Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.593
Filtrer
1.
Nat Commun ; 15(1): 5515, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951495

RÉSUMÉ

Like many other viruses, KSHV has two life cycle modes: the latent phase and the lytic phase. The RTA protein from KSHV is essential for lytic reactivation, but how this protein's activity is regulated is not fully understood. Here, we report that linear ubiquitination regulates the activity of RTA during KSHV lytic reactivation and de novo infection. Overexpressing OTULIN inhibits KSHV lytic reactivation, whereas knocking down OTULIN or overexpressing HOIP enhances it. Intriguingly, we found that RTA is linearly polyubiquitinated by HOIP at K516 and K518, and these modifications control the RTA's nuclear localization. OTULIN removes linear polyubiquitin chains from cytoplasmic RTA, preventing its nuclear import. The RTA orthologs encoded by the EB and MHV68 viruses are also linearly polyubiquitinated and regulated by OTULIN. Our study establishes that linear polyubiquitination plays a critically regulatory role in herpesvirus infection, adding virus infection to the list of biological processes known to be controlled by linear polyubiquitination.


Sujet(s)
Herpèsvirus humain de type 8 , Protéines précoces immédiates , Transactivateurs , Ubiquitination , Réplication virale , Herpèsvirus humain de type 8/physiologie , Herpèsvirus humain de type 8/génétique , Herpèsvirus humain de type 8/métabolisme , Humains , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Cellules HEK293 , Transactivateurs/métabolisme , Transactivateurs/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Activation virale , Infections à Herpesviridae/métabolisme , Infections à Herpesviridae/virologie , Noyau de la cellule/métabolisme
2.
Cell Signal ; 120: 111241, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38825173

RÉSUMÉ

Cardiac fibroblasts (CF) are mesenchymal-type cells responsible for maintaining the homeostasis of the heart's extracellular matrix (ECM). Their dysfunction leads to excessive secretion of ECM proteins, tissue stiffening, impaired nutrient and oxygen exchange, and electrical abnormalities in the heart. Additionally, CF act as sentinel cells in the cardiac tissue microenvironment, responding to various stimuli that may affect heart function. Deleterious stimuli induce an inflammatory response in CF, increasing the secretion of cytokines such as IL-1ß and TNF-α and the expression of cell adhesion molecules like ICAM1 and VCAM1, initially promoting damage resolution by recruiting immune cells. However, constant harmful stimuli lead to a chronic inflammatory process and heart dysfunction. Therefore, it is necessary to study the mechanisms that govern CF inflammation. NFκB is a key regulator of the cardiac inflammatory process, making the search for mechanisms of NFκB regulation and CF inflammatory response crucial for developing new treatment options for cardiovascular diseases. SGK1, a serine-threonine protein kinase, is one of the regulators of NFκB and is involved in the fibrotic effects of angiotensin II and aldosterone, as well as in CF differentiation. However, its role in the CF inflammatory response is unknown. On the other hand, many bioactive natural products have demonstrated anti-inflammatory effects, but their role in CF inflammation is unknown. One such molecule is boldine, an alkaloid obtained from Boldo (Peumus boldus), a Chilean endemic tree with proven cytoprotective effects. However, its involvement in the regulation of SGK1 and CF inflammation is unknown. In this study, we evaluated the role of SGK1 and boldine in the inflammatory response in CF isolated from neonatal Sprague-Dawley rats. The involvement of SGK1 was analyzed using GSK650394, a specific SGK1 inhibitor. Our results demonstrate that SGK1 is crucial for LPS- and IFN-γ-induced inflammatory responses in CF (cytokine expression, cell adhesion molecule expression, and leukocyte adhesion). Furthermore, a conditioned medium (intracellular content of CF subject to freeze/thaw cycles) was used to simulate a sterile inflammation condition. The conditioned medium induced a potent inflammatory response in CF, which was completely prevented by the SGK1 inhibitor. Finally, our results indicate that boldine inhibits both SGK1 activation and the CF inflammatory response induced by LPS, IFN-γ, and CF-conditioned medium. Taken together, our results position SGK1 as an important regulator of the CF inflammatory response and boldine as a promising anti-inflammatory drug in the context of cardiovascular diseases.


Sujet(s)
Aporphines , Fibroblastes , Protéines précoces immédiates , Facteur de transcription NF-kappa B , Protein-Serine-Threonine Kinases , Transduction du signal , Animaux , Facteur de transcription NF-kappa B/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protéines précoces immédiates/métabolisme , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Aporphines/pharmacologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Myocarde/anatomopathologie , Myocarde/métabolisme , Cellules cultivées , Rat Sprague-Dawley
3.
BMC Med Genomics ; 17(1): 152, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831322

RÉSUMÉ

OBJECTIVE: To investigate the role of BTG2 in periodontitis and diabetic kidney disease (DKD) and its potential underlying mechanism. METHODS: Gene expression data for periodontitis and DKD were acquired from the Gene Expression Omnibus (GEO) database. Differential expression analysis identified co-expressed genes between these conditions. The Nephroseq V5 online nephropathy database validated the role of these genes in DKD. Pearson correlation analysis identified genes associated with our target gene. We employed Gene Set Enrichment Analysis (GSEA) and Protein-Protein Interaction (PPI) networks to elucidate potential mechanisms. Expression levels of BTG2 mRNA were examined using quantitative polymerase Chain Reaction (qPCR) and immunofluorescence assays. Western blotting quantified proteins involved in epithelial-to-mesenchymal transition (EMT), apoptosis, mTORC1 signaling, and autophagy. Additionally, wound healing and flow cytometric apoptosis assays evaluated podocyte migration and apoptosis, respectively. RESULTS: Analysis of GEO database data revealed BTG2 as a commonly differentially expressed gene in both DKD and periodontitis. BTG2 expression was reduced in DKD compared to normal conditions and correlated with proteinuria. GSEA indicated enrichment of BTG2 in the EMT and mTORC1 signaling pathways. The PPI network highlighted BTG2's relevance to S100A9, S100A12, and FPR1. Immunofluorescence assays demonstrated significantly lower BTG2 expression in podocytes under high glucose (HG) conditions. Reduced BTG2 expression in HG-treated podocytes led to increased levels of EMT markers (α-SMA, vimentin) and the apoptotic protein Bim, alongside a decrease in nephrin. Lower BTG2 levels were associated with increased podocyte mobility and apoptosis, as well as elevated RPS6KB1 and mTOR levels, but reduced autophagy marker LC3. CONCLUSION: Our findings suggest that BTG2 is a crucial intermediary gene linking DKD and periodontitis. Modulating autophagy via inhibition of the mTORC1 signaling pathway, and consequently suppressing EMT, may be pivotal in the interplay between periodontitis and DKD.


Sujet(s)
Apoptose , Néphropathies diabétiques , Transition épithélio-mésenchymateuse , Parodontite , Protéines suppresseurs de tumeurs , Parodontite/génétique , Parodontite/métabolisme , Parodontite/anatomopathologie , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/génétique , Néphropathies diabétiques/anatomopathologie , Humains , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Podocytes/métabolisme , Podocytes/anatomopathologie , Transduction du signal , Autophagie , Cartes d'interactions protéiques , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Mouvement cellulaire
4.
Int J Biol Sci ; 20(8): 2922-2942, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904021

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and the production of autoantibodies. Previous studies have indicated an association between high-salt diets (HSD) and an increased risk of RA, yet the underlying mechanisms remain unclear. Macrophage pyroptosis, a pro-inflammatory form of cell death, plays a pivotal role in RA. In this study, we demonstrate that HSD exacerbates the severity of arthritis in collagen-induced arthritis (CIA) mice, correlating with macrophage infiltration and inflammatory lesions. Given the significant alterations observed in macrophages from CIA mice subjected to HSD, we specifically investigate the impact of HSD on macrophage responses in the inflammatory milieu of RA. In our in vitro experiments, pretreatment with NaCl enhances LPS-induced pyroptosis in RAW.264.7 and THP-1 cells through the p38 MAPK/NF-κB signaling pathway. Subsequent experiments reveal that Slc6a12 inhibitors and SGK1 silencing inhibit sodium-induced activation of macrophage pyroptosis and the p38 MAPK/NF-κB signaling pathway, whereas overexpression of the SGK1 gene counteracts the effect of sodium on macrophages. In conclusion, our findings verified that high salt intake promotes the progression of RA and provided a detailed elucidation of the activation of macrophage pyroptosis induced by sodium transportation through the Slc6a12 channel.


Sujet(s)
Polyarthrite rhumatoïde , Macrophages , Protein-Serine-Threonine Kinases , Pyroptose , Animaux , Souris , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Macrophages/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Chlorure de sodium/pharmacologie , Cellules RAW 264.7 , Humains , Mâle , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Arthrite expérimentale/métabolisme , Transduction du signal , p38 Mitogen-Activated Protein Kinases/métabolisme , Souris de lignée DBA
5.
J Exp Med ; 221(8)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38861480

RÉSUMÉ

Guard proteins initiate defense mechanisms upon sensing pathogen-encoded virulence factors. Successful viral pathogens likely inhibit guard protein activity, but these interactions have been largely undefined. Here, we demonstrate that the human pathogen herpes simplex virus 1 (HSV-1) stimulates and inhibits an antiviral pathway initiated by NLRP1, a guard protein that induces inflammasome formation and pyroptotic cell death when activated. Notably, HSV-1 infection of human keratinocytes promotes posttranslational modifications to NLRP1, consistent with MAPK-dependent NLRP1 activation, but does not result in downstream inflammasome formation. We identify infected cell protein 0 (ICP0) as the critical HSV-1 protein that is necessary and sufficient for inhibition of the NLRP1 pathway. Mechanistically, ICP0's cytoplasmic localization and function as an E3 ubiquitin ligase prevents proteasomal degradation of the auto-inhibitory NT-NLRP1 fragment, thereby preventing inflammasome formation. Further, we demonstrate that inhibiting this inflammasome is important for promoting HSV-1 replication. Thus, we have established a mechanism by which HSV-1 overcomes a guard-mediated antiviral defense strategy in humans.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Herpèsvirus humain de type 1 , Inflammasomes , Protéines NLR , Ubiquitin-protein ligases , Humains , Inflammasomes/métabolisme , Ubiquitin-protein ligases/métabolisme , Herpèsvirus humain de type 1/physiologie , Protéines NLR/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines précoces immédiates/métabolisme , Cellules HEK293 , Réplication virale , Kératinocytes/virologie , Kératinocytes/métabolisme , Herpès/virologie , Herpès/immunologie , Herpès/métabolisme , Animaux
6.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38731826

RÉSUMÉ

Although Herpes simplex virus type 1 (HSV-1) has been deeply studied, significant gaps remain in the fundamental understanding of HSV-host interactions: our work focused on studying the Infected Cell Protein 27 (ICP27) as an inhibitor of the Absent-in-melanoma-2 (AIM 2) inflammasome pathway, leading to reduced pro-inflammatory cytokines that influence the activation of a protective innate immune response to infection. To assess the inhibition of the inflammasome by the ICP27, hTert-immortalized Retinal Pigment Epithelial cells (hTert-RPE 1) infected with HSV-1 wild type were compared to HSV-1 lacking functional ICP27 (HSV-1∆ICP27) infected cells. The activation of the inflammasome by HSV-1∆ICP27 was demonstrated by quantifying the gene and protein expression of the inflammasome constituents using real-time PCR and Western blot. The detection of the cleavage of the pro-caspase-1 into the active form was performed by using a bioluminescent assay, while the quantification of interleukins 1ß (IL-1ß) and 18 (IL-18)released in the supernatant was quantified using an ELISA assay. The data showed that the presence of the ICP27 expressed by HSV-1 induces, in contrast to HSV-1∆ICP27 vector, a significant downregulation of AIM 2 inflammasome constituent proteins and, consequently, the release of pro-inflammatory interleukins into the extracellular environment reducing an effective response in counteracting infection.


Sujet(s)
Cytokines , Herpèsvirus humain de type 1 , Protéines précoces immédiates , Inflammasomes , Épithélium pigmentaire de la rétine , Humains , Lignée cellulaire , Cytokines/métabolisme , Protéines de liaison à l'ADN/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/virologie , Herpès/immunologie , Herpès/métabolisme , Herpès/virologie , Herpèsvirus humain de type 1/physiologie , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Inflammasomes/métabolisme , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/virologie
7.
Biomed Pharmacother ; 176: 116837, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815290

RÉSUMÉ

Hyperglycemic stress can directly lead to neuronal damage. The mechanosensitive ion channel PIEZO1 can be activated in response to hyperglycemia, but its role in hyperglycemic neurotoxicity is unclear. The role of PIEZO1 in hyperglycemic neurotoxicity was explored by constructing a hyperglycemic mouse model and a high-glucose HT22 cell model. The results showed that PIEZO1 was significantly upregulated in response to high glucose stress. In vitro experiments have shown that high glucose stress induces changes in neuronal cell morphology and membrane tension, a key mechanism for PIEZO1 activation. In addition, high glucose stress upregulates serum/glucocorticoid-regulated kinase-1 (SGK1) and activates PIEZO1 through the Ca2+ pool and store-operated calcium entry (SOCE). PIEZO1-mediated Ca2+ influx further enhances SGK1 and SOCE, inducing intracellular Ca2+ peaks in neurons. PIEZO1 mediated intracellular Ca2+ elevation leads to calcium/calmodulin-dependent protein kinase 2α (CaMK2α) overactivation, which promotes oxidative stress and apoptosis signalling through p-CaMK2α/ERK/CREB and ox-CaMK2α/MAPK p38/NFκB p65 pathways, subsequently inducing synaptic damage and cognitive impairment in mice. The intron miR-107 of pantothenic kinase 1 (PANK1) is highly expressed in the brain and has been found to target PIEZO1 and SGK1. The PANK1 receptor is activated by peroxisome proliferator-activated receptor α (PPARα), an activator known to upregulate miR-107 levels in the brain. The clinically used lipid-lowering drug bezafibrate, a known PPARα activator, may upregulate miR-107 through the PPARɑ/PANK1 pathway, thereby inhibiting PIEZO1 and improving hyperglycemia-induced neuronal cell damage. This study provides a new idea for the pathogenesis and drug treatment of hyperglycemic neurotoxicity and diabetes-related cognitive dysfunction.


Sujet(s)
Bézafibrate , Hyperglycémie , Canaux ioniques , Animaux , Canaux ioniques/métabolisme , Souris , Hyperglycémie/traitement médicamenteux , Hyperglycémie/métabolisme , Mâle , Bézafibrate/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , Souris de lignée C57BL , Stress oxydatif/effets des médicaments et des substances chimiques , Calcium/métabolisme , Lignée cellulaire , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , microARN/métabolisme , microARN/génétique , Glucose/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques
8.
mBio ; 15(6): e0016224, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38695580

RÉSUMÉ

Herpesvirus genomes are maintained as extrachromosomal plasmids within the nuclei of infected cells. Some herpesviruses persist within dividing cells, putting the viral genome at risk of being lost to the cytoplasm during mitosis because karyokinesis (nuclear division) requires nuclear envelope breakdown. Oncogenic herpesviruses (and papillomaviruses) avoid genome loss during mitosis by tethering their genomes to cellular chromosomes, thereby ensuring viral genome uptake into newly formed nuclei. These viruses use viral proteins with DNA- and chromatin-binding capabilities to physically link viral and cellular genomes together in a process called tethering. The known viral tethering proteins of human papillomavirus (E2), Epstein-Barr virus (EBNA1), and Kaposi's sarcoma-associated herpesvirus (LANA) each contain two independent domains required for genome tethering, one that binds sequence specifically to the viral genome and another that binds to cellular chromatin. This latter domain is called a chromatin tethering domain (CTD). The human cytomegalovirus UL123 gene encodes a CTD that is required for the virus to productively infect dividing fibroblast cells within the S phase of the cell cycle, presumably by tethering the viral genome to cellular chromosomes during mitosis. The CTD-containing UL123 gene product that supports S-phase infections is the IE19 protein. Here, we define two motifs in IE19 required for S-phase infections: an N-terminal triple lysine motif and a C-terminal nucleosome-binding motif within the CTD.IMPORTANCEThe IE19 protein encoded by human cytomegalovirus (HCMV) is required for S-phase infection of dividing cells, likely because it tethers the viral genome to cellular chromosomes, thereby allowing them to survive mitosis. The mechanism through which IE19 tethers viral genomes to cellular chromosomes is not understood. For human papillomavirus, Epstein-Barr virus, and Kaposi's sarcoma-associated herpesvirus, viral genome tethering is required for persistence (latency) and pathogenesis (oncogenesis). Like these viruses, HCMV also achieves latency, and it modulates the properties of glioblastoma multiforme tumors. Therefore, defining the mechanism through which IE19 tethers viral genomes to cellular chromosomes may help us understand, and ultimately combat or control, HCMV latency and oncomodulation.


Sujet(s)
Cytomegalovirus , Nucléosomes , Humains , Cytomegalovirus/génétique , Cytomegalovirus/métabolisme , Cytomegalovirus/physiologie , Nucléosomes/métabolisme , Nucléosomes/génétique , Phase S , Lysine/métabolisme , Lysine/génétique , Infections à cytomégalovirus/virologie , Infections à cytomégalovirus/métabolisme , Protéines virales/métabolisme , Protéines virales/génétique , Liaison aux protéines , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Motifs d'acides aminés
9.
Cell Signal ; 120: 111216, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38729325

RÉSUMÉ

Lung adenocarcinoma (LUAD) is the most prevalent subtype of lung cancer worldwide. Structural maintenance of chromosomes 2 (SMC2) serves as a predictor of poor prognosis across various cancer types. This study aims to explore the role and underlying mechanisms of SMC2 in LUAD progression. The expression of SMC2 in LUAD tissues and its correlation with prognosis were analyzed by public databases. Knockdown of SMC2 was performed to assess the proliferation, migration and invasion ability of LUAD cells. Bulk RNA sequencing analysis identified enriched cellular pathways and remarkable upregulation of BTG anti-proliferation factor 2 (BTG2) expression after SMC2 knockdown in LUAD cells. Then, BTG2 was silenced to assess the malignant behavior of LUAD cells. Subcutaneous transplantation and intracranial tumor models of LUAD cells in BALB/c nude mice were established to assess the antineoplastic effect of SMC2 knockdown in vivo. Additionally, a lung metastasis model was created to evaluate the pro-metastatic effect of SMC2. Our findings indicated that SMC2 was upregulated in LUAD tissues and cell lines, with higher expression correlating with poor prognosis. SMC2 silencing suppressed the proliferation, migration and invasion ability of LUAD cells by upregulating BTG2 expression via p53 and inactivating ERK and AKT pathways. BTG2 silencing reversed the effects of SMC2 downregulation on malignant behaviors of LUAD cells and restored the phosphorylated ERK and AKT levels. Furthermore, SMC2 knockdown effectively prevented the formation of subcutaneous, intracranial and metastatic tumor in vivo, and upregulation of BTG2 expression after SMC2 knockdown was confirmed in tumor models. This study revealed that SMC2 knockdown restrained the malignant progression of LUAD through upregulation of BTG2 expression and inactivation of ERK and AKT pathways, and SMC2 could be a potential therapeutic target for LUAD treatment.


Sujet(s)
Adénocarcinome pulmonaire , Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Protéines précoces immédiates , Tumeurs du poumon , Souris de lignée BALB C , Souris nude , Protéines suppresseurs de tumeurs , Régulation positive , Animaux , Femelle , Humains , Souris , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Évolution de la maladie , Techniques de knock-down de gènes , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique
10.
Arch Virol ; 169(6): 127, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38789713

RÉSUMÉ

Herpesviruses adhere to a precise temporal expression model in which immediate-early (IE) genes play a crucial role in regulating the viral life cycle. However, there is a lack of functional research on the IE genes in Ictalurid herpesvirus 1 (IcHV-1). In this study, we identified the IcHV-1 ORF24 as an IE gene via a metabolic inhibition assay, and subcellular analysis indicated its predominant localisation in the nucleus. To investigate its function, we performed yeast reporter assays using an ORF24 fusion protein containing the Gal4-BD domain and found that BD-ORF24 was able to activate HIS3/lacZ reporter genes without the Gal4-AD domain. Our findings provide concrete evidence that ORF24 is indeed an IE gene that likely functions as a transcriptional regulator during IcHV-1 infection. This work contributes to our understanding of the molecular mechanisms underlying fish herpesvirus IE gene expression.


Sujet(s)
Régulation de l'expression des gènes viraux , Gènes précoces , Protéines précoces immédiates/génétique , Protéines précoces immédiates/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Transcription génétique , Protéines virales/génétique , Protéines virales/métabolisme
11.
Biochem Biophys Res Commun ; 719: 150075, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38749087

RÉSUMÉ

Abundant evidence has shown the protective effect of aerobic exercise on central neuronal system, however, research about resistance exercise remains limited. To evaluate the effect and potential molecular mechanisms of resistance exercise in improving cognition and mental health, three-month-old male C57BL/6J mice underwent resistance training for five weeks. Body parameters, cognitive performance and synaptic plasticity were then assessed. In both groups, total RNA from the frontal cortex, hippocampus and gastrocnemius was isolated and sequenced, GO term and KEGG analysis were performed to identify molecular mechanisms. The results from RNA sequencing were then verified by RT-PCR. Our data found that mice in training group showed reduced anxiety-like behavior and better spatial memory. Accordingly, resistance exercise specifically increased the number of thin spines without affecting the number of other kind of spines. mRNA sequence analysis showed that resistance exercise induced differential expression of hundreds of genes in the above three tissues. KEGG analysis indicated the FoxO signaling pathway the most significant changed pathway throughout the brain and muscle. GO terms analysis showed that Sgk1 was enriched in the three key cognition related BP, including long-term memory, learning or memory and memory, and the expression level of Sgk1 was positive related with cognitive performance in the water maze. In conclusion, resistance exercise improved the mental health, cognition and synaptic plasticity of mice. Integrating analysis of mRNA expression profiles in frontal cortex, hippocampus and muscle reveals Sgk1 as the key mediator in brain-muscle crosstalk.


Sujet(s)
Encéphale , Protéines précoces immédiates , Souris de lignée C57BL , Muscles squelettiques , Conditionnement physique d'animal , Protein-Serine-Threonine Kinases , ARN messager , Animaux , Mâle , Protéines précoces immédiates/génétique , Protéines précoces immédiates/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Encéphale/métabolisme , Souris , Muscles squelettiques/métabolisme , Entraînement en résistance , Cognition/physiologie , Transcriptome , Plasticité neuronale/génétique , Hippocampe/métabolisme , Anxiété/génétique , Anxiété/métabolisme
12.
J Virol ; 98(6): e0042324, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38771044

RÉSUMÉ

Bovine alphaherpesvirus 1 (BoHV-1) infection causes respiratory tract disorders and immune suppression and may induce bacterial pneumonia. BoHV-1 establishes lifelong latency in sensory neurons after acute infection. Reactivation from latency consistently occurs following stress or intravenous injection of the synthetic corticosteroid dexamethasone (DEX), which mimics stress. The immediate early transcription unit 1 (IEtu1) promoter drives expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators necessary for productive infection and reactivation from latency. The IEtu1 promoter contains two glucocorticoid receptor (GR) responsive elements (GREs) that are transactivated by activated GR. GC-rich motifs, including consensus binding sites for specificity protein 1 (Sp1), are in the IEtu1 promoter sequences. E2F family members bind a consensus sequence (TTTCCCGC) and certain specificity protein 1 (Sp1) sites. Consequently, we hypothesized that certain E2F family members activate IEtu1 promoter activity. DEX treatment of latently infected calves increased the number of E2F2+ TG neurons. GR and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivate a 436-bp cis-regulatory module in the IEtu1 promoter that contains both GREs. A luciferase reporter construct containing a 222-bp fragment downstream of the GREs was transactivated by E2F2 unless two adjacent Sp1 binding sites were mutated. Chromatin immunoprecipitation studies revealed that E2F2 occupied IEtu1 promoter sequences when the BoHV-1 genome was transfected into mouse neuroblastoma (Neuro-2A) or monkey kidney (CV-1) cells. In summary, these findings revealed that GR and E2F2 cooperatively transactivate IEtu1 promoter activity, which is predicted to influence the early stages of BoHV-1 reactivation from latency. IMPORTANCE: Bovine alpha-herpesvirus 1 (BoHV-1) acute infection in cattle leads to establishment of latency in sensory neurons in the trigeminal ganglia (TG). A synthetic corticosteroid dexamethasone consistently initiates BoHV-1 reactivation in latently infected calves. The BoHV-1 immediate early transcription unit 1 (IEtu1) promoter regulates expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators. Hence, the IEtu1 promoter must be activated for the reactivation to occur. The number of TG neurons expressing E2F2, a transcription factor and cell cycle regulator, increased during early stages of reactivation from latency. The glucocorticoid receptor (GR) and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivated a 436-bp cis-regulatory module (CRM) in the IEtu1 promoter that contains two GR responsive elements. Chromatin immunoprecipitation studies revealed that E2F2 occupies IEtu1 promoter sequences in cultured cells. GR and E2F2 mediate cooperative transactivation of IEtu1 promoter activity, which is predicted to stimulate viral replication following stressful stimuli.


Sujet(s)
Régulation de l'expression des gènes viraux , Herpèsvirus bovin de type 1 , Régions promotrices (génétique) , Récepteurs aux glucocorticoïdes , Herpèsvirus bovin de type 1/génétique , Herpèsvirus bovin de type 1/physiologie , Animaux , Récepteurs aux glucocorticoïdes/métabolisme , Récepteurs aux glucocorticoïdes/génétique , Bovins , Activation de la transcription , Protéines virales/métabolisme , Protéines virales/génétique , Dexaméthasone/pharmacologie , Activation virale , Latence virale , Lignée cellulaire , Infections à Herpesviridae/virologie , Infections à Herpesviridae/métabolisme , Infections à Herpesviridae/médecine vétérinaire , Infections à Herpesviridae/génétique , Souris , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Éléments de réponse , Sites de fixation , Transactivateurs , Ubiquitin-protein ligases
13.
J Virol ; 98(6): e0071224, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38780246

RÉSUMÉ

Within the first 15 minutes of infection, herpes simplex virus 1 immediate early proteins repurpose cellular RNA polymerase (Pol II) for viral transcription. An important role of the viral-infected cell protein 27 (ICP27) is to facilitate viral pre-mRNA processing and export viral mRNA to the cytoplasm. Here, we use precision nuclear run-on followed by deep sequencing (PRO-seq) to characterize transcription of a viral ICP27 null mutant. At 1.5 and 3 hours post infection (hpi), we observed increased total levels of Pol II on the mutant viral genome and accumulation of Pol II downstream of poly A sites indicating increased levels of initiation and processivity. By 6 hpi, Pol II accumulation on specific mutant viral genes was higher than that on wild-type virus either at or upstream of poly A signals, depending on the gene. The PRO-seq profile of the ICP27 mutant on late genes at 6 hpi was similar but not identical to that caused by treatment with flavopiridol, a known inhibitor of RNA processivity. This pattern was different from PRO-seq profiles of other α gene mutants and upon inhibition of viral DNA replication with PAA. Together, these results indicate that ICP27 contributes to the repression of aberrant viral transcription at 1.5 and 3 hpi by inhibiting initiation and decreasing RNA processivity. However, ICP27 is needed to enhance processivity on most late genes by 6 hpi in a mechanism distinguishable from its role in viral DNA replication.IMPORTANCEWe developed and validated the use of a processivity index for precision nuclear run-on followed by deep sequencing data. The processivity index calculations confirm infected cell protein 27 (ICP27) induces downstream of transcription termination on certain host genes. The processivity indices and whole gene probe data implicate ICP27 in transient immediate early gene-mediated repression, a process that also requires ICP4, ICP22, and ICP0. The data indicate that ICP27 directly or indirectly regulates RNA polymerase (Pol II) initiation and processivity on specific genes at specific times post infection. These observations support specific and varied roles for ICP27 in regulating Pol II activity on viral genes in addition to its known roles in post transcriptional mRNA processing and export.


Sujet(s)
Génome viral , Herpèsvirus humain de type 1 , Protéines précoces immédiates , Réplication virale , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/physiologie , Protéines précoces immédiates/génétique , Protéines précoces immédiates/métabolisme , Humains , Mutation , RNA polymerase II/métabolisme , RNA polymerase II/génétique , ARN viral/génétique , ARN viral/métabolisme , Transcription virale/génétique , Animaux , Régulation de l'expression des gènes viraux , Cellules Vero , Chlorocebus aethiops , Herpès/virologie , Herpès/génétique , ARN messager/génétique , ARN messager/métabolisme
14.
J Virol ; 98(6): e0000524, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38717113

RÉSUMÉ

TRIM32 is often aberrantly expressed in many types of cancers. Kaposi's sarcoma-associated herpesvirus (KSHV) is linked with several human malignancies, including Kaposi's sarcoma and primary effusion lymphomas (PELs). Increasing evidence has demonstrated the crucial role of KSHV lytic replication in viral tumorigenesis. However, the role of TRIM32 in herpesvirus lytic replication remains unclear. Here, we reveal that the expression of TRIM32 is upregulated by KSHV in latency, and reactivation of KSHV lytic replication leads to the inhibition of TRIM32 in PEL cells. Strikingly, RTA, the master regulator of lytic replication, interacts with TRIM32 and dramatically promotes TRIM32 for degradation via the proteasome systems. Inhibition of TRIM32 induces cell apoptosis and in turn inhibits the proliferation and colony formation of KSHV-infected PEL cells and facilitates the reactivation of KSHV lytic replication and virion production. Thus, our data imply that the degradation of TRIM32 is vital for the lytic activation of KSHV and is a potential therapeutic target for KSHV-associated cancers. IMPORTANCE: TRIM32 is associated with many cancers and viral infections; however, the role of TRIM32 in viral oncogenesis remains largely unknown. In this study, we found that the expression of TRIM32 is elevated by Kaposi's sarcoma-associated herpesvirus (KSHV) in latency, and RTA (the master regulator of lytic replication) induces TRIM32 for proteasome degradation upon viral lytic reactivation. This finding provides a potential therapeutic target for KSHV-associated cancers.


Sujet(s)
Herpèsvirus humain de type 8 , Protéines précoces immédiates , Transactivateurs , Facteurs de transcription , Protéines à motif tripartite , Ubiquitin-protein ligases , Réplication virale , Humains , Herpèsvirus humain de type 8/physiologie , Herpèsvirus humain de type 8/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Transactivateurs/métabolisme , Transactivateurs/génétique , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéolyse , Latence virale , Apoptose , Activation virale , Sarcome de Kaposi/virologie , Sarcome de Kaposi/métabolisme , Proteasome endopeptidase complex/métabolisme , Lignée cellulaire , Lymphome primitif des séreuses/virologie , Lymphome primitif des séreuses/métabolisme
15.
Molecules ; 29(9)2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38731543

RÉSUMÉ

Ribonuclease P (RNase P) complexed with an external guide sequence (EGS) represents a promising nucleic acid-based gene targeting approach for gene expression knock-down and modulation. The RNase P-EGS strategy is unique as an EGS can be designed to basepair any mRNA sequence and recruit intracellular RNase P for hydrolysis of the target mRNA. In this study, we provide the first direct evidence that the RNase P-based approach effectively blocks the gene expression and replication of herpes simplex virus 2 (HSV-2), the causative agent of genital herpes. We constructed EGSs to target the mRNA encoding HSV-2 single-stranded DNA binding protein ICP8, which is essential for viral DNA genome replication and growth. In HSV-2 infected cells expressing a functional EGS, ICP8 levels were reduced by 85%, and viral growth decreased by 3000 folds. On the contrary, ICP8 expression and viral growth exhibited no substantial differences between cells expressing no EGS and those expressing a disabled EGS with mutations precluding RNase P recognition. The anti-ICP8 EGS is specific in targeting ICP8 because it only affects ICP8 expression but does not affect the expression of the other viral immediate-early and early genes examined. This study shows the effective and specific anti-HSV-2 activity of the RNase P-EGS approach and demonstrates the potential of EGS RNAs for anti-HSV-2 applications.


Sujet(s)
Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 2 , Réplication virale , Herpèsvirus humain de type 2/génétique , Herpèsvirus humain de type 2/physiologie , Humains , Ribonuclease P/métabolisme , Ribonuclease P/génétique , Animaux , Protéines virales/génétique , Protéines virales/métabolisme , Chlorocebus aethiops , ARN messager/génétique , ARN messager/métabolisme , Cellules Vero , Protéines précoces immédiates/génétique , Protéines précoces immédiates/métabolisme , Protéines de liaison à l'ADN
16.
J Transl Med ; 22(1): 511, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38807184

RÉSUMÉ

BACKGROUND: Myopia is one of the eye diseases that can damage the vision of young people. This study aimed to explore the protective role of miR-92b-3p against DNA damage and apoptosis in retinal tissues of negative lens-induced myopic (LIM) guinea pigs by targeting BTG2. METHODS: Biometric measurements of ocular parameters, flash electroretinogram (FERG), and retinal thickness (RT) were performed after miR-92b-3p intravitreal injection in LIM guinea pigs. The apoptotic rate was detected by Annexin V-FITC/PI double staining, and the change in mitochondrial membrane potential was measured by JC-1 staining. Retinal apoptosis and expression of p53, BTG2, and CDK2 were explored by TdT-mediated dUTP-biotin nick labeling (TUNEL) and immunofluorescence staining assays, respectively. BTG2 and its upstream and downstream molecules at gene and protein levels in retinal tissues were measured by real-time quantitative PCR (qPCR) and Western blotting. RESULTS: Compared with normal controls (NC), the ocular axial length of LIM guinea pig significantly increased, whereas refraction decreased. Meanwhile, dMax-a and -b wave amplitudes of ERG declined, retinal thickness was decreased, the number of apoptotic cells and apoptotic rate in LIM eyes was exaggerated, and the mitochondrial membrane potential significantly decreased. In addition, results of qPCR and Western blot assays showed that the expression levels of p53, BTG2, CDK2, and BAX in LIM guinea pigs were higher than the levels of the NC group, whereas the BCL-2 expression level was decreased. By contrast, the miR-92b-3p intravitreal injection in LIM guinea pigs could significantly inhibit axial elongation, alleviate DNA damage and apoptosis, and thus protect guinea pigs against myopia. CONCLUSION: In conclusion, p53 and BTG2 were activated in the retinal tissue of myopic guinea pigs, and the activated BTG2 could elevate the expression of CDK2 and BAX, and attenuate the expression of BCL-2, which in turn promote apoptosis and eventually lead to retinal thinning and impaired visual function in myopic guinea pigs. The miR-92b-3p intravitreal injection can attenuate the elongation of ocular length and retinal thickness, and inhibit the CDK2, BAX, and p53 expression by targeting BTG2, thereby ameliorating DNA damage and apoptosis in LIM guinea pigs and protecting ocular tissues.


Sujet(s)
Apoptose , Altération de l'ADN , microARN , Myopie , Rétine , Animaux , Cochons d'Inde , microARN/génétique , microARN/métabolisme , Rétine/anatomopathologie , Rétine/métabolisme , Myopie/métabolisme , Myopie/génétique , Myopie/anatomopathologie , Potentiel de membrane mitochondriale , Séquence nucléotidique , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Électrorétinographie , Modèles animaux de maladie humaine
17.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38732158

RÉSUMÉ

Biological membranes are composed of a lipid bilayer with embedded proteins, including ion channels like the epithelial sodium channel (ENaC), which are critical for sodium homeostasis and implicated in arterial hypertension (HTN). Changes in the lipid composition of the plasma membrane can significantly impact cellular processes related to physiological functions. We hypothesized that the observed overexpression of ENaC in neutrophils from HTN patients might result from alterations in the structuring domains within the plasma membrane, disrupting the endocytic processes responsible for ENaC retrieval. This study assessed the structural lipid composition of neutrophil plasma membranes from HTN patients along with the expression patterns of key elements regulating ENaC at the plasma membrane. Our findings suggest alterations in microdomain structure and SGK1 kinase activity, which could prolong ENaC presence on the plasma membrane. Additionally, we propose that the proteasomal and lysosomal degradation pathways are insufficient to diminish ENaC presence at the plasma membrane in HTN. These results highlight the importance of understanding ENaC retrieval mechanisms and suggest that targeting these mechanisms could provide insights for developing drugs to prevent and treat HTN.


Sujet(s)
Membrane cellulaire , Endocytose , Canaux sodium épithéliaux , Hypertension artérielle , Granulocytes neutrophiles , Canaux sodium épithéliaux/métabolisme , Humains , Granulocytes neutrophiles/métabolisme , Hypertension artérielle/métabolisme , Hypertension artérielle/anatomopathologie , Membrane cellulaire/métabolisme , Lipides membranaires/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Mâle , Femelle , Protéines précoces immédiates/métabolisme , Adulte d'âge moyen , Microdomaines membranaires/métabolisme
18.
Arch Virol ; 169(5): 116, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38722402

RÉSUMÉ

In this study, we investigated the role of serum/glucocorticoid-regulated kinase 1 (SGK1) in varicella-zoster virus (VZV) replication. VZV DNA replication and plaque formation were inhibited by SGK1 knockout and treatment with an SGK1 inhibitor. Furthermore, SGK1 inhibition suppressed the increase in cyclin B1 expression induced by VZV infection. These results suggest that VZV infection induces SGK1 activation, which is required for efficient viral proliferation through the expression of cyclin B1. This is the first study to report that SGK1 is involved in the VZV life cycle.


Sujet(s)
Cycline B1 , Herpèsvirus humain de type 3 , Protéines précoces immédiates , Protein-Serine-Threonine Kinases , Réplication virale , Protéines précoces immédiates/génétique , Protéines précoces immédiates/métabolisme , Humains , Herpèsvirus humain de type 3/génétique , Herpèsvirus humain de type 3/physiologie , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Cycline B1/métabolisme , Cycline B1/génétique , Lignée cellulaire , Réplication de l'ADN
19.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38673987

RÉSUMÉ

Sodium chloride (NaCl) activates Th17 and dendritic cells in hypertension by stimulating serum/glucocorticoid kinase 1 (SGK1), a sodium sensor. Memory T cells also play a role in hypertension by infiltrating target organs and releasing proinflammatory cytokines. We tested the hypothesis that the role of T cell SGK1 extends to memory T cells. We employed mice with a T cell deletion of SGK1, SGK1fl/fl × tgCD4cre mice, and used SGK1fl/fl mice as controls. We treated the mice with L-NAME (0.5 mg/mL) for 2 weeks and allowed a 2-week washout interval, followed by a 3-week high-salt (HS) diet (4% NaCl). L-NAME/HS significantly increased blood pressure and memory T cell accumulation in the kidneys and bone marrow of SGK1fl/fl mice compared to knockout mice on L-NAME/HS or groups on a normal diet (ND). SGK1fl/fl mice exhibited increased albuminuria, renal fibrosis, and interferon-γ levels after L-NAME/HS treatment. Myography demonstrated endothelial dysfunction in the mesenteric arterioles of SGK1fl/fl mice. Bone marrow memory T cells were adoptively transferred from either mouse strain after L-NAME/HS administration to recipient CD45.1 mice fed the HS diet for 3 weeks. Only the mice that received cells from SGK1fl/fl donors exhibited increased blood pressure and renal memory T cell infiltration. Our data suggest a new therapeutic target for decreasing hypertension-specific memory T cells and protecting against hypertension.


Sujet(s)
Hypertension artérielle , Protéines précoces immédiates , L-NAME , Protein-Serine-Threonine Kinases , Chlorure de sodium alimentaire , Animaux , Mâle , Souris , Pression sanguine/effets des médicaments et des substances chimiques , Hypertension artérielle/induit chimiquement , Hypertension artérielle/métabolisme , Hypertension artérielle/anatomopathologie , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Rein/métabolisme , Rein/anatomopathologie , Souris de lignée C57BL , Souris knockout , L-NAME/pharmacologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Chlorure de sodium alimentaire/effets indésirables , Lymphocytes T/métabolisme , Lymphocytes T/immunologie
20.
Metabolism ; 154: 155831, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38431129

RÉSUMÉ

BACKGROUND: Excessive dietary salt intake increases vascular stiffness in humans, especially in salt-sensitive populations. While we recently suggested that the endothelial sodium channel (EnNaC) contributes to salt-sensitivity related endothelial cell (EC) and arterial stiffening, mechanistic understanding remains incomplete. This study therefore aimed to explore the role of EC-serum and glucocorticoid regulated kinase 1 (SGK1), as a reported regulator of sodium channels, in EC and arterial stiffening. METHODS AND RESULTS: A mouse model of salt sensitivity-associated vascular stiffening was produced by subcutaneous implantation of slow-release deoxycorticosterone acetate (DOCA) pellets, with salt (1 % NaCl, 0.2 % KCl) administered via drinking water. Preliminary data showed that global SGK1 deletion caused significantly decreased blood pressure (BP), EnNaC activity and aortic endothelium stiffness as compared to control mice following DOCA-salt treatment. To probe EC signaling pathways, selective deletion of EC-SGK1 was performed by cross-breeding cadherin 5-Cre mice with sgk1flox/flox mice. DOCA-salt treated control mice had significantly increased BP, EC and aortic stiffness in vivo and ex vivo, which were attenuated by EC-SGK1 deficiency. To demonstrate relevance to humans, human aortic ECs were cultured in the absence or presence of aldosterone and high salt with or without the SGK1 inhibitor, EMD638683 (10uM or 25uM). Treatment with aldosterone and high salt increased intrinsic stiffness of ECs, which was prevented by SGK1 inhibition. Further, the SGK1 inhibitor prevented aldosterone and high salt induced actin polymerization, a key mechanism in cellular stiffening. CONCLUSION: EC-SGK1 contributes to salt-sensitivity related EC and aortic stiffening by mechanisms appearing to involve regulation of actin polymerization.


Sujet(s)
Cellules endothéliales , Protéines précoces immédiates , Protein-Serine-Threonine Kinases , Rigidité vasculaire , Animaux , Humains , Souris , Actines/métabolisme , Aldostérone/métabolisme , Aldostérone/pharmacologie , Pression sanguine/physiologie , Acétate de désoxycorticostérone , Cellules endothéliales/métabolisme , Glucocorticoïdes/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protéines précoces immédiates/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...