Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 7(68): eabi6763, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35148201

RESUMO

Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αß) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor-induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum-associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.


Assuntos
Imunidade Inata/imunologia , Interleucinas/imunologia , eIF-2 Quinase/imunologia , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , eIF-2 Quinase/deficiência
2.
J Clin Invest ; 131(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34324437

RESUMO

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.


Assuntos
Arritmias Cardíacas/prevenção & controle , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , eIF-2 Quinase/antagonistas & inibidores , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/metabolismo , Regulação para Baixo , Feminino , Fatores de Risco de Doenças Cardíacas , Humanos , Indóis/farmacologia , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Cardiovasculares , Inibidores de Proteínas Quinases/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética
3.
Cell Metab ; 33(3): 598-614.e7, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33592173

RESUMO

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.


Assuntos
Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial/metabolismo , Proteínas Mitocondriais/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , eIF-2 Quinase/metabolismo , Adipócitos Marrons/citologia , Adipócitos Marrons/efeitos dos fármacos , Adipócitos Marrons/metabolismo , Animais , Caseína Quinase II/metabolismo , Temperatura Baixa , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Glicosilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial/genética , Proteínas Mitocondriais/genética , N-Acetilglucosaminiltransferases/genética , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , RNA Guia de Cinetoplastídeos/metabolismo , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética
4.
Immunity ; 52(4): 668-682.e7, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294407

RESUMO

The primary mechanisms supporting immunoregulatory polarization of myeloid cells upon infiltration into tumors remain largely unexplored. Elucidation of these signals could enable better strategies to restore protective anti-tumor immunity. Here, we investigated the role of the intrinsic activation of the PKR-like endoplasmic reticulum (ER) kinase (PERK) in the immunoinhibitory actions of tumor-associated myeloid-derived suppressor cells (tumor-MDSCs). PERK signaling increased in tumor-MDSCs, and its deletion transformed MDSCs into myeloid cells that activated CD8+ T cell-mediated immunity against cancer. Tumor-MDSCs lacking PERK exhibited disrupted NRF2-driven antioxidant capacity and impaired mitochondrial respiratory homeostasis. Moreover, reduced NRF2 signaling in PERK-deficient MDSCs elicited cytosolic mitochondrial DNA elevation and, consequently, STING-dependent expression of anti-tumor type I interferon. Reactivation of NRF2 signaling, conditional deletion of STING, or blockade of type I interferon receptor I restored the immunoinhibitory potential of PERK-ablated MDSCs. Our findings demonstrate the pivotal role of PERK in tumor-MDSC functionality and unveil strategies to reprogram immunosuppressive myelopoiesis in tumors to boost cancer immunotherapy.


Assuntos
Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Epitelial do Ovário/imunologia , Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/imunologia , Proteínas de Membrana/imunologia , Neoplasias Cutâneas/imunologia , eIF-2 Quinase/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Epitelial do Ovário/genética , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma Epitelial do Ovário/patologia , Feminino , Humanos , Terapia de Imunossupressão , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/imunologia , Mitocôndrias/metabolismo , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/patologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/imunologia , Receptores de Interferon/genética , Receptores de Interferon/imunologia , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Resposta a Proteínas não Dobradas/imunologia , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética
5.
Proc Natl Acad Sci U S A ; 116(47): 23671-23681, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31690657

RESUMO

Invariant NKT (iNKT) cells have the unique ability to shape immunity during antitumor immune responses and other forms of sterile and nonsterile inflammation. Recent studies have highlighted a variety of classes of endogenous and pathogen-derived lipid antigens that can trigger iNKT cell activation under sterile and nonsterile conditions. However, the context and mechanisms that drive the presentation of self-lipid antigens in sterile inflammation remain unclear. Here we report that endoplasmic reticulum (ER)-stressed myeloid cells, via signaling events modulated by the protein kinase RNA-like ER kinase (PERK) pathway, increase CD1d-mediated presentation of immunogenic endogenous lipid species, which results in enhanced iNKT cell activation both in vitro and in vivo. In addition, we demonstrate that actin cytoskeletal reorganization during ER stress results in an altered distribution of CD1d on the cell surface, which contributes to enhanced iNKT cell activation. These results define a previously unidentified mechanism that controls iNKT cell activation during sterile inflammation.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/imunologia , Estresse do Retículo Endoplasmático/imunologia , Ativação Linfocitária , Células T Matadoras Naturais/imunologia , Animais , Apresentação de Antígeno , Antígenos CD1d/biossíntese , Antígenos CD1d/imunologia , Autoantígenos/imunologia , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Citoesqueleto/ultraestrutura , Endossomos/imunologia , Glicoesfingolipídeos/imunologia , Glicoesfingolipídeos/metabolismo , Humanos , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Lipídeos/imunologia , Lisossomos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células THP-1 , Tapsigargina/farmacologia , Resposta a Proteínas não Dobradas/imunologia , eIF-2 Quinase/deficiência , eIF-2 Quinase/fisiologia
6.
Appl Microbiol Biotechnol ; 103(15): 6153-6167, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31154490

RESUMO

MoHrip2, identified from Magnaporthe oryzae as an elicitor, can activate plant defense responses either in the form of recombinant protein in vitro or ectopic expressed protein in rice. However, its intrinsic function in the infective interaction of M. oryzae-rice is largely unknown. Here, we found that mohrip2 expression was significantly induced at stages of fungal penetration and colonization. Meanwhile, the induced MoHrip2 mainly accumulated in the rice apoplast by outlining the entire invasive hyphae during infection, and its secretion was via the conventional endoplasmic reticulum (ER)-to-Golgi pathway, demonstrating the nature of MoHrip2 as an apoplastic effector. What's more, the disease facilitating function of MoHrip2 was revealed by the significantly compromised virulence of Δmohrip2 mutants on rice seedlings and even on the wounded rice leaves. Inoculations of these mutant strains on rice leaf sheaths showed a reduction in penetration and subsequent expansion of fungal growth, which is probably due to activated host immunity including the expression of certain defense-related genes and the production of certain phytoalexins. Altogether, these results demonstrated the necessity of MoHrip2 in suppression of host immunity and the full virulence of M. oryzae.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Magnaporthe/patogenicidade , Oryza/microbiologia , Doenças das Plantas/microbiologia , Fatores de Virulência/metabolismo , eIF-2 Quinase/metabolismo , Deleção de Genes , Magnaporthe/enzimologia , Oryza/imunologia , Virulência , Fatores de Virulência/deficiência , eIF-2 Quinase/deficiência
7.
Aging Cell ; 18(3): e12887, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30821420

RESUMO

Brain lesions in Alzheimer's disease (AD) include amyloid plaques made of Aß peptides and neurofibrillary tangles composed of hyperphosphorylated tau protein with synaptic and neuronal loss and neuroinflammation. Aß oligomers can trigger tau phosphorylation and neuronal alterations through activation of neuronal kinases leading to progressive cognitive decline. PKR is a ubiquitous pro-apoptotic serine/threonine kinase, and levels of activated PKR are increased in AD brains and AD CSF. In addition, PKR regulates negatively memory formation in mice. To assess the role of PKR in an AD in vivo model, we crossed 5xFAD transgenic mice with PKR knockout (PKRKO) mice and we explored the contribution of PKR on cognition and brain lesions in the 5xFAD mouse model of AD as well as in neuron-microglia co-cultures exposed to the innate immunity activator lipopolysaccharide (LPS). Nine-month-old double-mutant mice revealed significantly improved memory consolidation with the new object location test, starmaze test, and elevated plus maze test as compared to 5xFAD mice. Brain amyloid accumulation and BACE1 levels were statistically decreased in double-mutant mice. Apoptosis, neurodegeneration markers, and synaptic alterations were significantly reduced in double-mutant mice as well as neuroinflammation markers such as microglial load and brain cytokine levels. Using cocultures, we found that PKR in neurons was essential for LPS microglia-induced neuronal death. Our results demonstrate the clear involvement of PKR in abnormal spatial memory and brain lesions in the 5xFAD model and underline its interest as a target for neuroprotection in AD.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Memória Espacial , eIF-2 Quinase/metabolismo , Doença de Alzheimer/patologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/patologia , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , eIF-2 Quinase/deficiência
8.
PLoS One ; 13(5): e0196983, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29795582

RESUMO

Metabolic disorders including obesity and type 2 diabetes are known to be associated with chronic inflammation and are obvious risk factors for Alzheimer's disease. Recent evidences concerning obesity and diabetes suggest that the metabolic inflammasome ("metaflammasome") mediates chronic inflammation. The double-stranded RNA-dependent protein kinase (PKR) is a central component of the metaflammasome. In wild type (WT) and PKR-/- mice, blood glucose, insulin and lipid levels and the brain expression of the phosphorylated components of the metaflammasome-PKR, JNK, IRS1 and IKKbeta-were studied after the induction of obesity by a high fat diet (HFD). The results showed significant increased levels of activated brain metaflammasome proteins in exposed WT mice but the changes were not significant in PKR-/- mice. In addition, gain weight was observed in WT mice and also in PKR-/- mice exposed to HFD. Increased blood insulin level was more accentuated in PKR -/- mice. The modulation of PKR activity could be an appropriate therapeutic approach, aimed at reducing abnormal brain metabolism and inflammation linked to metabolic disorders in order to reduce the risk of neurodegeneration.


Assuntos
Glicemia/metabolismo , Encéfalo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Insulina/sangue , Obesidade/metabolismo , eIF-2 Quinase/genética , Animais , Encéfalo/patologia , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Regulação da Expressão Gênica , Teste de Tolerância a Glucose , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Inflamação , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia , Transdução de Sinais , Triglicerídeos/sangue , Aumento de Peso/genética , eIF-2 Quinase/deficiência
9.
Sci Rep ; 8(1): 773, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29335505

RESUMO

As chondrocytes are highly secretory and they experience a variety of stresses, physiological unfolded protein response (UPR) signalling is essential for extracellular matrix (ECM) secretion and chondrogenesis. In the three branches of the UPR pathway, PERK governs the translational attenuation and transcriptional upregulation of amino acid and redox metabolism and induction of apoptosis. It was previously demonstrated that a defect of the PERK branch of the UPR signalling pathway causes the accumulation of unfolded proteins, leading to cell death without perturbing endoplasmic reticulum (ER)-to-Golgi transport in pancreatic ß cells. However, little is known about the role of PERK in chondrocytes. In this study, we found that PERK signalling is activated in chondrocytes, and inhibition of PERK reduces collagen secretion despite causing excessive collagen synthesis in the ER. Perk -/- mice displayed reduced collagen in articular cartilage but no differences in chondrocyte proliferation or apoptosis compared to the findings in wild-type mice. PERK inhibition increases misfolded protein levels in the ER, which largely hinder ER-to-Golgi transport. These results suggest that the translational control mediated by PERK is a critical determinant of ECM secretion in chondrocytes.


Assuntos
Condrócitos/metabolismo , Colágeno/metabolismo , eIF-2 Quinase/metabolismo , Animais , Linhagem Celular , Camundongos , Camundongos Knockout , eIF-2 Quinase/deficiência
10.
Biol Cell ; 110(2): 27-32, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29168198

RESUMO

In humans, the pathogenesis of diabetes is characterised by two major pancreatic ß cell defects: a reduction in ß cell mass and the failure of ß cells to produce enough insulin. Over the past two decades, multiple studies involving cell cultures, animal models and human subjects have established the importance of the protein kinase RNA-like endoplasmic reticulum kinase (PERK) in the adaptive functional capacity of pancreatic ß cells during embryonic development and into adulthood. In this review, we will highlight major findings identifying PERK as a crucial player in ß cell physiology and in diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Epífises/anormalidades , Fator de Iniciação 2 em Eucariotos/genética , Homeostase/genética , Células Secretoras de Insulina/enzimologia , Osteocondrodisplasias/genética , eIF-2 Quinase/genética , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/imunologia , Adulto , Animais , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/patologia , Epífises/enzimologia , Epífises/imunologia , Epífises/patologia , Fator de Iniciação 2 em Eucariotos/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Homeostase/imunologia , Humanos , Recém-Nascido , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/imunologia , Osteocondrodisplasias/enzimologia , Osteocondrodisplasias/imunologia , Osteocondrodisplasias/patologia , Transdução de Sinais , eIF-2 Quinase/deficiência , eIF-2 Quinase/imunologia
11.
RNA Biol ; 14(11): 1570-1579, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-28594311

RESUMO

While lentiviral expression vectors are widely used in many facets of molecular biology, due to their ability to stably express heterologous genes in both dividing and non-dividing cells, they suffer from the disadvantage that introns inserted into the vector genome are generally rapidly lost by splicing in packaging cell lines. The presence of an intron, if achievable, has the potential to facilitate the expression of transgene cDNAs, as splicing has been extensively shown to facilitate mRNA biogenesis and function. Moreover, if a stable intron could be introduced into a lentiviral vector, this could greatly facilitate the expression of microRNAs (miRNAs), and especially miRNA clusters, as the introduction of pri-miRNA stems into the exonic region of a lentiviral vector can strongly reduce both vector titer and the expression of any miRNA-linked indicator gene due to cleavage of the vector RNA genome by cellular Drosha. Here, we describe a novel lentiviral vector design in which transgenes and/or miRNAs are expressed using an antisense-orientated, inducible promoter driving an expression cassette bearing a functional intron. We demonstrate that this lentiviral vector, called pTREX, is able to express higher levels of both transgenes and pri-miRNA clusters when compared with a closely similar conventional lentiviral vector.


Assuntos
Engenharia Genética/métodos , Íntrons , Lentivirus/genética , MicroRNAs/genética , Splicing de RNA , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Éxons , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Insulina/genética , Insulina/metabolismo , Lentivirus/metabolismo , MicroRNAs/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Regiões Promotoras Genéticas , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Ribonuclease III/genética , Ribonuclease III/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Transgenes , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética
12.
RNA ; 23(2): 153-160, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27837013

RESUMO

While mammalian somatic cells are incapable of mounting an effective RNA interference (RNAi) response to viral infections, plants and invertebrates are able to generate high levels of viral short interfering RNAs (siRNAs) that can control many infections. In Drosophila, the RNAi response is mediated by the Dicer 2 enzyme (dDcr2) acting in concert with two cofactors called Loqs-PD and R2D2. To examine whether a functional RNAi response could be mounted in human somatic cells, we expressed dDcr2, in the presence or absence of Loqs-PD and/or R2D2, in a previously described human cell line, NoDice/ΔPKR, that lacks functional forms of human Dicer (hDcr) and PKR. We observed significant production of ∼21-nt long siRNAs, derived from a cotransfected double stranded RNA (dsRNA) expression vector, that were loaded into the human RNA-induced silencing complex (RISC) and were able to significantly reduce the expression of a cognate indicator gene. Surprisingly, dDcr2 was able to produce siRNAs even in the absence of Loqs-PD, which is thought to be required for dsRNA cleavage by dDcr2. This result may be explained by our finding that dDcr2 is able to bind the human Loqs-PD homolog TRBP when expressed in human cells in the absence of Loqs-PD. We conclude that it is possible to at least partially rescue the ability of mammalian somatic cells to express functional siRNAs using gene products of invertebrate origin.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , RNA Helicases/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Ribonuclease III/genética , Animais , Engenharia Celular , Linhagem Celular , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Teste de Complementação Genética , Humanos , Coativadores de Receptor Nuclear/genética , Coativadores de Receptor Nuclear/metabolismo , Ligação Proteica , RNA Helicases/metabolismo , RNA Interferente Pequeno/biossíntese , Proteínas de Ligação a RNA/metabolismo , Complexo de Inativação Induzido por RNA/biossíntese , Complexo de Inativação Induzido por RNA/genética , Ribonuclease III/deficiência , Ribonuclease III/metabolismo , Transgenes , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética
13.
J Biol Chem ; 291(29): 15093-107, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226571

RESUMO

The sensing of double-stranded RNA (dsRNA) in the liver is important for antiviral defenses but can also contribute to sterile inflammation during liver injury. Hepatocytes are often the target of viral infection and are easily injured by inflammatory insults. Here we sought to establish the pathways involved in the production of type I interferons (IFN-I) in response to extracellular poly(I:C), a dsRNA mimetic, in hepatocytes. This was of interest because hepatocytes are long-lived and, unlike most immune cells that readily die after activation with dsRNA, are not viewed as cells with robust antimicrobial capacity. We found that poly(I:C) leads to rapid up-regulation of inducible nitric oxide synthase (iNOS), double-stranded RNA-dependent protein kinase (PKR), and Src. The production of IFN-ß was dependent on iNOS, PKR, and Src and partially dependent on TLR3/Trif. iNOS and Src up-regulation was partially dependent on TLR3/Trif but entirely dependent on PKR. The phosphorylation of TLR3 on tyrosine 759 was shown to increase in parallel to IFN-ß production in an iNOS- and Src-dependent manner, and Src was found to directly interact with TLR3 in the endosomal compartment of poly(I:C)-treated cells. Furthermore, we identified a robust NO/cGMP/PKG-dependent feedforward pathway for the amplification of iNOS expression. These data identify iNOS/NO as an integral component of IFN-ß production in response to dsRNA in hepatocytes in a pathway that involves the coordinated activities of TLR3/Trif and PKR.


Assuntos
Hepatócitos/imunologia , Hepatócitos/metabolismo , Interferon beta/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/farmacologia , Receptor 3 Toll-Like/metabolismo , eIF-2 Quinase/metabolismo , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Células Cultivadas , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Hepatócitos/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Fosforilação/efeitos dos fármacos , Poli I-C/farmacologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética , Tirosina/química , Regulação para Cima/efeitos dos fármacos , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
14.
Cell Signal ; 28(8): 880-6, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27079961

RESUMO

To prevent excess accumulation of unfolded proteins in endoplasmic reticulum (ER), eukaryotic cells have signaling pathways from the ER to the cytosol or nucleus. These processes are known as the endoplasmic reticulum stress (ERS) response. Protein kinase R like endoplasmic reticulum kinase (PERK) is a major transducer of the ERS response and it directly phosphorylate α-subunit of eukaryotic initiation factor 2 (eIF2α), resulting in translational attenuation. Phosphorylated eIF2α specifically promoted the translation of the activating transcription factor 4 (ATF4). ATF4 is a known important transcription factor which plays a pivotal role in osteoblast differentiation and bone formation. Furthermore, ATF4 is a downstream target of PERK. Studies have shown that PERK-eIF2α-ATF4 signal pathway mediated by ERS was involved in osteoblastic differentiation of osteoblasts. We have known that orthodontic tooth movement is a process of periodontal ligament cells (PDLCs) osteodifferentiation and alveolar bone remodeling under mechanical force. However, the involvement of PERK-eIF2α-ATF4 signal pathway mediated by ERS in osteogenic differentiation of PDLCs under mechanical force has not been unclear. In our study, we applied the cyclic mechanical force at 10% elongation with 0.5Hz to mimic occlusal force, and explored whether PERK-eIF2α-ATF4 signaling pathway mediated by ERS involved in osteogenic differentiation of PDLCs under mechanical force. Firstly, cyclic mechanical force will induce ERS and intensify several osteoblast marker genes (ATF4, OCN, and BSP). Next, we found that PERK overexpression increased eIF2α phosphorylation and expression of ATF4, furthermore induced BSP, OCN expression, thus it will promote osteodifferentiation of hPDLCs; mechanical force could promote this effect. However, PERK(-/-) cells showed the opposite changes, which will inhibit osteodifferentiation of hPDLCs. Taken together, our study proved that PERK-eIF2α-ATF4 signaling pathway mediated by ERS involved in osteoblast differentiation of PDLCs under cyclic mechanical force.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Diferenciação Celular , Estresse do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos/metabolismo , Ligamento Periodontal/patologia , Transdução de Sinais , Estresse Mecânico , eIF-2 Quinase/metabolismo , Adolescente , Diferenciação Celular/genética , Criança , Estresse do Retículo Endoplasmático/genética , Regulação da Expressão Gênica , Humanos , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , eIF-2 Quinase/deficiência
15.
ASN Neuro ; 8(2)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27095827

RESUMO

In peripheral nerves, P0 glycoprotein accounts for more than 20% of myelin protein content. P0 is synthesized by Schwann cells, processed in the endoplasmic reticulum (ER) and enters the secretory pathway. However, the mutant P0 with S63 deleted (P0S63del) accumulates in the ER lumen and induces a demyelinating neuropathy in Charcot-Marie-Tooth disease type 1B (CMT1B)-S63del mice. Accumulation of P0S63del in the ER triggers a persistent unfolded protein response. Protein kinase RNA-like endoplasmic reticulum kinase (PERK) is an ER stress sensor that phosphorylates eukaryotic initiation factor 2 alpha (eIF2alpha) in order to attenuate protein synthesis. We have shown that increasing phosphophorylated-eIF2alpha (P-eIF2alpha) is a potent therapeutic strategy, improving myelination and motor function in S63del mice. Here, we explore the converse experiment:Perkhaploinsufficiency reduces P-eIF2alpha in S63del nerves as expected, but surprisingly, ameliorates, rather than worsens S63del neuropathy. Motor performance and myelin abnormalities improved in S63del//Perk+/- compared with S63del mice. These data suggest that mechanisms other than protein translation might be involved in CMT1B/S63del neuropathy. In addition,Perkdeficiency in other cells may contribute to demyelination in a non-Schwann-cell autonomous manner.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/fisiopatologia , Mutação/genética , eIF-2 Quinase/deficiência , Fatores Etários , Animais , Animais Recém-Nascidos , Células Cultivadas , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Técnicas de Cocultura , Modelos Animais de Doenças , Embrião de Mamíferos , Gânglios Espinais/citologia , Regulação da Expressão Gênica/genética , Imunoprecipitação , Camundongos , Camundongos Transgênicos , Proteína Básica da Mielina/metabolismo , Proteína P0 da Mielina/genética , Proteína P0 da Mielina/metabolismo , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/genética , Neurônios/efeitos dos fármacos , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , eIF-2 Quinase/genética
16.
J Virol ; 90(2): 1144-7, 2016 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-26512090

RESUMO

Murine cytomegalovirus (MCMV) proteins m142 and m143 are essential for viral replication. They bind double-stranded RNA and prevent protein kinase R-induced protein synthesis shutoff. Whether the two viral proteins have additional functions such as their homologs in human cytomegalovirus do remained unknown. We show that MCMV m142 and m143 knockout mutants attain organ titers equivalent to those attained by wild-type MCMV in Pkr knockout mice, suggesting that these viral proteins do not encode additional PKR-independent functions relevant for pathogenesis in vivo.


Assuntos
Muromegalovirus/fisiologia , Mutação , Proteínas Virais/genética , Replicação Viral , eIF-2 Quinase/deficiência , Animais , Camundongos Knockout , Muromegalovirus/genética , Carga Viral
17.
J Neuropathol Exp Neurol ; 74(8): 850-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26172286

RESUMO

We report the first detailed examination of the brain of a patient with Wolcott-Rallison syndrome. Wolcott-Rallison syndrome is an extremely rare clinical manifestation of a lack of protein kinase R-like endoplasmic reticulum kinase (PERK) function caused by mutations in the PERK gene EIF2AK3. Protein kinase R-like endoplasmic reticulum kinase is thought to play a significant pathogenetic role in several neurodegenerative diseases, including Alzheimer disease, other tauopathies, and Parkinson disease. The brain of a male patient aged 4 years 7 months showed pathologic and immunohistochemical evidence that the absence of PERK for several years is sufficient to induce early changes reminiscent of various neurodegenerative conditions. These include neurofibrillary tangles (as in progressive supranuclear palsy), FUS-immunopositive and p62-immunopositive neurons, and reactive glial changes. We also detected an increased amount of p62-positive puncta coimmunostaining for LC3 and ubiquitin, suggesting changes in autophagic flux. Studying a human brain with absent PERK function presents the opportunity to assess the long-term consequences of nonfunctioning of PERK in the presence of all of the compensatory mechanisms that are normally active in a living human, thereby confirming the importance of PERK for autophagy in the brain and for neurodegeneration.


Assuntos
Encéfalo/patologia , Diabetes Mellitus Tipo 1/patologia , Epífises/anormalidades , Degeneração Neural/patologia , Osteocondrodisplasias/patologia , eIF-2 Quinase/deficiência , Pré-Escolar , Diabetes Mellitus Tipo 1/genética , Epífises/patologia , Imunofluorescência , Humanos , Imuno-Histoquímica , Masculino , Degeneração Neural/genética , Osteocondrodisplasias/genética
18.
Cell Death Dis ; 6: e1594, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25590804

RESUMO

Brain thiamine homeostasis has an important role in energy metabolism and displays reduced activity in Alzheimer's disease (AD). Thiamine deficiency (TD) induces regionally specific neuronal death in the animal and human brains associated with a mild chronic impairment of oxidative metabolism. These features make the TD model amenable to investigate the cellular mechanisms of neurodegeneration. Once activated by various cellular stresses, including oxidative stress, PKR acts as a pro-apoptotic kinase and negatively controls the protein translation leading to an increase of BACE1 translation. In this study, we used a mouse TD model to assess the involvement of PKR in neuronal death and the molecular mechanisms of AD. Our results showed that the TD model activates the PKR-eIF2α pathway, increases the BACE1 expression levels of Aß in specific thalamus nuclei and induces motor deficits and neurodegeneration. These effects are reversed by PKR downregulation (using a specific inhibitor or in PKR knockout mice).


Assuntos
Peptídeos beta-Amiloides/biossíntese , Regulação para Baixo , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Tiamina/metabolismo , eIF-2 Quinase/metabolismo , Amiloide/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Caspase 3/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Inflamação/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Atividade Motora , Degeneração Neural/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo , Transporte Proteico , Transdução de Sinais , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/deficiência
19.
Cerebellum ; 14(4): 386-97, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25592072

RESUMO

Ethanol-induced neuronal loss is closely related to the pathogenesis of fetal alcohol spectrum disorders. The cerebellum is one of the brain areas that are most sensitive to ethanol. The mechanism underlying ethanol neurotoxicity remains unclear. Our previous in vitro studies have shown that the double-stranded RNA (dsRNA)-activated protein kinase (PKR) regulates neuronal apoptosis upon ethanol exposure and ethanol activates PKR through association with its intracellular activator RAX. However, the role of PKR and its interaction with RAX in vivo have not been investigated. In the current study, by utilizing N-PKR-/- mice, C57BL/6J mice with a deficient RAX-binding domain in PKR, we determined the critical role of RAX/PKR association in PKR-regulated ethanol neurotoxicity in the developing cerebellum. Our data indicate that while N-PKR-/- mice have a similar BAC profile as wild-type mice, ethanol induces less brain/body mass reduction as well as cerebellar neuronal loss. In addition, ethanol promotes interleukin-1ß (IL-1ß) secretion, and IL-1ß is a master cytokine regulating inflammatory response. Importantly, ethanol-promoted IL-1ß secretion is inhibited in the developing cerebellum of N-PKR-/- mice. Thus, RAX/PKR interaction and PKR activation regulate ethanol neurotoxicity in the developing cerebellum, which may involve ethanol-induced neuroinflammation. Further, PKR could be a possible target for pharmacological intervention to prevent or treat fetal alcohol spectrum disorder (FASD).


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Cerebelo , Etanol/toxicidade , Síndromes Neurotóxicas/etiologia , eIF-2 Quinase/deficiência , Fatores Etários , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/genética , Células Cultivadas , Cerebelo/efeitos dos fármacos , Cerebelo/crescimento & desenvolvimento , Cerebelo/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndromes Neurotóxicas/patologia , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/genética , Sincalida/farmacologia , Fatores de Tempo , eIF-2 Quinase/genética
20.
J Immunol ; 193(3): 1459-67, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24990083

RESUMO

Pseudomonas aeruginosa secrete N-(3-oxododecanoyl)-homoserine lactone (HSL-C12) as a quorum-sensing molecule to regulate bacterial gene expression. Because HSL-C12 is membrane permeant, multiple cell types in P. aeruginosa-infected airways may be exposed to HSL-C12, especially adjacent to biofilms where local (HSL-C12) may be high. Previous reports showed that HSL-C12 causes both pro- and anti-inflammatory effects. To characterize HSL-C12's pro- and anti-inflammatory effects in host cells, we measured protein synthesis, NF-κB activation, and KC (mouse IL-8) and IL-6 mRNA and protein secretion in wild-type mouse embryonic fibroblasts (MEF). To test the role of the endoplasmic reticulum stress inducer, PERK we compared these responses in PERK(-/-) and PERK-corrected PERK(-/-) MEF. During 4-h treatments of wild-type MEF, HSL-C12 potentially activated NF-κB p65 by preventing the resynthesis of IκB and increased transcription of KC and IL-6 genes (quantitative PCR). HSL-C12 also inhibited secretion of KC and/or IL-6 into the media (ELISA) both in control conditions and also during stimulation by TNF-α. HSL-C12 also activated PERK (as shown by increased phosphorylation of eI-F2α) and inhibited protein synthesis (as measured by incorporation of [(35)S]methionine by MEF). Comparisons of PERK(-/-) and PERK-corrected MEF showed that HSL-C12's effects were explained in part by activation of PERK→phosphorylation of eI-F2α→inhibition of protein synthesis→reduced IκBα production→activation of NF-κB→increased transcription of the KC gene but reduced translation and secretion of KC. HSL-C12 may be an important modulator of early (up to 4 h) inflammatory signaling in P. aeruginosa infections.


Assuntos
4-Butirolactona/análogos & derivados , Fator de Iniciação 2 em Eucariotos/fisiologia , Mediadores da Inflamação/fisiologia , Pseudomonas aeruginosa/imunologia , Percepção de Quorum/imunologia , Transdução de Sinais/imunologia , eIF-2 Quinase/fisiologia , 4-Butirolactona/fisiologia , Animais , Linhagem Celular , Estresse do Retículo Endoplasmático/imunologia , Camundongos , eIF-2 Quinase/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...