulatory mechanisms in the future will help develop new therapeutic targets for each disease.

3.
Materials (Basel) ; 14(12)2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34207994

ABSTRACT

This study evaluated the in vivo behavior and accumulation of silica particles in the form of wires, which were actively studied as drug carriers along with spheres, using positron emission tomography (PET). Wire-shaped silicon dioxide (SiO2) was synthesized at micro-size, using anodic aluminum oxide (AAO), a template, and folic acid (FA), which specifically binds folate receptors (FR) which are overexpressed in many cancers, and which was bound to the wire's surface to confirm its possible use as a cancer diagnostic agent. In addition, for evaluation using PET, the positron-emitting nuclide 89Zr (t1/2 = 3.3 days) was directly bonded to the hydroxyl group (-OH) on the particle surface. The diameter and shape of the synthesized silica microwires (SMWs) were confirmed using SEM and TEM, the chemical bonding of FA was confirmed through FT-IR and NMR, and the labeling of 89Zr was measured by means of radio-thin-layer chromatography (TLC) measurement. Folic acid-conjugated SMWs (FA-SMWs) were found to have a low receptor-mediated uptake in cell internalization evaluation, but in PET studies, FA-SMWs stayed longer at the tumor site. In conclusion, we successfully synthesized a homogeneous silica microwire for drug delivery, we confirmed that the FA-conjugated sample remains at the tumor site for a relatively longer time, and we have reported the characteristic in vivo behavior of 89Zr-FA-SMWs.

4.
Front Cell Neurosci ; 15: 663092, 2021.
Article in English | MEDLINE | ID: mdl-34149360

ABSTRACT

Glioblastoma (GBM) is the most common and malignant form of primary brain tumor with a median survival time of 14-16 months in GBM patients. Surgical treatment with chemotherapy and radiotherapy may help increase survival by removing GBM from the brain. However, complete surgical resection to eliminate GBM is almost impossible due to its high invasiveness. When GBM cells migrate to the brain, they interact with various cells, including astrocytes, neurons, endothelial cells, and the extracellular matrix (ECM). They can also make their cell body shrink to infiltrate into narrow spaces in the brain; thereby, they can invade regions of the brain and escape from surgery. Brain tumor cells create an appropriate microenvironment for migration and invasion by modifying and degrading the ECM. During those processes, the Ca2+ signaling pathway and other signaling cascades mediated by various ion channels contribute mainly to gene expression, motility, and invasion of GBM cells. Furthermore, GBM cells release glutamate, affecting migration via activation of ionotropic glutamate receptors in an autocrine manner. This review focuses on the cellular mechanisms of glioblastoma invasion and motility related to ECM, Ca2+ signaling, and glutamate. Finally, we discuss possible therapeutic interventions to inhibit invasion by GBM cells.

5.
BMC Cancer ; 19(1): 1113, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727003

ABSTRACT

Following publication of the original article [1], the authors have re-evaluated the authorship for this article. The updated author group is.

6.
Mol Cells ; 42(6): 501, 2019 06 30.
Article in English | MEDLINE | ID: mdl-31250623
7.
Mol Cells ; 41(8): 705-716, 2018 Aug 31.
Article in English | MEDLINE | ID: mdl-30078231

ABSTRACT

The endoplasmic reticulum (ER) is a critical organelle for protein synthesis, folding and modification, and lipid synthesis and calcium storage. Dysregulation of ER functions leads to the accumulation of misfolded- or unfolded-protein in the ER lumen, and this triggers the unfolded protein response (UPR), which restores ER homeostasis. The UPR is characterized by three distinct downstream signaling pathways that promote cell survival or apoptosis depending on the stressor, the intensity and duration of ER stress, and the cell type. Mammalian cells express the UPR transducers IRE1, PERK, and ATF6, which control transcriptional and translational responses to ER stress. Direct links between ER stress and immune responses are also evident, but the mechanisms by which UPR signaling cascades are coordinated with immunity remain unclear. This review discusses recent investigations of the roles of ER stress in immune responses that lead to differentiation, maturation, and cytokine expression in immune cells. Further understanding of how ER stress contributes to the pathogenesis of immune disorders will facilitate the development of novel therapies that target UPR pathways.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Unfolded Protein Response/immunology , X-Box Binding Protein 1/immunology , Cell Differentiation , Humans
8.
Hepatology ; 68(6): 2167-2181, 2018 12.
Article in English | MEDLINE | ID: mdl-29698569

ABSTRACT

Integrated stress response (ISR) is a signaling system in which phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) by stress-specific kinases and subsequent activation of activation transcription factor (ATF) 4 help restore cellular homeostasis following exposure to environmental stresses. ISR activation has been observed in metabolic diseases, including hepatic steatosis (HS), steatohepatitis (SH), and insulin resistance (IR), but it remains unclear whether ISR contributes to disease pathogenesis or represents an innate defense mechanism against metabolic stresses. Constitutive repressor of eIF2α phosphorylation (CReP) is a critical regulatory subunit of the eIF2α phosphatase complex. Here, we show that CReP ablation causes constitutive eIF2α phosphorylation in the liver, which leads to activation of the ATF4 transcriptional program including increased fibroblast growth factor 21 (FGF21) production. Liver-specific CReP knockout (CRePLKO ) mice exhibited marked browning of white adipose tissue (WAT) and increased energy expenditure and insulin sensitivity in an FGF21-dependent manner. Furthermore, CRePLKO mice were protected from high-fat diet (HFD)-induced obesity, HS, and IR. Acute CReP ablation in liver of HFD-induced obese mice also reduced adiposity and improved glucose homeostasis. Conclusion: These data suggest that CReP abundance is a critical determinant for eIF2α phosphorylation and ensuing ISR activation in the liver. Constitutive ISR activation in the liver induces FGF21 and confers protection from HFD-induced adiposity, IR, and HS in mice. Augmenting hepatic ISR may represent a therapeutic approach to treat metabolic disorders.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Fatty Liver/etiology , Fibroblast Growth Factors/metabolism , Protein Phosphatase 1/physiology , Stress, Physiological , Activating Transcription Factor 4/metabolism , Adipocytes, Beige/physiology , Adiposity , Animals , Diet, High-Fat/adverse effects , Energy Metabolism , Homeostasis , Insulin Resistance , Mice , Mice, Knockout , Obesity/etiology
9.
Sci Rep ; 7(1): 14940, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29097726

ABSTRACT

Long chain fatty acids (LCFAs) exert pro-inflammatory effects in vivo. However, little is known regarding the effect of LCFAs on invariant (i) NKT cell functions. Here, we report an inhibitory effect of saturated LCFAs on transcription factors in iNKT cells. Among the saturated LCFAs, palmitic acid (PA) specifically inhibited IL-4 and IFN-γ production and reduced gata-3 and t-bet transcript levels in iNKT cells during TCR-mediated activation. In iNKT cells, PA was localized and induced dilation in the endoplasmic reticulum and increased the mRNA levels of downstream molecules of IRE1α RNase. Moreover, PA increased the degradation rates of gata-3 and t-bet mRNA, which was restored by IRE1α inhibition or transfection with mutant gata-3 or t-bet, indicating that gata-3 and t-bet are cleaved via regulated IRE1α-dependent decay (RIDD). A PA-rich diet and PA injection suppressed IL-4 and IFN-γ production by iNKT cells in C57BL/6, but not Jα18 knockout mice, which was restored by injection of STF083010, an IRE1α-specific inhibitor. Furthermore, a PA-rich diet and PA injection attenuated arthritis in an iNKT cell-dependent manner. Taken together, our experiments demonstrate that a saturated LCFA induced RIDD-mediated t-bet and gata-3 mRNA degradation in iNKT cells, thereby suppressing arthritis.


Subject(s)
Arthritis/drug therapy , Endoribonucleases/immunology , GATA3 Transcription Factor/genetics , Natural Killer T-Cells/drug effects , Palmitic Acid/therapeutic use , Protein Serine-Threonine Kinases/immunology , RNA Stability/drug effects , T-Box Domain Proteins/genetics , Animals , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , Cells, Cultured , GATA3 Transcription Factor/immunology , Mice, Inbred C57BL , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Palmitic Acid/pharmacology , Signal Transduction/drug effects , T-Box Domain Proteins/immunology
10.
Mol Cell Biol ; 35(16): 2761-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26031337

ABSTRACT

The unfolded protein response (UPR) regulates endoplasmic reticulum (ER) homeostasis and protects cells from ER stress. IRE1α is a central regulator of the UPR that activates the transcription factor XBP1s through an unconventional splicing mechanism using its endoribonuclease activity. IRE1α also cleaves certain mRNAs containing XBP1-like secondary structures to promote the degradation of these mRNAs, a process known as regulated IRE1α-dependent decay (RIDD). We show here that the mRNA of CReP/Ppp1r15b, a regulatory subunit of eukaryotic translation initiation factor 2α (eIF2α) phosphatase, is a RIDD substrate. eIF2α plays a central role in the integrated stress response by mediating the translational attenuation to decrease the stress level in the cell. CReP expression was markedly suppressed in XBP1-deficient mice livers due to hyperactivated IRE1α. Decreased CReP expression caused the induction of eIF2α phosphorylation and the attenuation of protein synthesis in XBP1-deficient livers. ER stress also suppressed CReP expression in an IRE1α-dependent manner, which increased eIF2α phosphorylation and consequently attenuated protein synthesis. Taken together, the results of our study reveal a novel function of IRE1α in the regulation of eIF2α phosphorylation and the translational control.


Subject(s)
Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Protein Phosphatase 1/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Stability , RNA, Messenger/metabolism , Animals , DNA-Binding Proteins/genetics , Down-Regulation , Endoplasmic Reticulum Stress , Gene Knockout Techniques , Liver/metabolism , Mice, Inbred C57BL , Phosphorylation , RNA, Messenger/chemistry , RNA, Messenger/genetics , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , X-Box Binding Protein 1
11.
J Clin Invest ; 125(5): 1955-67, 2015 May.
Article in English | MEDLINE | ID: mdl-25844898

ABSTRACT

The HSP40 cochaperone SEC63 is associated with the SEC61 translocon complex in the ER. Mutations in the gene encoding SEC63 cause polycystic liver disease in humans; however, it is not clear how altered SEC63 influences disease manifestations. In mice, loss of SEC63 induces cyst formation both in liver and kidney as the result of reduced polycystin-1 (PC1). Here we report that inactivation of SEC63 induces an unfolded protein response (UPR) pathway that is protective against cyst formation. Specifically, using murine genetic models, we determined that SEC63 deficiency selectively activates the IRE1α-XBP1 branch of UPR and that SEC63 exists in a complex with PC1. Concomitant inactivation of both SEC63 and XBP1 exacerbated the polycystic kidney phenotype in mice by markedly suppressing cleavage at the G protein-coupled receptor proteolysis site (GPS) in PC1. Enforced expression of spliced XBP1 (XBP1s) enhanced GPS cleavage of PC1 in SEC63-deficient cells, and XBP1 overexpression in vivo ameliorated cystic disease in a murine model with reduced PC1 function that is unrelated to SEC63 inactivation. Collectively, the findings show that SEC63 function regulates IRE1α/XBP1 activation, SEC63 and XBP1 are required for GPS cleavage and maturation of PC1, and activation of XBP1 can protect against polycystic disease in the setting of impaired biogenesis of PC1.


Subject(s)
DNA Helicases/physiology , DNA-Binding Proteins/physiology , Endoribonucleases/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Recessive/genetics , Protein Serine-Threonine Kinases/metabolism , TRPP Cation Channels/deficiency , Transcription Factors/physiology , Unfolded Protein Response/physiology , Animals , Cell Line , DNA Helicases/deficiency , DNA Helicases/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Glucosidases/deficiency , Glucosidases/genetics , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Chaperones , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Recessive/metabolism , Protein Structure, Tertiary , RNA Splicing , RNA, Small Interfering/genetics , RNA-Binding Proteins , Receptors, G-Protein-Coupled/metabolism , Recombinant Fusion Proteins/metabolism , Regulatory Factor X Transcription Factors , TRPP Cation Channels/biosynthesis , TRPP Cation Channels/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transfection , X-Box Binding Protein 1
12.
Hepatology ; 61(3): 857-69, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25125366

ABSTRACT

UNLABELLED: Fat-specific protein 27 (Fsp27) is a lipid droplet-associated protein that promotes lipid droplet (LD) growth and triglyceride (TG) storage in white adipocytes. Fsp27 is also highly expressed in the steatotic liver and contributes to TG accumulation. In this study we discovered that the liver produces Fsp27ß, an alternative Fsp27 isoform, which contains 10 additional amino acids at the N-terminus of the original Fsp27 (Fsp27α). White adipose tissue (WAT) and the liver specifically expressed Fsp27α and Fsp27ß transcripts, respectively, which were driven by distinct promoters. The Fsp27ß promoter was activated by the liver-enriched transcription factor cyclic-AMP-responsive-element-binding protein H (CREBH) but not by peroxisome proliferator-activated receptor gamma (PPARγ), which activated the Fsp27α promoter. Enforced expression of the constitutively active CREBH strongly induced Fsp27ß and the human ortholog CIDEC2 in mouse hepatocytes and HepG2 cells, respectively. In contrast, loss of CREBH decreased hepatic Fsp27ß in fasted mice, suggesting that CREBH plays a critical role in Fsp27ß expression in the liver. Similar to Fsp27α, Fsp27ß localized on the surface of lipid droplets and suppressed lipolysis. Consequently, enforced expression of Fsp27ß or CREBH promoted lipid droplet enlargement and TG accumulation in the liver. CONCLUSION: The CREBH-Fsp27ß axis is important for regulating lipid droplet dynamics and TG storage in the liver.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Fatty Liver/etiology , Lipid Droplets/physiology , Liver/metabolism , Proteins/genetics , Transcriptional Activation , Animals , Mice , Mice, Inbred C57BL , Organ Specificity , PPAR gamma/physiology , Proteins/physiology , Triglycerides/metabolism
13.
J Immunol ; 193(6): 2772-83, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25114106

ABSTRACT

NFAT plays a crucial role in the immune system by regulating the transcription of inducible genes during immune responses. In T cells, NFAT proteins govern various cellular events related to T cell development, activation, tolerance induction, and differentiation. We previously reported the NFAT1-dependent enhancer activity of conserved noncoding sequence (CNS)-9, a distal cis-acting element, in the regulation of IL-10 transcription in T cells. In this study, we developed a T cell-based reporter system to identify compounds that modulate the regulatory activity of CNS-9. Among the identified candidates, 6-methoxyflavone (6-MF) significantly inhibited the enhancer activity of CNS-9, thereby reducing IL-10 expression in T cells without affecting cell viability. 6-MF also downregulated the transcription of NFAT1 target genes such as IL-4, IL-13, and IFN-γ. Treatment of 6-MF inhibited the translocation of NFAT1 into the nucleus, which consequently interrupted NFAT1 binding to the target loci, without affecting the expression or dephosphorylation of NFAT1. Treatment of 6-MF to CD4(+) T cells or B cells isolated from mice with atopic dermatitis significantly reduced disease-associated cytokine production, as well as the levels of IgE. In addition, oral administration of 6-MF to atopic dermatitis mice ameliorated disease symptoms by reducing serum IgE levels and infiltrating lymphocytes. Conclusively, our results suggest that 6-MF can be a potential candidate for the development of an effective immunomodulator via the suppression of NFAT-mediated T cell activation.


Subject(s)
Active Transport, Cell Nucleus/immunology , Flavones/pharmacology , Lymphocyte Activation/drug effects , NFATC Transcription Factors/immunology , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Nucleus , Conserved Sequence/drug effects , Conserved Sequence/genetics , Cytokines/biosynthesis , DNA-Binding Proteins/genetics , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , HEK293 Cells , Humans , Immunoglobulin E/blood , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-13/biosynthesis , Interleukin-13/genetics , Interleukin-4/biosynthesis , Interleukin-4/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NFATC Transcription Factors/antagonists & inhibitors , Phosphorylation , Protein Binding/drug effects , RNA, Untranslated/drug effects , RNA, Untranslated/genetics , Transcription, Genetic
14.
J Lipid Res ; 55(5): 850-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24598141

ABSTRACT

cAMP responsive element-binding protein H (CREBH) is an endoplasmic reticulum (ER) anchored transcription factor that is highly expressed in the liver and small intestine and implicated in nutrient metabolism and proinflammatory response. ApoA-IV is a glycoprotein secreted primarily by the intestine and to a lesser degree by the liver. ApoA-IV expression is suppressed in CREBH-deficient mice and strongly induced by enforced expression of the constitutively active form of CREBH, indicating that CREBH is the major transcription factor regulating Apoa4 gene expression. Here, we show that CREBH directly controls Apoa4 expression through two tandem CREBH binding sites (5'-CCACGTTG-3') located on the promoter, which are conserved between human and mouse. Chromatin immunoprecipitation and electrophoretic mobility-shift assays demonstrated specific association of CREBH with the CREBH binding sites. We also demonstrated that a substantial amount of CREBH protein was basally processed to the active nuclear form in normal mouse liver, which was further increased in steatosis induced by high-fat diet or fasting, increasing apoA-IV expression. However, we failed to find significant activation of CREBH in response to ER stress, arguing against the critical role of CREBH in ER stress response.


Subject(s)
Apolipoproteins A/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cell Line , Fatty Liver/genetics , Hepatocytes/metabolism , Humans , Mice , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Eur J Immunol ; 44(3): 867-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24242955

ABSTRACT

Inositol-requiring enzyme 1 (IRE1) is a kinase and ribonuclease that executes the splicing of X box binding protein 1 (XBP-1) mRNA in response to the accumulation of unfolded protein in the ER, a signal cascade termed the unfolded protein response. Recently, IRE1 has been implicated in mRNA and miRNA cleavage and degradation, a pathway termed regulated IRE1-dependent decay (RIDD). Deletion of XBP-1 in the liver and pancreas strongly enhances RIDD by upregulating IRE1 protein levels and enhancing its ribo-nuclease activity. Because XBP-1 is essential for generating plasma cells with developed secretory capacity, we sought to evaluate the contribution of RIDD to this regulation. Mice were conditionally deleted for XBP-1 and/or IRE1 in their B-cell lineage. Similarly to the liver, deletion of XBP-1 induces IRE1 expression in LPS-treated B cells. In vitro, IRE1 cleaves the mRNA of secretory µ chains, which explains the reduction in secretory µ mRNA and its synthesis in XBP-1 KO plasma cells. In accordance, the IgM response is partially restored in XBP-1/IRE1 double KO mice relative to XBP-1 KO mice. Interestingly, the IgG1 response is reduced to a similar level in XBP-1 KO, IRE1 KO, and their double knockout animals. Our data demonstrate a specific contribution by RIDD in curtailing immunoglobulin synthesis and secretion.


Subject(s)
Antibody Formation/physiology , Immunoglobulins/biosynthesis , Membrane Proteins/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , DNA-Binding Proteins/metabolism , Gene Expression , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Membrane Proteins/genetics , Mice , Mice, Knockout , Plasma Cells/ultrastructure , Protein Serine-Threonine Kinases/genetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , T-Lymphocytes/immunology , Transcription Factors/metabolism , X-Box Binding Protein 1
16.
Semin Liver Dis ; 33(4): 301-11, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24222088

ABSTRACT

The liver is a central organ that controls systemic energy homeostasis and nutrient metabolism. Dietary carbohydrates and lipids, and fatty acids derived from adipose tissue are delivered to the liver, and utilized for gluconeogenesis, lipogenesis, and ketogenesis, which are tightly regulated by hormonal and neural signals. Hepatic lipogenesis is activated primarily by insulin that is secreted from the pancreas after a high-carbohydrate meal. Sterol regulatory element binding protein-1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP) are major transcriptional regulators that induce key lipogenic enzymes to promote lipogenesis in the liver. Sterol regulatory element binding protein-1c is activated by insulin through complex signaling cascades that control SREBP-1c at both transcriptional and posttranslational levels. Carbohydrate-responsive element-binding protein is activated by glucose independently of insulin. Here, the authors attempt to summarize the current understanding of the molecular mechanism for the transcriptional regulation of hepatic lipogenesis, focusing on recent studies that explore the signaling pathways controlling SREBPs and ChREBP.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fatty Liver/metabolism , Lipogenesis , Liver/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Transcription, Genetic , Active Transport, Cell Nucleus , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Fatty Liver/genetics , Gene Expression Regulation , Humans , Lipogenesis/genetics , MicroRNAs/metabolism , Protein Stability , Signal Transduction , Sterol Regulatory Element Binding Proteins/genetics
17.
Cell Metab ; 16(4): 487-99, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23040070

ABSTRACT

XBP1 is a key regulator of the unfolded protein response (UPR), which is involved in a wide range of physiological and pathological processes. XBP1 ablation in liver causes profound hypolipidemia in mice, highlighting its critical role in lipid metabolism. XBP1 deficiency triggers feedback activation of its upstream enzyme IRE1α, instigating regulated IRE1-dependent decay (RIDD) of cytosolic mRNAs. Here, we identify RIDD as a crucial control mechanism of lipid homeostasis. Suppression of RIDD by RNA interference or genetic ablation of IRE1α reversed hypolipidemia in XBP1-deficient mice. Comprehensive microarray analysis of XBP1 and/or IRE1α-deficient liver identified genes involved in lipogenesis and lipoprotein metabolism as RIDD substrates, which might contribute to the suppression of plasma lipid levels by activated IRE1α. Ablation of XBP1 ameliorated hepatosteatosis, liver damage, and hypercholesterolemia in dyslipidemic animal models, suggesting that direct targeting of either IRE1α or XBP1 might be a feasible strategy to treat dyslipidemias.


Subject(s)
Endoribonucleases/metabolism , Lipid Metabolism/genetics , Lipids/blood , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cholesterol/blood , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Profiling , Lipogenesis , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , X-Box Binding Protein 1
18.
PLoS One ; 7(7): e40828, 2012.
Article in English | MEDLINE | ID: mdl-22815835

ABSTRACT

Type II collagen is the major component of articular cartilage and is mainly synthesized by chondrocytes. Repeated sub-culturing of primary chondrocytes leads to reduction of type II collagen gene (Col2a1) expression, which mimics the process of chondrocyte dedifferentiation. Although the functional importance of Col2a1 expression has been extensively investigated, mechanism of transcriptional regulation during chondrocyte dedifferentiation is still unclear. In this study, we have investigated the crosstalk between cis-acting DNA element and transcription factor on Col2a1 expression in primary chondrocytes. Bioinformatic analysis revealed the potential regulatory regions in the Col2a1 genomic locus. Among them, promoter and 3' untranslated region (UTR) showed highly accessible chromatin architecture with enriched recruitment of active chromatin markers in primary chondrocytes. 3' UTR has a potent enhancer function which recruits Lef1 (Lymphoid enhancer binding factor 1) transcription factor, leading to juxtaposition of the 3' UTR with the promoter through gene looping resulting in up-regulation of Col2a1 gene transcription. Knock-down of endogenous Lef1 level significantly reduced the gene looping and subsequently down-regulated Col2a1 expression. However, these regulatory loci become inaccessible due to condensed chromatin architecture as chondrocytes dedifferentiate which was accompanied by a reduction of gene looping and down-regulation of Col2a1 expression. Our results indicate that Lef1 mediated looping between promoter and 3' UTR under the permissive chromatin architecture upregulates Col2a1 expression in primary chondrocytes.


Subject(s)
3' Untranslated Regions/genetics , Chondrocytes/metabolism , Collagen Type II/genetics , Gene Expression Regulation , Nucleic Acid Conformation , Promoter Regions, Genetic , Animals , Cell Dedifferentiation/genetics , Cells, Cultured , Chondrocytes/cytology , Chromatin/metabolism , Collagen Type II/metabolism , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Genetic Loci/genetics , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mice, Inbred ICR , Protein Binding/genetics , Transcription, Genetic , Up-Regulation/genetics
19.
J Exp Med ; 209(2): 307-18, 2012 Feb 13.
Article in English | MEDLINE | ID: mdl-22291093

ABSTRACT

The mammalian stress sensor IRE1α plays a central role in the unfolded protein, or endoplasmic reticulum (ER), stress response by activating its downstream transcription factor XBP1 via an unconventional splicing mechanism. IRE1α can also induce the degradation of a subset of mRNAs in a process termed regulated IRE1-dependent decay (RIDD). Although diverse mRNA species can be degraded by IRE1α in vitro, the pathophysiological functions of RIDD are only beginning to be explored. Acetaminophen (APAP) overdose is the most frequent cause of acute liver failure in young adults in the United States and is primarily caused by CYP1A2-, CYP2E1-, and CYP3A4-driven conversion of APAP into hepatotoxic metabolites. We demonstrate here that genetic ablation of XBP1 results in constitutive IRE1α activation in the liver, leading to RIDD of Cyp1a2 and Cyp2e1 mRNAs, reduced JNK activation, and protection of mice from APAP-induced hepatotoxicity. A pharmacological ER stress inducer that activated IRE1α suppressed the expression of Cyp1a2 and Cyp2e1 in WT, but not IRE1α-deficient mouse liver, indicating the essential role of IRE1α in the down-regulation of these mRNAs upon ER stress. Our study reveals an unexpected function of RIDD in drug metabolism.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/metabolism , Enzyme Activation/physiology , Gene Expression Regulation/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Stability/genetics , Animals , Blotting, Western , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , DNA Primers/genetics , DNA-Binding Proteins/deficiency , Endoplasmic Reticulum Stress/genetics , Gene Deletion , HEK293 Cells , Humans , Mice , RNA Stability/physiology , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Regulatory Factor X Transcription Factors , Transcription Factors/deficiency , X-Box Binding Protein 1
20.
J Immunol ; 188(5): 2244-53, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22266280

ABSTRACT

IL-10 is a multifunctional cytokine that plays a crucial role in immunity and tolerance. IL-10 is produced by diverse immune cell types, including B cells and subsets of T cells. Although Th1 produce IL-10, their expression levels are much lower than Th2 cells under conventional stimulation conditions. The potential role of E26 transformation-specific 1 (Ets-1) transcription factor as a negative regulator for Il10 gene expression in CD4(+) T cells has been implicated previously. In this study, we investigated the underlying mechanism of Ets-1-mediated Il10 gene repression in Th1 cells. Compared with wild type Th1 cells, Ets-1 knockout Th1 cells expressed a significantly higher level of IL-10, which is comparable with that of wild type Th2 cells. Upregulation of IL-10 expression in Ets-1 knockout Th1 cells was accompanied by enhanced chromatin accessibility and increased recruitment of histone H3 acetylation at the Il10 regulatory regions. Reciprocally, Ets-1 deficiency significantly decreased histone deacetylase 1 (HDAC1) enrichment at the Il10 regulatory regions. Treatment with trichostatin A, an inhibitor of HDAC family, significantly increased Il10 gene expression by increasing histone H3 acetylation recruitment. We further demonstrated a physical interaction between Ets-1 and HDAC1. Coexpression of Ets-1 with HDAC1 synergistically repressed IL-10 transcription activity. In summary, our data suggest that an interaction of Ets-1 with HDAC1 represses the Il10 gene expression in Th1 cells.


Subject(s)
Down-Regulation/immunology , Gene Expression Regulation/immunology , Histone Deacetylase 1/physiology , Interleukin-10/antagonists & inhibitors , Interleukin-10/biosynthesis , Proto-Oncogene Protein c-ets-1/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Down-Regulation/genetics , HEK293 Cells , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Humans , Interleukin-10/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Protein c-ets-1/deficiency , Proto-Oncogene Protein c-ets-1/metabolism , Th1 Cells/cytology , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...