Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 77(4): 875-886.e7, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31836389

ABSTRACT

Dysregulation of cellular protein synthesis is linked to a variety of diseases. Mutations in EIF2S3, encoding the γ subunit of the heterotrimeric eukaryotic translation initiation factor eIF2, cause MEHMO syndrome, an X-linked intellectual disability disorder. Here, using patient-derived induced pluripotent stem cells, we show that a mutation at the C terminus of eIF2γ impairs CDC123 promotion of eIF2 complex formation and decreases the level of eIF2-GTP-Met-tRNAiMet ternary complexes. This reduction in eIF2 activity results in dysregulation of global and gene-specific protein synthesis and enhances cell death upon stress induction. Addition of the drug ISRIB, an activator of the eIF2 guanine nucleotide exchange factor, rescues the cell growth, translation, and neuronal differentiation defects associated with the EIF2S3 mutation, offering the possibility of therapeutic intervention for MEHMO syndrome.


Subject(s)
Acetamides/pharmacology , Cyclohexylamines/pharmacology , Epilepsy/genetics , Eukaryotic Initiation Factor-2/genetics , Genitalia/abnormalities , Hypogonadism/genetics , Mental Retardation, X-Linked/genetics , Microcephaly/genetics , Mutation , Obesity/genetics , Protein Biosynthesis/drug effects , Apoptosis , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology
2.
Ann Hematol ; 103(8): 2853-2863, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38842564

ABSTRACT

Acute Myeloid Leukemia (AML) is a life-threatening disease whose induction treatment consists of combination chemotherapy with Idarubicin and Cytarabine for fit patients. Treatment failures are frequent, urging the need for novel treatments for this disease. The DNA Damage Response Mechanism (DDR) comprises numerous molecules and pathways intended to arrest the cell cycle until DNA damage is repaired or else drive the cell to apoptosis. AML-derived cell lines after treatment with Idarubicin and Cytarabine were used for studying the expression profile of 84 DDR genes, through PCR arrays. Utilizing de novo AML patient and control samples we studied the expression of PPP1R15A, CDKN1A, GADD45A, GADD45G, and EXO1. Next, we performed PPP1R15A silencing in AML cell lines in two separate experiments using siRNA and CRISPR-cas9, respectively. Our findings highlight that DDR regulators demonstrate increased expression in patients with high cytogenetic risk possibly reflecting increased genotoxic stress. Especially, PPP1R15A is mainly involved in the recovery of the cells from stress and it was the only DDR gene upregulated in AML patients. The PPP1R15A silencing resulted in decreased viability of Idarubicin and Cytarabine-treated cell lines, in contrast to untreated cells. These findings shed light on new strategies to enhance chemotherapy efficacy and demonstrate that PPP1R15A is an important DDR regulator in AML and its downregulation might be a safe and effective way to increase sensitivity to chemotherapy in this disease.


Subject(s)
Cytarabine , DNA Damage , Gene Silencing , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , DNA Damage/drug effects , Cytarabine/pharmacology , Cell Line, Tumor , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Idarubicin/pharmacology , Idarubicin/administration & dosage , Male , Female , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Repair/drug effects , Middle Aged , Adult , Aged , Gene Expression Regulation, Leukemic/drug effects
3.
Biochem Biophys Res Commun ; 654: 112-119, 2023 04 30.
Article in English | MEDLINE | ID: mdl-36907138

ABSTRACT

Alzheimer's disease (AD) causes neurodegeneration, leading to cognitive impairment and memory loss. Our previous studies have demonstrated that the induction of growth arrest and DNA damage-inducible gene 34 (GADD34) by quercetin can affect eukaryotic translation initiation factor 2α (eIF2α) phosphorylation-activated transcription factor 4 (ATF4) signaling. However, the relationship between GADD34 expression and cognitive function has not been clarified. In this study, we determined the direct effect of GADD34 on memory. To achieve this, truncated GADD34 (GADD34.5) was injected into the mouse brain to suppress eIF2α phosphorylation and evaluate memory. The injection of GADD34.5 into the hippocampus in AD-model mice did not improve novel object recognition but improved novel object location. The injection of GADD34.5 into the amygdala also resulted in the maintenance of contextual fear memory based on the fear condition test. These results suggest that GADD34 is effective in improving memory for spatial cognition and contextual fear conditioning in AD by inhibiting eIF2α phosphorylation. In summary, GADD34 suppresses eIF2α phosphorylation in the brain and prevents memory loss. As quercetin feeding increases GADD34 expression, it might be used in preventative applications for AD.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/metabolism , Phosphorylation , Quercetin , Eukaryotic Initiation Factor-2/metabolism , Cognition , Memory Disorders , Protein Phosphatase 1/metabolism
4.
Cell Commun Signal ; 21(1): 326, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37957724

ABSTRACT

BACKGROUND: The placentas from newborns that are small for gestational age (SGA; birth weight < -2 SD for gestational age) may display multiple pathological characteristics. A key determinant of fetal growth and, therefore, birth weight is placental amino acid transport, which is under the control of the serine/threonine kinase mechanistic target of rapamycin (mTOR). The effects of endoplasmic reticulum (ER) stress on the mTOR pathway and the levels of amino acid transporters are not well established. METHODS: Placentas from SGA and appropriate for gestational age (AGA) newborns and the human placental BeWo cell line exposed to the ER stressor tunicamycin were used. RESULTS: We detected a significant increase in the levels of C/EBP homologous protein (CHOP) in the placentas from SGA newborns compared with those from AGA newborns, while the levels of other ER stress markers were barely affected. In addition, placental mTOR Complex 1 (mTORC1) activity and the levels of the mature form of the amino acid transporter sodium-coupled neutral amino acid transporter 2 (SNAT2) were also reduced in the SGA group. Interestingly, CHOP has been reported to upregulate growth arrest and DNA damage-inducible protein 34 (GADD34), which in turn suppresses mTORC1 activity. The GADD34 inhibitor guanabenz attenuated the increase in CHOP protein levels and the reduction in mTORC1 activity caused by the ER stressor tunicamycin in the human placental cell line BeWo, but it did not recover mature SNAT2 protein levels, which might be reduced as a result of defective glycosylation. CONCLUSIONS: Collectively, these data reveal that GADD34A activity and glycosylation are key factors controlling mTORC1 signaling and mature SNAT2 levels in trophoblasts, respectively, and might contribute to the SGA condition. Video Abstract.


Subject(s)
Amino Acid Transport System A , Placenta , TOR Serine-Threonine Kinases , Transcription Factor CHOP , Female , Humans , Infant, Newborn , Pregnancy , Birth Weight , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Gestational Age , Mechanistic Target of Rapamycin Complex 1/metabolism , Placenta/metabolism , TOR Serine-Threonine Kinases/metabolism , Tunicamycin/pharmacology , Up-Regulation , Transcription Factor CHOP/genetics , Amino Acid Transport System A/genetics
5.
Int J Mol Sci ; 24(24)2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38139150

ABSTRACT

The vertebrate PPP1R15 family consists of the proteins GADD34 (growth arrest and DNA damage-inducible protein 34, the product of the PPP1R15A gene) and CReP (constitutive repressor of eIF2α phosphorylation, the product of the PPP1R15B gene), both of which function as targeting/regulatory subunits for protein phosphatase 1 (PP1) by regulating subcellular localization, modulating substrate specificity and assembling complexes with target proteins. The primary cellular function of these proteins is to facilitate the dephosphorylation of eukaryotic initiation factor 2-alpha (eIF2α) by PP1 during cell stress. In this review, we will provide a comprehensive overview of the cellular function, biochemistry and pharmacology of GADD34 and CReP, starting with a brief introduction of eIF2α phosphorylation via the integrated protein response (ISR). We discuss the roles GADD34 and CReP play as feedback inhibitors of the unfolded protein response (UPR) and highlight the critical function they serve as inhibitors of the PERK-dependent branch, which is particularly important since it can mediate cell survival or cell death, depending on how long the stressful stimuli lasts, and GADD34 and CReP play key roles in fine-tuning this cellular decision. We briefly discuss the roles of GADD34 and CReP homologs in model systems and then focus on what we have learned about their function from knockout mice and human patients, followed by a brief review of several diseases in which GADD34 and CReP have been implicated, including cancer, diabetes and especially neurodegenerative disease. Because of the potential importance of GADD34 and CReP in aspects of human health and disease, we will discuss several pharmacological inhibitors of GADD34 and/or CReP that show promise as treatments and the controversies as to their mechanism of action. This review will finish with a discussion of the biochemical properties of GADD34 and CReP, their regulation and the additional interacting partners that may provide insight into the roles these proteins may play in other cellular pathways. We will conclude with a brief outline of critical areas for future study.


Subject(s)
Neurodegenerative Diseases , Protein Phosphatase 1 , Animals , Humans , Mice , Eukaryotic Initiation Factor-2/metabolism , Mice, Knockout , Phosphorylation , Protein Biosynthesis , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proteins/metabolism
6.
Int J Mol Sci ; 23(22)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36430227

ABSTRACT

The UPR is sustainably activated in degenerating retinas, leading to translational inhibition via p-eIF2α. Recent findings have demonstrated that ablation of growth arrest and DNA damage-inducible protein 34 (GADD34), a protein phosphatase 1 regulatory subunit permitting translational machinery operation through p-eIF2α elevation, does not impact the rate of translation in fast-degenerating rd16 mice. The current study aimed to validate whether P23H RHO mice degenerating at a slower pace manifest translational attenuation and whether GADD34 ablation impacts the rate of retinal degeneration via further suppression of retinal protein synthesis and apoptotic cell death. For this study, mice were examined with ERG and histological analyses. The molecular assessment was conducted in the naïve and LPS-challenged mice using Western blot and qRT-PCR analyses. Thus, this study demonstrates that the P23H RHO retinas manifest translational attenuation. However, GADD34 ablation resulted in a more prominent p-eIF2a increase without impacting the translation rate. GADD34 deficiency also led to a reduction in scotopic ERG amplitudes and an increased number of TUNEL-positive cells. Molecular analysis revealed that GADD34 deficiency reduces the expression of p-STAT3 and Il-6 while increasing the expression of Tnfa. Overall, the data indicate that GADD34 plays a multifunctional role. Under chronic UPR activation, GADD34 acts as a feedback player, dephosphorylating p-eIF2a, although this role does not seem to be critical. Additionally, GADD34 controls cytokine expression and STAT3 activation. Perhaps these molecular events are particularly important in controlling the pace of retinal degeneration.


Subject(s)
Retinal Degeneration , Animals , Mice , Eukaryotic Initiation Factor-2/metabolism , Mice, Inbred C57BL , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Retina/metabolism , Retinal Degeneration/metabolism
7.
Vet Res ; 52(1): 148, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34930429

ABSTRACT

Viruses have evolved multiple strategies to manipulate their host's translational machinery for the synthesis of viral proteins. A common viral target is the alpha subunit of eukaryotic initiation factor 2 (eIF2α). In this study, we show that global protein synthesis was increased but the eIF2α phosphorylation level was markedly decreased in porcine kidney 15 (PK15) cells infected with pseudorabies virus (PRV), a swine herpesvirus. An increase in the eIF2α phosphorylation level by salubrinal treatment or transfection of constructs expressing wild-type eIF2α or an eIF2α phosphomimetic [eIF2α(S51D)] attenuated global protein synthesis and suppressed PRV replication. To explore the mechanism involved in the inhibition of eIF2α phosphorylation during PRV infection, we examined the phosphorylation status of protein kinase R-like endoplasmic reticulum kinase (PERK) and double-stranded RNA-dependent protein kinase R (PKR), two kinases that regulate eIF2α phosphorylation during infection with numerous viruses. We found that the level of neither phosphorylated (p)-PERK nor p-PKR was altered in PRV-infected cells or the lungs of infected mice. However, the expression of growth arrest and DNA damage-inducible protein 34 (GADD34), which promotes eIF2α dephosphorylation by recruiting protein phosphatase 1 (PP1), was significantly induced both in vivo and in vitro. Knockdown of GADD34 and inhibition of PP1 activity by okadaic acid treatment led to increased eIF2α phosphorylation but significantly suppressed global protein synthesis and inhibited PRV replication. Collectively, these results demonstrated that PRV induces GADD34 expression to promote eIF2α dephosphorylation, thereby maintaining de novo protein synthesis and facilitating viral replication.


Subject(s)
Eukaryotic Initiation Factor-2 , Herpesvirus 1, Suid , Protein Phosphatase 1 , Pseudorabies , Viral Proteins , Virus Replication , Animals , Eukaryotic Initiation Factor-2/metabolism , Herpesvirus 1, Suid/physiology , Mice , Phosphorylation , Protein Phosphatase 1/metabolism , Pseudorabies/virology , Swine , Viral Proteins/genetics , Virus Replication/physiology
8.
J Biol Chem ; 294(15): 5945-5955, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30782845

ABSTRACT

GADD34 (growth arrest and DNA damage-inducible gene 34) plays a critical role in responses to DNA damage and endoplasmic reticulum stress. GADD34 has opposing effects on different stimuli-induced cell apoptosis events, but the reason for this is unclear. Here, using immunoblotting analyses and various molecular genetic approaches in HepG2 and SMMC-7721 cells, we demonstrate that GADD34 protects hepatocellular carcinoma (HCC) cells from tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by stabilizing a BCL-2 family member, myeloid cell leukemia 1 (MCL-1). We found that GADD34 knockdown decreased MCL-1 levels and that GADD34 overexpression up-regulated MCL-1 expression in HCC cells. GADD34 did not affect MCL-1 transcription but enhanced MCL-1 protein stability. The proteasome inhibitor MG132 abrogated GADD34 depletion-induced MCL-1 down-regulation, suggesting that GADD34 inhibits the proteasomal degradation of MCL-1. Furthermore, GADD34 overexpression promoted extracellular signal-regulated kinase (ERK) phosphorylation through a signaling axis that consists of the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6) and transforming growth factor-ß-activated kinase 1 (MAP3K7)-binding protein 1 (TAB1), which mediated the up-regulation of MCL-1 by GADD34. Of note, TRAIL up-regulated both GADD34 and MCL-1 levels, and knockdown of GADD34 and TRAF6 suppressed the induction of MCL-1 by TRAIL. Correspondingly, GADD34 knockdown potentiated TRAIL-induced apoptosis, and MCL-1 overexpression rescued TRAIL-treated and GADD34-depleted HCC cells from cell death. Taken together, these findings suggest that GADD34 inhibits TRAIL-induced HCC cell apoptosis through TRAF6- and ERK-mediated stabilization of MCL-1.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Proteins/metabolism , Protein Phosphatase 1/metabolism , TNF Receptor-Associated Factor 6/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasm Proteins/genetics , Protein Phosphatase 1/genetics , Protein Stability , TNF Receptor-Associated Factor 6/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics
9.
Neurobiol Dis ; 136: 104702, 2020 03.
Article in English | MEDLINE | ID: mdl-31837419

ABSTRACT

Mutations in Cu/Zn superoxide dismutase (SOD1) cause ~20% of familial ALS (FALS), which comprises 10% of total ALS cases. In mutant SOD1- (mtSOD1-) induced ALS, misfolded aggregates of SOD1 lead to activation of the unfolded protein response/integrated stress response (UPR/ISR). Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), a kinase that phosphorylates eukaryotic translation initiator factor 2α (p-eIF2α), coordinates the response by causing a global suppression of protein synthesis. Growth arrest and DNA damage 34 (GADD34) dephosphorylates p-eIF2α, allowing protein synthesis to return to normal. If the UPR/ISR is overwhelmed by the amount of misfolded protein, CCAAT/enhancer-binding homologous protein (CHOP) is activated leading to apoptosis. In the current study we investigated the effect of knocking down CHOP and GADD34 on disease of G93A and G85R mtSOD1 mice. Although a CHOP antisense oligonucleotide had no effect on survival, an intravenous injection of GADD34 shRNA encoded in adeno-associated virus 9 (AAV9) into neonatal G93A as well as neonatal G85R mtSOD1 mice led to a significantly increased survival. G85R mtSOD1 mice had a reduction in SOD1 aggregates/load, astrocytosis, and microgliosis. In contrast, there was no change in disease phenotype when GADD34 shRNA was delivered to older G93A mtSOD1 mice. Our current study shows that GADD34 shRNA is effective in ameliorating disease when administered to neonatal mtSOD1 mice. Targeting the UPR/ISR may be beneficial in mtSOD1-induced ALS as well as other neurodegenerative diseases in which misfolded proteins and ER stress have been implicated.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Gene Knockdown Techniques/methods , Protein Phosphatase 1/deficiency , Protein Phosphatase 1/genetics , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/prevention & control , Animals , Animals, Newborn , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase-1/metabolism
10.
RNA ; 24(6): 841-852, 2018 06.
Article in English | MEDLINE | ID: mdl-29632131

ABSTRACT

Mutations in eIF2B genes cause vanishing white matter disease (VWMD), a fatal leukodystrophy that can manifest following physical trauma or illness, conditions that activate the integrated stress response (ISR). EIF2B is the guanine exchange factor for eIF2, facilitating ternary complex formation and translation initiation. During the ISR, eIF2α is phosphorylated and inhibits eIF2B, causing global translation suppression and stress-induced gene translation, allowing stress adaptation and recovery. We demonstrate that VWMD patient cells hypersuppress translation during the ISR caused by acute ER stress, delaying stress-induced gene expression and interrupting a negative feedback loop that allows translational recovery by GADD34-mediated dephosphorylation of phospho-eIF2α. Thus, cells from VWMD patients undergo a prolonged state of translational hyperrepression and fail to recover from stress. We demonstrate that small molecules targeting eIF2B or the eIF2α kinase PERK rescue translation defects in patient cells. Therefore, defects in the ISR could contribute to white matter loss in VWMD.


Subject(s)
Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2/genetics , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Mutation , Protein Biosynthesis , Stress, Physiological , Adult , Case-Control Studies , Cells, Cultured , Female , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Phosphorylation , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Young Adult
11.
Inflamm Res ; 69(8): 779-787, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32435966

ABSTRACT

INTRODUCTION: Neuroinflammation is a key aspect of various injuries and diseases of the central nervous system and brain, including stroke, Alzheimer's, Parkinson's, multiple sclerosis, etc. Phoenixin-14 is a naturally occurring pleiotropic peptide involved in reproduction, anxiety, pain, and other functions. MATERIALS AND METHODS: Primary astrocytes were isolated from new-born pups of c57bl/6 mice. The gene expression of GPR173, CHOP, and GADD34 was measured by real-time PCR. Protein expression was assessed by western blot analysis. Secretions of IL-1ß and IL-18 were determined by ELISA. RESULTS: Phoenixin-14 (PNX-14) is a ligand for the G protein-coupled receptor GPR173, which we demonstrate to be expressed in astrocytes and suppressed by exposure to lipopolysaccharide (LPS). Endoplasmic reticulum (ER) stress resulting from injury or disease leads to the unfolded protein response, which is mediated by the activation of transcription factors including eIF-2α, ATF4, and CHOP, and regulated by GADD34. ER stress also leads to a robust neuroinflammatory response, which is mediated by HMGB1-induced activation of the NLRP3 inflammasome and subsequent production of IL-1ß and IL-18. In the present study, we demonstrate that PNX-14 could attenuate LPS-induced ER stress response and NLRP3 inflammasome activation in mouse cerebral astrocytes. Our findings show that PNX-14 could suppress the production of ROS as well as the decrease in SOD induced by LPS. PNX-14 also inhibited HMGB1-mediated NLRP3 inflammasome activation and production of IL-1ß and IL-18. Through a GPR173 siRNA knockdown experiment, we further demonstrate that GPR173 knockdown abolished the effects of PNX-14 on LPS-induced NLRP3 expression and IL-18 production. CONCLUSION: These findings suggest that PNX-14 may have potential in the treatment of neuroinflammation.


Subject(s)
Astrocytes/drug effects , Inflammasomes/antagonists & inhibitors , Inflammation/prevention & control , Peptides/pharmacology , Animals , Astrocytes/metabolism , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Oxidative Stress/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology
12.
J Biol Chem ; 293(1): 163-176, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29109149

ABSTRACT

Oxidative and endoplasmic reticulum (ER) stresses are hallmarks of the pathophysiology of ALS and other neurodegenerative diseases. In these stresses, different kinases phosphorylate eukaryotic initiation factor eIF2α, enabling the translation of stress response genes; among these is GADD34, the protein product of which recruits the α-isoform of protein phosphatase 1 catalytic subunit (PP1α) and eIF2α to assemble a phosphatase complex catalyzing eIF2α dephosphorylation and resumption of protein synthesis. Aberrations in this pathway underlie the aforementioned disorders. Previous observations indicating that GADD34 is induced by arsenite, a thiol-directed oxidative stressor, in the absence of eIF2α phosphorylation suggest other roles for GADD34. Here, we report that arsenite-induced oxidative stress differs from thapsigargin- or tunicamycin-induced ER stress in promoting GADD34 transcription and the preferential translation of its mRNA in the absence of eIF2α phosphorylation. Arsenite also stabilized GADD34 protein, slowing its degradation. In response to oxidative stress, but not ER stress, GADD34 recruited TDP-43, and enhanced cytoplasmic distribution and cysteine modifications of TDP-43 promoted its binding to GADD34. Arsenite also recruited a TDP-43 kinase, casein kinase-1ϵ (CK1ϵ), to GADD34. Concomitant with TDP-43 aggregation and proteolysis after prolonged arsenite exposure, GADD34-bound CK1ϵ catalyzed TDP-43 phosphorylations at serines 409/410, which were diminished or absent in GADD34-/- cells. Our findings highlight that the phosphatase regulator, GADD34, also functions as a kinase scaffold in response to chronic oxidative stress and recruits CK1ϵ and oxidized TDP-43 to facilitate its phosphorylation, as seen in TDP-43 proteinopathies.


Subject(s)
DNA-Binding Proteins/metabolism , Oxidative Stress/physiology , Protein Phosphatase 1/metabolism , TDP-43 Proteinopathies/metabolism , Animals , Arsenites/pharmacology , Casein Kinase 1 epsilon/metabolism , Cell Cycle Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Transgenic , Oxidative Stress/drug effects , Phosphorylation , Protein Phosphatase 1/deficiency
13.
Neurobiol Dis ; 127: 527-544, 2019 07.
Article in English | MEDLINE | ID: mdl-30923003

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease, characterized by motor neuron death in the brain and spinal cord. Mutations in the Cu/Zn superoxide dismutase (SOD1) gene account for ~20% of all familial ALS forms, corresponding to 1%-2% of all ALS cases. One of the suggested mechanisms by which mutant SOD1 (mtSOD1) exerts its toxic effects involves intracellular accumulation of abnormal mtSOD1 aggregates, which trigger endoplasmic reticulum (ER) stress and activate its adaptive signal transduction pathways, including the unfolded protein response (UPR). PERK, an eIF2α kinase, is central to the UPR and is the most rapidly activated pathway in response to ER stress. Previous reports using mtSOD1 transgenic mice indicated that genetic or pharmacological enhancement of the UPR-PERK pathway may be effective in treating ALS. We investigated the response to PERK haploinsufficiency, and the response to deficiency of its downstream effectors GADD34 and CHOP, in five distinct lines of mtSOD1 mice. We demonstrate that, in contrast to a previously published study, PERK haploinsufficiency has no effect on disease in all mtSOD1 lines examined. We also show that deficiency of GADD34, which enhances the UPR by prolonging the phosphorylation of eIF2α, does not ameliorate disease in these mtSOD1 mouse lines. Finally, we demonstrate that genetic ablation of CHOP transcription factor, which is known to be pro-apoptotic, does not ameliorate disease in mtSOD1 mice. Cumulatively, our studies reveal that neither genetic inhibition of the UPR via ablation of PERK, nor genetic UPR enhancement via ablation of GADD34, is beneficial for mtSOD1-induced motor neuron disease. Therefore, the PERK pathway is not a likely target for therapeutic intervention in mtSOD1-induced ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Signal Transduction/physiology , Superoxide Dismutase-1/metabolism , Unfolded Protein Response/physiology , eIF-2 Kinase/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Motor Neurons/metabolism , Superoxide Dismutase-1/genetics , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , eIF-2 Kinase/genetics
14.
Int J Mol Sci ; 20(11)2019 Jun 05.
Article in English | MEDLINE | ID: mdl-31195662

ABSTRACT

The initiation of protein synthesis is suppressed under several stress conditions, inducing phosphorylation of the α-subunit of the eukaryotic initiation factor 2 (eIF2α), thereby inactivating the GTP-GDP recycling protein eIF2B. By contrast, the mammalian activating transcription factor 4 (ATF4, also known as cAMP response element binding protein 2 (CREB2)) is still translated under stress conditions. Four protein kinases (general control nonderepressible-2 (GCN2) kinase, double-stranded RNA-activated protein kinase (PKR), PKR-endoplasmic reticulum (ER)-related kinase (PERK), and heme-regulated inhibitor kinase (HRI)) phosphorylate eIF2α in the presence of stressors such as amino acid starvation, viral infection, ER stress, and heme deficiency. This signaling reaction is known as the integrated stress response (ISR). Here, we review ISR signaling in the brain in a mouse model of Alzheimer's disease (AD). We propose that targeting ISR signaling with quercetin has therapeutic potential, because it suppresses amyloid-ß (Aß) production in vitro and prevents cognitive impairments in a mouse model of AD.


Subject(s)
Memory/drug effects , Quercetin/pharmacology , Stress, Physiological/drug effects , Alzheimer Disease/physiopathology , Animals , Humans , Neuronal Plasticity/drug effects , Signal Transduction/drug effects
15.
Cell Physiol Biochem ; 50(6): 2203-2215, 2018.
Article in English | MEDLINE | ID: mdl-30415257

ABSTRACT

BACKGROUND/AIMS: Tonsillectomy may be an important method to achieve a long-term remission of IgAN, but patients' physical status may limit their access to this surgery. We proposed an encouraging solution through inhibiting GADD34 expression in order to promote tonsillar mononuclear cells (TMCs) apoptosis and reduce nephropathic IgA secretion. METHODS: A total of 12 IgAN and 9 non-IgAN patients were involved from March 2015 to May 2016. After TMCs were extracted by density gradient centrifugation and stimulated by inactivated hemolytic streptococcus, the mRNA and protein expression of GADD34, GRP78, CHOP, Bcl-2, Bcl-XL, AID, Iα-Cα, and cleaved caspase-3 were examined by fluorescent RT-PCR and Western blotting. Guanabenz treatment and siRNA interference were applied to downregulate GADD34 in tonsillar mononuclear cells from IgAN patients, and P-eIF2α expression was examined by Western Blotting. Cell apoptosis was evaluated by Annexin V FITC/PI flowcytometry, and IgA secretion in cultural supernatant was inspected by enzyme linked immunosorbent assay. RESULTS: After stimulation, the expression of GADD34 was significantly increased in IgAN patients (P< 0.05). Cell apoptosis was mitigated and IgA secretion level was elevated (P< 0.05). To be noticed, CHOP expression had no significant difference between two groups. After guanabenz treatment and siRNA interference, a prolonged elevation of P-eIF2α expression was observed. Cell apoptosis was reinforced and IgA secretion level was decreased (P< 0.05). CONCLUSION: GADD34 may be a potential therapeutic target for IgAN treatment due to its effect on cell apoptosis.


Subject(s)
Apoptosis , Eukaryotic Initiation Factor-2/metabolism , Protein Phosphatase 1/metabolism , Adolescent , Adult , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Female , Glomerulonephritis, IGA/metabolism , Glomerulonephritis, IGA/pathology , Guanabenz/pharmacology , Heat-Shock Proteins/metabolism , Humans , Immunoglobulin A/metabolism , Male , Middle Aged , Palatine Tonsil/cytology , Palatine Tonsil/drug effects , Palatine Tonsil/metabolism , Phosphorylation , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/genetics , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factor CHOP/metabolism , Young Adult
16.
Mol Cell Biochem ; 440(1-2): 65-75, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28825160

ABSTRACT

Activating transcription factor 4 (ATF4), which is ubiquitously expressed, plays a crucial role in regulating various stress-responsive genes under pathophysiological conditions. Further, growth arrest and DNA damage-inducible gene 34 (GADD34), a downstream target of ATF4, has been reported to negatively regulate ATF4 expression. To understand the relationship between intrinsic ATF4 and GADD34 under resting and ER stress conditions, we used a novel gene editing approach, CRISPR/Cas9, to integrate antibiotic-resistant genes into the target genes, ATF4 and GADD34. First, we manipulated the ATF4 gene in the mouse neuroblastoma cell line, Neuro2a, and compared the ER stress responses between parental and ATF4-edited Neuro2a cells. Next, we established Neuro2a cells with edited GADD34 and ATF4/GADD34 genes and found that ATF4 acts as a proapoptotic factor, but GADD34 depletion did not attenuate the expression of cleaved caspase-3 induced by tunicamycin treatment. These findings provide new insights into the ATF4 signaling cascades. Additionally, the rapid establishment of cells lacking multiple genes using this CRISPR/Cas9 system will be a powerful tool for exploring various cellular issues under pathophysiological conditions.


Subject(s)
Activating Transcription Factor 4 , CRISPR-Cas Systems , Gene Editing , Gene Expression Regulation/genetics , Protein Phosphatase 1 , Signal Transduction/genetics , Activating Transcription Factor 4/biosynthesis , Activating Transcription Factor 4/genetics , Animals , Cell Line , Humans , Mice , Protein Phosphatase 1/biosynthesis , Protein Phosphatase 1/genetics
17.
Am J Respir Cell Mol Biol ; 57(4): 477-486, 2017 10.
Article in English | MEDLINE | ID: mdl-28594573

ABSTRACT

Intermittent hypoxia (IH) induces activation of the integrated stress response (ISR), but its role in IH-induced visceral white adipose tissue (vWAT) insulin resistance is unknown. CHOP is activated by chronic ISR, whereas GADD34 dephosphorylates the subunit of translation initiation factor 2 (eIF2α), leading to termination of the ISR. We hypothesized that CHOP/Gadd34 null mice would not manifest evidence of insulin resistance after IH exposures. Eight-week-old CHOP/GADD34-/- (double mutant [DM]) and wild-type (WT) littermates were randomly assigned to IH or room air (RA) exposures for 6 weeks. Glucose and insulin tolerance tests were performed, and regulatory T cells (Tregs) and macrophages in vWAT were assessed. Phosphorylated eIF2α:total eIF2α, ATF4, XBP1 expression, and insulin-induced pAKT/AKT expression changes were examined in vWATs. Single GADD34-/- and PERK+/- mice were also evaluated. Body weight and vWAT mass were reduced in DM and WT mice after IH. M1/M2 macrophages and inflammatory macrophages (Ly-6chigh) were significantly increased in WT vWAT but remained unchanged in DM mice. Tregs were significantly decreased in WT vWAT but not in DM mice. Systemic insulin and glucose tolerance tests revealed insulin resistance in IH-WT but not in IH-DM mice. Similarly, decreased pAKT/AKT responses to exogenous insulin emerged in IH-WT compared with RA-WT mice, whereas no significant differences emerged in IH-DM compared with DM-RA. Chronic ISR activation appears to contribute to the insulin resistance and vWAT inflammation that characteristically emerge after long-term IH exposures in a murine model of obstructive sleep apnea.


Subject(s)
Insulin Resistance/genetics , Intra-Abdominal Fat , Macrophages , Signal Transduction/genetics , Sleep Apnea Syndromes , T-Lymphocytes, Regulatory , Animals , Disease Models, Animal , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/physiopathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sleep Apnea Syndromes/genetics , Sleep Apnea Syndromes/metabolism , Sleep Apnea Syndromes/pathology , Sleep Apnea Syndromes/physiopathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
18.
Acta Virol ; 61(4): 445-452, 2017.
Article in English | MEDLINE | ID: mdl-29186961

ABSTRACT

Protein phosphatase 1, regulatory subunit 15A (PPP1R15A), also known as growth arrest and DNA damage-inducible protein GADD34, plays a vital role in promoting cell death and the unfolded protein response (UPR). In order to explore whether the SNP (rs611251) of PPP1R15A gene has a role in different types of Epstein-Barr virus (EBV) - associated tumors, we detected the PPP1R15A gene rs611251 polymorphism in 195 cases of EBV positive tumors (93 lymphomas, 48 gastric carcinomas, 54 nasopharyngeal carcinomas), 208 cases of EBV-negative tumors (136 gastric carcinoma, 19 nasopharyngeal carcinomas, 53 lymphomas) and 113 peripheral blood samples from healthy individuals. Compared with normal controls, the wild type TT and allele T of rs611251 showed higher frequency in gastric carcinoma (GCs), nasopharyngeal carcinomas (NPCs) and lymphomas. However, there was no significant difference between EBV-associated gastric (EBVaGC) and EBVnGC, EBV-positive NPCs and EBV-negative NPCs, EBV-related lymphomas and EBV-negative lymphomas in rs611251 of PPP1R15A. In conclusion, the PPP1R15A rs611251 polymorphism was significantly related to three kinds of tumors. Nevertheless, EBV has no obvious effect on PPP1R15A rs611251 polymorphism of NPC, GC and lymphoma. What's more, the genotype TT and allele T could be risk factors for NPC, GC and lymphoma. Our study explores the relationship between PPP1R15A gene polymorphism (rs611251) and Epstein-Barr virus-associated tumors for the first time. PPP1R15A gene SNP (rs611251) have association with multiple tumor types, which may provide some new clues to the detection and treatment of tumors.


Subject(s)
Carcinoma/genetics , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/physiology , Lymphoma/genetics , Nasopharyngeal Neoplasms/genetics , Polymorphism, Single Nucleotide , Protein Phosphatase 1/genetics , Stomach Neoplasms/genetics , Carcinoma/virology , Epstein-Barr Virus Infections/virology , Genotype , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Humans , Lymphoma/virology , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/virology , Stomach Neoplasms/virology
19.
J Biol Chem ; 290(47): 28257-28271, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26446796

ABSTRACT

In the integrated stress response, phosphorylation of eIF2α (eIF2α-P) reduces protein synthesis to conserve resources and facilitate preferential translation of transcripts that promote stress adaptation. Preferentially translated GADD34 (PPP1R15A) and constitutively expressed CReP (PPP1R15B) function to dephosphorylate eIF2α-P and restore protein synthesis. The 5'-leaders of GADD34 and CReP contain two upstream ORFs (uORFs). Using biochemical and genetic approaches we show that features of these uORFs are central for their differential expression. In the absence of stress, translation of an inhibitory uORF in GADD34 acts as a barrier that prevents reinitiation at the GADD34 coding region. Enhanced eIF2α-P during stress directs ribosome bypass of the uORF, facilitating translation of the GADD34 coding region. CReP expression occurs independent of eIF2α-P via an uORF that allows for translation reinitiation at the CReP coding region independent of stress. Importantly, alterations in the GADD34 uORF affect the status of eIF2α-P, translational control, and cell adaptation to stress. These results show that properties of uORFs that permit ribosome reinitiation are critical for directing gene-specific translational control in the integrated stress response.


Subject(s)
Protein Biosynthesis/physiology , Ribosomes/physiology , Stress, Physiological , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , DNA , Eukaryotic Initiation Factor-2/metabolism , Humans , Mice , Molecular Sequence Data , Open Reading Frames , Phosphorylation , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Sequence Homology, Amino Acid , Transcription, Genetic/physiology
20.
J Cell Biochem ; 116(7): 1300-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25737469

ABSTRACT

Cells respond to ER-stress via ER-stress sensors, leading to the UPR and subsequent apoptosis; however, occasionally, they activate autophagy without subsequent apoptosis in response to ER-stress. We previously showed that the induction of apoptosis by ER-stress was related to the presence or absence of CHOP expression; nevertheless, how ATF4 expression is elicited without downstream CHOP expression is unknown. We studied the role of GADD34 on the induction of autophagy and/or apoptosis by NaF- or tunicamycin-induced ER-stress in HepG2 cells transfected with GADD34 siRNA. Although NaF and tunicamycin both induced PERK activation followed by eIF2α phosphorylation and ATF4 expression, CHOP expression was only induced by tunicamycin. Concomitant with the signaling change, autophagy was activated both by NaF and tunicamycin, and apoptosis was induced only by tunicamycin. After 4 h, GADD34 mRNA expression was also increased by NaF and tunicamycin. Suppression of GADD34 by GADD34 siRNA increased ATF4 expression in both NaF- and tunicamycin-treated cells. The GADD34 siRNA increased CHOP expression, which corresponded to increased ATF4 in tunicamycin-treated cells; however, the increased ATF4 did not induce CHOP expression in NaF-treated cells. In concert with signal changes, siRNA treatment additively increased the autophagic activity of both NaF- and tunicamycin-treated cells; however, apoptosis was produced and accelerated only for tunicamycin-treated cells. These findings indicate that GADD34 expression induced by ER-stress delays CHOP expression and retards apoptotic cell death, and that an ATF4-signal-modulating machine other than GADD34 acts on ATF4-to-CHOP signaling to block ATF4-induced CHOP expression in ER-stress related autophagy.


Subject(s)
Activating Transcription Factor 4/genetics , Autophagy , Endoplasmic Reticulum Stress , Signal Transduction , Hep G2 Cells , Humans , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Sodium Fluoride/pharmacology , Transcription Factor CHOP/genetics , Tunicamycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL