Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
Biochem Biophys Res Commun ; 719: 150103, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38761636

ABSTRACT

The RNA-binding protein PKR serves as a crucial antiviral innate immune factor that globally suppresses translation by sensing viral double-stranded RNA (dsRNA) and by phosphorylating the translation initiation factor eIF2α. Recent findings have unveiled that single-stranded RNAs (ssRNAs), including in vitro transcribed (IVT) mRNA, can also bind to and activate PKR. However, the precise mechanism underlying PKR activation by ssRNAs, remains incompletely understood. Here, we developed a NanoLuc Binary Technology (NanoBiT)-based in vitro PKR dimerization assay to assess the impact of ssRNAs on PKR dimerization. Our findings demonstrate that, akin to double-stranded polyinosinic:polycytidylic acid (polyIC), an encephalomyocarditis virus (EMCV) RNA, as well as NanoLuc luciferase (Nluc) mRNA, can induce PKR dimerization. Conversely, homopolymeric RNA lacking secondary structure fails to promote PKR dimerization, underscoring the significance of secondary structure in this process. Furthermore, adenovirus VA RNA 1, another ssRNA, impedes PKR dimerization by competing with Nluc mRNA. Additionally, we observed structured ssRNAs capable of forming G-quadruplexes induce PKR dimerization. Collectively, our results indicate that ssRNAs have the ability to either induce or inhibit PKR dimerization, thus representing potential targets for the development of antiviral and anti-inflammatory agents.


Subject(s)
Encephalomyocarditis virus , Protein Multimerization , RNA, Double-Stranded , RNA, Viral , eIF-2 Kinase , eIF-2 Kinase/metabolism , eIF-2 Kinase/chemistry , Humans , RNA, Viral/metabolism , RNA, Viral/genetics , RNA, Viral/chemistry , Encephalomyocarditis virus/genetics , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/chemistry , Poly I-C/pharmacology , Nucleic Acid Conformation
2.
J Biol Chem ; 299(2): 102821, 2023 02.
Article in English | MEDLINE | ID: mdl-36563857

ABSTRACT

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


Subject(s)
Protein Aggregates , Protein Aggregation, Pathological , eIF-2 Kinase , tau Proteins , Humans , Disease Susceptibility , eIF-2 Kinase/chemistry , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Mutation , Risk Factors , tau Proteins/chemistry , tau Proteins/metabolism , Tauopathies/metabolism , Tauopathies/pathology
3.
Proc Natl Acad Sci U S A ; 119(33): e2204235119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35939694

ABSTRACT

Mammalian cells respond to dsRNA in multiple manners. One key response to dsRNA is the activation of PKR, an eIF2α kinase, which triggers translational arrest and the formation of stress granules. However, the process of PKR activation in cells is not fully understood. In response to increased endogenous or exogenous dsRNA, we observed that PKR forms novel cytosolic condensates, referred to as dsRNA-induced foci (dRIFs). dRIFs contain dsRNA, form in proportion to dsRNA, and are enhanced by longer dsRNAs. dRIFs enrich several other dsRNA-binding proteins, including ADAR1, Stau1, NLRP1, and PACT. Strikingly, dRIFs correlate with and form before translation repression by PKR and localize to regions of cells where PKR activation is initiated. We hypothesize that dRIF formation is a mechanism that cells use to enhance the sensitivity of PKR activation in response to low levels of dsRNA or to overcome viral inhibitors of PKR activation.


Subject(s)
RNA, Double-Stranded , RNA, Viral , Virus Diseases , eIF-2 Kinase , Enzyme Activation , Humans , Immunity, Innate , Phosphorylation , Protein Biosynthesis , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/immunology , RNA, Viral/chemistry , RNA, Viral/immunology , RNA-Binding Proteins/chemistry , Stress Granules , Virus Diseases/enzymology , Virus Diseases/immunology , eIF-2 Kinase/chemistry
4.
Fish Shellfish Immunol ; 112: 74-80, 2021 May.
Article in English | MEDLINE | ID: mdl-33667675

ABSTRACT

The dsRNA-activated protein kinase R (PKR) is one of key antiviral effectors induced by interferons (IFNs), and its functions are largely unknown in tilapia, an important commercial fish species suffering from several viral infectious diseases. In the present study, a PKR gene named On-PKR was identified and cloned from Nile tilapia, Oreochromis niloticus. On-PKR gene was constitutively expressed in all tissues examined, with the highest expression level observed in head kidney and liver, and was rapidly induced in all organs/tissues tested following the stimulation of poly(I:C). Importantly, the expression of On-PKR is induced by group I and group II IFNs with distinct induction kinetics in vivo: group I IFN elicits a relative delayed but sustained induction of On-PKR, whereas group II IFN triggers a rapid and transient expression of On-PKR. Moreover, the overexpression of On-PKR has been proven to inhibit the protein translation and virus replication in fish cells. The present study thus contributes to a better understanding of the functions of antiviral effectors in tilapia, and may provide clues for the prevention and therapy of viral diseases in fish.


Subject(s)
Cichlids/genetics , Cichlids/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology , Amino Acid Sequence , Animals , Fish Diseases/virology , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Immunity/genetics , Phylogeny , Poly I-C/pharmacology , Reoviridae/physiology , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Reoviridae Infections/virology , Sequence Alignment/veterinary , eIF-2 Kinase/chemistry
5.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562773

ABSTRACT

NSCLC (non-small cell lung cancer) is a leading cause of cancer-related deaths worldwide. Clinical trials showed that Hiltonol, a stable dsRNA representing an advanced form of polyI:C (polyinosinic-polycytidilic acid), is an adjuvant cancer-immunomodulator. However, its mechanisms of action and effect on lung cancer have not been explored pre-clinically. Here, we examined, for the first time, how a novel Hiltonol cocktail kills NSCLC cells. By retrospective analysis of NSCLC patient tissues obtained from the tumor biobank; pre-clinical studies with Hiltonol alone or Hiltonol+++ cocktail [Hiltonol+anti-IL6+AG490 (JAK2 inhibitor)+Stattic (STAT3 inhibitor)]; cytokine analysis; gene knockdown and gain/loss-of-function studies, we uncovered the mechanisms of action of Hiltonol+++. We demonstrated that Hiltonol+++ kills the cancer cells and suppresses the metastatic potential of NSCLC through: (i) upregulation of pro-apoptotic Caspase-9 and Caspase-3, (ii) induction of cytosolic cytochrome c, (iii) modulation of pro-inflammatory cytokines (GRO, MCP-1, IL-8, and IL-6) and anticancer IL-24 in NSCLC subtypes, and (iv) upregulation of tumor suppressors, PKR (protein kinase R) and OAS (2'5' oligoadenylate synthetase). In silico analysis showed that Lys296 of PKR and Lys66 of OAS interact with Hiltonol. These Lys residues are purportedly involved in the catalytic/signaling activity of the tumor suppressors. Furthermore, knockdown of PKR/OAS abrogated the anticancer action of Hiltonol, provoking survival of cancer cells. Ex vivo analysis of NSCLC patient tissues corroborated that loss of PKR and OAS is associated with cancer advancement. Altogether, our findings unraveled the significance of studying tumor biobank tissues, which suggests PKR and OAS as precision oncological suppressor candidates to be targeted by this novel Hiltonol+++ cocktail which represents a prospective drug for development into a potent and tailored therapy for NSCLC subtypes.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Antineoplastic Agents, Immunological/pharmacology , Carboxymethylcellulose Sodium/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/metabolism , Cyclic S-Oxides/pharmacology , Lung Neoplasms/metabolism , Poly I-C/pharmacology , Polylysine/analogs & derivatives , Tyrphostins/pharmacology , eIF-2 Kinase/metabolism , 2',5'-Oligoadenylate Synthetase/chemistry , 2',5'-Oligoadenylate Synthetase/genetics , A549 Cells , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Binding Sites , Carboxymethylcellulose Sodium/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Models, Molecular , Polylysine/pharmacology , Tumor Microenvironment/drug effects , eIF-2 Kinase/chemistry , eIF-2 Kinase/genetics
6.
PLoS Comput Biol ; 16(5): e1007864, 2020 05.
Article in English | MEDLINE | ID: mdl-32453748

ABSTRACT

Interactions between disordered proteins involve a wide range of changes in the structure and dynamics of the partners involved. These changes can be classified in terms of binding modes, which include disorder-to-order (DO) transitions, when proteins fold upon binding, as well as disorder-to-disorder (DD) transitions, when the conformational heterogeneity is maintained in the bound states. Furthermore, systematic studies of these interactions are revealing that proteins may exhibit different binding modes with different partners. Proteins that exhibit this context-dependent binding can be referred to as fuzzy proteins. Here we investigate amino acid code for fuzzy binding in terms of the entropy of the probability distribution of transitions towards decreasing order. We implement these entropy calculations into the FuzPred (http://protdyn-fuzpred.org) algorithm to predict the range of context-dependent binding modes of proteins from their amino acid sequences. As we illustrate through a variety of examples, this method identifies those binding sites that are sensitive to the cellular context or post-translational modifications, and may serve as regulatory points of cellular pathways.


Subject(s)
Binding Sites , Protein Binding , Protein Processing, Post-Translational , Proteins/chemistry , Algorithms , Computational Biology/methods , Databases, Protein , Eukaryotic Initiation Factor-2/chemistry , Fuzzy Logic , Humans , Intrinsically Disordered Proteins/chemistry , Probability , Protein Domains , Protein Folding , ROC Curve , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Tumor Suppressor Protein p53/chemistry , eIF-2 Kinase/chemistry
7.
Biochemistry ; 59(12): 1252-1260, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32176479

ABSTRACT

Protein kinase R (PKR) is a key antiviral component of the innate immune pathway and is activated by viral double-stranded RNAs (dsRNAs). Adenovirus-associated RNA 1 (VAI) is an abundant, noncoding viral RNA that functions as a decoy by binding PKR but not inducing activation, thereby inhibiting the antiviral response. In VAI, coaxial stacking produces an extended helix that mediates high-affinity PKR binding but is too short to result in activation. Like adenovirus, Epstein-Barr virus produces high concentrations of a noncoding RNA, EBER1. Here, we compare interactions of PKR with VAI and EBER1 and present a structural model of EBER1. Both RNAs function as inhibitors of dsRNA-mediated PKR activation. However, EBER1 weakly activates PKR whereas VAI does not. PKR binds EBER1 more weakly than VAI. Assays at physiological ion concentrations indicate that both RNAs can accommodate two PKR monomers and induce PKR dimerization. A structural model of EBER1 was obtained using constraints derived from chemical structure probing and small-angle X-ray scattering experiments. The central stem of EBER1 coaxially stacks with stem loop 4 and stem loop 1 to form an extended RNA duplex of ∼32 bp that binds PKR and promotes activation. Our observations that EBER1 binds PKR much more weakly than VAI and exhibits weak PKR activation suggest that EBER1 is less well suited to function as an RNA decoy.


Subject(s)
Herpesvirus 4, Human/genetics , Host Microbial Interactions/genetics , RNA, Viral/metabolism , eIF-2 Kinase/genetics , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/immunology , Humans , Immunity, Innate/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Multimerization/genetics , Protein Multimerization/immunology , RNA Stability , RNA, Viral/chemistry , Scattering, Small Angle , X-Ray Diffraction , eIF-2 Kinase/chemistry , eIF-2 Kinase/immunology , eIF-2 Kinase/metabolism
8.
Nat Struct Mol Biol ; 26(11): 1053-1062, 2019 11.
Article in English | MEDLINE | ID: mdl-31695187

ABSTRACT

BiP is a major endoplasmic reticulum (ER) chaperone and is suggested to act as primary sensor in the activation of the unfolded protein response (UPR). How BiP operates as a molecular chaperone and as an ER stress sensor is unknown. Here, by reconstituting components of human UPR, ER stress and BiP chaperone systems, we discover that the interaction of BiP with the luminal domains of UPR proteins IRE1 and PERK switch BiP from its chaperone cycle into an ER stress sensor cycle by preventing the binding of its co-chaperones, with loss of ATPase stimulation. Furthermore, misfolded protein-dependent dissociation of BiP from IRE1 is primed by ATP but not ADP. Our data elucidate a previously unidentified mechanistic cycle of BiP function that explains its ability to act as an Hsp70 chaperone and ER stress sensor.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Heat-Shock Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism , Adenosine Triphosphate/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/chemistry , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/chemistry , Humans , Models, Molecular , Protein Folding , Protein Interaction Domains and Motifs , Protein Interaction Maps , Protein Serine-Threonine Kinases/chemistry , Unfolded Protein Response , eIF-2 Kinase/chemistry
9.
Biochemistry ; 58(27): 2967-2977, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31246429

ABSTRACT

The RNA-activated protein kinase, PKR, is a key mediator of the innate immunity response to viral infection. Viral double-stranded RNAs induce PKR dimerization and autophosphorylation. The PKR kinase domain forms a back-to-back dimer. However, intermolecular ( trans) autophosphorylation is not feasible in this arrangement. We have obtained PKR kinase structures that resolves this dilemma. The kinase protomers interact via the known back-to-back interface as well as a front-to-front interface that is formed by exchange of activation segments. Mutational analysis of the front-to-front interface support a functional role in PKR activation. Molecular dynamics simulations reveal that the activation segment is highly dynamic in the front-to-front dimer and can adopt conformations conducive to phosphoryl transfer. We propose a mechanism where back-to-back dimerization induces a conformational change that activates PKR to phosphorylate a "substrate" kinase docked in a front-to-front geometry. This mechanism may be relevant to related kinases that phosphorylate the eukaryotic initiation factor eIF2α.


Subject(s)
eIF-2 Kinase/chemistry , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Phosphorylation , Protein Conformation , Protein Domains , Protein Multimerization
10.
J Virol ; 93(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31189710

ABSTRACT

Accumulated evidence demonstrates that Japanese encephalitis virus (JEV) infection triggers endoplasmic reticulum (ER) stress and neuron apoptosis. ER stress sensor protein kinase R-like endoplasmic reticulum kinase (PERK) has been reported to induce apoptosis under acute or prolonged ER stress. However, the precise role of PERK in JEV-induced apoptosis and encephalitis remains unknown. Here, we report that JEV infection activates the PERK-ATF4-CHOP apoptosis pathway both in vitro and in vivo PERK activation also promotes the formation of stress granule, which in turn represses JEV-induced apoptosis. However, PERK inhibitor reduces apoptosis, indicating that JEV-activated PERK predominantly induces apoptosis via the PERK-ATF4-CHOP apoptosis pathway. Among JEV proteins that have been reported to induce ER stress, only JEV NS4B can induce PERK activation. PERK has been reported to form an active molecule by dimerization. The coimmunoprecipitation assay shows that NS4B interacts with PERK. Moreover, glycerol gradient centrifugation shows that NS4B induces PERK dimerization. Both the LIG-FHA and the LIG-WD40 domains within NS4B are required to induce PERK dimerization, suggesting that JEV NS4B pulls two PERK molecules together by simultaneously interacting with them via different motifs. PERK deactivation reduces brain cell damage and encephalitis during JEV infection. Furthermore, expression of JEV NS4B is sufficient to induce encephalitis via PERK in mice, indicating that JEV activates PERK primarily via its NS4B to cause encephalitis. Taken together, our findings provide a novel insight into JEV-caused encephalitis.IMPORTANCE Japanese encephalitis virus (JEV) infection triggers endoplasmic reticulum (ER) stress and neuron apoptosis. ER stress sensor protein kinase R-like endoplasmic reticulum kinase (PERK) has been reported to induce apoptosis under acute or prolonged ER stress. However, whether the PERK pathway of ER stress response plays important roles in JEV-induced apoptosis and encephalitis remains unknown. Here, we found that JEV infection activates ER stress sensor PERK in neuronal cells and mouse brains. PERK activation induces apoptosis via the PERK-ATF4-CHOP apoptosis pathway upon JEV infection. Among the JEV proteins prM, E, NS1, NS2A, NS2B, and NS4B, only NS4B activates PERK. Moreover, activated PERK participates in apoptosis and encephalitis induced by JEV and NS4B. These findings provide a novel therapeutic approach for JEV-caused encephalitis.


Subject(s)
Encephalitis Virus, Japanese/pathogenicity , Encephalitis, Japanese/metabolism , Neurons/cytology , Viral Nonstructural Proteins/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Animals , Apoptosis , Binding Sites , Cell Line , Disease Models, Animal , Encephalitis Virus, Japanese/metabolism , Encephalitis, Japanese/virology , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Indoles/pharmacology , Indoles/therapeutic use , Mice , Neurons/metabolism , Neurons/virology , Protein Multimerization , Signal Transduction , Transcription Factor CHOP/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , eIF-2 Kinase/chemistry
11.
Autophagy ; 15(7): 1214-1233, 2019 07.
Article in English | MEDLINE | ID: mdl-30741620

ABSTRACT

Mutations in the ER-associated VAPB/ALS8 protein cause amyotrophic lateral sclerosis and spinal muscular atrophy. Previous studies have argued that ER stress may underlie the demise of neurons. We find that loss of VAP proteins (VAPs) leads to an accumulation of aberrant lysosomes and impairs lysosomal degradation. VAPs mediate ER to Golgi tethering and their loss may affect phosphatidylinositol-4-phosphate (PtdIns4P) transfer between these organelles. We found that loss of VAPs elevates PtdIns4P levels in the Golgi, leading to an expansion of the endosomal pool derived from the Golgi. Fusion of these endosomes with lysosomes leads to an increase in lysosomes with aberrant acidity, contents, and shape. Importantly, reducing PtdIns4P levels with a PtdIns4-kinase (PtdIns4K) inhibitor, or removing a single copy of Rab7, suppress macroautophagic/autophagic degradation defects as well as behavioral defects observed in Drosophila Vap33 mutant larvae. We propose that a failure to tether the ER to the Golgi when VAPs are lost leads to an increase in Golgi PtdIns4P levels, and an expansion of endosomes resulting in an accumulation of dysfunctional lysosomes and a failure in proper autophagic lysosomal degradation. Abbreviations: ALS: amyotrophic lateral sclerosis; CSF: cerebrospinal fluid; CERT: ceramide transfer protein; FFAT: two phenylalanines in an acidic tract; MSP: major sperm proteins; OSBP: oxysterol binding protein; PH: pleckstrin homology; PtdIns4P: phosphatidylinositol-4-phosphate; PtdIns4K: phosphatidylinositol 4-kinase; UPR: unfolded protein response; VAMP: vesicle-associated membrane protein; VAPA/B: mammalian VAPA and VAPB proteins; VAPs: VAMP-associated proteins (referring to Drosophila Vap33, and human VAPA and VAPB).


Subject(s)
Autophagy/genetics , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , R-SNARE Proteins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy/drug effects , Carrier Proteins/chemistry , Carrier Proteins/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endosomes/drug effects , Endosomes/genetics , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomes/drug effects , Lysosomes/genetics , Lysosomes/ultrastructure , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation , R-SNARE Proteins/genetics , eIF-2 Kinase/chemistry , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
12.
RNA ; 25(5): 539-556, 2019 05.
Article in English | MEDLINE | ID: mdl-30770398

ABSTRACT

The interferon-inducible protein kinase R (PKR) is a key component of host innate immunity that restricts viral replication and propagation. As one of the four eIF2α kinases that sense diverse stresses and direct the integrated stress response (ISR) crucial for cell survival and proliferation, PKR's versatile roles extend well beyond antiviral defense. Targeted by numerous host and viral regulators made of RNA and proteins, PKR is subject to multiple layers of endogenous control and external manipulation, driving its rapid evolution. These versatile regulators include not only the canonical double-stranded RNA (dsRNA) that activates the kinase activity of PKR, but also highly structured viral, host, and artificial RNAs that exert a full spectrum of effects. In this review, we discuss our deepening understanding of the allosteric mechanism that connects the regulatory and effector domains of PKR, with an emphasis on diverse structured RNA regulators in comparison to their protein counterparts. Through this analysis, we conclude that much of the mechanistic details that underlie this RNA-regulated kinase await structural and functional elucidation, upon which we can then describe a "PKR code," a set of structural and chemical features of RNA that are both descriptive and predictive for their effects on PKR.


Subject(s)
Host-Pathogen Interactions/genetics , RNA, Double-Stranded/genetics , RNA, Untranslated/genetics , Virus Diseases/genetics , eIF-2 Kinase/genetics , Allosteric Regulation , Animals , Base Sequence , Binding Sites , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Interferons/genetics , Interferons/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/immunology , RNA, Untranslated/chemistry , RNA, Untranslated/immunology , Virus Diseases/immunology , Virus Diseases/virology , Virus Replication , eIF-2 Kinase/chemistry , eIF-2 Kinase/immunology
13.
Mol Biol Rep ; 46(1): 1275-1284, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30694453

ABSTRACT

Recent in vitro studies have shown that vitamin C (Vit C) with pro-oxidative properties causes cytotoxicity of breast cancer cells by selective oxidative stress. However, the effect of Vit C in itself at different concentration levels on MCF-7 breast cancer cell line after 24 h, has not yet been described. We aimed to examine the effect of Vit C on the viability and signalling response of MCF-7/WT (MCF-7 wild-type) cells that were exposed to various concentrations (0.125-4 mM) of Vit C during 24 h. The cytotoxic effect of Vit C on MCF-7/VitC (MCF-7/WT after added 2 mM Vit C) was observed, resulting in a decrease of cell index after 12 h. Also, the cytotoxicity of Vit C (2 mM) after 24 h was confirmed by flow cytometry, i.e., increase of dead, late apoptotic, and depolarized dead MCF-7/VitC cells compared to MCF-7/WT cells. Moreover, changes in proteomic profile of MCF-7/VitC cells compared to the control group were investigated via label-free quantitative mass spectrometry and post-translational modification. Using bioinformatics assessment (i.e., iPathwayGuide and SPIA R packages), a significantly impacted pathway in MCF-7/VitC was identified, namely the protein processing in endoplasmic reticulum. The semi-quantitative change (log2fold change = 4.5) and autophosphorylation at Thr-446 of protein kinase (PKR) (involved in this pathway) indicates that PKR protein could be responsible for the unfolded protein response and inhibition of the cell translation during endoplasmic reticulum stress, and eventually, for cell apoptosis. These results suggest that increased activity of PKR (Thr-446 autophosphorylation) related to cytotoxic effect of Vit C (2 mM) may cause the MCF-7 cells death.


Subject(s)
Ascorbic Acid/pharmacology , Endoplasmic Reticulum Stress/drug effects , Unfolded Protein Response/drug effects , Apoptosis/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Eukaryotic Initiation Factor-2/chemistry , Eukaryotic Initiation Factor-2/metabolism , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Models, Molecular , Phosphorylation/drug effects , Phosphothreonine/metabolism , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects , eIF-2 Kinase/chemistry , eIF-2 Kinase/metabolism
14.
J Biomol Struct Dyn ; 37(7): 1715-1723, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29663856

ABSTRACT

The Heme-Regulated Inhibitor (HRI) kinase regulates globin synthesis in a heme-dependent manner in reticulocytes and erythroid cells in bone marrow. Inhibitors of HRI have been proposed to lead to an increased amount of haemoglobin, benefitting anaemia patients. A series of indeno[1,2-c]pyrazoles were discovered to be the first known in vitro inhibitors of HRI. However, the structural mechanism of inhibition is yet to be understood. The aim of this study was to unravel the binding mechanism of these inhibitors using molecular dynamic simulations and docking. The docking scores were observed to correlate well with experimentally determined pIC50 values. The inhibitors were observed to bind in the ATP-binding site forming hydrogen bonds with the hinge region and van der Waals interactions with non-polar residues in the binding site. Further, quantitative structure-activity relationship (QSAR) studies were performed to correlate the structural features of the inhibitors with their biological activity. The developed QSAR models were found to be statistically significant in terms of internal and external predictabilities. The presence of chlorine atoms and the hydroxymethyl groups were found to correlate with higher activity. The identified binding modes and the descriptors can support future rational identification of more potent and selective small molecule inhibitors for this kinase which are of therapeutic importance in the context of various human pathological disorders.


Subject(s)
Binding Sites , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/chemistry , eIF-2 Kinase/chemistry , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Interaction Domains and Motifs/drug effects , Protein Kinase Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , eIF-2 Kinase/antagonists & inhibitors
15.
J Interferon Cytokine Res ; 38(8): 333-340, 2018 08.
Article in English | MEDLINE | ID: mdl-30052119

ABSTRACT

Double-stranded RNA-dependent protein kinase (PKR) is an important antiviral IFN-stimulated gene (ISGs) that recognizes double-stranded RNA (dsRNA) and mediates inhibition of translation initiation and protein synthesis in various types of viral infection. In this study, the complete coding sequence (CDS) of goose PKR (goPKR) is identified and characterized. The open reading frame (ORF) of goPKR is 1668 bp, which encodes a polypeptide of 555 amino acids. The sequence identity results demonstrate that the goose PKR is most closely related to duck PKR gene, with nucleotide identities of 91.6%, whereas nucleotide identity of the goose PKR to chicken, human, and mouse PKR is 76.4%, 51.9%, and 52.0%, respectively. Interestingly, the deduced amino acid sequence of goose PKR contains 3 main structure domains, including 2 double-strand RNA-binding motif (dsRBM) domains and one serine/threonine protein kinase domain. This is similar to the chicken and mammals, whereas it is different from duck PKR protein, which contains only one dsRBM1 domain and one serine/threonine protein kinase domain. Quantitative real-time PCR analysis indicates that goose PKR mRNA is widely expressed in all sampled tissues. It is highly expressed in the blood, spleen, lung, and bursa of Fabricius and jejunum and is slightly expressed in heart, muscle, trachea, and brain. The results of confocal microscopy suggest that PKR-EGFP is mainly localized in the cytoplasm, and overexpression of goPKR protein significantly reduces Newcastle disease virus (NDV) replication (viral copies and viral titer) in goose embryo fibroblasts. These findings show that goose PKR is an important antiviral ISG, involved in the antiviral innate immune defense to NDV in geese.


Subject(s)
Antiviral Agents/pharmacology , Geese/genetics , Gene Expression Profiling , Newcastle disease virus/drug effects , Peptides/pharmacology , eIF-2 Kinase/genetics , eIF-2 Kinase/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Newcastle disease virus/metabolism , Peptides/chemistry , Peptides/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Virus Replication/drug effects , eIF-2 Kinase/chemistry , eIF-2 Kinase/metabolism
16.
Mol Immunol ; 101: 65-73, 2018 09.
Article in English | MEDLINE | ID: mdl-29879548

ABSTRACT

The human protein kinase R (PKR) recognizes invading RNA viruses and mediates the antiviral immune response by phosphorylating the eukaryotic translation initiation factor 2α (eIF-2α), thus blocking protein translation in infected cells and thus preventing viral replication. The observation that individuals show different degrees of susceptibility to viral infections gives rise to the hypothesis that single nucleotide polymorphisms (SNPs) in the protein kinase R may alter the response to an infection. Using different available servers (e.g. SIFT, PROVEAN, Polyphen2, SNAP2, SNP&GOs, SNP-PhD, I-Mutant Suite), 14 SNPs were identified that were predicted to have deleterious effects on the protein kinase R. Five SNPs, namely D266Y, Y323D, I398 K, Y465C and Y472C, were selected for homology modeling and the generated models were investigated with regard to their secondary structure, residue fluctuations and eIF-2α binding. Analysis with computational tools POLYVIEW-MM, SAAPdap, SRIDE, CMView, elNémo, NMsim and PatchDock revealed structural changes in all mutants yielding a more stable structure at the cost of reduced flexibility (except Y465C) and less conformational freedom compared to the native protein. The conformational changes in the mutant protein structures and the displacement of functional residues from their strategic positions are predicted to affect the functionality of PKR, and consequently will affect the efficiency of the individual's antiviral immune response negatively. This study will aid the physicians in precision medicine field to tailor optimal treatment for the patients.


Subject(s)
Computational Biology/methods , Polymorphism, Single Nucleotide/genetics , eIF-2 Kinase/genetics , Amino Acid Sequence , Databases, Genetic , Eukaryotic Initiation Factor-2/metabolism , Humans , Molecular Docking Simulation , Mutant Proteins/chemistry , Phosphorylation , Protein Binding , Protein Domains , Protein Structure, Secondary , eIF-2 Kinase/chemistry , eIF-2 Kinase/metabolism
17.
Biotechnol Genet Eng Rev ; 34(1): 33-59, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29716441

ABSTRACT

The innate immune system offers a first line of defense by neutralizing foreign pathogens such as bacteria, fungi, and viruses. These pathogens express molecules (RNA and proteins) that have discrete structures, known as the pathogen-associated molecular patterns that are recognized by a highly specialized class of host proteins called pattern recognition receptors to facilitate the host's immune response against infection. The RNA-dependent Protein Kinase R (PKR) is one of the host's pattern recognition receptors that is a key component of an innate immune system. PKR recognizes imperfectly double-stranded non-coding viral RNA molecules via its N-terminal double-stranded RNA binding motifs, undergoes phosphorylation of the C-terminal kinase domain, ultimately resulting in inhibition of viral protein translation by inhibiting the guanine nucleotide exchange activity of eukaryotic initiation factor 2α. Not surprisingly, viruses have evolved mechanisms by which viral non-coding RNA or protein molecules inhibit PKR's activation and/or its downstream activity to allow viral replication. In this review, we will highlight the role of viral proteins in inhibiting PKR's activity and summarize currently known mechanisms by which viral proteins execute such inhibitory activity.


Subject(s)
RNA, Double-Stranded/metabolism , Viral Proteins/metabolism , Virus Diseases/immunology , eIF-2 Kinase/metabolism , Binding Sites , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Humans , Immunity, Innate , Protein Binding , Protein Biosynthesis , RNA, Viral/metabolism , Virus Diseases/virology , Virus Replication , Viruses/genetics , Viruses/immunology , eIF-2 Kinase/chemistry
18.
PLoS One ; 13(3): e0194335, 2018.
Article in English | MEDLINE | ID: mdl-29538447

ABSTRACT

Eukaryotic translation initiation factor 2-alpha kinase (EIF2AK) proteins inhibit protein synthesis at translation initiation level, in response to various stress conditions, including oxidative stress, heme deficiency, osmotic shock, and heat shock. Origin and functional diversification of EIF2AK sequences remain ambiguous. Here we determine the origin and molecular evolution of EIF2AK proteins in lower eukaryotes and studied the molecular basis of divergence among sub-family sequences. Present work emphasized primitive origin of EIF2AK4 sub-family gene in lower eukaryotes of protozoan lineage. Phylogenetic analysis supported common origin and sub-family based classification of EIF2AKs. Functional divergence studies across sub-families revealed several putative amino acid sites, which assist in altered protein interactions of kinase domains. The data can facilitate designing site-directed experimental studies aiming at elucidating diverse functional aspects of kinase domains regarding down-regulation of protein synthesis.


Subject(s)
Evolution, Molecular , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Amino Acid Sequence , Animals , Binding Sites , Computational Biology/methods , Databases, Genetic , Multigene Family , Phylogeny , Protein Binding , Protein Interaction Domains and Motifs , Sequence Alignment , Sequence Analysis, DNA , eIF-2 Kinase/chemistry
19.
J Biol Chem ; 293(11): 4110-4121, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29386355

ABSTRACT

PRKR-like endoplasmic reticulum kinase (PERK) is one of the major sensor proteins that detect protein folding imbalances during endoplasmic reticulum (ER) stress. However, it remains unclear how ER stress activates PERK to initiate a downstream unfolded protein response (UPR). Here, we found that PERK's luminal domain can recognize and selectively interact with misfolded proteins but not with native proteins. Screening a phage-display library, we identified a peptide substrate, P16, of the PERK luminal domain and confirmed that P16 efficiently competes with misfolded proteins for binding this domain. To unravel the mechanism by which the PERK luminal domain interacts with misfolded proteins, we determined the crystal structure of the bovine PERK luminal domain complexed with P16 to 2.8-Å resolution. The structure revealed that PERK's luminal domain binds the peptide through a conserved hydrophobic groove. Substitutions within hydrophobic regions of the PERK luminal domain abolished the binding between PERK and misfolded proteins. We also noted that peptide binding results in major conformational changes in the PERK luminal domain that may favor PERK oligomerization. The structure of the PERK luminal domain-P16 complex suggested stacking of the luminal domain that leads to PERK oligomerization and activation via autophosphorylation after ligand binding. Collectively, our structural and biochemical results strongly support a ligand-driven model in which the PERK luminal domain interacts directly with misfolded proteins to induce PERK oligomerization and activation, resulting in ER stress signaling and the UPR.


Subject(s)
Peptide Fragments/metabolism , Protein Folding , Protein Multimerization , eIF-2 Kinase/chemistry , eIF-2 Kinase/metabolism , Animals , Binding Sites , Cattle , Cells, Cultured , Crystallography, X-Ray , Endoplasmic Reticulum Stress , Mice , Mice, Knockout , Peptide Fragments/chemistry , Peptide Library , Phosphorylation , Protein Binding , Protein Conformation , Signal Transduction
20.
J Biomol Struct Dyn ; 36(11): 2845-2861, 2018 Aug.
Article in English | MEDLINE | ID: mdl-28814160

ABSTRACT

The eIF2α kinase activity of the heme-regulated inhibitor (HRI) is regulated by heme which makes it a unique member of the family of eIF2α kinases. Since heme concentrations create an equilibrium for the kinase to be active/inactive, it becomes important to study the heme binding effects upon the kinase and understanding its mechanism of functionality. In the present study, we report the thermostability achieved by the catalytic kinase domain of HRI (HRI.CKD) upon ligand (heme) binding. Our CD data demonstrates that the HRI.CKD retains its secondary structure at higher temperatures when it is in ligand bound state. HRI.CKD when incubated with hemin loses its monomeric state and attains a higher order oligomeric form resulting in its stability. The HRI.CKD fails to refold into its native conformation upon mutation of H377A/H381A, thereby confirming the necessity of these His residues for correct folding, stability, and activity of the kinase. Though our in silico study demonstrated these His being the ligand binding sites in the kinase insert region, the spectra-based study did not show significant difference in heme affinity for the wild type and His mutant HRI.CKD.


Subject(s)
Catalytic Domain , Heme/chemistry , Hemin/chemistry , Models, Molecular , eIF-2 Kinase/chemistry , Binding Sites , Calorimetry, Differential Scanning , Gene Expression , Heme/metabolism , Hemin/metabolism , Ligands , Molecular Conformation , Mutation , Protein Binding , Protein Stability , Recombinant Proteins , Structure-Activity Relationship , Thermodynamics , eIF-2 Kinase/genetics , eIF-2 Kinase/isolation & purification , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL