Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Genes Dev ; 31(14): 1417-1438, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28860159

ABSTRACT

Numerous environmental, physiological, and pathological insults disrupt protein-folding homeostasis in the endoplasmic reticulum (ER), referred to as ER stress. Eukaryotic cells evolved a set of intracellular signaling pathways, collectively termed the unfolded protein response (UPR), to maintain a productive ER protein-folding environment through reprogramming gene transcription and mRNA translation. The UPR is largely dependent on transcription factors (TFs) that modulate expression of genes involved in many physiological and pathological conditions, including development, metabolism, inflammation, neurodegenerative diseases, and cancer. Here we summarize the current knowledge about these mechanisms, their impact on physiological/pathological processes, and potential therapeutic applications.


Subject(s)
Transcription Factors/metabolism , Unfolded Protein Response/genetics , Disease/genetics , Endoplasmic Reticulum Stress , Humans , Protein Biosynthesis , Transcription Factors/antagonists & inhibitors , Transcriptional Activation
2.
J Biol Chem ; 299(2): 102821, 2023 02.
Article in English | MEDLINE | ID: mdl-36563857

ABSTRACT

Tauopathies are neurodegenerative diseases caused by pathologic misfolded tau protein aggregation in the nervous system. Population studies implicate EIF2AK3 (eukaryotic translation initiation factor 2 alpha kinase 3), better known as PERK (protein kinase R-like endoplasmic reticulum kinase), as a genetic risk factor in several tauopathies. PERK is a key regulator of intracellular proteostatic mechanisms-unfolded protein response and integrated stress response. Previous studies found that tauopathy-associated PERK variants encoded functional hypomorphs with reduced signaling in vitro. But, it remained unclear how altered PERK activity led to tauopathy. Here, we chemically or genetically modulated PERK signaling in cell culture models of tau aggregation and found that PERK pathway activation prevented tau aggregation, whereas inhibition exacerbated tau aggregation. In primary tauopathy patient brain tissues, we found that reduced PERK signaling correlated with increased tau neuropathology. We found that tauopathy-associated PERK variants targeted the endoplasmic reticulum luminal domain; and two of these variants damaged hydrogen bond formation. Our studies support that PERK activity protects against tau aggregation and pathology. This may explain why people carrying hypomorphic PERK variants have increased risk for developing tauopathies. Finally, our studies identify small-molecule augmentation of PERK signaling as an attractive therapeutic strategy to treat tauopathies by preventing tau pathology.


Subject(s)
Protein Aggregates , Protein Aggregation, Pathological , eIF-2 Kinase , tau Proteins , Humans , Disease Susceptibility , eIF-2 Kinase/chemistry , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Mutation , Risk Factors , tau Proteins/chemistry , tau Proteins/metabolism , Tauopathies/metabolism , Tauopathies/pathology
3.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062980

ABSTRACT

Mitochondrial stress, resulting from dysfunction and proteostasis disturbances, triggers the mitochondrial unfolded protein response (UPRMT), which activates gene encoding chaperones and proteases to restore mitochondrial function. Although ATFS-1 mediates mitochondrial stress UPRMT induction in C. elegans, the mechanisms relaying mitochondrial stress signals to the nucleus in mammals remain poorly defined. Here, we explored the role of protein kinase R (PKR), an eIF2α kinase activated by double-stranded RNAs (dsRNAs), in mitochondrial stress signaling. We found that UPRMT does not occur in cells lacking PKR, indicating its crucial role in this process. Mechanistically, we observed that dsRNAs accumulate within mitochondria under stress conditions, along with unprocessed mitochondrial transcripts. Furthermore, we demonstrated that accumulated mitochondrial dsRNAs in mouse embryonic fibroblasts (MEFs) deficient in the Bax/Bak channels are not released into the cytosol and do not induce the UPRMT upon mitochondrial stress, suggesting a potential role of the Bax/Bak channels in mediating the mitochondrial stress response. These discoveries enhance our understanding of how cells maintain mitochondrial integrity, respond to mitochondrial dysfunction, and communicate stress signals to the nucleus through retrograde signaling. This knowledge provides valuable insights into prospective therapeutic targets for diseases associated with mitochondrial stress.


Subject(s)
Mitochondria , RNA, Double-Stranded , Unfolded Protein Response , eIF-2 Kinase , Animals , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Mitochondria/metabolism , RNA, Double-Stranded/metabolism , Mice , Stress, Physiological , Signal Transduction , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics , Fibroblasts/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , Humans
4.
J Biol Chem ; 298(9): 102277, 2022 09.
Article in English | MEDLINE | ID: mdl-35863436

ABSTRACT

La-related protein 1 (LARP1) has been identified as a key translational inhibitor of terminal oligopyrimidine (TOP) mRNAs downstream of the nutrient sensing protein kinase complex, mTORC1. LARP1 exerts this inhibitory effect on TOP mRNA translation by binding to the mRNA cap and the adjacent 5'TOP motif, resulting in the displacement of the cap-binding protein eIF4E from TOP mRNAs. However, the involvement of additional signaling pathway in regulating LARP1-mediated inhibition of TOP mRNA translation is largely unexplored. In the present study, we identify a second nutrient sensing kinase GCN2 that converges on LARP1 to control TOP mRNA translation. Using chromatin-immunoprecipitation followed by massive parallel sequencing (ChIP-seq) analysis of activating transcription factor 4 (ATF4), an effector of GCN2 in nutrient stress conditions, in WT and GCN2 KO mouse embryonic fibroblasts, we determined that LARP1 is a GCN2-dependent transcriptional target of ATF4. Moreover, we identified GCN1, a GCN2 activator, participates in a complex with LARP1 on stalled ribosomes, suggesting a role for GCN1 in LARP1-mediated translation inhibition in response to ribosome stalling. Therefore, our data suggest that the GCN2 pathway controls LARP1 activity via two mechanisms: ATF4-dependent transcriptional induction of LARP1 mRNA and GCN1-mediated recruitment of LARP1 to stalled ribosomes.


Subject(s)
Amino Acids , Protein Biosynthesis , Protein Serine-Threonine Kinases , RNA 5' Terminal Oligopyrimidine Sequence , RNA, Messenger , RNA-Binding Proteins , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Animals , Cell Culture Techniques , Chromatin Immunoprecipitation , Eukaryotic Initiation Factor-4E/metabolism , Fibroblasts , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
J Biol Chem ; 295(1): 237-249, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31792031

ABSTRACT

Endoplasmic reticulum (ER) stress activates the unfolded protein response (UPR), which reduces levels of misfolded proteins. However, if ER homeostasis is not restored and the UPR remains chronically activated, cells undergo apoptosis. The UPR regulator, PKR-like endoplasmic reticulum kinase (PERK), plays an important role in promoting cell death when persistently activated; however, the underlying mechanisms are poorly understood. Here, we profiled the microRNA (miRNA) transcriptome in human cells exposed to ER stress and identified miRNAs that are selectively induced by PERK signaling. We found that expression of a PERK-induced miRNA, miR-483, promotes apoptosis in human cells. miR-483 induction was mediated by a transcription factor downstream of PERK, activating transcription factor 4 (ATF4), but not by the CHOP transcription factor. We identified the creatine kinase brain-type (CKB) gene, encoding an enzyme that maintains cellular ATP reserves through phosphocreatine production, as being repressed during the UPR and targeted by miR-483. We found that ER stress, selective PERK activation, and CKB knockdown all decrease cellular ATP levels, leading to increased vulnerability to ER stress-induced cell death. Our findings identify miR-483 as a downstream target of the PERK branch of the UPR. We propose that disruption of cellular ATP homeostasis through miR-483-mediated CKB silencing promotes ER stress-induced apoptosis.


Subject(s)
Adenosine Triphosphate/metabolism , MicroRNAs/metabolism , Unfolded Protein Response , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Apoptosis , Creatine Kinase, BB Form/genetics , Creatine Kinase, BB Form/metabolism , HEK293 Cells , HeLa Cells , Homeostasis , Humans , MicroRNAs/genetics , eIF-2 Kinase/genetics
6.
Int J Mol Sci ; 22(21)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34769253

ABSTRACT

Protein synthesis is important for maintaining cellular homeostasis under various stress responses. In this study, we screened an anticancer drug library to select compounds with translational repression functions. AZD8055, an ATP-competitive mechanistic target of rapamycin complex 1/2 (mTORC1/2) inhibitor, was selected as a translational suppressor. AZD8055 inhibited protein synthesis in mouse embryonic fibroblasts and hepatocellular carcinoma HepG2 cells. Extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) were activated during the early phase of mTORC1/2 inhibition by AZD8055 treatment. Combined treatment of AZD8055 with the MAPK kinase1/2 (MEK1/2) inhibitor refametinib or the p38 inhibitor SB203580 markedly decreased translation in HepG2 cells. Thus, the inhibition of ERK1/2 or p38 may enhance the efficacy of AZD8055-mediated inhibition of protein synthesis. In addition, AZD8055 down-regulated the phosphorylation of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), and AZD8055-induced phosphorylation of ERK1/2 and p38 had no effect on phosphorylation status of 4E-BP1. Interestingly, AZD8055 modulated the 4E-BP1 mRNA pool by up-regulating ERK1/2 and p38 pathways. Together, these results suggest that AZD8055-induced activation of MAPKs interferes with inhibition of protein synthesis at an early stage of mTORC1/2 inhibition, and that it may contribute to the development of resistance to mTORC1/2 inhibitors.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MAP Kinase Signaling System , Morpholines/pharmacology , Neoplasm Proteins/metabolism , Protein Biosynthesis/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/pathology , Enzyme Activation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/pathology
7.
Int J Mol Sci ; 21(3)2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32013014

ABSTRACT

Environmental high-temperature heat exposure is linked to physiological stress such as disturbed protein homeostasis caused by endoplasmic reticulum (ER) stress. Abnormal proteostasis in neuronal cells is a common pathological factor of Parkinson's disease (PD). Chronic heat stress is thought to induce neuronal cell death during the onset and progression of PD, but the exact role and mechanism of ER stress and the activation of the unfolded protein response (UPR) remains unclear. Here, we showed that chronic heat exposure induces ER stress mediated by the PKR-like eukaryotic initiation factor 2α kinase (PERK)/eIF2α phosphorylation signaling pathway in Drosophila neurons. Chronic heat-induced eIF2α phosphorylation was regulated by PERK activation and required for neuroprotection from chronic heat stress. Moreover, the attenuated protein synthesis by eIF2α phosphorylation was a critical factor for neuronal cell survival during chronic heat stress. We further showed that genetic downregulation of PERK, specifically in dopaminergic (DA) neurons, impaired motor activity and led to DA neuron loss. Therefore, our findings provide in vivo evidence demonstrating that chronic heat exposure may be a critical risk factor in the onset of PD, and eIF2α phosphorylation mediated by PERK may contribute to the protection of DA neurons against chronic heat stress in Drosophila.


Subject(s)
Dopaminergic Neurons/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Eukaryotic Initiation Factor-2/metabolism , eIF-2 Kinase/genetics , Animals , Down-Regulation , Drosophila melanogaster/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Heat-Shock Response , Locomotion , Male , Phosphorylation , Signal Transduction , eIF-2 Kinase/metabolism
8.
Int J Mol Sci ; 21(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731602

ABSTRACT

Stress granules are membraneless organelles composed of numerous components including ribonucleoproteins. The stress granules are characterized by a dynamic complex assembly in response to various environmental stressors, which has been implicated in the coordinated regulation of diverse biological pathways, to exert a protective role against stress-induced cell death. Here, we show that stress granule formation is induced by morusin, a novel phytochemical displaying antitumor capacity through barely known mechanisms. Morusin-mediated induction of stress granules requires activation of protein kinase R (PKR) and subsequent eIF2α phosphorylation. Notably, genetic inactivation of stress granule formation mediated by G3BP1 knockout sensitized cancer cells to morusin treatment. This protective function against morusin-mediated cell death can be attributed at least in part to the sequestration of receptors for activated C kinase-1 (RACK1) within the stress granules, which reduces caspase-3 activation. Collectively, our study provides biochemical evidence for the role of stress granules in suppressing the antitumor capacity of morusin, proposing that morusin treatment, together with pharmacological inhibition of stress granules, could be an efficient strategy for targeting cancer.


Subject(s)
Apoptosis/drug effects , Cytoplasmic Granules/metabolism , Flavonoids/pharmacology , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Receptors for Activated C Kinase/metabolism , eIF-2 Kinase/metabolism , Cytoplasmic Granules/pathology , HCT116 Cells , HeLa Cells , Humans , PC-3 Cells
9.
Curr Top Microbiol Immunol ; 414: 103-130, 2018.
Article in English | MEDLINE | ID: mdl-28900680

ABSTRACT

The endoplasmic reticulum (ER) is a cellular organelle responsible for folding of secretory and membrane proteins. Perturbance in ER homeostasis caused by various intrinsic/extrinsic stimuli challenges the protein-folding capacity of the ER, leading to an ER dysfunction, called ER stress. Cells have developed a defensive response to adapt and/or survive in the face of ER stress that may be detrimental to cell function and survival. When exposed to ER stress, the cell activates a complex and elaborate signaling network that includes translational modulation and transcriptional induction of genes. In addition to these autonomous responses, recent studies suggest that the stressed tissue secretes peptides or unknown factors that transfer the signal to other cells in the same or different organs, leading the organism as a whole to cope with challenges in a non-autonomous manner. In this review, we discuss the mechanisms by which cells adapt to ER stress challenges autonomously and transfer the stress signal to non-stressed cells in different organs.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Folding , Unfolded Protein Response/physiology , Adaptation, Physiological , Adipose Tissue/metabolism , Animals , Endoplasmic Reticulum Stress , Humans , Insulin-Secreting Cells/metabolism , Liver/metabolism , Signal Transduction/physiology
10.
Int J Mol Sci ; 20(24)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847234

ABSTRACT

Endoplasmic reticulum (ER) stress is known to influence various cellular functions, including cell cycle progression. Although it is well known how ER stress inhibits cell cycle progression at the G1 phase, the molecular mechanism underlying how ER stress induces G2/M cell cycle arrest remains largely unknown. In this study, we found that ER stress and subsequent induction of the UPR led to cell cycle arrest at the G2/M phase by reducing the amount of cyclin B1. Pharmacological inhibition of the IRE1α or ATF6α signaling did not affect ER stress-induced cell cycle arrest at the G2/M phase. However, when the alpha subunit of eukaryotic translation initiation factor 2 (eIF2α) phosphorylation was genetically abrogated, the cell cycle progressed without arresting at the G2/M phase after ER stress. GEO database analysis showed that growth arrest and DNA-damage-inducible protein α (Gadd45α) were induced in an eIF2a phosphorylation-dependent manner, which was confirmed in this study. Knockdown of GADD45α abrogated cell cycle arrest at the G2/M phase upon ER stress. Finally, the cell death caused by ER stress significantly reduced when GADD45α expression was knocked down. In conclusion, GADD45α is a key mediator of ER stress-induced growth arrest via regulation of the G2/M transition and cell death through the eIF2α signaling pathway.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Division , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , G2 Phase Cell Cycle Checkpoints , Signal Transduction , Cell Cycle Proteins/genetics , Cyclin B1/genetics , Cyclin B1/metabolism , Databases, Genetic , Eukaryotic Initiation Factor-2/genetics , Humans , Phosphorylation
11.
Int J Mol Sci ; 20(7)2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30925759

ABSTRACT

Hepatic intrasinusoidal (HI) natural killer (NK) cells from liver perfusate have unique features that are similar to those of liver-resident NK cells. Previously, we have reported that HI CD56bright NK cells effectively degranulate against SNU398 hepatocellular carcinoma (HCC) cells. Thus, the aim of this study was to further investigate the phenotype and function of HI NK cells. We found that HI CD56bright NK cells degranulated much less to Huh7 cells. HI CD56bright NK cells expressed NKG2D, NKp46, TNF-related apoptosis-inducing ligand (TRAIL), and FAS ligand (FASL) at higher levels than CD56dim cells. SNU398 cells expressed more NKG2D ligands and FAS and less PD-L1 than Huh7 cells. Blockade of NKG2D, TRAIL, and FASL significantly reduced the cytotoxicity of HI NK cells against SNU398 cells, but blockade of PD-L1 did not lead to any significant change. However, HI NK cells produced IFN-γ well in response to Huh7 cells. In conclusion, the cytotoxicity of HI CD56bright NK cells was attributed to the expression of NKG2D, TRAIL, and FASL. The results suggest the possible use of HI NK cells for cancer immunotherapy and prescreening of HCC cells to help identify the most effective NK cell therapy recipients.


Subject(s)
CD56 Antigen/immunology , Carcinoma, Hepatocellular/immunology , Killer Cells, Natural/immunology , Liver Neoplasms/immunology , Adult , CD56 Antigen/analysis , Cell Line, Tumor , Fas Ligand Protein/analysis , Fas Ligand Protein/immunology , Female , GPI-Linked Proteins/analysis , GPI-Linked Proteins/immunology , Humans , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/immunology , Interferon-gamma/analysis , Interferon-gamma/immunology , Male , TNF-Related Apoptosis-Inducing Ligand/analysis , TNF-Related Apoptosis-Inducing Ligand/immunology
12.
PLoS Biol ; 13(10): e1002277, 2015 10.
Article in English | MEDLINE | ID: mdl-26469762

ABSTRACT

Although glucose uniquely stimulates proinsulin biosynthesis in ß cells, surprisingly little is known of the underlying mechanism(s). Here, we demonstrate that glucose activates the unfolded protein response transducer inositol-requiring enzyme 1 alpha (IRE1α) to initiate X-box-binding protein 1 (Xbp1) mRNA splicing in adult primary ß cells. Using mRNA sequencing (mRNA-Seq), we show that unconventional Xbp1 mRNA splicing is required to increase and decrease the expression of several hundred mRNAs encoding functions that expand the protein secretory capacity for increased insulin production and protect from oxidative damage, respectively. At 2 wk after tamoxifen-mediated Ire1α deletion, mice develop hyperglycemia and hypoinsulinemia, due to defective ß cell function that was exacerbated upon feeding and glucose stimulation. Although previous reports suggest IRE1α degrades insulin mRNAs, Ire1α deletion did not alter insulin mRNA expression either in the presence or absence of glucose stimulation. Instead, ß cell failure upon Ire1α deletion was primarily due to reduced proinsulin mRNA translation primarily because of defective glucose-stimulated induction of a dozen genes required for the signal recognition particle (SRP), SRP receptors, the translocon, the signal peptidase complex, and over 100 other genes with many other intracellular functions. In contrast, Ire1α deletion in ß cells increased the expression of over 300 mRNAs encoding functions that cause inflammation and oxidative stress, yet only a few of these accumulated during high glucose. Antioxidant treatment significantly reduced glucose intolerance and markers of inflammation and oxidative stress in mice with ß cell-specific Ire1α deletion. The results demonstrate that glucose activates IRE1α-mediated Xbp1 splicing to expand the secretory capacity of the ß cell for increased proinsulin synthesis and to limit oxidative stress that leads to ß cell failure.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Adolescent , Adult , Animals , Cells, Cultured , Crosses, Genetic , DNA-Binding Proteins/genetics , Endoribonucleases/genetics , Female , Humans , Hyperglycemia/blood , Hyperglycemia/pathology , Insulin Secretion , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/ultrastructure , Male , Mice, Knockout , Mice, Transgenic , Middle Aged , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction , Tissue Donors , Transcription Factors/genetics , X-Box Binding Protein 1 , Young Adult
13.
Biochem Biophys Res Commun ; 493(2): 957-963, 2017 11 18.
Article in English | MEDLINE | ID: mdl-28942141

ABSTRACT

Low dose of carbon monoxide (CO) has anti-inflammatory role through various signaling pathways. Cellular metabolism has been implicated in the activation of inflammation in immune cells. However, the mechanisms by which CO-dependent metabolic regulation affect the immune response remain unclear. Here we show that CO-dependent metabolic pathway regulates the activation of the nucleotide-binding domain, leucine-rich-repeat-containing receptor (NLR), pyrin-domain-containing 3 (NLRP3) inflammasome. CO-releasing molecule-3 (CORM-3) resulted in reduced glycolysis-dependent NLRP3 inflammasome activation in macrophages. The reduced mTORC1 activation by CORM-3 resulted in less glycolysis during NLRP3 inflammasome activation. CORM-3 suppressed caspase-1 activation and the secretion of interleukin (IL)-1ß and IL-18 in macrophages in response to lipopolysaccharide (LPS) and ATP. Moreover, CORM-3 inhibits the oligomerization of the adaptor protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), which is required for NLRP3-dependent caspase-1 activation. Furthermore, CORM-3-treated mice showed substantial reduction in IL-1ß production by hyperglycemia in a mouse model of streptozotocin (STZ)-induced diabetes. Our results suggest that CO regulates glycolysis-dependent NLRP3 inflammasome activation and may provide a therapeutic approach for inflammation in metabolic diseases.


Subject(s)
Carbon Monoxide/immunology , Inflammasomes/immunology , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Glycolysis/drug effects , Hyperglycemia/complications , Hyperglycemia/drug therapy , Hyperglycemia/immunology , Inflammation/drug therapy , Inflammation/immunology , Macrophages/drug effects , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes/immunology , Organometallic Compounds/pharmacology , Organometallic Compounds/therapeutic use , TOR Serine-Threonine Kinases/immunology
14.
Circ Res ; 116(11): 1736-43, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25872946

ABSTRACT

RATIONALE: Myeloid-derived C/EBP-homologous protein (CHOP), an effector of the endoplasmic reticulum stress-induced unfolded protein response, promotes macrophage apoptosis in advanced atherosclerosis, but the role of CHOP in vascular smooth muscle cells (VSMCs) in atherosclerosis is not known. OBJECTIVE: To investigate the role of CHOP in SM22α(+) VSMCs in atherosclerosis. METHODS AND RESULTS: Chop(fl/fl) mice were generated and crossed into the Apoe(-/-) and SM22α-CreKI(+) backgrounds. SM22α-CreKI causes deletion of floxed genes in adult SMCs. After 12 weeks of Western-type diet feeding, the content of α-actin-positive cells in aortic root lesions was decreased in Chop(fl/fl)SM22α-CreKI(+)Apoe(-/-) versus control Chop(fl/fl)Apoe(-/-) mice, and aortic explant-derived VSMCs from the VSMC-CHOP-deficient mice displayed reduced proliferation. Krüppel-like factor 4 (KLF4), a key suppressor of VSMC proliferation, was increased in lesions and aortic VSMCs from Chop(fl/fl)SM22α-CreKI(+)Apoe(-/-) mice, and silencing Klf4 in CHOP-deficient VSMCs restored proliferation. CHOP deficiency in aortic VSMCs increased KLF4 through 2 mechanisms mediated by the endoplasmic reticulum stress effector activating transcription factor 4: transcriptional induction of Klf4 mRNA and decreased proteasomal degradation of KLF4 protein. CONCLUSIONS: These findings in SM22α-CHOP-deficient mice imply that CHOP expression in SM22α(+) VSMCs promotes cell proliferation by downregulating KLF4. The mechanisms involve newly discovered roles of CHOP in the transcriptional and post-translational regulation of KLF4.


Subject(s)
Atherosclerosis/metabolism , Cell Proliferation , Myocytes, Smooth Muscle/metabolism , Transcription Factor CHOP/deficiency , Actins/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Aorta/cytology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Immunoblotting , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/cytology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques , Transcription Factor CHOP/genetics
15.
PLoS Genet ; 10(5): e1004348, 2014 May.
Article in English | MEDLINE | ID: mdl-24809345

ABSTRACT

The endoplasmic reticulum (ER) responds to changes in intracellular homeostasis through activation of the unfolded protein response (UPR). Yet, it is not known how UPR-signaling coordinates adaptation versus cell death. Previous studies suggested that signaling through PERK/ATF4 is required for cell death. We show that high levels of ER stress (i.e., ischemia-like conditions) induce transcription of the ubiquitin ligases Siah1/2 through the UPR transducers PERK/ATF4 and IRE1/sXBP1. In turn, Siah1/2 attenuates proline hydroxylation of ATF4, resulting in its stabilization, thereby augmenting ER stress output. Conversely, ATF4 activation is reduced upon Siah1/2 KD in cultured cells, which attenuates ER stress-induced cell death. Notably, Siah1a(+/-)::Siah2(-/-) mice subjected to neuronal ischemia exhibited smaller infarct volume and were protected from ischemia-induced death, compared with the wild type (WT) mice. In all, Siah1/2 constitutes an obligatory fine-tuning mechanism that predisposes cells to death under severe ER stress conditions.


Subject(s)
Isoenzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Endoplasmic Reticulum/metabolism , Enzyme Activation , Humans , Mice , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stress, Physiological , Transcription, Genetic , Ubiquitin-Protein Ligases/genetics
16.
J Lipid Res ; 57(8): 1329-38, 2016 08.
Article in English | MEDLINE | ID: mdl-27146479

ABSTRACT

The endoplasmic reticulum (ER) is a cellular organelle important for regulating calcium homeostasis, lipid metabolism, protein synthesis, and posttranslational modification and trafficking. Numerous environmental, physiological, and pathological insults disturb ER homeostasis, referred to as ER stress, in which a collection of conserved intracellular signaling pathways, termed the unfolded protein response (UPR), are activated to maintain ER function for cell survival. However, excessive and/or prolonged UPR activation leads to initiation of self-destruction through apoptosis. Excessive accumulation of lipids and their intermediate products causes metabolic abnormalities and cell death, called lipotoxicity, in peripheral organs, including the pancreatic islets, liver, muscle, and heart. Because accumulating evidence links chronic ER stress and defects in UPR signaling to lipotoxicity in peripheral tissues, understanding the role of ER stress in cell physiology is a topic under intense investigation. In this review, we highlight recent findings that link ER stress and UPR signaling to the pathogenesis of peripheral organs due to lipotoxicity.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Lipid Metabolism , Animals , Apoptosis , Humans , Liver/metabolism , Liver/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myocardium/metabolism , Myocardium/pathology , Unfolded Protein Response
17.
Proc Natl Acad Sci U S A ; 109(21): 8079-84, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22562797

ABSTRACT

The unfolded protein response (UPR) is an intracellular stress-signaling pathway that counteracts the accumulation of misfolded proteins in the endoplasmic reticulum (ER). Because defects in ER protein folding are associated with many pathological states, including metabolic, neurologic, genetic, and inflammatory diseases, it is important to understand how the UPR maintains ER protein-folding homeostasis. All metazoans have conserved the fundamental UPR transducers IRE1, ATF6, and PERK. In Caenorhabditis elegans, the UPR is required to prevent larval lethality and intestinal degeneration. Although ire-1-null worms are viable, they are particularly sensitive to ER stress. To identify genes that are required for development of ire-1-null worms, we performed a comprehensive RNA interference screen to find 10 genes that exhibit synthetic growth and intestinal defects with the ire-1(v33) mutant but not with atf-6(tm1153) or pek-1(ok275) mutants. The expression of two of these genes, exos-3 and F48E8.6, was induced by ER stress, and their knockdown in a wild-type strain caused ER stress. Because these genes encode subunits of the exosome complex that functions in mRNA surveillance, we analyzed other gene products required for nonsense-mediated mRNA decay (NMD). Our results demonstrate that defects in smg-1, smg-4, and smg-6 in C. elegans and SMG6 in mammalian cells cause ER stress and sensitize to the lethal effects of ER stress. Although ER stress did not activate mRNA surveillance complex assembly, ER stress did induce SMG6 expression, and NMD regulators were constitutively localized to the ER. Importantly, the findings demonstrate a unique and fundamental interaction where NMD-mediated mRNA quality control is required to prevent ER stress.


Subject(s)
Caenorhabditis elegans/genetics , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/physiology , Homeostasis/physiology , RNA/genetics , Unfolded Protein Response/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/physiology , Cell Line, Transformed , Cell Survival/physiology , Codon, Nonsense/genetics , HeLa Cells , Hepatocytes/cytology , Humans , Larva/physiology , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , RNA Interference/physiology , RNA-Binding Proteins , Telomerase/genetics , Telomerase/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
18.
J Biol Chem ; 288(26): 18624-42, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23720735

ABSTRACT

Nonalcoholic fatty liver disease is a heterogeneous disorder characterized by liver steatosis; inflammation and fibrosis are features of the progressive form nonalcoholic steatohepatitis. The endoplasmic reticulum stress response is postulated to play a role in the pathogenesis of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. In particular, C/EBP homologous protein (CHOP) is undetectable under normal conditions but is induced by cellular stress, including endoplasmic reticulum stress. Chop wild type (Chop(+/+)) and knock-out (Chop(-/-)) mice were used in these studies to elucidate the role of CHOP in the pathogenesis of fatty liver disease. Paradoxically, Chop(-/-) mice developed greater liver injury, inflammation, and fibrosis than Chop(+/+) mice, with greater macrophage activation. Primary, bone marrow-derived, and peritoneal macrophages from Chop(+/+) and Chop(-/-) were challenged with palmitic acid, an abundant saturated free fatty acid in plasma and liver lipids. Where palmitic acid treatment activated Chop(+/+) and Chop(-/-) macrophages, Chop(-/-) macrophages were resistant to its lipotoxicity. Chop(-/-) mice were sensitized to liver injury in a second model of dietary steatohepatitis using the methionine-choline-deficient diet. Analysis of bone marrow chimeras between Chop(-/-) and Chop(+/+) mice demonstrated that Chop in macrophages protects from liver injury and inflammation when fed the methionine-choline-deficient diet. We conclude that Chop deletion has a proinflammatory effect in fatty liver injury apparently due to decreased cell death of activated macrophages, resulting in their net accumulation in the liver. Thus, macrophage CHOP plays a key role in protecting the liver from steatohepatitis likely by limiting macrophage survival during lipotoxicity.


Subject(s)
Apoptosis , Fatty Liver/prevention & control , Liver/metabolism , Macrophages/pathology , Transcription Factor CHOP/metabolism , Animals , Blood Glucose/metabolism , Cell Survival , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Fatty Liver/metabolism , Female , Fibrosis/metabolism , Inflammation , Liver/injuries , Macrophages/cytology , Male , Mice , Mice, Transgenic , Obesity , PPAR gamma/metabolism
19.
J Biol Chem ; 288(24): 17202-13, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23645676

ABSTRACT

Endoplasmic reticulum (ER) stress-induced responses are associated with the loss of insulin-producing ß-cells in type 2 diabetes mellitus. ß-Cell survival during ER stress is believed to depend on decreased protein synthesis rates that are mediated via phosphorylation of the translation initiation factor eIF2α. It is reported here that chronic ER stress correlated with increased islet protein synthesis and apoptosis in ß-cells in vivo. Paradoxically, chronic ER stress in ß-cells induced an anabolic transcription program to overcome translational repression by eIF2α phosphorylation. This program included expression of amino acid transporter and aminoacyl-tRNA synthetase genes downstream of the stress-induced ATF4-mediated transcription program. The anabolic response was associated with increased amino acid flux and charging of tRNAs for branched chain and aromatic amino acids (e.g. leucine and tryptophan), the levels of which are early serum indicators of diabetes. We conclude that regulation of amino acid transport in ß-cells during ER stress involves responses leading to increased protein synthesis, which can be protective during acute stress but can lead to apoptosis during chronic stress. These studies suggest that the increased expression of amino acid transporters in islets can serve as early diagnostic biomarkers for the development of diabetes.


Subject(s)
Amino Acids/metabolism , Apoptosis , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Insulin-Secreting Cells/physiology , Activating Transcription Factor 4/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cell Survival , Diabetes Mellitus, Type 2/pathology , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , RNA, Transfer/metabolism , Transcriptional Activation
20.
Mol Cells ; 46(11): 688-699, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37968983

ABSTRACT

We set up this study to understand the underlying mechanisms of reduced ceramides on immune cells in acute rejection (AR). The concentrations of ceramides and sphingomyelins were measured in the sera from hepatic transplant patients, skin graft mice and hepatocyte transplant mice by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Serum concentrations of C24 ceramide, C24:1 ceramide, C16:0 sphingomyelin, and C18:1 sphingomyelin were lower in liver transplantation (LT) recipients with than without AR. Comparisons with the results of LT patients with infection and cardiac transplant patients with cardiac allograft vasculopathy in humans and in mouse skin graft and hepatocyte transplant models suggested that the reduced C24 and C24:1 ceramides were specifically involved in AR. A ceramide synthase inhibitor, fumonisin B1 exacerbated allogeneic immune responses in vitro and in vivo, and reduced tolerogenic dendritic cells (tDCs), while increased P3-like plasmacytoid DCs (pDCs) in the draining lymph nodes from allogeneic skin graft mice. The results of mixed lymphocyte reactions with ceranib-2, an inhibitor of ceramidase, and C24 ceramide also support that increasing ceramide concentrations could benefit transplant recipients with AR. The results suggest increasing ceramides as novel therapeutic target for AR, where reduced ceramides were associated with the changes in DC subsets, in particular tDCs.


Subject(s)
Ceramides , Liver Transplantation , Humans , Mice , Animals , Sphingomyelins , Chromatography, Liquid , Skin Transplantation , Tandem Mass Spectrometry , Hepatocytes , Dendritic Cells
SELECTION OF CITATIONS
SEARCH DETAIL