Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.853
Filter
1.
Cell Death Dis ; 15(8): 587, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138189

ABSTRACT

The unfolded protein response (UPR) is a conserved and adaptive intracellular pathway that relieves the endoplasmic reticulum (ER) stress by activating ER transmembrane stress sensors. As a consequence of ER stress, the inhibition of nonsense-mediated mRNA decay (NMD) is due to an increase in the phosphorylation of eIF2α, which has the effect of inhibiting translation. However, the role of NMD in maintaining ER homeostasis remains unclear. In this study, we found that the three NMD factors, up-frameshift (UPF)1, UPF2, or UPF3B, were required to negate the UPR. Among these three NMD factors, only UPF3B interacted with inositol-requiring enzyme-1α (IRE1α). This interaction inhibited the kinase activity of IRE1α, abolished autophosphorylation, and reduced IRE1α clustering for ER stress. BiP and UPF3B jointly control the activation of IRE1α on both sides of the ER membrane. Under stress conditions, the phosphorylation of UPF3B was increased and the phosphorylated sites were identified. Both the UPF3BY160D genetic mutation and phosphorylation at Thr169 of UPF3B abolished its interaction with IRE1α and UPF2, respectively, leading to activation of ER stress and NMD dysfunction. Our study reveals a key physiological role for UPF3B in the reciprocal regulatory relationship between NMD and ER stress.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Endoribonucleases/metabolism , Phosphorylation , HeLa Cells , Nonsense Mediated mRNA Decay , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Unfolded Protein Response , HEK293 Cells , Protein Binding , Endoplasmic Reticulum/metabolism
2.
Front Immunol ; 15: 1358462, 2024.
Article in English | MEDLINE | ID: mdl-39100663

ABSTRACT

The double-stranded DNA (dsDNA) sensor STING has been increasingly implicated in responses to "sterile" endogenous threats and pathogens without nominal DNA or cyclic di-nucleotide stimuli. Previous work showed an endoplasmic reticulum (ER) stress response, known as the unfolded protein response (UPR), activates STING. Herein, we sought to determine if ER stress generated a STING ligand, and to identify the UPR pathways involved. Induction of IFN-ß expression following stimulation with the UPR inducer thapsigargin (TPG) or oxygen glucose deprivation required both STING and the dsDNA-sensing cyclic GMP-AMP synthase (cGAS). Furthermore, TPG increased cytosolic mitochondrial DNA, and immunofluorescence visualized dsDNA punctae in murine and human cells, providing a cGAS stimulus. N-acetylcysteine decreased IFN-ß induction by TPG, implicating reactive oxygen species (ROS). However, mitoTEMPO, a mitochondrial oxidative stress inhibitor did not impact TPG-induced IFN. On the other hand, inhibiting the inositol requiring enzyme 1 (IRE1) ER stress sensor and its target transcription factor XBP1 decreased the generation of cytosolic dsDNA. iNOS upregulation was XBP1-dependent, and an iNOS inhibitor decreased cytosolic dsDNA and IFN-ß, implicating ROS downstream of the IRE1-XBP1 pathway. Inhibition of the PKR-like ER kinase (PERK) pathway also attenuated cytoplasmic dsDNA release. The PERK-regulated apoptotic factor Bim was required for both dsDNA release and IFN-ß mRNA induction. Finally, XBP1 and PERK pathways contributed to cytosolic dsDNA release and IFN-induction by the RNA virus, Vesicular Stomatitis Virus (VSV). Together, our findings suggest that ER stressors, including viral pathogens without nominal STING or cGAS ligands such as RNA viruses, trigger multiple canonical UPR pathways that cooperate to activate STING and downstream IFN-ß via mitochondrial dsDNA release.


Subject(s)
Cytosol , Endoplasmic Reticulum Stress , Interferon-beta , Membrane Proteins , Nucleotidyltransferases , Unfolded Protein Response , Humans , Animals , Mice , Nucleotidyltransferases/metabolism , Cytosol/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Interferon-beta/metabolism , DNA/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , eIF-2 Kinase/metabolism , Endoribonucleases/metabolism , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Thapsigargin/pharmacology , Reactive Oxygen Species/metabolism , Transcriptional Activation , DNA, Mitochondrial/metabolism
3.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39103227

ABSTRACT

The complex multistep activation cascade of Ire1 involves changes in the Ire1 conformation and oligomeric state. Ire1 activation enhances ER folding capacity, in part by overexpressing the ER Hsp70 molecular chaperone BiP; in turn, BiP provides tight negative control of Ire1 activation. This study demonstrates that BiP regulates Ire1 activation through a direct interaction with Ire1 oligomers. Particularly, we demonstrated that the binding of Ire1 luminal domain (LD) to unfolded protein substrates not only trigger conformational changes in Ire1-LD that favour the formation of Ire1-LD oligomers but also exposes BiP binding motifs, enabling the molecular chaperone BiP to directly bind to Ire1-LD in an ATP-dependent manner. These transient interactions between BiP and two short motifs in the disordered region of Ire1-LD are reminiscent of interactions between clathrin and another Hsp70, cytoplasmic Hsc70. BiP binding to substrate-bound Ire1-LD oligomers enables unfolded protein substrates and BiP to synergistically and dynamically control Ire1-LD oligomerisation, helping to return Ire1 to its deactivated state when an ER stress response is no longer required.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Endoribonucleases , Heat-Shock Proteins , Protein Binding , Protein Serine-Threonine Kinases , Protein Serine-Threonine Kinases/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/chemistry , Endoribonucleases/metabolism , Endoribonucleases/chemistry , Humans , Endoplasmic Reticulum/metabolism , Unfolded Protein Response , Protein Multimerization , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , Protein Folding , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Protein Domains
4.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126106

ABSTRACT

RNase Y is a key endoribonuclease that regulates global mRNA turnover and processing in Bacillus subtilis and likely many other bacteria. This enzyme is anchored to the cell membrane, creating a pseudo-compartmentalization that aligns with its role in initiating the decay of mRNAs primarily translated at the cell periphery. However, the reasons behind and the consequences of RNase Y's membrane attachment remain largely unknown. In our study, we examined a strain expressing wild-type levels of a cytoplasmic form of RNase Y from its chromosomal locus. This strain exhibits a slow-growth phenotype, similar to that of an RNase Y null mutant. Genome-wide data reveal a significant impact on the expression of hundreds of genes. While certain RNA substrates clearly depend on RNase Y's membrane attachment, others do not. We observed no correlation between mRNA stabilization in the mutant strains and the cellular location or function of the encoded proteins. Interestingly, the Y-complex, a specificity factor for RNase Y, also appears also recognize the cytoplasmic form of the enzyme, restoring wild-type levels of the corresponding transcripts. We propose that membrane attachment of RNase Y is crucial for its functional interaction with many coding and non-coding RNAs, limiting the cleavage of specific substrates, and potentially avoiding unfavorable competition with other ribonucleases like RNase J, which shares a similar evolutionarily conserved cleavage specificity.


Subject(s)
Bacillus subtilis , Bacterial Proteins , Cell Membrane , Gene Expression Regulation, Bacterial , Bacillus subtilis/genetics , Bacillus subtilis/enzymology , Bacillus subtilis/metabolism , Cell Membrane/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , RNA Stability , Endoribonucleases/metabolism , Endoribonucleases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
PLoS Genet ; 20(8): e1011349, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39088561

ABSTRACT

Cellular processes require precise and specific gene regulation, in which continuous mRNA degradation is a major element. The mRNA degradation mechanisms should be able to degrade a wide range of different RNA substrates with high efficiency, but should at the same time be limited, to avoid killing the cell by elimination of all cellular RNA. RNase Y is a major endoribonuclease found in most Firmicutes, including Bacillus subtilis and Staphylococcus aureus. However, the molecular interactions that direct RNase Y to cleave the correct RNA molecules at the correct position remain unknown. In this work we have identified transcripts that are homologs in S. aureus and B. subtilis, and are RNase Y targets in both bacteria. Two such transcript pairs were used as models to show a functional overlap between the S. aureus and the B. subtilis RNase Y, which highlighted the importance of the nucleotide sequence of the RNA molecule itself in the RNase Y targeting process. Cleavage efficiency is driven by the primary nucleotide sequence immediately downstream of the cleavage site and base-pairing in a secondary structure a few nucleotides downstream. Cleavage positioning is roughly localised by the downstream secondary structure and fine-tuned by the nucleotide immediately upstream of the cleavage. The identified elements were sufficient for RNase Y-dependent cleavage, since the sequence elements from one of the model transcripts were able to convert an exogenous non-target transcript into a target for RNase Y.


Subject(s)
Bacillus subtilis , Gene Expression Regulation, Bacterial , RNA Cleavage , RNA Stability , RNA, Bacterial , Staphylococcus aureus , Staphylococcus aureus/genetics , Staphylococcus aureus/enzymology , Bacillus subtilis/genetics , Bacillus subtilis/enzymology , Bacillus subtilis/metabolism , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Nucleic Acid Conformation , Base Sequence
6.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000233

ABSTRACT

The pathogenesis of non-alcoholic fatty liver disease (NAFLD) is influenced by a number of variables, including endoplasmic reticulum stress (ER). Thioredoxin domain-containing 5 (TXNDC5) is a member of the protein disulfide isomerase family and acts as an endoplasmic reticulum (ER) chaperone. Nevertheless, the function of TXNDC5 in hepatocytes under ER stress remains largely uncharacterized. In order to identify the role of TXNDC5 in hepatic wild-type (WT) and TXNDC5-deficient (KO) AML12 cell lines, tunicamycin, palmitic acid, and thapsigargin were employed as stressors. Cell viability, mRNA, protein levels, and mRNA splicing were then assayed. The protein expression results of prominent ER stress markers indicated that the ERN1 and EIF2AK3 proteins were downregulated, while the HSPA5 protein was upregulated. Furthermore, the ATF6 protein demonstrated no significant alterations in the absence of TXNDC5 at the protein level. The knockout of TXNDC5 has been demonstrated to increase cellular ROS production and its activity is required to maintain normal mitochondrial function during tunicamycin-induced ER stress. Tunicamycin has been observed to disrupt the protein levels of HSPA5, ERN1, and EIF2AK3 in TXNDC5-deficient cells. However, palmitic acid has been observed to disrupt the protein levels of ATF6, HSPA5, and EIF2AK3. In conclusion, TXNDC5 can selectively activate distinct ER stress pathways via HSPA5, contingent on the origin of ER stress. Conversely, the absence of TXNDC5 can disrupt the EIF2AK3 cascade.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Hepatocytes , Protein Disulfide-Isomerases , Signal Transduction , Tunicamycin , Endoplasmic Reticulum Chaperone BiP/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/genetics , Hepatocytes/metabolism , Animals , Tunicamycin/pharmacology , Endoplasmic Reticulum/metabolism , Mice , Reactive Oxygen Species/metabolism , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/genetics , Cell Line , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Palmitic Acid/pharmacology , Palmitic Acid/metabolism , Thapsigargin/pharmacology , Humans , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Thioredoxins/metabolism , Thioredoxins/genetics , Cell Survival/drug effects
7.
Front Immunol ; 15: 1427859, 2024.
Article in English | MEDLINE | ID: mdl-39026685

ABSTRACT

Endoplasmic reticulum stress occurs due to large amounts of misfolded proteins, hypoxia, nutrient deprivation, and more. The unfolded protein is a complex intracellular signaling network designed to operate under this stress. Composed of three individual arms, inositol-requiring enzyme 1, protein kinase RNA-like ER kinase, and activating transcription factor-6, the unfolded protein response looks to resolve stress and return to proteostasis. The CD8+ T cell is a critical cell type for the adaptive immune system. The unfolded protein response has been shown to have a wide-ranging spectrum of effects on CD8+ T cells. CD8+ T cells undergo cellular stress during activation and due to environmental insults. However, the magnitude of the effects this response has on CD8+ T cells is still understudied. Thus, studying these pathways is important to unraveling the inner machinations of these powerful cells. In this review, we will highlight the recent literature in this field, summarize the three pathways of the unfolded protein response, and discuss their roles in CD8+ T cell biology and functionality.


Subject(s)
CD8-Positive T-Lymphocytes , Endoplasmic Reticulum Stress , Signal Transduction , Unfolded Protein Response , Unfolded Protein Response/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Animals , Endoplasmic Reticulum Stress/immunology , Protein Serine-Threonine Kinases/metabolism , Activating Transcription Factor 6/metabolism , Endoribonucleases/metabolism , Endoribonucleases/immunology , Lymphocyte Activation/immunology
8.
Zhen Ci Yan Jiu ; 49(7): 686-692, 2024 Jul 25.
Article in English, Chinese | MEDLINE | ID: mdl-39020486

ABSTRACT

OBJECTIVES: To investigate the mechanism of the effect of acupuncture and moxibustion on improving liver injury in cisplatin (DDP) induced liver injury model mice by observing the changes of inositol-requiring enzyme (IRE) -1 signaling pathway. METHODS: Forty KM mice were randomly divided into control, model, acupuncture and moxibustion groups, with 10 mice in each group. The liver injury model was replicated by intraperitoneal injection of DDP (10 mg/kg). In the acupuncture group and the moxibustion group, acupuncture and moxibustion were performed at "Dazhui"(GV14), and bilateral "Ganshu"(BL18), "Shenshu" (BL23), and "Zusanli"(ST36), respectively for 6 min, once per day for 7 d. The apoptosis of hepatocytes was detected by TUNEL staining. The expression of phosphorylation(p)-IRE-1α, glucose-regulated protein (Grp) 78 and cysteine aspartic protease (Caspase) -12 in liver tissue were detected by immunohistochemistry and Western blot, respectively. The expression levels of Grp78 and Caspase-12 mRNA in liver tissue were detected by quantitative real-time PCR. RESULTS: Compared with the control group, the apoptosis rate of hepatocytes was increased (P<0.05), the positive expression and protein expression of p-IRE-1α, Grp78, and Caspase-12 were increased (P<0.05), the expression levels of Grp78 and Caspase-12 mRNA were increased (P<0.05) in the model group. Compared with the model group, all these indicators showed opposite trends (P<0.05) in the acupuncture and moxibustion groups. CONCLUSIONS: Acupuncture and moxibustion can reduce liver injury due to DDP chemotherapy by modulating IRE-1 signaling pathway, inhibiting the excessive activation of endoplasmic reticulum stress, and reducing liver cell apoptosis.


Subject(s)
Acupuncture Therapy , Apoptosis , Cisplatin , Endoplasmic Reticulum Chaperone BiP , Liver , Moxibustion , Protein Serine-Threonine Kinases , Signal Transduction , Animals , Mice , Male , Humans , Liver/metabolism , Liver/injuries , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Chemical and Drug Induced Liver Injury/therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/genetics , Acupuncture Points , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Caspase 12/metabolism , Caspase 12/genetics , Hepatocytes/metabolism
9.
J Clin Invest ; 134(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949019

ABSTRACT

Type 3 innate lymphoid cells (ILC3s) are key regulators of intestinal homeostasis and epithelial barrier integrity. In this issue of the JCI, Cao and colleagues found that a sensor of endoplasmic reticulum (ER) stress, the inositol-requiring kinase 1α/X-box-binding protein 1 (IRE1α/XBP1) pathway, fine-tuned the functions of ILC3s. Activation of IRE1α and XBP1 in ILC3s limited intestinal inflammation in mice and correlated with the efficacy of ustekinumab, an IL-12/IL-23 blocker, in patients with Crohn's disease. These results advance our understanding in the use of ILCs as biomarkers not only to predict disease outcomes but also to indicate the response to biologicals in patients with inflammatory bowel disease.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/immunology , Animals , Endoribonucleases/metabolism , Endoribonucleases/genetics , Endoribonucleases/immunology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Humans , Mice , Endoplasmic Reticulum Stress/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Signal Transduction/immunology , Crohn Disease/immunology , Crohn Disease/pathology , Crohn Disease/metabolism , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology
10.
Discov Med ; 36(186): 1370-1377, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054708

ABSTRACT

BACKGROUND: Glucose and fatty acid overload-induced glucolipid toxicity of pancreatic ß-cells is associated with the development of diabetes. Endoplasmic reticulum stress (ERS) plays an essential role in this process. Ghrelin, a peptide secreted by the pancreas, negatively correlates with oxidative stress. The study aimed to investigate ghrelin's role in glycolipid-induced ß-cell dysfunction and its possible mechanism. METHODS: Mouse insulinoma ß-cell, NIT-1 cells, were stimulated with high fat and high glucose to induce glucolipid toxicity. High fat and high glucose-induced NIT-1 cells were treated with acylated ghrelin (AG) or [d-Lys3]-growth hormone releasing peptide (GHRP)-6. Flow cytometry and Cell Counting Kit-8 (CCK-8) assay were performed to assess apoptosis and cell viability. The protein expression related to apoptosis, inositol-requiring kinase 1 (IRE1)/c-Jun N-terminal kinase (JNK) signaling, and ERS were investigated using western blot. Enzyme-linked immunosorbent assay (ELISA) was adopted to examine insulin's synthesis and secretion levels. RESULTS: Ghrelin treatment improved cell viability while inhibiting cell glucolipotoxicity-induced NIT-1 cell apoptosis. Ghrelin can promote the synthesis and secretion of insulin in NIT-1 cells. Mechanistically, ghrelin attenuates ERS and inhibits the IRE1/JNK signaling pathway in NIT-1 cells induced by glucolipotoxicity. CONCLUSION: Ghrelin improves ß-cellular dysfunction induced by glucolipotoxicity by inhibiting the IRE1/JNK pathway induced by ERS. It could be an effective treatment for ß-cellular dysfunction.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Endoribonucleases , Ghrelin , Insulin-Secreting Cells , Protein Serine-Threonine Kinases , Animals , Mice , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Ghrelin/pharmacology , Ghrelin/metabolism , Glucose , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , MAP Kinase Signaling System/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects
11.
J Am Chem Soc ; 146(31): 21525-21534, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39047145

ABSTRACT

Various approaches have been developed to target RNA and modulate its function with modes of action including binding and cleavage. Herein, we explored how small molecule binding is correlated with cleavage induced by heterobifunctional ribonuclease targeting chimeras (RiboTACs), where RNase L is recruited to cleave the bound RNA target, in a transcriptome-wide, unbiased fashion. Only a fraction of bound targets was cleaved by RNase L, induced by RiboTAC binding. Global analysis suggested that (i) cleaved targets generally form a region of stable structure that encompasses the small molecule binding site; (ii) cleaved targets have preferred RNase L cleavage sites nearby small molecule binding sites; (iii) RiboTACs facilitate a cellular interaction between cleaved targets and RNase L; and (iv) the expression level of the target influences the extent of cleavage observed. In one example, we converted a binder of LGALS1 (galectin-1) mRNA into a RiboTAC. In MDA-MB-231 cells, the binder had no effect on galectin-1 protein levels, while the RiboTAC cleaved LGALS1 mRNA, reduced galectin-1 protein abundance, and affected galectin-1-associated oncogenic cellular phenotypes. Using LGALS1, we further assessed additional factors including the length of the linker that tethers the two components of the RiboTAC, cellular uptake, and the RNase L-recruiting module on RiboTAC potency. Collectively, these studies may facilitate triangulation of factors to enable the design of RiboTACs.


Subject(s)
Transcriptome , Humans , Cell Line, Tumor , Endoribonucleases/metabolism , Endoribonucleases/chemistry , RNA, Messenger/metabolism , RNA, Messenger/genetics , Ribonucleases/metabolism , Ribonucleases/chemistry
12.
EMBO Rep ; 25(8): 3627-3650, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38982191

ABSTRACT

Skeletal muscle regeneration involves a signaling network that regulates the proliferation, differentiation, and fusion of muscle precursor cells to injured myofibers. IRE1α, one of the arms of the unfolded protein response, regulates cellular proteostasis in response to ER stress. Here, we demonstrate that inducible deletion of IRE1α in satellite cells of mice impairs skeletal muscle regeneration through inhibiting myoblast fusion. Knockdown of IRE1α or its downstream target, X-box protein 1 (XBP1), also inhibits myoblast fusion during myogenesis. Transcriptome analysis revealed that knockdown of IRE1α or XBP1 dysregulates the gene expression of molecules involved in myoblast fusion. The IRE1α-XBP1 axis mediates the gene expression of multiple profusion molecules, including myomaker (Mymk). Spliced XBP1 (sXBP1) transcription factor binds to the promoter of Mymk gene during myogenesis. Overexpression of myomaker in IRE1α-knockdown cultures rescues fusion defects. Inducible deletion of IRE1α in satellite cells also inhibits myoblast fusion and myofiber hypertrophy in response to functional overload. Collectively, our study demonstrates that IRE1α promotes myoblast fusion through sXBP1-mediated up-regulation of the gene expression of multiple profusion molecules, including myomaker.


Subject(s)
Cell Fusion , Endoribonucleases , Muscle Development , Muscle, Skeletal , Myoblasts , Protein Serine-Threonine Kinases , Signal Transduction , X-Box Binding Protein 1 , Animals , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mice , Myoblasts/metabolism , Myoblasts/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle Development/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Satellite Cells, Skeletal Muscle/metabolism , Regeneration/genetics , Cell Differentiation/genetics , Gene Expression Regulation , Membrane Proteins , Muscle Proteins
13.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062922

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder which affects dopaminergic neurons of the midbrain. Accumulation of α-synuclein or exposure to neurotoxins like 6-hydroxydopamine (6-OHDA) induces endoplasmic reticulum (ER) stress along with the unfolded protein response (UPR), which executes apoptosis via activation of PERK/CHOP or IRE1/JNK signaling. The present study aimed to determine which of these pathways is a major contributor to neurodegeneration in an 6-OHDA-induced in vitro model of PD. For this purpose, we have applied pharmacological PERK and JNK inhibitors (AMG44 and JNK V) in differentiated SH-SY5Y cells exposed to 6-OHDA. Inhibition of PERK and JNK significantly decreased genotoxicity and improved mitochondrial respiration, but only JNK inhibition significantly increased cell viability. Gene expression analysis revealed that the effect of JNK inhibition was dependent on a decrease in MAPK10 and XBP1 mRNA levels, whereas inhibition of either PERK or JNK significantly reduced the expression of DDIT3 mRNA. Western blot has shown that JNK inhibition strongly induced the XBP1s protein, and inhibition of each pathway attenuated the phosphorylation of eIF2α and JNK, as well as the expression of CHOP. Collectively, our data suggests that targeting the IRE1/JNK pathway of the UPR is a more effective option for PD treatment as it simultaneously affects more than one pro-apoptotic pathway.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Oxidopamine , Protein Serine-Threonine Kinases , Transcription Factor CHOP , Unfolded Protein Response , eIF-2 Kinase , Humans , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , eIF-2 Kinase/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Endoribonucleases/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 10/genetics , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Unfolded Protein Response/drug effects , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics
14.
Arch Biochem Biophys ; 759: 110104, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39059599

ABSTRACT

BACKGROUND: Endoplasmic reticulum stress and synthesis of serine are essential for tumor growth, but the mechanism of their interaction is not clarified yet. The overarching goal of this work was to investigate the impact of ERN1 (endoplasmic reticulum to nucleus signaling 1) inhibition on the expression of serine synthesis genes in U87MG glioblastoma cells concerning the suppression of cell proliferation. METHODS: Wild type U87MG glioblastoma cells and their clones with overexpression of transgenes dnERN1 (without cytoplasmic domain of ERN1) and dnrERN1 (with mutation in endoribonuclease of ERN1), and empty vector (as control) were used. The silencing of ERN1 and XBP1 was also used to inhibition of ERN1 and its function. Gene expression was measured by qPCR. RESULTS: We show that the expression of PSAT1 and several other related to serine synthesis genes is suppressed in cells with ERN1 inhibition by dissimilar mechanisms: PHGDH gene through ERN1 protein kinase, because its expression was resistant to inhibition of ERN1 endoribonuclease, but ATF4 gene via endoribonuclease of ERN1. However, in the control of PSAT1 and PSPH genes both enzymatic activities of ERN1 signaling protein are involved. At the same time, ERN1 knockdown strongly increased SHMT1 expression, which controls serine metabolism and enhances the proliferation and invasiveness of glioma cells. The level of microRNAs, which have binding sites in PSAT1, SHMT1, and PSPH mRNAs, was also changed in cells harboring dnERN1 transgene. Inhibition of ERN1 suppressed cell proliferation and enzymatic activity of PHGDH, a rate-limiting enzyme for serine synthesis. CONCLUSION: Changes in the expression of phosphoserine aminotransferase 1 and other genes related to serine synthesis are mediated by diverse ERN1-dependent mechanisms and contributed to suppressed proliferation and enhanced invasiveness of ERN1 knockdown glioblastoma cell.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma , Protein Serine-Threonine Kinases , Transaminases , Humans , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Transaminases/genetics , Transaminases/metabolism , Endoribonucleases/metabolism , Endoribonucleases/genetics , Gene Knockdown Techniques , Serine/metabolism , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics
15.
Sci Total Environ ; 949: 175040, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39079638

ABSTRACT

Trichloromethane (TCM), a commonly recognized disinfection by-product formed during the chlorination of water, has been associated with the onset of colorectal cancer (CRC) in humans. Despite this, the impact of TCM on the progression of CRC remains uncertain. In this investigation, it was observed that exposure to TCM could augment the migratory capabilities of CRC cells and facilitate the advancement of colorectal tumors. To delve deeper into the mechanism responsible for TCM-induced CRC progression, we performed RNA-Seq analysis at cellular and animal levels after TCM exposure. Both the KEGG and GO enrichment analyses indicated the activation of endoplasmic reticulum stress (ERS) and the regulation of the cytoskeleton. Subsequently, we confirmed the activation of the IRE1α/XBP1 pathway of ERS through western blot and RT-qPCR. Additionally, we observed the aggregation of cytoskeletal proteins F-actin and ß-tubulin at the cell membrane periphery and the development of cellular pseudopods using immunofluorescence following exposure to TCM in vitro. The downregulation of IRE1α and XBP1 through siRNA interference resulted in the disruption of cell cytoskeleton rearrangement and impaired cell migration capability. Conversely, treatment with TCM mitigated this inhibitory effect. Moreover, chronic exposure to low concentration of TCM also triggered CRC cell migration by causing cytoskeletal reorganization, a process controlled by the IRE1α/XBP1 axis. Our study concludes that TCM exposure induces cell migration through the activation of ERS, which in turn regulates cytoskeleton rearrangement. This study offers novel insights into the mechanism through which TCM facilitates the progression of CRC.


Subject(s)
Chloroform , Colorectal Neoplasms , Endoplasmic Reticulum Stress , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , Animals , Humans , Mice , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Endoribonucleases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Water Pollutants, Chemical/toxicity , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Chloroform/toxicity , Drinking Water
16.
Antiviral Res ; 228: 105921, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38825019

ABSTRACT

The SARS-CoV-2 pandemic has bolstered unprecedented research efforts to better understand the pathogenesis of coronavirus (CoV) infections and develop effective therapeutics. We here focus on non-structural protein nsp15, a hexameric component of the viral replication-transcription complex (RTC). Nsp15 possesses uridine-specific endoribonuclease (EndoU) activity for which some specific cleavage sites were recently identified in viral RNA. By preventing accumulation of viral dsRNA, EndoU helps the virus to evade RNA sensors of the innate immune response. The immune-evading property of nsp15 was firmly established in several CoV animal models and makes it a pertinent target for antiviral therapy. The search for nsp15 inhibitors typically proceeds via compound screenings and is aided by the rapidly evolving insight in the protein structure of nsp15. In this overview, we broadly cover this fascinating protein, starting with its structure, biochemical properties and functions in CoV immune evasion. Next, we summarize the reported studies in which compound screening or a more rational method was used to identify suitable leads for nsp15 inhibitor development. In this way, we hope to raise awareness on the relevance and druggability of this unique CoV protein.


Subject(s)
Antiviral Agents , COVID-19 , Endoribonucleases , SARS-CoV-2 , Viral Nonstructural Proteins , Antiviral Agents/pharmacology , Endoribonucleases/metabolism , Endoribonucleases/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Humans , SARS-CoV-2/drug effects , Animals , COVID-19/virology , Virus Replication/drug effects , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Betacoronavirus/drug effects , COVID-19 Drug Treatment , Pandemics , RNA, Viral/genetics , Immune Evasion
17.
Mol Biol Cell ; 35(8): ar104, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38865176

ABSTRACT

RNase E is the most common RNA decay nuclease in bacteria, setting the global mRNA decay rate and scaffolding formation of the RNA degradosome complex and BR-bodies. To properly set the global mRNA decay rate, RNase E from Escherichia coli and neighboring γ-proteobacteria were found to autoregulate RNase E levels via the decay of its mRNA's 5' untranslated region (UTR). While the 5' UTR is absent from other groups of bacteria in the Rfam database, we identified that the α-proteobacterium Caulobacter crescentus RNase E contains a similar 5' UTR structure that promotes RNase E autoregulation. In both bacteria, the C-terminal intrinsically disordered region (IDR) of RNase E is required for proper autoregulation to occur, and this IDR is also necessary and sufficient for RNase E to phase-separate, generating BR-bodies. Using in vitro purified RNase E, we find that the IDR's ability to promote phase separation correlates with enhanced 5' UTR cleavage, suggesting that phase separation of RNase E with the 5' UTR enhances autoregulation. Finally, using growth competition experiments, we find that a strain capable of autoregulation rapidly outcompetes a strain with a 5' UTR mutation that cannot autoregulate, suggesting autoregulation promotes optimal cellular fitness.


Subject(s)
5' Untranslated Regions , Caulobacter crescentus , Endoribonucleases , Homeostasis , RNA Stability , Caulobacter crescentus/metabolism , Caulobacter crescentus/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , 5' Untranslated Regions/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Polyribonucleotide Nucleotidyltransferase/metabolism , Polyribonucleotide Nucleotidyltransferase/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Multienzyme Complexes , RNA Helicases
18.
Endocr Regul ; 58(1): 144-152, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38861539

ABSTRACT

Objective. Serine hydroxymethyltransferase (SHMT2) plays a multifunctional role in mitochondria (folate-dependent tRNA methylation, translation, and thymidylate synthesis). The endoplasmic reticulum stress, hypoxia, and glucose and glutamine supply are significant factors of malignant tumor growth including glioblastoma. Previous studies have shown that the knockdown of the endoplasmic reticulum to nucleus signaling 1 (ERN1) pathway of endoplasmic reticulum stress strongly suppressed glioblastoma cell proliferation and modified the sensitivity of these cells to hypoxia and glucose or glutamine deprivations. The present study aimed to investigate the regulation of the SHMT2 gene in U87MG glioblastoma cells by ERN1 knockdown, hypoxia, and glucose or glutamine deprivations with the intent to reveal the role of ERN1 signaling in sensitivity of this gene expression to hypoxia and nutrient supply. Methods. The control U87MG glioblastoma cells (transfected by an empty vector) and ERN1 knockdown cells with inhibited ERN1 endoribonuclease and protein kinase (dnERN1) or only ERN1 endoribonuclease (dnrERN1) were used. Hypoxia was introduced by dimethyloxalylglycine (500 ng/ml for 4 h). For glucose and glutamine deprivations, cells were exposed in DMEM without glucose and glutamine, respectively for 16 h. RNA was extracted from cells and reverse transcribed. The expression level of the SHMT2 gene was studied by real-time qPCR and normalized to ACTB. Results. It was found that inhibition of ERN1 endoribonuclease and protein kinase in glioblastoma cells led to a down-regulation of SHMT2 gene expression in U87MG cells. At the same time, the expression of this gene did not significantly change in cells with inhibited ERN1 endoribonuclease, but tunicamycin strongly increased its expression. Moreover, the expression of the SHMT2 gene was not affected in U87MG cells after silencing of XBP1. Hypoxia up-regulated the expression level of the SHMT2 gene in both control and ERN1 knockdown U87MG cells. The expression of this gene was significantly up-regulated in glioblastoma cells under glucose and glutamine deprivations and ERN1 knockdown significantly increased the sensitivity of the SHMT2 gene to these nutrient deprivation conditions. Conclusion. The results of the present study demonstrate that the expression of the SHMT2 gene responsible for serine metabolism and formation of folate one-carbon is controlled by ERN1 protein kinase and induced by hypoxia as well as glutamine and glucose deprivation conditions in glioblastoma cells and reflects the ERN1-mediated reprogramming of sensitivity this gene expression to nutrient deprivation.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Gene Expression Regulation, Neoplastic , Glioblastoma , Glycine Hydroxymethyltransferase , Humans , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum Stress/genetics , Cell Line, Tumor , Endoribonucleases/genetics , Endoribonucleases/metabolism , Glucose/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Hypoxia/physiology , Cell Hypoxia/genetics , Glutamine/metabolism , Gene Knockdown Techniques
19.
Nucleic Acids Res ; 52(13): 7809-7824, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38874491

ABSTRACT

RNA helicases-central enzymes in RNA metabolism-often feature intrinsically disordered regions (IDRs) that enable phase separation and complex molecular interactions. In the bacterial pathogen Pseudomonas aeruginosa, the non-redundant RhlE1 and RhlE2 RNA helicases share a conserved REC catalytic core but differ in C-terminal IDRs. Here, we show how the IDR diversity defines RhlE RNA helicase specificity of function. Both IDRs facilitate RNA binding and phase separation, localizing proteins in cytoplasmic clusters. However, RhlE2 IDR is more efficient in enhancing REC core RNA unwinding, exhibits a greater tendency for phase separation, and interacts with the RNase E endonuclease, a crucial player in mRNA degradation. Swapping IDRs results in chimeric proteins that are biochemically active but functionally distinct as compared to their native counterparts. The RECRhlE1-IDRRhlE2 chimera improves cold growth of a rhlE1 mutant, gains interaction with RNase E and affects a subset of both RhlE1 and RhlE2 RNA targets. The RECRhlE2-IDRRhlE1 chimera instead hampers bacterial growth at low temperatures in the absence of RhlE1, with its detrimental effect linked to aberrant RNA droplets. By showing that IDRs modulate both protein core activities and subcellular localization, our study defines the impact of IDR diversity on the functional differentiation of RNA helicases.


Subject(s)
Bacterial Proteins , Endoribonucleases , Intrinsically Disordered Proteins , Pseudomonas aeruginosa , RNA Helicases , RNA Helicases/metabolism , RNA Helicases/genetics , RNA Helicases/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Endoribonucleases/metabolism , Endoribonucleases/chemistry , Endoribonucleases/genetics , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/genetics , Protein Binding
20.
Cell Rep ; 43(6): 114287, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38823018

ABSTRACT

Viral infection triggers several double-stranded RNA (dsRNA) sensors that lead to changes in gene expression in the cell. One of these sensors activates an endonuclease, ribonuclease L (RNase L), that cleaves single-stranded RNA. However, how the resultant widespread RNA fragmentation affects gene expression is not fully understood. Here, we show that this fragmentation induces the ribotoxic stress response via ZAKα, potentially through stalled ribosomes and/or ribosome collisions. The p38 and JNK pathways that are activated as part of this response promote outcomes that inhibit the virus, such as programmed cell death. We also show that RNase L limits the translation of stress-responsive genes. Intriguingly, we found that the activity of the generic endonuclease, RNase A, recapitulates many of the same molecular phenotypes as activated RNase L, demonstrating how widespread RNA cleavage can evoke an antiviral program.


Subject(s)
Endoribonucleases , Immunity, Innate , Endoribonucleases/metabolism , Endoribonucleases/genetics , Humans , RNA Cleavage , Animals , RNA, Double-Stranded/metabolism , Mice , Ribonuclease, Pancreatic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL