Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.415
Filtrar
1.
Methods Mol Biol ; 2854: 265-282, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39192136

RESUMEN

Protein kinase R (PKR), a key double-stranded RNA (dsRNA)-activated sensor, is pivotal for cellular responses to diverse stimuli. This protocol delineates a comprehensive methodological framework employing single luciferase assays, yeast assays, immunoblot assays, and quantitative PCR (qPCR) to discern and validate PKR activities and their downstream impacts on NF-κB-activating signaling pathways. These methodologies furnish a systematic approach to unraveling the role of PKR as a dsRNA sensor and effector in antiviral innate immunity, enabling in-depth analyses of dsRNA sensor activities.


Asunto(s)
Inmunidad Innata , ARN Bicatenario , eIF-2 Quinasa , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , ARN Bicatenario/inmunología , ARN Bicatenario/genética , Humanos , FN-kappa B/metabolismo , Transducción de Señal , Animales
2.
Biochim Biophys Acta Mol Cell Res ; 1871(8): 119852, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39357547

RESUMEN

Endoplasmic reticulum stress-induced cell apoptosis is a pivotal mechanism underlying the progression of cardiac hypertrophy. NLRX1, a member of the NOD-like receptor family, modulates various cellular processes, including STING, NF-κB, MAPK pathways, reactive oxygen species production, essential metabolic pathways, autophagy and cell death. Emerging evidence suggests that NLRX1 may offer protection against diverse cardiac diseases. However, the impacts and mechanisms of NLRX1 on endoplasmic reticulum stress in cardiac hypertrophy remains largely unexplored. In our study, we observed that the NLRX1 and phosphorylated STING (p-STING) were highly expressed in both hypertrophic mouse heart and cellular model of cardiac hypertrophy. Whereas over-expression of NLRX1 mitigated the expression levels of p-STING, as well as the endoplasmic reticulum stress markers, including transcription activating factor 4 (ATF4), C/EBP homologous protein (CHOP) and the ratios of phosphorylated PERK to PERK, phosphorylated IRE1 to IRE1 and phosphorylated eIF2α to eIF2α in an Angiotensin II (Ang II)-induced cellular model of cardiac hypertrophy. Importantly, the protective effects of NLRX1 were attenuated upon pretreatment with the STING agonist, DMXAA. Our findings provide the evidence that NLRX1 attenuates the PERK-eIF2α-ATF4-CHOP axis of endoplasmic reticulum stress response via inhibition of p-STING in Ang II-treated cardiomyocytes, thereby ameliorating the development of cardiac hypertrophy.


Asunto(s)
Cardiomegalia , Estrés del Retículo Endoplásmico , Proteínas de la Membrana , Animales , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/genética , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Masculino , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Fosforilación , Ratones Endogámicos C57BL , Angiotensina II/farmacología , Apoptosis , Transducción de Señal , Proteínas Mitocondriales
3.
Sci Rep ; 14(1): 23533, 2024 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-39384561

RESUMEN

Recognition of viral infection often relies on the detection of double-stranded RNA (dsRNA), a process that is conserved in many different organisms. In mammals, proteins such as MDA5, RIG-I, OAS, and PKR detect viral dsRNA, but struggle to differentiate between viral and endogenous dsRNA. This study investigates an shRNA targeting DDX54's potential to activate PKR, a key player in the immune response to dsRNA. Knockdown of DDX54 by a specific shRNA induced robust PKR activation in human cells, even when DDX54 is overexpressed, suggesting an off-target mechanism. Activation of PKR by the shRNA was enhanced by knockdown of ADAR1, a dsRNA binding protein that suppresses PKR activation, indicating a dsRNA-mediated mechanism. In vitro assays confirmed direct PKR activation by the shRNA. These findings emphasize the need for rigorous controls and alternative methods to validate gene function and minimize unintended immune pathway activation.


Asunto(s)
ARN Bicatenario , ARN Interferente Pequeño , Proteínas de Unión al ARN , eIF-2 Quinasa , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Humanos , ARN Bicatenario/metabolismo , ARN Bicatenario/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/genética , Activación Enzimática , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/genética , Células HEK293 , Técnicas de Silenciamiento del Gen
4.
Sci Rep ; 14(1): 23812, 2024 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-39394239

RESUMEN

Common single-nucleotide variants (SNVs) of eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3) slightly increase the risk of disorders in the periphery and the central nervous system. EIF2AK3 encodes protein kinase RNA-like endoplasmic reticulum kinase (PERK), a key regulator of ER stress. Three exonic EIF2AK3 SNVs form the PERK-B haplotype, which is present in 28% of the global population. Importantly, the precise impact of these SNVs on PERK activity remains elusive. In this study, we demonstrate that PERK-B SNVs do not alter PERK expression or basal activity in vitro and in the novel triple knock-in mice expressing the exonic PERK-B SNVs in vivo. However, the kinase activity of PERK-B protein is higher than that of PERK-A in a cell-free assay and in mouse liver homogenates. Pancreatic tissue in PERK-B/B mice also exhibit increased susceptibility to apoptosis under acute ER stress. Monocyte-derived macrophages from PERK-B/B mice exhibit higher PERK activity than those from PERK-A/A mice, albeit with minimal functional consequences at acute timepoints. The subtle PERK-B-driven effects observed in liver and pancreas during acute stress implicate PERK as a contributor to disease susceptibility. The novel PERK-B mouse model provides valuable insights into ER stress-induced PERK activity, aiding the understanding of the genetic basis of disorders associated with ER stress.


Asunto(s)
Estrés del Retículo Endoplásmico , Técnicas de Sustitución del Gen , Hígado , Páncreas , eIF-2 Quinasa , Animales , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Estrés del Retículo Endoplásmico/genética , Hígado/metabolismo , Ratones , Páncreas/metabolismo , Apoptosis/genética , Polimorfismo de Nucleótido Simple
5.
Food Funct ; 15(19): 10179-10189, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39301672

RESUMEN

Green tea polyphenols (GTP), an important phytochemical in the daily human diet, bind to various cellular receptors and exert anti-inflammatory and antioxidant benefits. The environmental contaminant tetrabromobisphenol A (TBBPA) enters the digestive system through multiple pathways, resulting in oxidative stress (OS), gastroenteritis, and mucosal injury. The aim of this study was to explore the molecular mechanisms of TBBPA-induced gastritis in mice treated with GTP in vivo and in an in vitro model. The results showed that exposure to TBBPA increased reactive oxygen species (ROS) levels, activated oxidative stress (OS) induced endoplasmic reticulum stress (ERS), and the expression of endoplasmic reticulum stress-related factors (e.g., GRP78, PERK, IRE-1, ATF-6, etc.) increased. The inflammatory pathway NF-κB was activated, and the pro-inflammatory factors TNF-α, IL-1ß, and IL-6 increased, while triggering a cascade reaction mediated by caspase-3. However, the addition of GTP could inhibit OS, restore the balance of endoplasmic reticulum homeostasis, and improve the inflammatory infiltration and apoptosis of gastric mucosal epithelial cells. Therefore, GTP alleviated ERS, reduced inflammation and apoptosis, and restored the gastric mucosal barrier by alleviating TBBPA-induced OS in mouse gastric tissues and GES-1 cells. This provides basic information for exploring the antioxidant mechanism of GTP and further investigating the toxic effects of TBBPA on mouse gastric mucosa.


Asunto(s)
Factor de Transcripción Activador 6 , Apoptosis , Chaperón BiP del Retículo Endoplásmico , Gastritis , Bifenilos Polibrominados , Polifenoles , Especies Reactivas de Oxígeno , , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Polifenoles/farmacología , Apoptosis/efectos de los fármacos , Té/química , Gastritis/inducido químicamente , Gastritis/tratamiento farmacológico , Gastritis/metabolismo , Factor de Transcripción Activador 6/metabolismo , Masculino , Estrés del Retículo Endoplásmico/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Estrés Oxidativo/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
6.
Commun Biol ; 7(1): 1091, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237614

RESUMEN

Cisplatin is a common anticancer drug, but its frequent nephrotoxicity limits its clinical use. Small GTP-binding protein GDP dissociation stimulator (smgGDS), a small GTPase chaperone protein, was considerably downregulated during cisplatin-induced acute kidney injury (CDDP-AKI), especially in renal tubular epithelial cells. SmgGDS-knockdown mice was established and found that smgGDS knockdown promoted CDDP-AKI, as demonstrated by an increase in serum creatine, blood urea nitrogen levels and the appearance of tubular patterns. RNA sequencing suggested that protein kinase RNA-like ER kinase (PERK), which bridges mitochondria-associated ER membranes, was involved in smgGDS knockdown following CDDP-AKI, and then identified that smgGDS knockdown increased phosphorylated-PERK in vivo and in vitro. Furthermore, we confirmed that smgGDS deficiency aggravated apoptosis and ER stress in vivo and in vitro. And the ER stress inhibitor 4-Phenylbutyric acid and the inhibition of PERK phosphorylation mitigated smgGDS deficiency-induced ER stress related apoptosis following cisplatin treatment, while the eIF2α phosphorylation inhibitor could not reverse the smgGDS deficiency accelerated cell death. Furthermore, the over-expression of smgGDS could reverse the ER stress and apoptosis caused by CDDP. Overall, smgGDS regulated PERK-dependent ER stress and apoptosis, thereby influencing renal damage. This study identified a target for diagnosing and treating cisplatin-induced acute kidney injury.


Asunto(s)
Lesión Renal Aguda , Cisplatino , Estrés del Retículo Endoplásmico , eIF-2 Quinasa , Cisplatino/efectos adversos , Cisplatino/toxicidad , Animales , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/genética , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ratones , Masculino , Apoptosis/efectos de los fármacos , Ratones Endogámicos C57BL , Antineoplásicos/efectos adversos , Antineoplásicos/toxicidad , Fosforilación
7.
Cell Commun Signal ; 22(1): 424, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39223663

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) is characterized by the abnormal proliferation of myeloid precursor cells and presents significant challenges in treatment due to its heterogeneity. Recently, the NLRP3 inflammasome has emerged as a potential contributor to AML pathogenesis, although its precise mechanisms remain poorly understood. METHODS: Public genome datasets were utilized to evaluate the expression of NLRP3 inflammasome-related genes (IL-1ß, IL-18, ASC, and NLRP3) in AML patients compared to healthy individuals. CRISPR/Cas9 technology was employed to generate NLRP3-deficient MOLM-13 AML cells, followed by comprehensive characterization using real-time PCR, western blotting, FACS analysis, and transmission electron and immunofluorescence microscopy. Proteomic analyses were conducted to identify NLRP3-dependent alterations in protein levels, with a focus on the eIF2 kinase PERK-mediated signaling pathways. Additionally, in vivo studies were performed using a leukemic mouse model to elucidate the pathogenic role of NLRP3 in AML. RESULTS: Elevated expression of NLRP3 was significantly associated with diminished overall survival in AML patients. Genetic deletion, pharmacological inhibition and silencing by RNA interference of NLRP3 led to decreased AML cell survival through the induction of apoptosis. Proteomic analyses uncovered NLRP3-dependent alterations in protein translation, characterized by enhanced eIF2α phosphorylation in NLRP3-deficient AML cells. Moreover, inhibition of PERK-mediated eIF2α phosphorylation reduced apoptosis by downregulating pro-apoptotic Bcl-2 family members. In vivo studies demonstrated reduced leukemic burden in mice engrafted with NLRP3 knockout AML cells, as evidenced by alleviated leukemic symptoms. CONCLUSION: Our findings elucidate the involvement of the NLRP3/PERK/eIF2 axis as a novel driver of AML cell survival. Targeting NLRP3-induced signaling pathways, particularly through the PERK/eIF2 axis, presents a promising therapeutic strategy for AML intervention. These insights into the role of the NLRP3 inflammasome offer potential avenues for improving the prognosis and treatment outcomes of AML patients.


Asunto(s)
Apoptosis , Factor 2 Eucariótico de Iniciación , Leucemia Mieloide Aguda , Proteína con Dominio Pirina 3 de la Familia NLR , eIF-2 Quinasa , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Humanos , Apoptosis/genética , Animales , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Ratones , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Transducción de Señal , Línea Celular Tumoral , Progresión de la Enfermedad , Inflamasomas/metabolismo
8.
Stroke ; 55(10): 2510-2521, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39224971

RESUMEN

BACKGROUND: In ischemia, acidosis occurs in/around injured tissue and parallels disease progression. Therefore, targeting an acid-sensitive receptor offers unique advantages in achieving the spatial and temporal specificity required for therapeutic interventions. We previously demonstrated that increased expression of GPR68 (G protein-coupled receptor 68), a proton-sensitive G protein-coupled receptor, mitigates ischemic brain injury. Here, we investigated the mechanism underlying GPR68-dependent protection. METHODS: We performed biochemical and molecular analyses to examine poststroke signaling. We used in vitro brain slice cultures and in vivo mouse transient middle cerebral artery occlusion (tMCAO) models to investigate ischemia-induced injuries. RESULTS: GPR68 deletion reduced PERK (protein kinase R-like ER kinase) expression in mouse brain. Compared with the wild-type mice, the GPR68-/- (knockout) mice exhibited a faster decline in eIF2α (eukaryotic initiation factor-2α) phosphorylation after tMCAO. Ogerin, a positive modulator of GPR68, stimulated eIF2α phosphorylation at 3 to 6 hours after tMCAO, primarily in the ipsilateral brain tissue. Consistent with the changes in eIF2α phosphorylation, Ogerin enhanced tMCAO-induced reduction in protein synthesis in ipsilateral brain tissue. In organotypic cortical slices, Ogerin reduced pH 6 and oxygen-glucose deprivation-induced neurotoxicity. Following tMCAO, intravenous delivery of Ogerin reduced brain infarction in wild-type but not knockout mice. Coapplication of a PERK inhibitor abolished Ogerin-induced protection. Delayed Ogerin delivery at 5 hours after tMCAO remained protective, and Ogerin has a similar protective effect in females. Correlated with these findings, tMCAO induced GPR68 expression at 6 hours, and Ogerin alters post-tMCAO proinflammatory/anti-inflammatory cytokine/chemokine expression profile. CONCLUSIONS: These data demonstrate that GPR68 potentiation leads to neuroprotection, at least in part, through enhancing PERK-eIF2α activation in ischemic tissue but has little impact on healthy tissue.


Asunto(s)
Isquemia Encefálica , Ratones Noqueados , Receptores Acoplados a Proteínas G , eIF-2 Quinasa , Animales , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Ratones , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/genética , Masculino , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/genética , Fosforilación , Ratones Endogámicos C57BL , Factores de Tiempo
9.
Int Immunopharmacol ; 142(Pt B): 113209, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39340998

RESUMEN

The endothelial-mesenchymal transition (EndMT) is involved in the development of atherosclerosis (AS) and is a key process in vascular endothelial injury. Oxidative stress, inflammation, and apoptosis are common causes of EndMT, and EndMT progression can further accelerate the development of AS. The metabolite trimethylamine N-oxide (TMAO) is produced by the gut microbiome and is implicated in the development of several diseases, including diabetes and chronic kidney disease. However, the impact of TMAO on transforming growth factor ß1(TGF-ß1)-induced EndMT remains unclear. We hypothesize that TMAO exacerbates plaque formation and cardiac function impairment by promoting EndMT. Herein, we showed that high serum TMAO levels caused plaque formation, cardiac function damage and haemodynamic changes in ApoE-/- mice. In vitro, TMAO upregulated mesenchymal markers and downregulated endothelial markers in HAECs. Furthermore, TMAO increased the migratory capacity of EndMT cells. Mechanistically, we found that PERK downregulation could alleviate TMAO-induced oxidative stress, EndMT, plaque formation and cardiac function damage. Further study showed that activated transcription factor 3 (ATF3), the downstream molecule of protein kinase RNA-like endoplasmic reticulum kinase (PERK), could bind with TGF-ß1/2 and affect EndMT. Overall, TMAO promotes EndMT, possibly through the PERK-eIF2α-ATF4-CHOP or the PERk-eIF2α-ATF3-TGF-ß signalling pathways.


Asunto(s)
Apoptosis , Aterosclerosis , Metilaminas , eIF-2 Quinasa , Animales , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Apoptosis/efectos de los fármacos , Ratones , Humanos , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta1/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Cultivadas , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ratones Noqueados para ApoE , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Transición Endotelial-Mesenquimatosa
10.
Int J Biol Macromol ; 279(Pt 2): 135274, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39226976

RESUMEN

Stress granules (SGs) are cytoplasmic aggregates of proteins and mRNA that form in response to diverse environmental stressors, including viral infections. Several viruses possess the ability to block the formation of stress granules by targeting the SGs marker protein G3BP. However, the molecular functions and mechanisms underlying the regulation of SGs formation by Getah virus (GETV) remain unclear. In this study, we found that GETV infection triggered the formation of Nsp3-G3BP aggregates, which differed in composition from SGs. Further studies revealed that the presence of these aggregates was dependent on the activation of the PKR/eIF2α signaling pathway. Interestingly, we found that Nsp3 HVD domain blocked the formation of SGs by binding to G3BP NTF2 domain. Moreover, knockout of G3BP in NCI-H1299 cells had no effect on GETV replication, while overexpression of G3BP to form the genuine SGs significantly inhibited GETV replication. Overall, our study elucidates a novel role GETV Nsp3 to change the composition of SG as well as cellular stress response.


Asunto(s)
ADN Helicasas , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , Gránulos de Estrés , Proteínas no Estructurales Virales , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Humanos , Gránulos de Estrés/metabolismo , ARN Helicasas/metabolismo , ADN Helicasas/metabolismo , ADN Helicasas/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Replicación Viral , Transducción de Señal , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Gránulos Citoplasmáticos/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética
11.
In Vivo ; 38(5): 2228-2238, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39187325

RESUMEN

BACKGROUND/AIM: Breast cancer is the most predominant type of cancer affecting women worldwide and the current therapeutic treatment for breast cancer patients is not adequately effective. This study aimed to investigate the mechanism of 17-AAG, a heat shock protein (HSP90) inhibitor, as a treatment for inducing breast cancer cell apoptosis. MATERIALS AND METHODS: The pharmacology network was employed to examine the correlation of 17-AAG with the gene expression profiles of breast cancer, obtained by Gene Expression Profiling Interactive Analysis (GEPIA). MTT and flow cytometry were utilized to investigate cell proliferation and cell apoptosis, respectively. Dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay and western blot analysis were employed to examine the correlation between cellular oxidant levels and protein expression. Immunofluorescence staining was utilized to confirm the protein localization and assess DNA damage. RESULTS: The pharmacological network analysis revealed that HSP90 serves as the common target connecting 17-AAG and breast cancer genes. Treatment with 17-AAG significantly increased cell apoptosis. Moreover, the treatment resulted in up-regulation of cellular oxidant levels and PERK/eIF2α expression. In line with these, protein localization after treatment revealed an increase in DNA damage, correlating with higher ER stress levels. Furthermore, GEPIA demonstrated that PERK and eIF2α expression were significantly higher in breast invasive carcinoma compared to other tumor types. CONCLUSION: HSP90 emerges as a potential target for inducing apoptosis in breast cancer cells by disrupting protein homeostasis in the endoplasmic reticulum, possibly through PERK/eIF2α up-regulation. 17-AAG, an HSP90 inhibitor, may therefore potentially hold an alternative therapeutic strategy for breast cancer treatment.


Asunto(s)
Apoptosis , Benzoquinonas , Neoplasias de la Mama , Estrés del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación , Lactamas Macrocíclicas , eIF-2 Quinasa , Humanos , Benzoquinonas/farmacología , Lactamas Macrocíclicas/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Femenino , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/genética , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
12.
Elife ; 122024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39207915

RESUMEN

HIV disease remains prevalent in the USA and chronic kidney disease remains a major cause of morbidity in HIV-1-positive patients. Host double-stranded RNA (dsRNA)-activated protein kinase (PKR) is a sensor for viral dsRNA, including HIV-1. We show that PKR inhibition by compound C16 ameliorates the HIV-associated nephropathy (HIVAN) kidney phenotype in the Tg26 transgenic mouse model, with reversal of mitochondrial dysfunction. Combined analysis of single-nucleus RNA-seq and bulk RNA-seq data revealed that oxidative phosphorylation was one of the most downregulated pathways and identified signal transducer and activator of transcription (STAT3) as a potential mediating factor. We identified in Tg26 mice a novel proximal tubular cell cluster enriched in mitochondrial transcripts. Podocytes showed high levels of HIV-1 gene expression and dysregulation of cytoskeleton-related genes, and these cells dedifferentiated. In injured proximal tubules, cell-cell interaction analysis indicated activation of the pro-fibrogenic PKR-STAT3-platelet-derived growth factor (PDGF)-D pathway. These findings suggest that PKR inhibition and mitochondrial rescue are potential novel therapeutic approaches for HIVAN.


Asunto(s)
Nefropatía Asociada a SIDA , Ratones Transgénicos , Mitocondrias , eIF-2 Quinasa , Animales , Humanos , Ratones , Nefropatía Asociada a SIDA/genética , Nefropatía Asociada a SIDA/metabolismo , Nefropatía Asociada a SIDA/patología , Modelos Animales de Enfermedad , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , VIH-1/genética , VIH-1/fisiología , Mitocondrias/metabolismo , Podocitos/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética
13.
J Cell Biol ; 223(10)2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39150520

RESUMEN

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


Asunto(s)
Obesidad , Fenotipo , Piperidinas , Proteínas Serina-Treonina Quinasas , Quinazolinonas , Transducción de Señal , eIF-2 Quinasa , Animales , Quinazolinonas/farmacología , Piperidinas/farmacología , Ratones , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Obesidad/patología , Obesidad/metabolismo , Obesidad/prevención & control , Obesidad/genética , Transducción de Señal/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ratones Endogámicos C57BL , Masculino , Resistencia a la Insulina , Insulina/metabolismo , Insulina/sangre , Estrés Fisiológico/efectos de los fármacos , Modelos Animales de Enfermedad , Dieta Alta en Grasa/efectos adversos , Diabetes Mellitus/patología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/prevención & control , Intolerancia a la Glucosa/tratamiento farmacológico , Adenina/análogos & derivados , Indoles
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(8): 704-709, 2024 Aug.
Artículo en Chino | MEDLINE | ID: mdl-39215668

RESUMEN

Objective To explore the effects of Myxovirus resistance protein A (MxA) on the Janus kinase/Signal transducer and activator of transcription (JAK/STAT) pathway in HepG2 cells. Methods HepG2 cells were transfected with the pcDNA3.1-Flag-MxA construct, and subsequent localization and expression of the MxA protein were detected through immunofluorescence cytochemistry. The presence of MxA protein was further confirmed by using Western blot analysis. Following transfection with MxA small interfering RNA (si-MxA) and subsequent treatment with alpha interferon (IFN-α), real-time fluorescent quantitative PCR was employed to measure the mRNA levels of myxovirus resistance protein A (MxA), protein kinase R (PKR), and oligoadenylate synthase (OAS). Western blot analysis was used to detect the protein expression of MxA, PKR, OAS, signal transducer and activator of transcription 1 (STAT1), phosphorylated STAT1 (pSTAT1), STAT2, phosphorylated STAT2 (p-STAT2) and interferon regulatory factor 9 (IRF9). Additionally, pcDNA3.1-Flag-MxA and pISRE-TA-luc were co-transfected into HepG2 and HepG2.2.15 cells, respectively, to assess the activity of the interferon-stimulated response element (ISRE) by using a luciferase activity assay. Results MxA protein was expressed in both the cytoplasm and nucleus of HepG2 cells, with higher expression levels in the cytoplasm than in the nucleus. Knocking down MxA expression in HepG2 cells did not affect the expression of STAT1, p-STAT1, STAT2, p-STAT2, and IRF9 proteins induced by IFN-α, but significantly reduced the expression of antiviral proteins PKR and OAS. Overexpression of MxA in HepG2 cells enhanced ISRE activity and increased the expression of PKR and OAS proteins, but this effect was inhibited in HepG2.2.15 cells. Conclusion MxA induces the expression of antiviral proteins by enhancing the activity of the JAK/STAT signaling pathway ISRE.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Proteínas de Resistencia a Mixovirus , Factor de Transcripción STAT1 , eIF-2 Quinasa , Humanos , Células Hep G2 , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Interferón-alfa/farmacología , Interferón-alfa/genética , Interferón-alfa/metabolismo , Elementos de Respuesta/genética , Transducción de Señal , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Interferones/genética , Interferones/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Regulación de la Expresión Génica
15.
J Virol ; 98(9): e0102024, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39194235

RESUMEN

Some negative-sense RNA viruses, including measles virus (MeV), share the characteristic that during their infection cycle, cytoplasmic inclusion bodies (IBs) are formed where components of the viral replication machinery are concentrated. As a foci of viral replication, how IBs act to enhance the efficiency of infection by affecting virus-host interactions remains an important topic of investigation. We previously established that upon MeV infection, the epigenetic host protein, WD repeat-containing protein 5 (WDR5), translocates to cytoplasmic viral IBs and facilitates MeV replication. We now show that WDR5 is recruited to IBs by forming a complex with IB-associated MeV phosphoprotein via a conserved binding motif located on the surface of WDR5. Furthermore, we provide evidence that WDR5 promotes viral replication by suppressing a major innate immune response pathway, the double-stranded RNA-mediated activation of protein kinase R and integrated stress response. IMPORTANCE: MeV is a pathogen that remains a global concern, with an estimated 9 million measles cases and 128,000 measles deaths in 2022 according to the World Health Organization. A large population of the world still has inadequate access to the effective vaccine against the exceptionally transmissible MeV. Measles disease is characterized by a high morbidity in children and in immunocompromised individuals. An important area of research for negative-sense RNA viruses, including MeV, is the characterization of the complex interactome between virus and host occurring at cytoplasmic IBs where viral replication occurs. Despite the progress made in understanding IB structures, little is known regarding the virus-host interactions within IBs and the role of these interactions in promoting viral replication and antagonizing host innate immunity. Herein we provide evidence suggesting a model by which MeV IBs utilize the host protein WDR5 to suppress the protein kinase R-integrated stress response pathway.


Asunto(s)
Inmunidad Innata , Virus del Sarampión , Sarampión , Replicación Viral , Virus del Sarampión/fisiología , Virus del Sarampión/genética , Humanos , Sarampión/virología , Sarampión/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Interacciones Huésped-Patógeno , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Células HEK293 , Estrés Fisiológico , ARN Bicatenario/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Animales
16.
Sci Adv ; 10(32): eado7464, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39110805

RESUMEN

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


Asunto(s)
Evolución Molecular , Filogenia , Vertebrados , eIF-2 Quinasa , Animales , Vertebrados/genética , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , ARN Bicatenario/metabolismo , Inmunidad Innata , Humanos , Ácidos Nucleicos/metabolismo , Evolución Biológica
17.
Eur J Pharmacol ; 982: 176899, 2024 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-39153651

RESUMEN

Colorectal cancer (CRC) ranks among the leading causes of cancer-related deaths worldwide. Enhancing CRC diagnosis and prognosis requires the development of improved biomarkers and therapeutic targets. Emerging evidence suggests that the unfolded protein response (UPR) plays a pivotal role in CRC progression, presenting new opportunities for diagnosis, treatment, and prevention. This study hypothesizes that genetic variants in endoplasmic reticulum (ER) stress response genes influence CRC susceptibility. We examined the frequencies of SNPs in PERK (rs13045) and GRP78/BiP (rs430397) within a South Iranian cohort. We mapped the cellular and molecular features of PERK and GRP78 genes in colorectal cancer, observing their differential expressions in tumor and metastatic tissues. We constructed co-expression and protein-protein interaction networks and performed gene set enrichment analysis, highlighting autophagy as a significant pathway through KEGG. Furthermore, the study included 64 CRC patients and 60 control subjects. DNA extraction and genotyping were conducted using high-resolution melting (HRM) analysis. Significant differences in PERK and GRP78 expressions were observed between CRC tissues and controls. Variations in PERK and GRP78 genotypes were significantly correlated with CRC risk. Utilizing a Multi-Target Directed Ligands approach, a dual PERK/GRP78 inhibitor was designed and subjected to molecular modeling studies. Docking experiments indicated high-affinity binding between the proposed inhibitor and both genes, PERK and GRP78, suggesting a novel therapy for CRC. These findings highlight the importance of understanding genetic backgrounds in different populations to assess CRC risk. Polymorphisms in UPR signaling pathway elements may serve as potential markers for predicting CRC susceptibility, paving the way for personalized therapeutic strategies.


Asunto(s)
Neoplasias Colorrectales , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico , Polimorfismo de Nucleótido Simple , eIF-2 Quinasa , Humanos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Masculino , Femenino , Proteínas de Choque Térmico/genética , Persona de Mediana Edad , Simulación del Acoplamiento Molecular , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Terapia Molecular Dirigida , Anciano , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mapas de Interacción de Proteínas/genética
18.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3837-3847, 2024 Jul.
Artículo en Chino | MEDLINE | ID: mdl-39099357

RESUMEN

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


Asunto(s)
Factor de Transcripción Activador 4 , Lesión Pulmonar Aguda , Células Epiteliales Alveolares , Apoptosis , Factor 2 Eucariótico de Iniciación , Ginsenósidos , Sepsis , Factor de Transcripción CHOP , eIF-2 Quinasa , Animales , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/genética , Ginsenósidos/farmacología , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , Ratones , Apoptosis/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Sepsis/tratamiento farmacológico , Sepsis/complicaciones , Sepsis/metabolismo , Sepsis/genética , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Masculino , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Humanos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ratones Endogámicos C57BL
19.
Int J Mol Sci ; 25(14)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39062980

RESUMEN

Mitochondrial stress, resulting from dysfunction and proteostasis disturbances, triggers the mitochondrial unfolded protein response (UPRMT), which activates gene encoding chaperones and proteases to restore mitochondrial function. Although ATFS-1 mediates mitochondrial stress UPRMT induction in C. elegans, the mechanisms relaying mitochondrial stress signals to the nucleus in mammals remain poorly defined. Here, we explored the role of protein kinase R (PKR), an eIF2α kinase activated by double-stranded RNAs (dsRNAs), in mitochondrial stress signaling. We found that UPRMT does not occur in cells lacking PKR, indicating its crucial role in this process. Mechanistically, we observed that dsRNAs accumulate within mitochondria under stress conditions, along with unprocessed mitochondrial transcripts. Furthermore, we demonstrated that accumulated mitochondrial dsRNAs in mouse embryonic fibroblasts (MEFs) deficient in the Bax/Bak channels are not released into the cytosol and do not induce the UPRMT upon mitochondrial stress, suggesting a potential role of the Bax/Bak channels in mediating the mitochondrial stress response. These discoveries enhance our understanding of how cells maintain mitochondrial integrity, respond to mitochondrial dysfunction, and communicate stress signals to the nucleus through retrograde signaling. This knowledge provides valuable insights into prospective therapeutic targets for diseases associated with mitochondrial stress.


Asunto(s)
Mitocondrias , ARN Bicatenario , Respuesta de Proteína Desplegada , eIF-2 Quinasa , Animales , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Mitocondrias/metabolismo , ARN Bicatenario/metabolismo , Ratones , Estrés Fisiológico , Transducción de Señal , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Fibroblastos/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Humanos
20.
EMBO J ; 43(17): 3587-3603, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38951609

RESUMEN

Transposable elements (TEs) are mobile genetic modules of viral derivation that have been co-opted to become modulators of mammalian gene expression. TEs are a major source of endogenous dsRNAs, signaling molecules able to coordinate inflammatory responses in various physiological processes. Here, we provide evidence for a positive involvement of TEs in inflammation-driven bone repair and mineralization. In newly fractured mice bone, we observed an early transient upregulation of repeats occurring concurrently with the initiation of the inflammatory stage. In human bone biopsies, analysis revealed a significant correlation between repeats expression, mechanical stress and bone mineral density. We investigated a potential link between LINE-1 (L1) expression and bone mineralization by delivering a synthetic L1 RNA to osteoporotic patient-derived mesenchymal stem cells and observed a dsRNA-triggered protein kinase (PKR)-mediated stress response that led to strongly increased mineralization. This response was associated with a strong and transient inflammation, accompanied by a global translation attenuation induced by eIF2α phosphorylation. We demonstrated that L1 transfection reshaped the secretory profile of osteoblasts, triggering a paracrine activity that stimulated the mineralization of recipient cells.


Asunto(s)
Inflamación , Elementos de Nucleótido Esparcido Largo , Células Madre Mesenquimatosas , eIF-2 Quinasa , Animales , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Ratones , Humanos , Inflamación/metabolismo , Inflamación/genética , Inflamación/patología , Células Madre Mesenquimatosas/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Osteoblastos/metabolismo , Calcificación Fisiológica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...