Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 426
Filter
1.
Sci Rep ; 14(1): 9029, 2024 04 19.
Article in English | MEDLINE | ID: mdl-38641657

ABSTRACT

Double-stranded RNA-activated protein kinase R (PKR) is highly expressed in colorectal cancer (CRC). However, the role of PKR in CRC remains unclear. The aim of this study was to clarify whether C16 (a PKR inhibitor) exhibits antitumor effects and to identify its target pathway in CRC. We evaluated the effects of C16 on CRC cell lines using the MTS assay. Enrichment analysis was performed to identify the target pathway of C16. The cell cycle was analyzed using flow cytometry. Finally, we used immunohistochemistry to examine human CRC specimens. C16 suppressed the proliferation of CRC cells. Gene Ontology (GO) analysis revealed that the cell cycle-related GO category was substantially enriched in CRC cells treated with C16. C16 treatment resulted in G1 arrest and increased p21 protein and mRNA expression. Moreover, p21 expression was associated with CRC development as observed using immunohistochemical analysis of human CRC tissues. C16 upregulates p21 expression in CRC cells to regulate cell cycle and suppress tumor growth. Thus, PKR inhibitors may serve as a new treatment option for patients with CRC.


Subject(s)
Colorectal Neoplasms , Protein Kinase Inhibitors , Humans , Apoptosis , Cell Cycle , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Protein Kinase Inhibitors/pharmacology , Indoles/pharmacology , Thiazoles/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism
2.
Int J Antimicrob Agents ; 63(5): 107124, 2024 May.
Article in English | MEDLINE | ID: mdl-38412930

ABSTRACT

For successful viral propagation within infected cells, the virus needs to overcome the cellular integrated stress response (ISR), triggered during viral infection, which, in turn, inhibits general protein translation. This paper reports a tactic employed by viruses to suppress the ISR by upregulating host cell polyribonucleotide nucleotidyltransferase 1 (PNPT1). The propagation of adenovirus, murine cytomegalovirus and hepatovirus within their respective host cells induces PNPT1 expression. Notably, when PNPT1 is knocked down, the propagation of all three viruses is prevented. Mechanistically, the inhibition of PNPT1 facilitates the relocation of mitochondrial double-stranded RNAs (mt-dsRNAs) to the cytoplasm, where they activate RNA-activated protein kinase (PKR). This activation leads to eukaryotic initiation factor 2α (eIF2α) phosphorylation, resulting in the suppression of translation. Furthermore, by scrutinizing the PNPT1 recognition element and screening 17,728 drugs and bioactive compounds approved by the US Food and Drug Administration, lanatoside C was identified as a potent PNPT1 inhibitor. This compound impedes the propagation of adenovirus, murine cytomegalovirus and hepatovirus, and suppresses production of the severe acute respiratory syndrome coronavirus-2 spike protein. These discoveries shed light on a novel strategy to impede pan-viral propagation by activating the host cell mt-dsRNA-PKR-eIF2α signalling axis.


Subject(s)
eIF-2 Kinase , Humans , Animals , eIF-2 Kinase/metabolism , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , Antiviral Agents/pharmacology , Muromegalovirus/physiology , Muromegalovirus/drug effects , Mice , Eukaryotic Initiation Factor-2/metabolism , Virus Replication/drug effects , RNA, Double-Stranded/genetics , Adenoviridae/genetics , Adenoviridae/drug effects , Phosphorylation , SARS-CoV-2/drug effects
3.
Int Immunopharmacol ; 111: 109092, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35940075

ABSTRACT

Neuroinflammation plays a key role in the development of depression-like behaviors.Endoplasmic reticulum (ER) stress,defined as accumulation of unfolded proteins in the ER,is suggested tocollaboratewithinflammation process to drive sustained neuroinflammation. Protein kinase R-like endoplasmic reticulum kinase (PERK) is ofparticularly attractive target because it plays key rolein the regulation of ER stress-induced neuroinflammation, however, little isknown whether PERKmediatedER stress is implicated in LPS-induced depression-like behaviors.Thus, we aimed to evaluate the induction of PERK pathwayin mice with depression-like behaviors induced by LPS, as well as the alterations in depression-like behaviorsfollowing the blocking of PERK pathway.We found that LPS challenges resulted in enhanced PERK in the hippocampus, with no alteration in the prefrontal cortex. Importantly, we found that PERKinhibitorISRIB reducedthe proinflammatory responsesof microglia in the context of acute LPS-induced brain inflammation, and subsequent the preserved hippocampal neurogenesis, and improvement in depression-like behavioroutcomes following LPS challenges.It was also worth mentioning thatISRIB treatmentreduced the peripheral pro-inflammatory cytokines includingIL-1ß, IL-6 and IL-18. Thus, targetingPERK mediated Endoplasmic reticulum stress may be a promising antidepressant and anti-inflammatory candidate drug for the alleviation of neuroinflammationmediated depression, and PERKinhibitorISRIBmay havebenefits for combating major depressive disorder.


Subject(s)
Depressive Disorder, Major , Lipopolysaccharides , eIF-2 Kinase , Animals , Anti-Inflammatory Agents/therapeutic use , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Depressive Disorder, Major/metabolism , Endoplasmic Reticulum Stress , Lipopolysaccharides/pharmacology , Male , Mice , Neuroinflammatory Diseases , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism
4.
Exp Neurol ; 347: 113899, 2022 01.
Article in English | MEDLINE | ID: mdl-34678230

ABSTRACT

BACKGROUND AND PURPOSE: Traumatic brain injury (TBI) destroys white matter, and this destruction is aggravated by secondary neuroinflammatory reactions. Although white matter injury (WMI) is strongly correlated with poor neurological function, understanding of white matter integrity maintenance is limited, and no available therapies can effectively protect white matter. One candidate approach that may fulfill this goal is cannabinoid receptor 2 (CB2) agonist treatment. Here, we confirmed that a selective CB2 agonist, JWH133, protected white matter after TBI. METHODS: The motor evoked potentials (MEPs), open field test, and Morris water maze test were used to assess neurobehavioral outcomes. Brain tissue loss, WM damage, Endoplasmic reticulum stress (ER stress), microglia responses were evaluated after TBI. The functional integrity of WM was measured by diffusion tensor imaging (DTI) and transmission electron microscopy (TEM). Primary microglia and oligodendrocyte cocultures were used for additional mechanistic studies. RESULTS: JWH133 increased myelin basic protein (MBP) and neurofilament heavy chain (NF200) levels and anatomic preservation of myelinated axons revealed by DTI and TEM. JWH133 also increased the numbers of oligodendrocyte precursor cells and mature oligodendrocytes. Furthermore, JWH133 drove microglial polarization toward the protective M2 phenotype and modulated the redistribution of microglia in the striatum. Further investigation of the underlying mechanism revealed that JWH133 downregulated phosphorylation of the protein kinase R (PKR)-like endoplasmic reticulum (ER) kinase (PERK) signaling pathway and its downstream signals eukaryotic translation initiation factor 2 α (eIF2α), activating transcription factor 4 (ATF4) and Growth arrest and DNA damage-inducible protein (GADD34); this downregulation was followed by p-Protein kinase B(p-Akt) upregulation. In primary cocultures of microglia and oligodendrocytes, JWH133 decreased phosphorylated PERK expression in microglia stimulated with tunicamycin and facilitated oligodendrocyte survival. These data reveal that JWH133 ultimately alleviates WMI and improves neurological behavior following TBI. However, these effects were prevented by SR144528, a selective CB2 antagonist. CONCLUSIONS: This work illustrates the PERK-mediated interaction between microglia and oligodendrocytes. In addition, the results are consistent with recent findings that microglial polarization switching accelerates WMI, highlighting a previously unexplored role for CB2 agonists. Thus, CB2 agonists are potential therapeutic agents for TBI and other neurological conditions involving white matter destruction.


Subject(s)
Cannabinoids/pharmacology , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction/physiology , White Matter/metabolism , eIF-2 Kinase/biosynthesis , Animals , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Agonists/therapeutic use , Cannabinoids/therapeutic use , Cells, Cultured , Disease Models, Animal , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/physiology , Male , Microglia/drug effects , Microglia/metabolism , Rats , Rats, Sprague-Dawley , White Matter/diagnostic imaging , White Matter/drug effects , White Matter/injuries , eIF-2 Kinase/antagonists & inhibitors
5.
Mol Cell ; 82(2): 420-434.e6, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34951963

ABSTRACT

Exon back-splicing-generated circular RNAs, as a group, can suppress double-stranded RNA (dsRNA)-activated protein kinase R (PKR) in cells. We have sought to synthesize immunogenicity-free, short dsRNA-containing RNA circles as PKR inhibitors. Here, we report that RNA circles synthesized by permuted self-splicing thymidylate synthase (td) introns from T4 bacteriophage or by Anabaena pre-tRNA group I intron could induce an immune response. Autocatalytic splicing introduces ∼74 nt td or ∼186 nt Anabaena extraneous fragments that can distort the folding status of original circular RNAs or form structures themselves to provoke innate immune responses. In contrast, synthesized RNA circles produced by T4 RNA ligase without extraneous fragments exhibit minimized immunogenicity. Importantly, directly ligated circular RNAs that form short dsRNA regions efficiently suppress PKR activation 103- to 106-fold higher than reported chemical compounds C16 and 2-AP, highlighting the future use of circular RNAs as potent inhibitors for diseases related to PKR overreaction.


Subject(s)
Protein Kinase Inhibitors/pharmacology , RNA, Circular/pharmacology , eIF-2 Kinase/antagonists & inhibitors , A549 Cells , Bacteriophage T4/enzymology , Bacteriophage T4/genetics , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/drug effects , Introns , Nucleic Acid Conformation , Protein Kinase Inhibitors/immunology , RNA Ligase (ATP)/genetics , RNA Ligase (ATP)/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Circular/genetics , RNA, Circular/immunology , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , eIF-2 Kinase/metabolism
6.
Mediators Inflamm ; 2021: 1805147, 2021.
Article in English | MEDLINE | ID: mdl-34790063

ABSTRACT

Mycobacterium tuberculosis (Mtb) remains a significant threat to global health as it induces granuloma and systemic inflammatory responses during active tuberculosis. Mtb can induce macrophage pyroptosis, leading to the release of IL-1ß and tissue damage, promoting its spread. Here, we established an in vitro Mtb-infected macrophage model to seek an effective antipyroptosis agent. Baicalin, isolated from Radix Scutellariae, was found to reduce pyroptosis in Mtb-infected macrophages. Baicalin could inhibit activation of the PERK/eIF2α pathway and thus downregulates TXNIP expression and subsequently reduces activation of the NLRP3 inflammasome, resulting in reduced pyroptosis in Mtb-infected macrophages. In conclusion, baicalin reduced pyroptosis by inhibiting the PERK/TXNIP/NLRP3 axis and might thus be a new adjuvant host-directed therapy (HDT) drug.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Flavonoids/pharmacology , Mycobacterium tuberculosis/pathogenicity , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Pyroptosis/drug effects , Thioredoxins/antagonists & inhibitors , eIF-2 Kinase/antagonists & inhibitors , Animals , Carrier Proteins/physiology , Endoplasmic Reticulum Stress/drug effects , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , RAW 264.7 Cells , Thioredoxins/physiology , eIF-2 Kinase/physiology
7.
JCI Insight ; 6(18)2021 09 22.
Article in English | MEDLINE | ID: mdl-34549728

ABSTRACT

ER stress and activation of the unfolded protein response in the periphery as well as the central nervous system have been linked to various metabolic abnormalities. Chemically lowering protein kinase R-like ER kinase (PERK) activity within the hypothalamus leads to decreased food intake and body weight. However, the cell populations required in this response remain undefined. In the current study, we investigated the effects of proopiomelanocortin-specific (POMC-specific) PERK deficiency on energy balance and glucose metabolism. Male mice deficient for PERK in POMC neurons exhibited improvements in energy balance on a high-fat diet, showing decreased food intake and body weight, independent of changes in glucose and insulin tolerances. The plant-based inhibitor of PERK, celastrol, increases leptin sensitivity, resulting in decreased food intake and body weight in a murine model of diet-induced obesity (DIO). Our data extend these observations by demonstrating that celastrol-induced improvements in leptin sensitivity and energy balance were attenuated in mice with PERK deficiency in POMC neurons. Altogether, these data suggest that POMC-specific PERK deficiency in male mice confers protection against DIO, possibly providing a new therapeutic target for the treatment of diabetes and metabolic syndrome.


Subject(s)
Energy Metabolism , Glucose/metabolism , Leptin/pharmacology , Pentacyclic Triterpenes/pharmacology , Pro-Opiomelanocortin/physiology , eIF-2 Kinase/antagonists & inhibitors , Animals , Arcuate Nucleus of Hypothalamus/cytology , Body Weight/drug effects , Diet, High-Fat/adverse effects , Eating/drug effects , Endoplasmic Reticulum Stress , Insulin Resistance , Male , Mice , Mice, Knockout , Neurons , Obesity/etiology , Obesity/prevention & control , Pro-Opiomelanocortin/metabolism , eIF-2 Kinase/genetics
8.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360909

ABSTRACT

Neurodegenerative diseases are an ever-increasing problem for the rapidly aging population. Despite this, our understanding of how these neurodegenerative diseases develop and progress, is in most cases, rudimentary. Protein kinase RNA (PKR)-like ER kinase (PERK) comprises one of three unfolded protein response pathways in which cells attempt to manage cellular stress. However, because of its role in the cellular stress response and the far-reaching implications of this pathway, error within the PERK pathway has been shown to lead to a variety of pathologies. Genetic and clinical studies show a correlation between failure of the PERK pathway in neural cells and the development of neurodegeneration, but the wide array of methodology of these studies is presenting conflicting narratives about the role of PERK in these affected systems. Because of the connection between PERK and pathology, PERK has become a high value target of study for understanding neurodegenerative diseases and potentially how to treat them. Here, we present a review of the literature indexed in PubMed of the PERK pathway and some of the complexities involved in investigating the protein's role in the development of neurodegenerative diseases as well as how it may act as a target for therapeutics.


Subject(s)
Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Signal Transduction/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Aged , Aging/genetics , Aging/metabolism , Animals , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Humans , Molecular Targeted Therapy/methods , Neurodegenerative Diseases/drug therapy , Polymorphism, Single Nucleotide , Signal Transduction/drug effects , Unfolded Protein Response , eIF-2 Kinase/antagonists & inhibitors
9.
J Clin Invest ; 131(18)2021 09 15.
Article in English | MEDLINE | ID: mdl-34324437

ABSTRACT

Ischemic cardiomyopathy is associated with an increased risk of sudden death, activation of the unfolded protein response (UPR), and reductions in multiple cardiac ion channels. When activated, the protein kinase-like ER kinase (PERK) branch of the UPR reduces protein translation and abundance. We hypothesized that PERK inhibition could prevent ion channel downregulation and reduce arrhythmia risk after myocardial infarct (MI). MI induced in mice by coronary artery ligation resulted in reduced ion channel levels, ventricular tachycardia (VT), and prolonged corrected intervals between the Q and T waves on the ECGs (QTc). Protein levels of major cardiac ion channels were decreased. MI cardiomyocytes showed significantly prolonged action potential duration and decreased maximum upstroke velocity. Cardiac-specific PERK KO reduced electrical remodeling in response to MI, with shortened QTc intervals, fewer VT episodes, and higher survival rates. Pharmacological PERK inhibition had similar effects. In conclusion, we found that activated PERK during MI contributed to arrhythmia risk by the downregulation of select cardiac ion channels. PERK inhibition prevented these changes and reduced arrhythmia risk. These results suggest that ion channel downregulation during MI is a fundamental arrhythmia mechanism and that maintenance of ion channel levels is antiarrhythmic.


Subject(s)
Arrhythmias, Cardiac/prevention & control , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Unfolded Protein Response/physiology , eIF-2 Kinase/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Down-Regulation , Female , Heart Disease Risk Factors , Humans , Indoles/pharmacology , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Protein Kinase Inhibitors/pharmacology , Unfolded Protein Response/drug effects , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
10.
Sci Adv ; 7(25)2021 06.
Article in English | MEDLINE | ID: mdl-34134991

ABSTRACT

Infection by highly pathogenic coronaviruses results in substantial apoptosis. However, the physiological relevance of apoptosis in the pathogenesis of coronavirus infections is unknown. Here, with a combination of in vitro, ex vivo, and in vivo models, we demonstrated that protein kinase R-like endoplasmic reticulum kinase (PERK) signaling mediated the proapoptotic signals in Middle East respiratory syndrome coronavirus (MERS-CoV) infection, which converged in the intrinsic apoptosis pathway. Inhibiting PERK signaling or intrinsic apoptosis both alleviated MERS pathogenesis in vivo. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-CoV induced apoptosis through distinct mechanisms but inhibition of intrinsic apoptosis similarly limited SARS-CoV-2- and SARS-CoV-induced apoptosis in vitro and markedly ameliorated the lung damage of SARS-CoV-2-inoculated human angiotensin-converting enzyme 2 (hACE2) mice. Collectively, our study provides the first evidence that virus-induced apoptosis is an important disease determinant of highly pathogenic coronaviruses and demonstrates that this process can be targeted to attenuate disease severity.


Subject(s)
Antiviral Agents/pharmacology , Apoptosis/drug effects , COVID-19 Drug Treatment , Coronavirus Infections/drug therapy , eIF-2 Kinase/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Angiotensin-Converting Enzyme 2/genetics , Animals , Apoptosis/physiology , COVID-19/etiology , COVID-19/pathology , Cell Line , Coronavirus Infections/etiology , Coronavirus Infections/pathology , Dipeptidyl Peptidase 4/genetics , Epithelial Cells/virology , Female , Humans , Indoles/pharmacology , Lung/virology , Male , Mice, Transgenic , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics
11.
Eur J Pharmacol ; 906: 174223, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34081906

ABSTRACT

Protein kinase R (PKR) plays a main role in inflammation, insulin resistance, and glucose balance. It is activated by various stress signals and is key mediators of diabetes and associated complications. In the present study, we investigated the effect of PKR inhibition on myocardial dysfunction, inflammatory, cell death and interrelated signalling pathways in isoproterenol induced myocardial ischemia in vivo in wistar rats and in vitro in cultured cardiomyocytes. H9C2 rat cardiomyocytes were treated with 10 µM Isoproterenol (ISO). For in vivo studies, rats were divided into 4 groups: control, ischemic group (ISO), preventive group, curative group and each group consist of 8 rats. Myocardial Ischemia (MI) was induced with two subsequent doses of ISO (100 mg/kg, s.c.). The rats were treated with PKR inhibitor, C16 (166.5 µg/kg, i.p.) for 14 days. Heart rate, systolic, diastolic and mean arterial pressures were measured by non-invasive BP apparatus. Cardiac biomarkers were measured by commercial kits. Ischemic Zone, Morphological abnormalities and fibrosis of heart was detected by TTC, haematoxylin & eosin staining, Masson's and Sirius red staining respectively. Protein expression was done by western blotting and immune histochemistry. mRNA expression was done by RT-PCR. MI was characterized by declined myocardial performance along with elevation of cardiac biomarkers and associated with increased expression of PKR, oxidative-nitrosative stress, activated various inflammatory pathways (nuclear factor kappa light chain enhancer of activated B cells -NF-κB); Mitogen-activated protein kinases-MAPK; c-Jun N-terminal kinase-JNK), increased expression of inflammatory markers (Tumour necrosis factor alpha-TNF-α), markers of fibrosis (Alpha smooth muscle actin -α-SMA; Transforming growth factor beta-TGF-ß), enhanced cell death (Ischemic zone) and increased expression of extracellular regulated-kinases (ERK-1/2) and advanced glycation end products (AGE's). Interestingly, inhibition of PKR attenuated myocardial dysfunction, cardiac fibrosis, oxidative/nitrosative stress, inflammation, cell death, and inter-related signalling pathways. Our findings report that inhibition of PKR improves the ischemic mediated inflammation, apoptosis, cardiac hypertrophy and fibrosis in MI induced rats. Hence, inhibition of PKR might be one of intervention therapy for the treatment of myocardial ischemia.


Subject(s)
Heart/drug effects , Myocardial Infarction/drug therapy , Myocardium/pathology , Protein Kinase Inhibitors/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Animals , Cell Line , Disease Models, Animal , Fibrosis , Humans , Isoproterenol/administration & dosage , Isoproterenol/toxicity , Male , Myocardial Infarction/chemically induced , Myocardial Infarction/diagnosis , Myocardial Infarction/pathology , Myocardium/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Protein Kinase Inhibitors/therapeutic use , Rats , Rats, Wistar , eIF-2 Kinase/metabolism
12.
Drug Des Devel Ther ; 15: 1717-1724, 2021.
Article in English | MEDLINE | ID: mdl-33935494

ABSTRACT

BACKGROUND: Osteonecrosis of the femoral head (ONFH) seriously affects the quality of life and labor ability of patients. It is urgent and vital to find the methods for necrosis clinical treatment. OBJECTIVE: This study aims to study the potential protective effects of Alendronate in the early stage of femur head necrosis. METHODS: Ten clinal ONFH tissue samples were employed. H&E staining was employed for the observation of the pathological characteristics of ONFH. The rat model (n=12) was established by the treatment of liquid nitrogen and then treated with Alendronate. The protein expression of BMP2, EIF2AK3, EIF2A and ATF4 were detected via Western blotting and IHC. RESULTS: Fibrin and necrotizing granulation tissue were observed in ONFH tissues with lymphocytes and plasma cells infiltrating in the necrotic area, exhibiting the inflammatory muscle with abnormal shape and color. In the Model group, the BMP2 and ATF4 were mainly distributed in the cell boundaries. The relative protein expression of BMP2, EIF2AK3, EIF2A, ATF4 was decreased in the Model group, compared to the NC group, which was partially recovered by the Alendronate application. CONCLUSION: Alendronate application partially reversed the suppression of expression of BMP2, EIF2AK3, EIF2A, ATF4 caused by liquid nitrogen. Alendronate could be a promising strategy of curing ONFH via targeting BMP2/EIF2AK3/EIF2A/ATF4 pathway.


Subject(s)
Activating Transcription Factor 4/metabolism , Alendronate/pharmacology , Bone Morphogenetic Protein 2/metabolism , Femur Head Necrosis/drug therapy , Up-Regulation/drug effects , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Female , Femur Head Necrosis/metabolism , Femur Head Necrosis/pathology , Humans , Male , Middle Aged , Nitrogen/pharmacology , Osteonecrosis/drug therapy , Osteonecrosis/metabolism , Osteonecrosis/pathology , Rats , Rats, Sprague-Dawley , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics
13.
Bioorg Med Chem Lett ; 43: 128058, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33895276

ABSTRACT

The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of the three endoplasmic reticulum (ER) transmembrane sensors of the unfolded protein response (UPR) that regulates protein synthesis, alleviates cellular ER stress and has been implicated in tumorigenesis and prolonged cancer cell survival. In this study, we report a series of 2-amino-3-amido-5-aryl-pyridines that we have identified as potent, selective, and orally bioavailable PERK inhibitors. Amongst the series studied herein, compound (28) a (R)-2-Amino-5-(4-(2-(3,5-difluorophenyl)-2-hydroxyacetamido)-2-ethylphenyl)-N-isopropylnicotinamide has demonstrated potent biochemical and cellular activity, robust pharmacokinetics and 70% oral bioavailability in mice. Given these data, this compound (28) was studied in the 786-O renal cell carcinoma xenograft model. We observed dose-dependent, statistically significant tumor growth inhibition, supporting the use of this tool compound in additional mechanistic studies.


Subject(s)
Drug Discovery , Pyridines/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Administration, Oral , Biological Availability , Dose-Response Relationship, Drug , Humans , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Structure-Activity Relationship , eIF-2 Kinase/metabolism
14.
Int J Mol Sci ; 22(9)2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33925820

ABSTRACT

Primary open-angle glaucoma (POAG) constitutes the most common type of glaucoma. Emerging evidence suggests that Endoplasmic Reticulum (ER) stress and the protein kinase RNA-like endoplasmic reticulum kinase (PERK)-mediated Unfolded Protein Response (UPR) signaling pathway play a key role in POAG pathogenesis. Thus, the main aim of the study was to evaluate the effectiveness of the PERK inhibitor LDN-0060609 in cellular model of glaucoma using primary human trabecular meshwork (HTM) cells. To evaluate the level of the ER stress marker proteins, Western blotting and TaqMan gene expression assay were used. The cytotoxicity was measured by XTT, LDH assays and Giemsa staining, whereas genotoxicity via comet assay. Changes in cell morphology were assessed by phase-contrast microscopy. Analysis of apoptosis was performed by caspase-3 assay and flow cytometry (FC), whereas cell cycle progression by FC. The results obtained have demonstrated that LDN-0060609 triggered a significant decrease of ER stress marker proteins within HTM cells with induced ER stress conditions. Moreover, LDN-0060609 effectively increased viability, reduced DNA damage, increased proliferation, restored normal morphology, reduced apoptosis and restored normal cell cycle distribution of HTM cells with induced ER stress conditions. Thereby, PERK inhibitors, such as LDN-0060609, may provide an innovative, ground-breaking treatment strategy against POAG.


Subject(s)
Glaucoma, Open-Angle , Protein Kinase Inhibitors/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , DNA Damage/drug effects , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , Glaucoma, Open-Angle/drug therapy , Glaucoma, Open-Angle/metabolism , Humans , Protein Kinase Inhibitors/adverse effects , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
15.
J Mol Endocrinol ; 66(4): 313-323, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33870911

ABSTRACT

We examined endoplasmic reticulum (ER) stress-related gene expression in orbital tissues from patients with Graves' orbitopathy (GO) and the effects of silencing protein kinase RNA-like endoplasmic reticulum kinase (PERK) in primary orbital fibroblast cultures to demonstrate the therapeutic potential of PERK-modulating agents in GO management. The expression of ER stress-related genes in orbital tissue harvested from individuals with or without GO was studied using real-time PCR. The role of PERK in GO pathogenesis was examined through small-interfering RNA (siRNA)-mediated silencing in cultured primary orbital fibroblasts. Intracellular reactive oxygen species (ROS) levels induced in response to cigarette smoke extract (CSE) or hydrogen peroxide were measured using 5-(and 6)-carboxy-20,70-dichlorodihydrofluorescein diacetate staining and flow cytometry. Cells were stained with Oil Red O, and adipogenesis-related transcription factor expression was evaluated through Wwestern blotting after adipogenic differentiation. PERK, activating transcription factor 4 (ATF4), and CCAAT-enhancer-binding protein (C/EBP)-homologous protein (CHOP) mRNA levels were significantly higher in GO orbital tissues than in non-GO orbital tissues. PERK silencing inhibited CSE- or hydrogen peroxide-induced ROS generation. After adipogenic differentiation, GO orbital fibroblasts revealed decreased lipid droplets and downregulation of C/EBPα, C/EBPß, and peroxisome proliferator-activator gamma (PPARγ) in PERK siRNA-transfected cells. The orbital tissues of patients with GO were exposed to chronic ER stress and subsequently exhibited enhanced unfolded protein response (especially through the PERK pathway). PERK silencing reduced oxidative stress and adipogenesis in GO orbital fibroblasts in vitro. Our results imply that PERK-modulating agents can potentially be used to manage GO.


Subject(s)
Activating Transcription Factor 4/genetics , Graves Ophthalmopathy/genetics , PPAR gamma/genetics , Transcription Factor CHOP/genetics , eIF-2 Kinase/genetics , Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Cell Line , Endoplasmic Reticulum Stress/genetics , Fibroblasts/metabolism , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/pathology , Humans , Oxidative Stress/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , eIF-2 Kinase/antagonists & inhibitors
16.
J Biol Chem ; 296: 100264, 2021.
Article in English | MEDLINE | ID: mdl-33837743

ABSTRACT

Recent studies have demonstrated that embryonic stem cells (ESCs) are deficient in expressing type I interferons (IFN), the cytokines that play key roles in antiviral responses. However, the underlying molecular mechanisms and biological implications of this finding are poorly understood. In this study, we developed a synthetic RNA-based assay that can simultaneously assess multiple forms of antiviral responses. Dicer is an enzyme essential for RNA interference (RNAi), which is used as a major antiviral mechanism in invertebrates. RNAi activity is detected in wild-type ESCs but is abolished in Dicer knockout ESCs (D-/-ESCs) as expected. Surprisingly, D-/-ESCs have gained the ability to express IFN, which is otherwise deficient in wild-type ESCs. Furthermore, D-/-ESCs have constitutively active double-stranded RNA (dsRNA)-activated protein kinase (PKR), an enzyme that is also involved in antiviral response. D-/-ESCs show increased sensitivity to the cytotoxicity resulting from RNA transfection. The effects of dsRNA can be partly replicated with a synthetic B2RNA corresponding to the retrotransposon B2 short interspersed nuclear element. B2RNA has secondary structure features of dsRNA and accumulates in D-/-ESCs, suggesting that B2RNA could be a cellular RNA that activates PKR and contributes to the decreased cell proliferation and viability of D-/-ESCs. Treatment of D-/-ESCs with a PKR inhibitor and IFNß-neutralizing antibodies increased cell proliferation rate and cell viability. Based on these findings, we propose that, in ESCs, Dicer acts as a repressor of antiviral responses and plays a key role in the maintenance of proliferation, viability, and pluripotency of ESCs.


Subject(s)
DEAD-box RNA Helicases/genetics , Interferon Type I/genetics , Interferon-gamma/genetics , Mouse Embryonic Stem Cells/drug effects , Ribonuclease III/genetics , eIF-2 Kinase/genetics , Animals , Antiviral Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference/drug effects , RNA, Double-Stranded/drug effects , RNA, Double-Stranded/genetics , Retroelements/genetics , eIF-2 Kinase/antagonists & inhibitors
17.
Eur J Pharm Biopharm ; 163: 179-187, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33771622

ABSTRACT

In this study, we developed a mesoporous silica nanoparticles - mRNA (MSN-mRNA) subcutaneous delivery system composed of naked mRNA and a subcutaneous depot of imidazolo-oxindole RNA-activated protein kinase (PKR) inhibitor C16. We show that C16 treatment during mRNA transfection is a potent immune evasion approach that non-linearly enhances translation of unmodified mRNA in both mouse fibroblasts and dendritic cells in vitro exceeding that of nucleoside-modified mRNA. Notably, C16 further enhances translation of nucleoside-modified mRNA and HPLC purified mRNA. However, translation enhancement is dependent on and potentiated by C16's continuous presence. C16 mediated translation enhancement is extended in vivo by employing MSN as an interface to sustain-release C16. Subcutaneously administered MSN-mRNA significantly enhanced in vivo translation and expression kinetics of naked mRNA in unmodified, nucleoside-modified, and HPLC purified formats. We applied a MSN-mRNA vaccine formulation composed of naked mRNA encoding ovalbumin and granulocyte macrophage colony stimulating factor, and C16@MSNs on a xenograft E.G7-OVA prophylactic tumor model, resulting in very potent tumor inhibition. The MSN-mRNA delivery system bears great translational potential in mRNA therapeutics.


Subject(s)
Cancer Vaccines/administration & dosage , Drug Carriers/chemistry , Indoles/administration & dosage , Neoplasms/therapy , Thiazoles/administration & dosage , Animals , Cancer Vaccines/genetics , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Dendritic Cells , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , Fibroblasts , Hep G2 Cells , Humans , Indoles/pharmacokinetics , Injections, Subcutaneous , Mice , NIH 3T3 Cells , Nanoparticles/chemistry , Neoplasms/pathology , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Silicon Dioxide/chemistry , Thiazoles/pharmacokinetics , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism
18.
Cell Metab ; 33(3): 598-614.e7, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33592173

ABSTRACT

The architecture of cristae provides a spatial mitochondrial organization that contains functional respiratory complexes. Several protein components including OPA1 and MICOS complex subunits organize cristae structure, but upstream regulatory mechanisms are largely unknown. Here, in vivo and in vitro reconstitution experiments show that the endoplasmic reticulum (ER) kinase PERK promotes cristae formation by increasing TOM70-assisted mitochondrial import of MIC19, a critical subunit of the MICOS complex. Cold stress or ß-adrenergic stimulation activates PERK that phosphorylates O-linked N-acetylglucosamine transferase (OGT). Phosphorylated OGT glycosylates TOM70 on Ser94, enhancing MIC19 protein import into mitochondria and promoting cristae formation and respiration. In addition, PERK-activated OGT O-GlcNAcylates and attenuates CK2α activity, which mediates TOM70 Ser94 phosphorylation and decreases MIC19 mitochondrial protein import. We have identified a cold-stress inter-organelle PERK-OGT-TOM70 axis that increases cell respiration through mitochondrial protein import and subsequent cristae formation. These studies have significant implications in cellular bioenergetics and adaptations to stress conditions.


Subject(s)
Mitochondrial Precursor Protein Import Complex Proteins/metabolism , Mitochondrial Proteins/metabolism , N-Acetylglucosaminyltransferases/metabolism , eIF-2 Kinase/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Animals , Casein Kinase II/metabolism , Cold Temperature , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Glycosylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Precursor Protein Import Complex Proteins/genetics , Mitochondrial Proteins/genetics , N-Acetylglucosaminyltransferases/genetics , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , RNA, Guide, Kinetoplastida/metabolism , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
19.
PLoS Pathog ; 17(1): e1009183, 2021 01.
Article in English | MEDLINE | ID: mdl-33444388

ABSTRACT

The antiviral protein kinase R (PKR) is an important host restriction factor, which poxviruses must overcome to productively infect host cells. To inhibit PKR, many poxviruses encode a pseudosubstrate mimic of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2), designated K3 in vaccinia virus. Although the interaction between PKR and eIF2α is highly conserved, some K3 orthologs from host-restricted poxviruses were previously shown to inhibit PKR in a species-specific manner. To better define this host range function, we compared the sensitivity of PKR from 17 mammals to inhibition by K3 orthologs from closely related orthopoxviruses, a genus with a generally broader host range. The K3 orthologs showed species-specific inhibition of PKR and exhibited three distinct inhibition profiles. In some cases, PKR from closely related species showed dramatic differences in their sensitivity to K3 orthologs. Vaccinia virus expressing the camelpox virus K3 ortholog replicated more than three orders of magnitude better in human and sheep cells than a virus expressing vaccinia virus K3, but both viruses replicated comparably well in cow cells. Strikingly, in site-directed mutagenesis experiments between the variola virus and camelpox virus K3 orthologs, we found that different amino acid combinations were necessary to mediate improved or diminished inhibition of PKR derived from different host species. Because there is likely a limited number of possible variations in PKR that affect K3-interactions but still maintain PKR/eIF2α interactions, it is possible that by chance PKR from some potential new hosts may be susceptible to K3-mediated inhibition from a virus it has never previously encountered. We conclude that neither the sensitivity of host proteins to virus inhibition nor the effectiveness of viral immune antagonists can be inferred from their phylogenetic relatedness but must be experimentally determined.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Host Specificity , Orthopoxvirus/classification , Orthopoxvirus/physiology , Poxviridae Infections/virology , Virus Replication , eIF-2 Kinase/antagonists & inhibitors , Amino Acid Sequence , Animals , Antiviral Agents/metabolism , HeLa Cells , Humans , Phosphorylation , Phylogeny , Poxviridae Infections/genetics , Poxviridae Infections/metabolism , Sequence Homology , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
20.
Prog Neurobiol ; 196: 101892, 2021 01.
Article in English | MEDLINE | ID: mdl-32795489

ABSTRACT

A major challenge in neurobiology is the identification of the mechanisms by which protein misfolding leads to cellular toxicity. Many neurodegenerative disorders, in which aberrant protein conformers aggregate into pathological inclusions, present the chronic activation of the PERK branch of the unfolded protein response. The adaptive effects of the PERK pathway include reduction of translation by transient inhibition of eIF2α and antioxidant protein production via induction of Nrf2 transcription factor. In contrast, PERK prolonged activation leads to sustained reduction in protein synthesis and induction of cell death pathways. To further investigate the role of the PERK pathway in neurodegenerative disorders, we focused on Down syndrome (DS), in which aging confers a high risk of Alzheimer disease (AD). By investigating human DS frontal cortices, we found early and sustained PERK activation associated with the induction of eIF2α and ATF4 downstream signals. We also observed that the Nrf2 response is uncoupled from PERK and its antioxidant effects are repressed in a mechanism implicating the transcription repressor Bach1. The pharmacological inhibition of PERK in DS mice reduced eIF2α-related translational repression and promoted Nrf2 nuclear translocation, favoring the rescue of Nrf2/Bach1 imbalance. The further analysis of peripheral cells from living DS individuals provided strong support of the pathological link between PERK and trisomy 21. Our results suggest that failure to regulate the PERK pathway is a peculiar characteristic of DS pathology and it may represent an essential step to promote cellular dysfunction, which actively contributes in the brain to the early development of AD.


Subject(s)
Alzheimer Disease/metabolism , Down Syndrome/metabolism , Protein Kinase Inhibitors/pharmacology , Unfolded Protein Response/physiology , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Autopsy , Basic-Leucine Zipper Transcription Factors/metabolism , Child , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , NF-E2-Related Factor 2/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...