Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Biofabrication ; 15(4)2023 09 22.
Article in English | MEDLINE | ID: mdl-37659401

ABSTRACT

One of the most promising techniques for treating severe peripheral artery disease is the use of cellular tissue-engineered vascular grafts (TEVGs). This study proposes an inverse-gravity (IG) extrusion technique for creating long double-layered cellular TEVGs with diameters over 3 mm. A three-layered coaxial laminar hydrogel flow in an 8 mm-diameter pipe was realised simply by changing the extrusion direction of the hydrogel from being aligned with the direction of gravity to against it. This technique produced an extruded mixture of human aortic smooth muscle cells (HASMCs) and type-I collagen as a tubular structure with an inner diameter of 3.5 mm. After a 21 day maturation period, the maximal burst pressure, longitudinal breaking force, and circumferential breaking force of the HASMC TEVG were 416 mmHg, 0.69 N, and 0.89 N, respectively. The HASMC TEVG was endothelialised with human umbilical vein endothelial cells to form a tunica intima that simulated human vessels. Besides subcutaneous implantability on mice, the double-layered blood vessels showed a considerably lower adherence of platelets and red blood cells once exposed to heparinised mouse blood and were considered nonhaemolytic. The proposed IG extrusion technique can be applied in various fields requiring multilayered materials with large diameters.


Subject(s)
Aorta , Blood Platelets , Humans , Animals , Mice , Blood Vessel Prosthesis , Human Umbilical Vein Endothelial Cells , Hydrogels
2.
Diabetes ; 72(10): 1384-1396, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37478284

ABSTRACT

Eukaryotic translation initiation factor 2α (eIF2α) is a key mediator of the endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR). In mammals, eIF2α is phosphorylated by overnutrition-induced ER stress and is related to the development of obesity. Here, we studied the function of phosphorylated eIF2α (p-eIF2α) in agouti-related peptide (AgRP) neurons using a mouse model (AgRPeIF2αA/A) with an AgRP neuron-specific substitution from Ser 51 to Ala in eIF2α, which impairs eIF2α phosphorylation in AgRP neurons. These AgRPeIF2αA/A mice had decreases in starvation-induced AgRP neuronal activity and food intake and an increased responsiveness to leptin. Intriguingly, impairment of eIF2α phosphorylation produced decreases in the starvation-induced expression of UPR and autophagy genes in AgRP neurons. Collectively, these findings suggest that eIF2α phosphorylation regulates AgRP neuronal activity by affecting intracellular responses such as the UPR and autophagy during starvation, thereby participating in the homeostatic control of whole-body energy metabolism. ARTICLE HIGHLIGHTS: This study examines the impact of eukaryotic translation initiation factor 2α (eIF2α) phosphorylation, triggered by an energy deficit, on hypothalamic AgRP neurons and its subsequent influence on whole-body energy homeostasis. Impaired eIF2α phosphorylation diminishes the unfolded protein response and autophagy, both of which are crucial for energy deficit-induced activation of AgRP neurons. This study highlights the significance of eIF2α phosphorylation as a cellular marker indicating the availability of energy in AgRP neurons and as a molecular switch that regulates homeostatic feeding behavior.


Subject(s)
Eukaryotic Initiation Factor-2 , eIF-2 Kinase , Animals , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , eIF-2 Kinase/metabolism , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Feeding Behavior , Mammals/metabolism , Neurons/metabolism , Peptides/metabolism , Phosphorylation , Mice
3.
Autophagy ; 19(7): 2111-2142, 2023 07.
Article in English | MEDLINE | ID: mdl-36719671

ABSTRACT

There are diverse links between macroautophagy/autophagy pathways and unfolded protein response (UPR) pathways under endoplasmic reticulum (ER) stress conditions to restore ER homeostasis. Phosphorylation of EIF2S1/eIF2α is an important mechanism that can regulate all three UPR pathways through transcriptional and translational reprogramming to maintain cellular homeostasis and overcome cellular stresses. In this study, to investigate the roles of EIF2S1 phosphorylation in regulation of autophagy during ER stress, we used EIF2S1 phosphorylation-deficient (A/A) cells in which residue 51 was mutated from serine to alanine. A/A cells exhibited defects in several steps of autophagic processes (such as autophagosome and autolysosome formation) that are regulated by the transcriptional activities of the autophagy master transcription factors TFEB and TFE3 under ER stress conditions. EIF2S1 phosphorylation was required for nuclear translocation of TFEB and TFE3 during ER stress. In addition, EIF2AK3/PERK, PPP3/calcineurin-mediated dephosphorylation of TFEB and TFE3, and YWHA/14-3-3 dissociation were required for their nuclear translocation, but were insufficient to induce their nuclear retention during ER stress. Overexpression of the activated ATF6/ATF6α form, XBP1s, and ATF4 differentially rescued defects of TFEB and TFE3 nuclear translocation in A/A cells during ER stress. Consequently, overexpression of the activated ATF6 or TFEB form more efficiently rescued autophagic defects, although XBP1s and ATF4 also displayed an ability to restore autophagy in A/A cells during ER stress. Our results suggest that EIF2S1 phosphorylation is important for autophagy and UPR pathways, to restore ER homeostasis and reveal how EIF2S1 phosphorylation connects UPR pathways to autophagy.Abbreviations: A/A: EIF2S1 phosphorylation-deficient; ACTB: actin beta; Ad-: adenovirus-; ATF6: activating transcription factor 6; ATZ: SERPINA1/α1-antitrypsin with an E342K (Z) mutation; Baf A1: bafilomycin A1; BSA: bovine serum albumin; CDK4: cyclin dependent kinase 4; CDK6: cyclin dependent kinase 6; CHX: cycloheximide; CLEAR: coordinated lysosomal expression and regulation; Co-IP: coimmunoprecipitation; CTSB: cathepsin B; CTSD: cathepsin D; CTSL: cathepsin L; DAPI: 4',6-diamidino-2-phenylindole dihydrochloride; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethyl sulfoxide; DTT: dithiothreitol; EBSS: Earle's Balanced Salt Solution; EGFP: enhanced green fluorescent protein; EIF2S1/eIF2α: eukaryotic translation initiation factor 2 subunit alpha; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; ER: endoplasmic reticulum; ERAD: endoplasmic reticulum-associated degradation; ERN1/IRE1α: endoplasmic reticulum to nucleus signaling 1; FBS: fetal bovine serum; gRNA: guide RNA; GSK3B/GSK3ß: glycogen synthase kinase 3 beta; HA: hemagglutinin; Hep: immortalized hepatocyte; IF: immunofluorescence; IRES: internal ribosome entry site; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LMB: leptomycin B; LPS: lipopolysaccharide; MAP1LC3A/B/LC3A/B: microtubule associated protein 1 light chain 3 alpha/beta; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MEFs: mouse embryonic fibroblasts; MFI: mean fluorescence intensity; MTORC1: mechanistic target of rapamycin kinase complex 1; NES: nuclear export signal; NFE2L2/NRF2: NFE2 like bZIP transcription factor 2; OE: overexpression; PBS: phosphate-buffered saline; PLA: proximity ligation assay; PPP3/calcineurin: protein phosphatase 3; PTM: post-translational modification; SDS: sodium dodecyl sulfate; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; SEM: standard error of the mean; TEM: transmission electron microscopy; TFE3: transcription factor E3; TFEB: transcription factor EB; TFs: transcription factors; Tg: thapsigargin; Tm: tunicamycin; UPR: unfolded protein response; WB: western blot; WT: wild-type; Xbp1s: spliced Xbp1; XPO1/CRM1: exportin 1.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Animals , Mice , Protein Serine-Threonine Kinases/metabolism , Phosphorylation , Endoribonucleases/metabolism , Prokaryotic Initiation Factor-2/metabolism , Autophagy/genetics , Calcineurin/metabolism , Endoplasmic Reticulum-Associated Degradation , Sodium Dodecyl Sulfate/metabolism , Fibroblasts/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Microtubule-Associated Proteins/metabolism , Lysosomes/metabolism
4.
Int J Mol Sci ; 22(21)2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34769510

ABSTRACT

The autophagy-lysosome pathway is a major protein degradation pathway stimulated by multiple cellular stresses, including nutrient or growth factor deprivation, hypoxia, misfolded proteins, damaged organelles, and intracellular pathogens. Recent studies have revealed that transcription factor EB (TFEB) and transcription factor E3 (TFE3) play a pivotal role in the biogenesis and functions of autophagosome and lysosome. Here we report that three translation inhibitors (cycloheximide, lactimidomycin, and rocaglamide A) can facilitate the nuclear translocation of TFEB/TFE3 via dephosphorylation and 14-3-3 dissociation. In addition, the inhibitor-mediated TFEB/TFE3 nuclear translocation significantly increases the transcriptional expression of their downstream genes involved in the biogenesis and function of autophagosome and lysosome. Furthermore, we demonstrated that translation inhibition increased autophagosome biogenesis but impaired the degradative autolysosome formation because of lysosomal dysfunction. These results highlight the previously unrecognized function of the translation inhibitors as activators of TFEB/TFE3, suggesting a novel biological role of translation inhibition in autophagy regulation.


Subject(s)
Autophagosomes/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Protein Biosynthesis , Animals , Autophagy/physiology , Cells, Cultured , Humans
5.
Metabolites ; 11(5)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922080

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a serious lung disease characterized by excessive collagen matrix deposition and extracellular remodeling. Signaling pathways mediated by fibrotic cytokine transforming growth factor ß1 (TGF-ß1) make important contributions to pulmonary fibrosis, but it remains unclear how TGF-ß1 alters metabolism and modulates the activation and differentiation of pulmonary fibroblasts. We found that TGF-ß1 lowers NADH and NADH/NAD levels, possibly due to changes in the TCA cycle, resulting in reductions in the ATP level and oxidative phosphorylation in pulmonary fibroblasts. In addition, we showed that butyrate (C4), a short chain fatty acid (SCFA), exhibits potent antifibrotic activity by inhibiting expression of fibrosis markers. Butyrate treatment inhibited mitochondrial elongation in TGF-ß1-treated lung fibroblasts and increased the mitochondrial membrane potential (MMP). Consistent with the mitochondrial observations, butyrate significantly increased ADP, ATP, NADH, and NADH/NAD levels in TGF-ß1-treated pulmonary fibroblasts. Collectively, our findings indicate that TGF-ß1 induces changes in mitochondrial dynamics and energy metabolism during myofibroblast differentiation, and that these changes can be modulated by butyrate, which enhances mitochondrial function.

6.
Biochem Biophys Res Commun ; 558: 1-7, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33894672

ABSTRACT

ATF6 has two isoforms, ATF6α and ATF6ß, which are ubiquitously expressed type II transmembrane glycoproteins in the endoplasmic reticulum (ER). While the regulatory mechanisms and transcriptional roles of ATF6α in response to ER stress have been well-studied, those of its paralogue ATF6ß are less understood. Moreover, there is no specific cell-based reporter assay to monitor ATF6ß activation. Here, we developed a new cell-based reporter system that can monitor activation of endogenous ATF6ß. This system expresses a chimeric protein containing a synthetic transcription factor followed by the transmembrane domain and C-terminal luminal domain of ATF6ß. Under ER stress conditions, the chimeric protein was cleaved by regulated intramembrane proteolysis (RIP) to liberate the N-terminal synthetic transcription factor, which induced luciferase expression in the HeLa Luciferase Reporter cell line. This new stable reporter cell line will be an innovative tool to investigate RIP of ATF6ß.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum Stress/physiology , Activating Transcription Factor 6/chemistry , Activating Transcription Factor 6/genetics , Cell Line , Dithiothreitol/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Genes, Reporter , Humans , Luciferases, Firefly/chemistry , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Proprotein Convertases/antagonists & inhibitors , Protein Domains , Pyrrolidines/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Gut ; 70(10): 1954-1964, 2021 10.
Article in English | MEDLINE | ID: mdl-33208407

ABSTRACT

OBJECTIVE: Lipotoxic hepatocyte injury is a primary event in non-alcoholic steatohepatitis (NASH), but the mechanisms of lipotoxicity are not fully defined. Sphingolipids and free cholesterol (FC) mediate hepatocyte injury, but their link in NASH has not been explored. We examined the role of free cholesterol and sphingomyelin synthases (SMSs) that generate sphingomyelin (SM) and diacylglycerol (DAG) in hepatocyte pyroptosis, a specific form of programmed cell death associated with inflammasome activation, and NASH. DESIGN: Wild-type C57BL/6J mice were fed a high fat and high cholesterol diet (HFHCD) to induce NASH. Hepatic SMS1 and SMS2 expressions were examined in various mouse models including HFHCD-fed mice and patients with NASH. Pyroptosis was estimated by the generation of the gasdermin-D N-terminal fragment. NASH susceptibility and pyroptosis were examined following knockdown of SMS1, protein kinase Cδ (PKCδ), or the NLR family CARD domain-containing protein 4 (NLRC4). RESULTS: HFHCD increased the hepatic levels of SM and DAG while decreasing the level of phosphatidylcholine. Hepatic expression of Sms1 but not Sms2 was higher in mouse models and patients with NASH. FC in hepatocytes induced Sms1 expression, and Sms1 knockdown prevented HFHCD-induced NASH. DAG produced by SMS1 activated PKCδ and NLRC4 inflammasome to induce hepatocyte pyroptosis. Depletion of Nlrc4 prevented hepatocyte pyroptosis and the development of NASH. Conditioned media from pyroptotic hepatocytes activated the NOD-like receptor family pyrin domain containing 3 inflammasome (NLRP3) in Kupffer cells, but Nlrp3 knockout mice were not protected against HFHCD-induced hepatocyte pyroptosis. CONCLUSION: SMS1 mediates hepatocyte pyroptosis through a novel DAG-PKCδ-NLRC4 axis and holds promise as a therapeutic target for NASH.


Subject(s)
Hepatocytes/enzymology , Non-alcoholic Fatty Liver Disease/enzymology , Pyroptosis , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
8.
Autophagy ; 17(3): 761-778, 2021 03.
Article in English | MEDLINE | ID: mdl-32167007

ABSTRACT

Lysosomal Ca2+ contributes to macroautophagy/autophagy, an intracellular process for the degradation of cytoplasmic material and organelles in the lysosomes to protect cells against stress responses. TMBIM6 (transmembrane BAX inhibitor motif containing 6) is a Ca2+ channel-like protein known to regulate ER stress response and apoptosis. In this study, we examined the as yet unknown role of TMBIM6 in regulating lysosomal Ca2+ levels. The Ca2+ efflux from the ER through TMBIM6 was found to increase the resting lysosomal Ca2+ level, in which ITPR-independent regulation of Ca2+ status was observed. Further, TMBIM6 regulated the local release of Ca2+ through lysosomal MCOLN1/TRPML1 channels under nutrient starvation or MTOR inhibition. The local Ca2+ efflux through MCOLN1 channels was found to activate PPP3/calcineurin, triggering TFEB (transcription factor EB) nuclear translocation, autophagy induction, and lysosome biogenesis. Upon genetic inactivation of TMBIM6, lysosomal Ca2+ and the associated TFEB nuclear translocation were decreased. Furthermore, autophagy flux was significantly enhanced in the liver or kidney from starved Tmbim6+/+ mice compared with that in the counter tmbim6-/- mice. Together, our observations indicated that under stress conditions, TMBIM6 increases lysosomal Ca2+ release, leading to PPP3/calcineurin-mediated TFEB activation and subsequently enhanced autophagy. Thus, TMBIM6, an ER membrane protein, is suggested to be a lysosomal Ca2+ modulator that coordinates with autophagy to alleviate metabolism stress.Abbreviations: AVs: autophagic vacuoles; CEPIA: calcium-measuring organelle-entrapped protein indicator; ER: endoplasmic reticulum; GPN: glycyl-L-phenylalanine-beta-naphthylamide; ITPR/IP3R: inositol 1,4,5-trisphosphate receptor; LAMP1: lysosomal associated membrane protein 1; MCOLN/TRPML: mucolipin; MEF: mouse embryonic fibroblast; ML-SA1: mucolipin synthetic agonist 1; MTORC1: mechanistic target of rapamycin kinase complex 1; RPS6KB1: ribosomal protein S6 kinase B1; SQSTM1: sequestosome 1; TFEB: transcription factor EB; TKO: triple knockout; TMBIM6/BI-1: transmembrane BAX inhibitor motif containing 6.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Apoptosis/physiology , Autophagosomes/metabolism , Calcineurin/metabolism , Fibroblasts/metabolism , Humans , Lysosomes/genetics
9.
Biofabrication ; 12(4): 045033, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32970614

ABSTRACT

Human umbilical vein endothelial cells (HUVECs) and human aortic smooth muscle cells (HASMCs) were coaxially and continuously extruded without ultraviolet illumination using a microfluidic-based nozzle. Type I collagen (3 mg ml-1) containing HUVECs and a crosslinking reagent (100 mM CaCl2) were supplied as the core material. A mixture of 3 mg ml-1 of type I collagen (25%) and 1.8% weight volume-1 of sodium alginate (75%) was provided as the shell layer material surrounding the core material. The HUVECs were well proliferated at the core and reshaped into a monolayer formation along the axial direction of the scaffold. The HASMCs showed more than 90% cell viability in the shell layer. Fluorescent beads were passed through the inside channel of the scaffold with the HUVEC core and HASMC shell using an in-house connector. This double-layered scaffold showed higher angiogenesis in growth factor-free medium than the scaffold with only a HUVEC core. The HASMCs in the shell layer affected angiogenesis, extracellular matrix secretion, and outer diameter. The proposed technique could be applied to three-dimensional bioprinting for the production of high-volume vascularised tissue.


Subject(s)
Bioprinting , Lighting , Tissue Scaffolds , Human Umbilical Vein Endothelial Cells , Humans , Printing, Three-Dimensional , Tissue Engineering
10.
Cell Struct Funct ; 45(1): 65-76, 2020 Jun 04.
Article in English | MEDLINE | ID: mdl-32350191

ABSTRACT

It is often assumed that α-subunit phosphorylation of the eukaryotic translation initiation factor 2 (eIF2) complex is just a mechanism to control protein synthesis. However, eIF2α phosphorylation induced by multiple kinases can recognize various intracellular and extracellular stress conditions, and it is involved in various other cellular processes beyond protein synthesis. This review introduces the roles of eIF2α phosphorylation in translational regulation, the generation of reactive oxygen species, changes in mitochondria structure and shape, and mitochondrial retrograde signaling pathways in response to diverse stress conditions.Key words: eIF2α phosphorylation, Translation, Unfolded Protein Response, Reactive Oxygen Species, Mitochondria.


Subject(s)
Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis/physiology , Stress, Physiological/physiology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Eukaryotic Initiation Factor-2/genetics , Humans , Phosphorylation
11.
Mol Cells ; 43(3): 264-275, 2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32150794

ABSTRACT

Reactive oxygen species (ROS) play a significant role in intracellular signaling and regulation, particularly when they are maintained at physiologic levels. However, excess ROS can cause cell damage and induce cell death. We recently reported that eIF2α phosphorylation protects hepatocytes from oxidative stress and liver fibrosis induced by fructose metabolism. Here, we found that hepatocyte-specific eIF2α phosphorylation-deficient mice have significantly reduced expression of the epidermal growth factor receptor (EGFR) and altered EGFR-mediated signaling pathways. EGFR-mediated signaling pathways are important for cell proliferation, differentiation, and survival in many tissues and cell types. Therefore, we studied whether the reduced amount of EGFR is responsible for the eIF2α phosphorylationdeficient hepatocytes' vulnerability to oxidative stress. ROS such as hydrogen peroxide and superoxides induce both EGFR tyrosine phosphorylation and eIF2α phosphorylation. eIF2α phosphorylation-deficient primary hepatocytes, or EGFR knockdown cells, have decreased ROS scavenging ability compared to normal cells. Therefore, these cells are particularly susceptible to oxidative stress. However, overexpression of EGFR in these eIF2α phosphorylationdeficient primary hepatocytes increased ROS scavenging ability and alleviated ROS-mediated cell death. Therefore, we hypothesize that the reduced EGFR level in eIF2α phosphorylation-deficient hepatocytes is one of critical factors responsible for their susceptibility to oxidative stress.


Subject(s)
ErbB Receptors/metabolism , Eukaryotic Initiation Factor-2/metabolism , Hepatocytes/metabolism , Oxidative Stress/physiology , Animals , Cell Line , Epidermal Growth Factor/metabolism , HEK293 Cells , Humans , Mice , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction
12.
FEBS J ; 287(10): 2055-2069, 2020 05.
Article in English | MEDLINE | ID: mdl-32167239

ABSTRACT

Endoplasmic reticulum (ER) stress-induced cell death of vascular smooth muscle cells (VSMCs) is extensively involved in atherosclerotic plaque stabilization. We previously reported that nucleotide-binding oligomerization domain protein 2 (NOD2) participated in vascular homeostasis and tissue injury. However, the role and underlying mechanisms of NOD2 remain unknown in ER stress-induced cell death of VSMC during vascular diseases, including advanced atherosclerosis. Here, we report that NOD2 specifically interacted with ER stress sensor activating transcription factor 6 (ATF6) and suppressed the expression of proapoptotic transcription factor CHOP (C/EBP homologous protein) during ER stress. CHOP-positive cells were increased in neointimal lesions after femoral artery injury in NOD2-deficient mice. In particular, a NOD2 ligand, MDP, and overexpression of NOD2 decreased CHOP expression in wild-type VSMCs. NOD2 interacted with an ER stress sensor molecule, ATF6, and acted as a negative regulator for ATF6 activation and its downstream target molecule, CHOP, that regulates ER stress-induced apoptosis. Moreover, NOD2 deficiency promoted disruption of advanced atherosclerotic lesions and CHOP expression in NOD2-/- ApoE-/- mice. Our findings indicate an unsuspected critical role for NOD2 in ER stress-induced cell death.


Subject(s)
Activating Transcription Factor 6/genetics , Atherosclerosis/genetics , Nod2 Signaling Adaptor Protein/genetics , Transcription Factor CHOP/genetics , Animals , Apolipoproteins E/genetics , Apoptosis/genetics , Atherosclerosis/pathology , Cell Death/genetics , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Stress/genetics , Humans , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology
13.
Mol Cells ; 42(11): 783-793, 2019 Nov 30.
Article in English | MEDLINE | ID: mdl-31707777

ABSTRACT

When endoplasmic reticulum (ER) functions are perturbed, the ER induces several signaling pathways called unfolded protein response to reestablish ER homeostasis through three ER transmembrane proteins: inositol-requiring enzyme 1 (IRE1), PKR-like ER kinase (PERK), and activating transcription factor 6 (ATF6). Although it is important to measure the activity of ATF6 that can indicate the status of the ER, no specific cell-based reporter assay is currently available. Here, we report a new cell-based method for monitoring ER stress based on the cleavage of ATF6α by sequential actions of proteases at the Golgi apparatus during ER stress. A new expressing vector was constructed by using fusion gene of GAL4 DNA binding domain (GAL4DBD) and activation domain derived from herpes simplex virus VP16 protein (VP16AD) followed by a human ATF6α N-terminal deletion variant. During ER stress, the GAL4DBD-VP16AD(GV)-hATF6α deletion variant was cleaved to liberate active transcription activator encompassing GV-hATF6α fragment which could translocate into the nucleus. The translocated GV-hATF6α fragment strongly induced the expression of firefly luciferase in HeLa Luciferase Reporter cell line containing a stably integrated 5X GAL4 site-luciferase gene. The established double stable reporter cell line HLR-GV-hATF6α(333) represents an innovative tool to investigate regulated intramembrane proteolysis of ATF6α. It can substitute active pATF6(N) binding motif-based reporter cell lines.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum/metabolism , Luciferases/metabolism , Membrane Proteins/metabolism , Unfolded Protein Response , eIF-2 Kinase/metabolism , Activating Transcription Factor 6/genetics , Cell Line, Tumor , Dithiothreitol/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gene Expression Regulation/drug effects , Genes, Reporter/genetics , HeLa Cells , Humans , Luciferases/genetics , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Signal Transduction/drug effects , Signal Transduction/genetics , eIF-2 Kinase/genetics
14.
Metabolites ; 9(9)2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31466271

ABSTRACT

Short chain fatty acids (SCFAs) are the main products of dietary fibers that are not digested by the human body, and they have been shown to affect human metabolism and inflammation. The amount of SCFAs in the body is related to many human diseases, and studies have focused on elucidating their roles and target molecules in both metabolic and immune responses. Thus, the quantitation of SCFAs in biological samples becomes crucial in understanding their important roles in the human body. Herein, a facile profiling method of SCFAs using liquid chromatography-tandem mass spectrometry (LC-MS/MS) was developed and then applied to biological samples. C2-C6 SCFAs were derivatized while using 4-acetamido-7-mercapto-2,1,3-benzoxadiazole for 5 min. at room temperature prior to LC-MS/MS analysis, and characteristic fragmentation patterns and increased hydrophobicity after chemical derivatization enabled specific discrimination among 12 SCFAs. Derivatization was fast and reliable, and the reaction products were stable for a week at 4 °C. The developed method was applied to measure SCFAs in mouse feces, plasma, and human exhaled breath condensates. This fast and simple method can save labor and effort to profile SCFAs from various biological samples.

15.
J Inflamm (Lond) ; 16: 17, 2019.
Article in English | MEDLINE | ID: mdl-31312114

ABSTRACT

BACKGROUND: Obesity-induced skeletal muscle inflammation is a major contributor of skeletal muscle loss/atrophy and is implicated in metabolic complications such as insulin resistance. Fibroblast growth factor 21 (FGF21) is known to be an important metabolic regulator with anti-inflammatory properties. However, the effect of FGF21 on skeletal muscle atrophy is unclear. In this study, we investigated the effect of FGF21 deficiency on obesity-induced skeletal muscle inflammation and atrophy in mice. RESULTS: The expression of atrophic factors (MuRF1 and Atrogin-1) was upregulated at the mRNA and/or protein levels in the skeletal muscle of FGF21-deficient obese mice compared with wild type obese control mice. This was accompanied by an increase in levels of inflammatory cytokines (TNFα and MCP-1) and a reduction in AMPK phosphorylation. FGF21 treatment markedly suppressed TNFα-mediated inflammatory and atrophic responses in cultured myotubes, and the actions of FGF21 were blunted by the AMPK inhibitor compound C. CONCLUSION: These findings suggest that FGF21 deficiency aggravates obesity-induced inflammation and atrophic responses in the skeletal muscle of obese mice, and FGF21 may protect inflammation-mediated atrophy through the AMPK pathway.

16.
Biochim Biophys Acta Mol Cell Res ; 1866(9): 1463-1474, 2019 09.
Article in English | MEDLINE | ID: mdl-31199931

ABSTRACT

The perinuclear stacks of the Golgi apparatus maintained by dynamic microtubules are essential for cell migration. Activation of Akt (protein kinase B, PKB) negatively regulates glycogen synthase kinase 3ß (GSK3ß)-mediated tau phosphorylation, which enhances tau binding to microtubules and microtubule stability. In this study, experiments were performed on developmentally regulated GTP-binding protein 2 (DRG2)-stably knockdown HeLa cells to determine whether knockdown of DRG2 in HeLa cells treated with epidermal growth factor (EGF) affects microtubule dynamics, perinuclear Golgi stacking, and cell migration. Here, we show that DRG2 plays a key role in regulating microtubule stability, perinuclear Golgi stack formation, and cell migration. DRG2 knockdown prolonged the EGF receptor (EGFR) localization in endosome, enhanced Akt activity and inhibitory phosphorylation of GSK3ß. Tau, a target of GSK3ß, was hypo-phosphorylated in DRG2-knockdown cells and showed greater association with microtubules, resulting in microtubule stabilization. DRG2-knockdown cells showed defects in microtubule growth and microtubule organizing centers (MTOC), Golgi fragmentation, and loss of directional cell migration. These results reveal a previously unappreciated role for DRG2 in the regulation of perinuclear Golgi stacking and cell migration via its effects on GSK3ß phosphorylation, and microtubule stability.


Subject(s)
GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Golgi Apparatus/metabolism , Microtubules/metabolism , Cell Movement , Gene Knockdown Techniques , HeLa Cells , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
17.
Mol Cells ; 41(12): 1045-1051, 2018 Dec 31.
Article in English | MEDLINE | ID: mdl-30453731

ABSTRACT

The developmentally regulated GTP binding protein 2 (DRG2) is involved in the control of cell growth and differentiation. Here, we demonstrate that DRG2 regulates microtubule dynamics in HeLa cells. Analysis of live imaging of the plus-ends of microtubules with EB1-EGFP showed that DRG2 deficiency (shDRG2) significantly reduced the growth rate of HeLa cells. Depletion of DRG2 increased 'slow and long-lived' subpopulations, but decreased 'fast and short-lived' subpopulations of microtubules. Microtubule polymerization inhibitor exhibited a reduced response in shDRG2 cells. Using immunoprecipitation, we show that DRG2 interacts with tau, which regulates microtubule polymerization. Collectively, these data demonstrate that DRG2 may aid in affecting microtubule dynamics in HeLa cells.


Subject(s)
GTP-Binding Proteins/deficiency , Microtubules/metabolism , Cell Proliferation/physiology , Gene Knockdown Techniques , HeLa Cells , Humans , Phosphorylation , Transfection , tau Proteins/metabolism
18.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 4198-4201, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30441280

ABSTRACT

By using the microfluidic spinning technology we generated tiny hydrogel tubular scaffolds. Fibroblast (NIH/3T3) cell cultures were performed for seventeen days to demonstrate the potential of cell attachment on surfaces and encapsulation in the wall of he microscopic scaffolds for blood vessel-like structure forming. Over theculture period, the NIH/3T3 confluence reached around 80\%, and 100\% on the inside and outside scaffolds' surface respectively while cells proliferated and coalesced in cell group in the hydrogel wall. These results could further be applied to endothelial co-culturing for forming engineered blood vessel.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Animals , Coculture Techniques , Humans , Male , Mice
19.
Bone ; 116: 279-289, 2018 11.
Article in English | MEDLINE | ID: mdl-30144578

ABSTRACT

The autophagy pathway has been suggested to influence skeletal structure by modulating bone metabolism. Recent findings suggest that microRNAs (miR) play a critical role in autophagy. We hypothesized that inflammation induces miR-155, which enhances autophagy in osteoclasts (OC), leading to inflammatory bone loss. The expression of miR-155 was elevated in tibiae from LPS-injected mice and in OC stimulated by lipopolysaccharide (LPS) compared with vehicle treatment. Overexpression of miR-155 enhanced autophagy as well as differentiation in OC, whereas inhibition of endogenous miR-155 decreased both. Transforming growth factor ß-activated kinase 1-binding protein 2 (TAB2) was identified as a target gene of miR-155 via binding to the 3'-UTR of TAB2, which directly interacts with BECLIN1. BECLIN1 was dissociated from TAB2, which started to associate with TAK1 when autophagy was induced. Our data demonstrate that LPS-induced miR-155 promoted autophagy to increase OC formation via decreased TAB2.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Lipopolysaccharides/pharmacology , MicroRNAs/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , 3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Female , Mice, Inbred C57BL , MicroRNAs/genetics
20.
Nutr Metab (Lond) ; 14: 48, 2017.
Article in English | MEDLINE | ID: mdl-28781602

ABSTRACT

BACKGROUND: Dietary fructose can rapidly cause fatty liver in animals through de novo lipogenesis (DNL) and contribute to the development and severity of nonalcoholic fatty liver disease (NAFLD). In response to diverse cellular insults including endoplasmic reticulum (ER) and oxidative stress, phosphorylation of the eukaryotic translation initiation factor 2 alpha subunit (eIF2α) attenuates general translation initiation, allowing cells to conserve resources and initiate adaptive gene expression to restore homeostasis. The present study aimed to investigate the role of eIF2α phosphorylation in protecting against NAFLD induced by high fructose ingestion in a hepatocyte-specific eIF2α-phosphorylation-deficient mouse model. METHODS: Hepatocyte-specific non-phosphorylatable (S51A) eIF2α knock-in (A/A;fTg/0;CreHep/0, A/AHep ) mice were generated by crossing A/A;fTg/fTg mice with the floxed WT eIF2α transgene (fTg) with Alfp-Cre recombinase transgenic S/A;CreHep/0 (S/A-CreHep ) mice. Hepatocyte-specific eIF2α-phosphorylation-deficient 3-month-old mice or 12-month-old mice were fed a 60% high fructose diet (HFrD) for 16 or 5 wks, and the effects of eIF2α-phosphorylation deficiency on NADP/NADPH and GSSG/GSH levels, ROS-defense gene expression, oxidative damage, cell death, and fibrosis were observed. RESULTS: Prolonged fructose feeding to mice caused dysregulation of the unfolded protein response (UPR) sensor activation and UPR gene expression, and then led to decreased expression of several ROS defense genes including glutathione biogenesis genes. Nonetheless, these changes were not sufficient to induce the death of eIF2α phosphorylation-sufficient hepatocytes. However, there was a substantial increase in hepatocyte death and liver fibrosis in fructose-fed middle-aged mice deficient in hepatocyte-specific eIF2α phosphorylation because of diminished antioxidant capacity due to reduced expression of antioxidant enzymes (GPX1 and HO-1) and lower NADPH and glutathione levels, as well as a possible increase in ROS-induced damage from infiltrating NOX2-expressing leukocytes; all this led to a vicious cycle of hepatocyte death and leukocyte infiltration. CONCLUSION: Our findings suggest that eIF2α phosphorylation maintains NADPH and GSH levels and controls the expression of ROS-defense genes, thereby protecting hepatocytes from oxidative stresses induced by fructose metabolism.

SELECTION OF CITATIONS
SEARCH DETAIL
...