Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.245
Filter
1.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3837-3847, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39099357

ABSTRACT

The study investigates the therapeutic effects and mechanisms of ginsenoside Rg_1(GRg_1) on sepsis-induced acute lung injury(SALI). A murine model of SALI was created using cecal ligation and puncture(CLP) surgery, and mice were randomly assigned to groups for GRg_1 intervention. Survival and body weight changes were recorded, lung function was assessed with a non-invasive lung function test system, and lung tissue damage was evaluated through HE staining. The content and expression of inflammatory factors were measured by ELISA and qRT-PCR. Apoptosis was examined using flow cytometry and TUNEL staining. The activation and expression of apoptosis-related molecules cysteinyl aspartate specific proteinase 3(caspase-3), B-cell lymphoma-2(Bcl-2), Bcl-2 associated X protein(Bax), and endoplasmic reticulum stress-related molecules protein kinase R-like endoplasmic reticulum kinase(PERK), eukaryotic initiation factor 2α(eIF2α), activating transcription factor 4(ATF4), and C/EBP homologous protein(CHOP) were studied using Western blot and qRT-PCR. In addition, an in vitro model of lipopolysaccharide(LPS)-induced lung alveolar epithelial cell injury was used, with the application of the endoplasmic reticulum stress inducer tunicamycin to validate the action mechanism of GRg_1. RESULTS:: indicated that, when compared to the model group, GRg_1 intervention significantly enhanced the survival time of CLP mice, mitigated body weight loss, and improved impaired lung function indices. The GRg_1-treated mice also displayed reduced lung tissue pathological scores, a reduced lung tissue wet-to-dry weight ratio, and lower protein content in the bronchoalveolar lavage fluid. Serum levels of interleukin-6(IL-6), interleukin-1ß(IL-1ß), and tumor necrosis factor-α(TNF-α), as well as the mRNA expressions of these cytokines in lung tissues, were decreased. There was a notable decrease in the proportion of apopto-tic alveolar epithelial cells, and down-regulated expressions of caspase-3, Bax, PERK, eIF2α, ATF4, and CHOP and up-regulated expression of Bcl-2 were observed. In vitro findings showed that the apoptosis-lowering and apoptosis-related protein down-regulating effects of GRg_1 were significantly inhibited with the co-application of tunicamycin. Altogether, GRg_1 reduces apoptosis of alveolar epithelial cells, inhibits inflammation in the lungs, alleviates lung injury, and enhances lung function, possibly through the PERK/eIF2α/ATF4/CHOP pathway.


Subject(s)
Activating Transcription Factor 4 , Acute Lung Injury , Alveolar Epithelial Cells , Apoptosis , Eukaryotic Initiation Factor-2 , Ginsenosides , Sepsis , Transcription Factor CHOP , eIF-2 Kinase , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Ginsenosides/pharmacology , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Mice , Apoptosis/drug effects , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , Sepsis/genetics , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/genetics , Male , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Humans , Endoplasmic Reticulum Stress/drug effects , Mice, Inbred C57BL
2.
Sci Adv ; 10(32): eado7464, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110805

ABSTRACT

Self and nonself discrimination is fundamental to immunity. However, it remains largely enigmatic how the mechanisms of distinguishing nonself from self originated. As an intracellular nucleic acid sensor, protein kinase R (PKR) recognizes double-stranded RNA (dsRNA) and represents a crucial component of antiviral innate immunity. Here, we combine phylogenomic and functional analyses to show that PKR proteins probably originated from a preexisting kinase protein through acquiring dsRNA binding domains at least before the last common ancestor of jawed vertebrates during or before the Silurian period. The function of PKR appears to be conserved across jawed vertebrates. Moreover, we repurpose a protein closely related to PKR proteins into a putative dsRNA sensor, recapturing the making of PKR. Our study illustrates how a nucleic acid sensor might have originated via molecular tinkering with preexisting proteins and provides insights into the origins of innate immunity.


Subject(s)
Evolution, Molecular , Phylogeny , Vertebrates , eIF-2 Kinase , Animals , Vertebrates/genetics , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , RNA, Double-Stranded/metabolism , Immunity, Innate , Humans , Nucleic Acids/metabolism , Biological Evolution
3.
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
4.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39150520

ABSTRACT

The integrated stress response (ISR) is a vital signaling pathway initiated by four kinases, PERK, GCN2, HRI and PKR, that ensure cellular resilience and protect cells from challenges. Here, we investigated whether increasing ISR signaling could rescue diabetes-like phenotypes in a mouse model of diet-induced obesity (DIO). We show that the orally available and clinically approved GCN2 activator halofuginone (HF) can activate the ISR in mouse tissues. We found that daily oral administration of HF increases glucose tolerance whilst reducing weight gain, insulin resistance, and serum insulin in DIO mice. Conversely, the ISR inhibitor GSK2656157, used at low doses to optimize its selectivity, aggravates glucose intolerance in DIO mice. Whilst loss of function mutations in mice and humans have revealed that PERK is the essential ISR kinase that protects from diabetes, our work demonstrates the therapeutic value of increasing ISR signaling by activating the related kinase GCN2 to reduce diabetes phenotypes in a DIO mouse model.


Subject(s)
Obesity , Phenotype , Piperidines , Protein Serine-Threonine Kinases , Quinazolinones , Signal Transduction , eIF-2 Kinase , Animals , Quinazolinones/pharmacology , Piperidines/pharmacology , Mice , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Obesity/pathology , Obesity/metabolism , Obesity/prevention & control , Obesity/genetics , Signal Transduction/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mice, Inbred C57BL , Male , Insulin Resistance , Insulin/metabolism , Insulin/blood , Stress, Physiological/drug effects , Disease Models, Animal , Diet, High-Fat/adverse effects , Diabetes Mellitus/pathology , Diabetes Mellitus/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/drug therapy , Diabetes Mellitus/prevention & control , Glucose Intolerance/drug therapy , Adenine/analogs & derivatives , Indoles
5.
Viruses ; 16(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39066190

ABSTRACT

Negative-strand RNA viruses form cytoplasmic inclusion bodies (IBs) representing virus replication foci through phase separation or biomolecular condensation of viral and cellular proteins, as a hallmark of their infection. Alternatively, mammalian cells form stalled mRNA containing antiviral stress granules (SGs), as a consequence of phosphorylation of eukaryotic initiation factor 2α (eIF2α) through condensation of several RNA-binding proteins including TIA-1. Whether and how Chandipura virus (CHPV), an emerging human pathogen causing influenza-like illness, coma and death, forms IBs and evades antiviral SGs remain unknown. By confocal imaging on CHPV-infected Vero-E6 cells, we found that CHPV infection does not induce formation of distinct canonical SGs. Instead, CHPV proteins condense and co-localize together with SG proteins to form heterogeneous IBs, which ensued independent of the activation of eIF2α and eIF2α kinase, protein kinase R (PKR). Interestingly, siRNA-mediated depletion of PKR or TIA-1 significantly decreased viral transcription and virion production. Moreover, CHPV infection also caused condensation and recruitment of PKR to IBs. Compared to SGs, IBs exhibited significant rapidity in disassembly dynamics. Altogether, our study demonstrating that CHPV replication co-optimizes with SG proteins and revealing an unprecedented proviral role of TIA-1/PKR may have implications in understanding the mechanisms regulating CHPV-IB formation and designing antiviral therapeutics. Importance: CHPV is an emerging tropical pathogen reported to cause acute influenza-like illness and encephalitis in children with a very high mortality rate of ~70%. Lack of vaccines and an effective therapy against CHPV makes it a potent pathogen for causing an epidemic in tropical parts of globe. Given these forewarnings, it is of paramount importance that CHPV biology must be understood comprehensively. Targeting of host factors offers several advantages over targeting the viral components due to the generally higher mutation rate in the viral genome. In this study, we aimed at understanding the role of SGs forming cellular RNA-binding proteins in CHPV replication. Our study helps understand participation of cellular factors in CHPV replication and could help develop effective therapeutics against the virus.


Subject(s)
Inclusion Bodies, Viral , T-Cell Intracellular Antigen-1 , Virus Replication , eIF-2 Kinase , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Animals , T-Cell Intracellular Antigen-1/metabolism , T-Cell Intracellular Antigen-1/genetics , Chlorocebus aethiops , Vero Cells , Inclusion Bodies, Viral/metabolism , Humans , Stress Granules/metabolism , Inclusion Bodies/metabolism , Host-Pathogen Interactions , Cytoplasmic Granules/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Phase Separation
6.
Viruses ; 16(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39066257

ABSTRACT

Yaba monkey tumor virus (YMTV) and Tanapox virus (TPV) are members of the Yatapoxvirus genus and can infect humans and other primates. Despite the threat posed by yatapoxviruses, the factors determining their host range are poorly understood. In this study, we analyzed the ability of YMTV and TPV orthologs of vaccinia virus K3 (called 012 in YMTV and TPV), which share 75% amino acid identity with one another, to inhibit PKR from 15 different primate species. We first used a luciferase-based reporter, and found that YMTV and TPV K3 orthologs inhibited PKR in a species-specific manner and showed distinct PKR inhibition profiles. TPV 012 inhibited PKR from 11 primates, including humans, substantially better than YMTV 012. In contrast, both K3 orthologs inhibited the other four primate PKRs comparably well. Using YMTV 012 and TPV 012 hybrids, we mapped the region responsible for the differential PKR inhibition to the C- terminus of the K3 orthologs. Next, we generated chimeric vaccinia virus strains to investigate whether TPV K3 and YMTV K3 orthologs could rescue the replication of a vaccinia virus strain that lacks PKR inhibitors K3L and E3L. Virus replication in primate-derived cells generally correlated with the patterns observed in the luciferase-based assay. Together, these observations demonstrate that yatapoxvirus K3 orthologs have distinct PKR inhibition profiles and inhibit PKR in a species-specific manner, which may contribute to the differential susceptibility of primate species to yatapoxvirus infections.


Subject(s)
Yatapoxvirus , eIF-2 Kinase , Animals , Humans , Cell Line , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , eIF-2 Kinase/antagonists & inhibitors , Host Specificity , Primates , Species Specificity , Vaccinia virus/genetics , Vaccinia virus/physiology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Yatapoxvirus/genetics
7.
Arch Insect Biochem Physiol ; 116(3): e22127, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38976652

ABSTRACT

Ubiquitin-fold modifier 1 (UFM1) is attached to protein substrates through the sequential activity of an E1 (UBA5)-E2 (UFC1)-E3 (UFL1) cascade. UFL1 is the E3 ligase for UFMylation in vertebrates. However, there have been no studies on UFL1 in silkworm to date. In this study, we identified a UFL1 ortholog in Bombyx mori genome. Spatio-temporal expression profiles showed that BmUFL1 expression was high in the midgut, epidermis, and testis and in the pupa-adult stage. BmUFL1 knockdown inhibited B. mori nucleopolyhedrovirus (BmNPV) proliferation, while BmUFL1 overexpression promoted BmNPV proliferation. Mechanically, protein kinase RNA-like endoplasmic reticulum kinase (PERK) signaling and cell apoptosis are involved in BmUFL1-regulated BmNPV proliferation. Overall, these results suggest that BmUFL1 facilitates BmNPV proliferation in silkworm.


Subject(s)
Apoptosis , Bombyx , Insect Proteins , Nucleopolyhedroviruses , eIF-2 Kinase , Animals , Bombyx/virology , Bombyx/genetics , Bombyx/growth & development , Nucleopolyhedroviruses/physiology , Insect Proteins/metabolism , Insect Proteins/genetics , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Virus Replication , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Larva/virology , Larva/growth & development , Larva/metabolism , Larva/genetics
8.
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
9.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(3): 425-434, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38953267

ABSTRACT

Alzheimer's disease (AD) is a severe threat to human health and one of the three major causes of human death.Double-stranded RNA-dependent protein kinase (PKR) is an interferon-induced protein kinase involved in innate immunity.In the occurrence and development of AD,PKR is upregulated and continuously activated.On the one hand,the activation of PKR triggers an integrated stress response in brain cells.On the other hand,it indirectly upregulates the expression of ß-site amyloid precursor protein cleaving enzyme 1 and facilitates the accumulation of amyloid-ß protein (Aß),which could activate PKR activator to further activate PKR,thus forming a sustained accumulation cycle of Aß.In addition,PKR can promote Tau phosphorylation,thereby reducing microtubule stability in nerve cells.Inflammation in brain tissue,neurotoxicity resulted from Aß accumulation,and disruption of microtubule stability led to the progression of AD and the declines of memory and cognitive function.Therefore,PKR is a key molecule in the development and progression of AD.Effective PKR detection can aid in the diagnosis and prediction of AD progression and provide opportunities for clinical treatment.The inhibitors targeting PKR are expected to control the activity of PKR,thereby controlling the progression of AD.Therefore,PKR could be a target for the development of therapeutic drugs for AD.


Subject(s)
Alzheimer Disease , eIF-2 Kinase , Alzheimer Disease/metabolism , Humans , eIF-2 Kinase/metabolism , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Phosphorylation , Brain/metabolism , Amyloid beta-Protein Precursor/metabolism
10.
Dokl Biochem Biophys ; 517(1): 264-268, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002013

ABSTRACT

Translation inhibition can activate two cell death pathways. The first pathway is activated by translational aberrations, the second by endoplasmic reticulum (ER) stress. In this work, the effect of ribosome-inactivating protein type II (RIP-II) viscumin on M1 macrophages derived from the THP-1 cell line was investigated. The number of modified ribosomes was evaluated by real-time PCR. Transcriptome analysis revealed that viscumin induces the ER stress activated by the PERK sensor.


Subject(s)
Activating Transcription Factor 4 , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2 , Macrophages , Signal Transduction , eIF-2 Kinase , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , Humans , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Macrophages/metabolism , Macrophages/drug effects , THP-1 Cells
11.
J Immunol ; 213(5): 700-717, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39058317

ABSTRACT

dsRNA-dependent protein kinase R (PKR) is a key factor of innate immunity. It is involved in translation inhibition, apoptosis, and enhancement of the proinflammatory and IFN responses. However, how these antiviral functions are conserved during evolution remains largely unknown. Overexpression and knockout studies in a Chinook salmon (Oncorhynchus tshawytscha) cell line were conducted to assess the role of salmonid PKR in the antiviral response. Three distinct mRNA isoforms from a unique pkr gene, named pkr-fl (full length), pkr-ml (medium length) and pkr-sl (short length), were cloned and a pkr-/- clonal fish cell line was developed using CRISPR/Cas9 genome editing. PKR-FL includes an N-terminal dsRNA-binding domain and a C-terminal kinase domain, whereas PKR-ML and PKR-SL display a truncated or absent kinase domain, respectively. PKR-FL is induced during IFNA2 stimulation but not during viral hemorrhagic septicemia virus (VHSV) infection. Overexpression experiments showed that only PKR-FL possesses antiviral functions, including activation of apoptosis and inhibition of de novo protein synthesis. Knockout experiments confirmed that PKR is involved in apoptosis activation during the late stage of VHSV infection. Endogenous PKR also plays a critical role in translation inhibition upon poly(I:C) transfection after IFNA2 treatment. It is, however, not involved in translational arrest during VHSV infection. Extra- and intracellular titrations showed that endogenous PKR does not directly inhibit viral replication but apparently favors virion release into the supernatant, likely by triggering late apoptosis. Altogether, our data confirm that salmonid PKR has conserved molecular functions that VHSV appears to bypass with subversion strategies.


Subject(s)
Apoptosis , Protein Biosynthesis , Salmon , eIF-2 Kinase , Animals , Apoptosis/immunology , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Salmon/immunology , Cell Line , Protein Biosynthesis/immunology , Immunity, Innate , Novirhabdovirus/physiology , Novirhabdovirus/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , RNA, Double-Stranded/immunology , Fish Diseases/immunology , CRISPR-Cas Systems
12.
Pharm Biol ; 62(1): 607-620, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39034914

ABSTRACT

CONTEXT: Ulcerative colitis has been clinically treated with Qing Hua Chang Yin (QHCY), a traditional Chinese medicine formula. However, its precise mechanisms in mitigating chronic colitis are largely uncharted. OBJECTIVE: To elucidate the therapeutic efficiency of QHCY on chronic colitis and explore its underlying molecular mechanisms. MATERIALS AND METHODS: A total ion chromatogram fingerprint of QHCY was analysed. Chronic colitis was induced in male C57BL/6 mice using 2% dextran sodium sulphate (DSS) over 49 days. Mice were divided into control, DSS, DSS + QHCY (0.8, 1.6 and 3.2 g/kg/d dose, respectively) and DSS + mesalazine (0.2 g/kg/d) groups (n = 6). Mice were intragastrically administered QHCY or mesalazine for 49 days. The changes of disease activity index (DAI), colon length, colon histomorphology and serum pro-inflammatory factors in mice were observed. RNA sequencing was utilized to identify the differentially expressed transcripts (DETs) in colonic tissues and the associated signalling pathways. The expression of endoplasmic reticulum (ER) stress-related protein and NF-κB signalling pathway-related proteins in colonic tissues was detected by immunohistochemistry staining. RESULTS: Forty-seven compounds were identified in QHCY. Compared with the DSS group, QHCY significantly improved symptoms of chronic colitis like DAI increase, weight loss, colon shortening and histological damage. It notably reduced serum levels of IL-6, IL-1ß and TNF-α. QHCY suppressed the activation of PERK-ATF4-CHOP pathway of ER stress and NF-κB signalling pathways in colonic tissues. DISCUSSION AND CONCLUSIONS: The findings in this study provide novel insights into the potential of QHCY in treating chronic colitis patients.


Subject(s)
Activating Transcription Factor 4 , Dextran Sulfate , Drugs, Chinese Herbal , Endoplasmic Reticulum Stress , Mice, Inbred C57BL , NF-kappa B , Signal Transduction , Transcription Factor CHOP , eIF-2 Kinase , Animals , Male , Signal Transduction/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , Drugs, Chinese Herbal/pharmacology , NF-kappa B/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/metabolism , Transcription Factor CHOP/metabolism , Chronic Disease , Colitis/drug therapy , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Dose-Response Relationship, Drug
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.
BMC Med ; 22(1): 229, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38853264

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive ages. Our previous study has implicated a possible link between RNA editing and PCOS, yet the actual role of RNA editing, its association with clinical features, and the underlying mechanisms remain unclear. METHODS: Ten RNA-Seq datasets containing 269 samples of multiple tissue types, including granulosa cells, T helper cells, placenta, oocyte, endometrial stromal cells, endometrium, and adipose tissues, were retrieved from public databases. Peripheral blood samples were collected from twelve PCOS and ten controls and subjected to RNA-Seq. Transcriptome-wide RNA-Seq data analysis was conducted to identify differential RNA editing (DRE) between PCOS and controls. The functional significance of DRE was evaluated by luciferase reporter assays and overexpression in human HEK293T cells. Dehydroepiandrosterone and lipopolysaccharide were used to stimulate human KGN granulosa cells to evaluate gene expression. RESULTS: RNA editing dysregulations across multiple tissues were found to be associated with PCOS in public datasets. Peripheral blood transcriptome analysis revealed 798 DRE events associated with PCOS. Through weighted gene co-expression network analysis, our results revealed a set of hub DRE events in PCOS blood. A DRE event in the eukaryotic translation initiation factor 2-alpha kinase 2 (EIF2AK2:chr2:37,100,559) was associated with PCOS clinical features such as luteinizing hormone (LH) and the ratio of LH over follicle-stimulating hormone. Luciferase assays, overexpression, and knockout of RNA editing enzyme adenosine deaminase RNA specific (ADAR) showed that the ADAR-mediated editing cis-regulated EIF2AK2 expression. EIAF2AK2 showed a higher expression after dehydroepiandrosterone and lipopolysaccharide stimulation, triggering changes in the downstrean MAPK pathway. CONCLUSIONS: Our study presented the first evidence of cross-tissue RNA editing dysregulation in PCOS and its clinical associations. The dysregulation of RNA editing mediated by ADAR and the disrupted target EIF2AK2 may contribute to PCOS development via the MPAK pathway, underlining such epigenetic mechanisms in the disease.


Subject(s)
Polycystic Ovary Syndrome , RNA Editing , eIF-2 Kinase , Humans , Polycystic Ovary Syndrome/genetics , Female , RNA Editing/genetics , eIF-2 Kinase/genetics , Adult , HEK293 Cells , Gene Expression Profiling , Clinical Relevance
15.
J Toxicol Sci ; 49(7): 313-319, 2024.
Article in English | MEDLINE | ID: mdl-38945842

ABSTRACT

Dihydropyrazines (DHPs) are formed by non-enzymatic glycation reactions in vivo and in food. We recently reported that 3-hydro-2,2,5,6-tetramethylpyrazine (DHP-3), which is a methyl-substituted DHP, caused severe oxidative stress and cytotoxicity. However, the molecular mechanisms underlying the cytotoxic pathways of the DHP response remain elusive. Because oxidative stress induces endoplasmic reticulum (ER) stress and autophagy, we investigated the ability of DHP-3 to modulate the ER stress and autophagy pathways. DHP-3 activated the ER stress pathway by increasing inositol-requiring enzyme 1 (IRE1) and PKR-like ER kinase (PERK) phosphorylation and transcription factor 6 (ATF6) expression. Moreover, DHP-3 increased the expression of activating transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP), which are downstream targets of PERK. In addition, DHP-3 inhibited the autophagy pathway by increasing the accumulation of microtubule-associated protein 1 light chain 3 alpha-phosphatidylethanolamine conjugate (LC3-II) and p62/sequestosome 1 (p62), while decreasing autophagic flux. Taken together, these results indicate that DHP-3 activates the ER stress pathway and inhibits the autophagy pathway, suggesting that the resulting removal of damaged organelles is inadequate.


Subject(s)
Activating Transcription Factor 4 , Activating Transcription Factor 6 , Autophagy , Endoplasmic Reticulum Stress , Protein Serine-Threonine Kinases , Pyrazines , eIF-2 Kinase , Humans , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Pyrazines/pharmacology , Hep G2 Cells , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , eIF-2 Kinase/metabolism , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Phosphorylation , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Oxidative Stress/drug effects , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Signal Transduction/drug effects , Microtubule-Associated Proteins/metabolism
16.
Cell Rep ; 43(6): 114358, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38865243

ABSTRACT

Despite the consensus that accumulation of unfolded proteins in the endoplasmic reticulum (ER) lumen, i.e. ER stress, activates the unfolded protein response (UPR), studies under physiological and pathophysiological conditions suggest that ER stress may not always trigger the UPR, and the UPR can be activated in an ER stress-independent way. To better understand how the UPR is regulated and its relationship with ER stress requires direct detection of unfolded proteins in the ER, a method that is still lacking. Here, we report a strategy of visualizing unfolded protein accumulation in the ER lumen in living cells by employing an engineered ER stress sensor, PERK, which forms fluorescence puncta upon unfolded protein binding, in a fast and reversible way. Our reporter enables us to clarify the involvement of unfolded proteins in UPR activation under several physiological conditions and suggests that persistent unfolded protein accumulation in the ER despite UPR attenuation predicts cell death.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Unfolded Protein Response , eIF-2 Kinase , Endoplasmic Reticulum/metabolism , Humans , eIF-2 Kinase/metabolism , HEK293 Cells , HeLa Cells , Animals
17.
Mol Med Rep ; 30(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38904207

ABSTRACT

Montelukast and zafirlukast, cysteinyl leukotriene receptor antagonists (LTRAs), trigger apoptosis and inhibit cell proliferation of triple­negative breast cancer MDA­MB­231 cells. By contrast, only zafirlukast induces G0/G1 cell cycle arrest. The present study compared the effects of these drugs on proteins regulating cell proliferation, apoptosis, autophagy, and endoplasmic reticulum (ER) and oxidative stress using reverse transcription­quantitative PCR, western blotting and flow cytometry. The expression of proliferating markers, Ki­67 and proliferating cell nuclear antigen, was decreased by both drugs. Zafirlukast, but not montelukast, decreased the expression of cyclin D1 and CDK4, disrupting progression from G1 to S phase. Zafirlukast also increased the expression of p27, a cell cycle inhibitor. Both drugs decreased the expression of anti­apoptotic protein Bcl­2 and ERK1/2 phosphorylation, and increased levels of the autophagy marker LC3­II and DNA damage markers, including cleaved PARP­1, phosphorylated (p)­ATM and p­histone H2AX. The number of caspase 3/7­positive cells was greater in montelukast­treated cells compared with zafirlukast­treated cells. Montelukast induced higher levels of the ER stress marker CHOP compared with zafirlukast. Montelukast activated PERK, activating transcription factor 6 (ATF6) and inositol­requiring enzyme type 1 (IRE1) pathways, while zafirlukast only stimulated ATF6 and IRE1 pathways. GSK2606414, a PERK inhibitor, decreased apoptosis mediated by montelukast, but did not affect zafirlukast­induced cell death. The knockdown of CHOP by small interfering RNA reduced apoptosis triggered by montelukast and zafirlukast. In conclusion, the effects on cell cycle regulator proteins may contribute to cell cycle arrest caused by zafirlukast. The greater apoptotic effects of montelukast may be caused by the higher levels of activated caspase enzymes and the activation of three pathways of ER stress: PERK, ATF6, and IRE1.


Subject(s)
Acetates , Apoptosis , Autophagy , Cyclopropanes , DNA Damage , Endoplasmic Reticulum Stress , Indoles , Quinolines , Sulfides , Sulfonamides , Humans , Sulfides/pharmacology , Cyclopropanes/pharmacology , Quinolines/pharmacology , Apoptosis/drug effects , Acetates/pharmacology , Endoplasmic Reticulum Stress/drug effects , Cell Line, Tumor , Autophagy/drug effects , Sulfonamides/pharmacology , Indoles/pharmacology , Female , DNA Damage/drug effects , Phenylcarbamates/pharmacology , Tosyl Compounds/pharmacology , Cell Proliferation/drug effects , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Cell Cycle Checkpoints/drug effects , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Cell Cycle/drug effects , Leukotriene Antagonists/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
18.
Cell Rep ; 43(7): 114402, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38943644

ABSTRACT

The unfolded protein response (UPR) relieves endoplasmic reticulum (ER) stress through multiple strategies, including reducing protein synthesis, increasing protein folding capabilities, and enhancing misfolded protein degradation. After a multi-omics analysis, we find that signal recognition particle 14 (SRP14), an essential component of the SRP, is markedly reduced in cells undergoing ER stress. Further experiments indicate that SRP14 reduction requires PRKR-like ER kinase (PERK)-mediated eukaryotic translation initiation factor 2α (eIF2α) phosphorylation but is independent of ATF4 or ATF3 transcription factors. The decrease of SRP14 correlates with reduced translocation of fusion proteins and endogenous cathepsin D. Enforced expression of an SRP14 variant with elongation arrest capability prevents the reduced translocation of cathepsin D in stressed cells, whereas an SRP14 mutant without the activity does not. Finally, overexpression of SRP14 augments the UPR and aggravates ER-stress-induced cell death. These data suggest that translocational attenuation mediated by the PERK-SRP14 axis is a protective measure for the UPR to mitigate ER stress.


Subject(s)
Endoplasmic Reticulum Stress , Unfolded Protein Response , eIF-2 Kinase , eIF-2 Kinase/metabolism , Humans , Phosphorylation , Protein Transport , Signal Recognition Particle/metabolism , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , HeLa Cells
19.
Biochem Pharmacol ; 226: 116372, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38885773

ABSTRACT

MicroRNA and mitofusin-2 (Mfn2) play an important role in the myocardial apoptosis induced by acute myocardial infarction (AMI). However, the target relationship and underlying mechanism associated with interorganelle interaction between endoplasmic reticulum (ER) and mitochondria under ischemic condition is not completely clear. MI-induced injury, Mfn2 expression, Mfn2-mediated mitochondrial function and ER stress, and target regulation by miRNA-15b (miR-15b) were evaluated by animal MI and cellular hypoxic models with advanced molecular techniques. The results confirmed that Mfn2 was down-regulated and miR-15b was up-regulated upon the target binding profile under ischemic/hypoxic condition. Our data showed that miR-15b caused cardiac apoptotic injury that was reversed by rAAV9-anti-miR-15b or AMO-15b. The damage effect of miR-15b on Mfn2 expression and mitochondrial function was observed and rescued by rAAV9-anti-miR-15b or AMO-15b. The targeted regulation of miR-15b on Mfn2 was verified by luciferase reporter and microRNA-masking. Importantly, miR-15b-mediated Mfn2 suppression activated PERK/CHOP pathway, by which leads to ER stress and mitochondrial dysfunction, and cardiac apoptosis eventually. In conclusion, our research, for the first time, revealed the missing molecular link in Mfn2 and apoptosis and elucidated that pro-apoptotic miR-15b plays crucial roles during the pathogenesis of AMI through down-regulation of Mfn2 and activation of PERK-mediated ER stress. These findings may provide an opportunity to develop new therapies for prophylaxis and treatment of ischemic heart disease.


Subject(s)
GTP Phosphohydrolases , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Male , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/antagonists & inhibitors , Signal Transduction/physiology , Myocardial Ischemia/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Mice , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum Stress/genetics , Apoptosis , Mice, Inbred C57BL
20.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L232-L249, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38860845

ABSTRACT

COVID-19 syndrome is characterized by acute lung injury, hypoxemic respiratory failure, and high mortality. Alveolar type 2 (AT2) cells are essential for gas exchange, repair, and regeneration of distal lung epithelium. We have shown that the causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and other members of the ß-coronavirus genus induce an endoplasmic reticulum (ER) stress response in vitro; however, the consequences for host AT2 cell function in vivo are less understood. To study this, two murine models of coronavirus infection were used-mouse hepatitis virus-1 (MHV-1) in A/J mice and a mouse-adapted SARS-CoV-2 strain. MHV-1-infected mice exhibited dose-dependent weight loss with histological evidence of distal lung injury accompanied by elevated bronchoalveolar lavage fluid (BALF) cell counts and total protein. AT2 cells showed evidence of both viral infection and increased BIP/GRP78 expression, consistent with activation of the unfolded protein response (UPR). The AT2 UPR included increased inositol-requiring enzyme 1α (IRE1α) signaling and a biphasic response in PKR-like ER kinase (PERK) signaling accompanied by marked reductions in AT2 and BALF surfactant protein (SP-B and SP-C) content, increases in surfactant surface tension, and emergence of a reprogrammed epithelial cell population (Krt8+ and Cldn4+). The loss of a homeostatic AT2 cell state was attenuated by treatment with the IRE1α inhibitor OPK-711. As a proof-of-concept, C57BL6 mice infected with mouse-adapted SARS-CoV-2 demonstrated similar lung injury and evidence of disrupted surfactant homeostasis. We conclude that lung injury from ß-coronavirus infection results from an aberrant host response, activating multiple AT2 UPR stress pathways, altering surfactant metabolism/function, and changing AT2 cell state, offering a mechanistic link between SARS-CoV-2 infection, AT2 cell biology, and acute respiratory failure.NEW & NOTEWORTHY COVID-19 syndrome is characterized by hypoxemic respiratory failure and high mortality. In this report, we use two murine models to show that ß-coronavirus infection produces acute lung injury, which results from an aberrant host response, activating multiple epithelial endoplasmic reticular stress pathways, disrupting pulmonary surfactant metabolism and function, and forcing emergence of an aberrant epithelial transition state. Our results offer a mechanistic link between SARS-CoV-2 infection, AT2 cell biology, and respiratory failure.


Subject(s)
COVID-19 , Endoplasmic Reticulum Stress , Endoribonucleases , Homeostasis , Murine hepatitis virus , SARS-CoV-2 , Animals , Mice , COVID-19/metabolism , COVID-19/pathology , COVID-19/virology , COVID-19/complications , Murine hepatitis virus/pathogenicity , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Alveolar Epithelial Cells/pathology , Endoplasmic Reticulum Chaperone BiP , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Coronavirus Infections/virology , Coronavirus Infections/complications , Pulmonary Surfactants/metabolism , Unfolded Protein Response , Betacoronavirus , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/virology , Respiratory Insufficiency/pathology , Disease Models, Animal , eIF-2 Kinase/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL