Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.572
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731826

RESUMEN

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.


Asunto(s)
Citocinas , Herpesvirus Humano 1 , Proteínas Inmediatas-Precoces , Inflamasomas , Epitelio Pigmentado de la Retina , Humanos , Inflamasomas/metabolismo , Herpesvirus Humano 1/fisiología , Citocinas/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/genética , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/virología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Línea Celular , Herpes Simple/inmunología , Herpes Simple/metabolismo , Herpes Simple/virología , Proteínas de Unión al ADN
2.
Arch Virol ; 169(5): 116, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38722402

RESUMEN

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.


Asunto(s)
Ciclina B1 , Herpesvirus Humano 3 , Proteínas Inmediatas-Precoces , Proteínas Serina-Treonina Quinasas , Replicación Viral , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Humanos , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ciclina B1/metabolismo , Ciclina B1/genética , Línea Celular , Replicación del ADN
3.
Molecules ; 29(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38731543

RESUMEN

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.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 2 , Replicación Viral , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiología , Humanos , Ribonucleasa P/metabolismo , Ribonucleasa P/genética , Animales , Proteínas Virales/genética , Proteínas Virales/metabolismo , Chlorocebus aethiops , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Vero , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de Unión al ADN
4.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732158

RESUMEN

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.


Asunto(s)
Membrana Celular , Endocitosis , Canales Epiteliales de Sodio , Hipertensión , Neutrófilos , Canales Epiteliales de Sodio/metabolismo , Humanos , Neutrófilos/metabolismo , Hipertensión/metabolismo , Hipertensión/patología , Membrana Celular/metabolismo , Lípidos de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Masculino , Femenino , Proteínas Inmediatas-Precoces/metabolismo , Persona de Mediana Edad , Microdominios de Membrana/metabolismo
5.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673987

RESUMEN

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.


Asunto(s)
Hipertensión , Proteínas Inmediatas-Precoces , NG-Nitroarginina Metil Éster , Proteínas Serina-Treonina Quinasas , Cloruro de Sodio Dietético , Animales , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/inducido químicamente , Hipertensión/patología , Ratones , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/genética , Cloruro de Sodio Dietético/efectos adversos , NG-Nitroarginina Metil Éster/farmacología , Ratones Noqueados , Linfocitos T/metabolismo , Linfocitos T/inmunología , Masculino , Ratones Endogámicos C57BL , Presión Sanguínea/efectos de los fármacos , Riñón/metabolismo , Riñón/patología
6.
Am J Med Sci ; 367(6): 397-405, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38437946

RESUMEN

BACKGROUND: Numerous studies have explored the therapeutic potential of microRNA (miR) in myocardial infarction (MI) treatment. This study focuses on the role of miR-322-5p in MI, particularly in its regulatory interaction with B-cell translocation gene 2 (BTG2). MATERIALS AND METHODS: Expression levels of miR-322-5p and BTG2 were assessed in a rat MI model. Adenovirus altering miR-322-5p or BTG2 expression were administered to MI rats. Evaluation included cardiac function, inflammation, myocardial injury, pathological changes, apoptosis, and NF-κB pathway-related genes in MI rats post-targeted treatment. The miR-322-5p and BTG2 targeting relationship was investigated. RESULTS: MI rats exhibited low miR-322-5p and high BTG2 expression in the myocardial tissues. Restoration of miR-322-5p enhanced cardiac function, alleviated inflammation and myocardial injury, mitigated pathological changes and apoptosis, and deactivated the NF-κB pathway in MI rats. BTG2 expression was negatively-regulated by miR-322-5p. Overexpressed BTG2 counteracted miR-322-5p-induced cardioprotection on MI rats. CONCLUSION: This study provides evidence that miR-322-5p protects against MI by suppressing BTG2 expression.


Asunto(s)
Proteínas Inmediatas-Precoces , MicroARNs , Infarto del Miocardio , Ratas Sprague-Dawley , Animales , MicroARNs/metabolismo , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Ratas , Masculino , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Apoptosis , FN-kappa B/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Modelos Animales de Enfermedad
7.
Sci Rep ; 14(1): 6423, 2024 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-38494504

RESUMEN

Hepatic ischemia-reperfusion injury (HIRI) elicits an immune-inflammatory response that may result in hepatocyte necrosis and apoptosis, ultimately culminating in postoperative hepatic dysfunction and hepatic failure. The precise mechanisms governing the pathophysiology of HIRI remain incompletely understood, necessitating further investigation into key molecules and pathways implicated in disease progression to guide drug discovery and potential therapeutic interventions. Gene microarray data was downloaded from the GEO expression profile database. Integrated bioinformatic analyses were performed to identify HIRI signature genes, which were subsequently validated for expression levels and diagnostic efficacy. Finally, the gene expression was verified in an experimental HIRI model and the effect of anti-IL17A antibody intervention in three time points (including pre-ischemic, post-ischemic, and at 1 h of reperfusion) on HIRI and the expression of these genes was investigated. Bioinformatic analyses of the screened characterized genes revealed that inflammation, immune response, and cell death modulation were significantly associated with HIRI pathophysiology. CCL2, BTG2, GADD45A, FOS, CXCL10, TNFRSF12A, and IL-17 pathway were identified as key components involved in the HIRI. Serum and liver IL-17A expression were significantly upregulated during the initial phase of HIRI. Pretreatment with anti-IL-17A antibody effectively alleviated the damage of liver tissue, suppressed inflammatory factors, and serum transaminase levels, and downregulated the mRNA expression of CCL2, GADD45A, FOS, CXCL10, and TNFRSF12A. Injection of anti-IL17A antibody after ischemia and at 1 h of reperfusion failed to demonstrate anti-inflammatory and attenuating HIRI benefits relative to earlier intervention. Our study reveals that the IL-17 pathway and related genes may be involved in the proinflammatory mechanism of HIRI, which may provide a new perspective and theoretical basis for the prevention and treatment of HIRI.


Asunto(s)
Proteínas Inmediatas-Precoces , Hepatopatías , Daño por Reperfusión , Humanos , Interleucina-17/metabolismo , Hígado/metabolismo , Daño por Reperfusión/metabolismo , Hepatopatías/metabolismo , Isquemia/metabolismo , Inflamación/genética , Inflamación/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Supresoras de Tumor/metabolismo
8.
Mol Biol Rep ; 51(1): 376, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38427115

RESUMEN

OBJECTIVE: Organisms and cellular viability are of paramount importance to living creatures. Disruption of the balance between cell survival and apoptosis results in compromised viability and even carcinogenesis. One molecule involved in keeping this homeostasis is serum-glucocorticoid regulated kinase (SGK) 1. Emerging evidence points to a significant role of SGK1 in cell growth and survival, cell metabolism, reproduction, and life span, particularly in prenatal programming and reproductive senescence by the same token. Whether the hormone inducible SGK1 kinase is a major driver in the pathophysiological processes of prenatal programming and reproductive senescence? METHOD: The PubMed/Medline, Web of Science, Embase/Ovid, and Elsevier Science Direct literature databases were searched for articles in English focusing on SGK1 published up to July 2023 RESULT: Emerging evidence is accumulating pointing to a pathophysiological role of the ubiquitously expressed SGK1 in the cellular and organismal viability. Under the regulation of specific hormones, extracellular stimuli, and various signals, SGK1 is involved in several biological processes relevant to viability, including cell proliferation and survival, cell migration and differentiation. In line, SGK1 contributes to the development of germ cells, embryos, and fetuses, whereas SGK1 inhibition leads to abnormal gametogenesis, embryo loss, and truncated reproductive lifespan. CONCLUTION: SGK1 integrates a broad spectrum of effects to maintain the homeostasis of cell survival and apoptosis, conferring viability to multiple cell types as well as both simple and complex organisms, and thus ensuring appropriate prenatal development and reproductive lifespan.


Asunto(s)
Glucocorticoides , Proteínas Inmediatas-Precoces , Embarazo , Femenino , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Reproducción
9.
Metabolism ; 154: 155831, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38431129

RESUMEN

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.


Asunto(s)
Células Endoteliales , Proteínas Inmediatas-Precoces , Proteínas Serina-Treonina Quinasas , Rigidez Vascular , Animales , Humanos , Ratones , Actinas/metabolismo , Aldosterona/metabolismo , Aldosterona/farmacología , Presión Sanguínea/fisiología , Acetato de Desoxicorticosterona , Células Endoteliales/metabolismo , Glucocorticoides/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Inmediatas-Precoces/metabolismo
10.
FEBS Lett ; 598(7): 758-773, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38436147

RESUMEN

The human Mediator complex subunit MED25 binds transactivation domains (TADs) present in various cellular and viral proteins using two binding interfaces, named H1 and H2, which are found on opposite sides of its ACID domain. Here, we use and compare deep learning methods to characterize human MED25-TAD interfaces and assess the predicted models to published experimental data. For the H1 interface, AlphaFold produces predictions with high-reliability scores that agree well with experimental data, while the H2 interface predictions appear inconsistent, preventing reliable binding modes. Despite these limitations, we experimentally assess the validity of MED25 interface predictions with the viral transcriptional activators Lana-1 and IE62. AlphaFold predictions also suggest the existence of a unique hydrophobic pocket for the Arabidopsis MED25 ACID domain.


Asunto(s)
Proteínas Inmediatas-Precoces , Complejo Mediador , Humanos , Complejo Mediador/genética , Complejo Mediador/metabolismo , Activación Transcripcional , Reproducibilidad de los Resultados , Factores de Transcripción/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Transactivadores/metabolismo , Proteínas Inmediatas-Precoces/metabolismo
11.
J Virol ; 98(4): e0193523, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38451085

RESUMEN

Placental infection plays a central role in the pathogenesis of congenital human cytomegalovirus (HCMV) infections and is a cause of fetal growth restriction and pregnancy loss. HCMV can replicate in some trophoblast cell types, but it remains unclear how the virus evades antiviral immunity in the placenta and how infection compromises placental development and function. Human trophoblast stem cells (TSCs) can be differentiated into extravillous trophoblasts (EVTs), syncytiotrophoblasts (STBs), and organoids, and this study assessed the utility of TSCs as a model of HCMV infection in the first-trimester placenta. HCMV was found to non-productively infect TSCs, EVTs, and STBs. Immunofluorescence assays and flow cytometry experiments further revealed that infected TSCs frequently only express immediate early viral gene products. Similarly, RNA sequencing found that viral gene expression in TSCs does not follow the kinetic patterns observed during lytic infection in fibroblasts. Canonical antiviral responses were largely not observed in HCMV-infected TSCs and TSC-derived trophoblasts. Rather, infection dysregulated factors involved in cell identity, differentiation, and Wingless/Integrated signaling. Thus, while HCMV does not replicate in TSCs, infection may perturb trophoblast differentiation in ways that could interfere with placental function. IMPORTANCE: Placental infection plays a central role in human cytomegalovirus (HCMV) pathogenesis during pregnancy, but the species specificity of HCMV and the limited availability and lifespan of primary trophoblasts have been persistent barriers to understanding how infection impacts this vital organ. Human trophoblast stem cells (TSCs) represent a new approach to modeling viral infection early in placental development. This study reveals that TSCs, like other stem cell types, restrict HCMV replication. However, infection perturbs the expression of genes involved in differentiation and cell fate determination, pointing to a mechanism by which HCMV could cause placental injury.


Asunto(s)
Citomegalovirus , Células Madre , Trofoblastos , Replicación Viral , Femenino , Humanos , Embarazo , Diferenciación Celular/genética , Linaje de la Célula/genética , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/fisiopatología , Infecciones por Citomegalovirus/virología , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Placenta/citología , Placenta/patología , Placenta/fisiopatología , Placenta/virología , Primer Trimestre del Embarazo , Células Madre/citología , Células Madre/virología , Trofoblastos/citología , Trofoblastos/virología
12.
Viruses ; 16(2)2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38400065

RESUMEN

Programmed necrosis is an integral part of intrinsic immunity, serving to combat invading pathogens and restricting viral dissemination. The orchestration of necroptosis relies on a precise interplay within the necrosome complex, which consists of RIPK1, RIPK3 and MLKL. Human cytomegalovirus (HCMV) has been found to counteract the execution of necroptosis during infection. In this study, we identify the immediate-early 1 (IE1) protein as a key antagonist of necroptosis during HCMV infection. Infection data obtained in a necroptosis-sensitive cell culture system revealed a robust regulation of post-translational modifications (PTMs) of the necrosome complex as well as the importance of IE1 expression for an effective counteraction of necroptosis. Interaction analyses unveiled an association of IE1 and RIPK3, which occurs in an RHIM-domain independent manner. We propose that this interaction manipulates the PTMs of RIPK3 by promoting its ubiquitination. Furthermore, IE1 was found to exert an indirect activity by modulating the levels of MLKL via antagonizing its interferon-mediated upregulation. Overall, we claim that IE1 performs a broad modulation of innate immune signaling to impede the execution of necroptotic cell death, thereby generating a favorable environment for efficient viral replication.


Asunto(s)
Citomegalovirus , Proteínas Inmediatas-Precoces , Humanos , Citomegalovirus/fisiología , Muerte Celular/fisiología , Apoptosis/fisiología , Necrosis , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
13.
Mol Cell Endocrinol ; 584: 112156, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38278341

RESUMEN

The imbalance between T helper 17 (Th17) and regulatory T (Treg) cells is an important mechanism in the pathogenesis of diabetic nephropathy (DN). Serum/glucocorticoid regulated kinase 1 (SGK1) is a serine-threonine kinase critical for stabilizing the Th17 cell phenotype. Sodium-glucose cotransporter 2 (SGLT2) is a glucose transporter that serves as a treatment target for diabetes. Our study investigated the regulatory role of SGLT2 in the development of DN. The results revealed that SGLT2 knockdown suppressed high glucose-induced excessive secretion of sodium (Na+) and inflammatory cytokines in mouse renal tubular epithelial TCMK-1 cells. High Na+ content induced Th17 differentiation and upregulated SGK1, phosphorylated forkhead box protein O1 (p-FoxO1), and the interleukin 23 receptor (IL-23 R) in primary mouse CD4+ T cells. Co-culture of CD4+ T cells with the culture medium of TCMK-1 cells with insufficient SGLT2 expression significantly suppressed cell migration ability, reduced the production of pro-inflammatory cytokines, and inhibited Th17 differentiation possibly by downregulating SGK1, p-FoxO1, and IL-23 R. In addition, in vivo data demonstrated that SGLT2 knockdown markedly downregulated SGK1 in db/db mice. Insufficient SGLT2 or SGK1 expression also ameliorated the Th17/Treg imbalance, suppressed the development of DN, and regulated the expression of IL-23 R and p-FoxO1. In conclusion, this study showed that SGLT2 knockdown restored the Th17/Treg balance and suppressed DN possibly by regulating the SGK1/p-FoxO1/IL-23 R axis by altering Na+ content in the local environment. These findings highlight the potential use of SGLT2 and SGK1 for the management of DN.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Proteínas Inmediatas-Precoces , Proteínas Serina-Treonina Quinasas , Transportador 2 de Sodio-Glucosa , Animales , Ratones , Citocinas/metabolismo , Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Glucocorticoides/metabolismo , Glucosa/metabolismo , Interleucina-23/metabolismo , Ratones Endogámicos , Sodio/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Linfocitos T Reguladores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Inmediatas-Precoces/metabolismo
14.
EMBO Rep ; 25(2): 725-744, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38177923

RESUMEN

Viral infection often trigger an ATM serine/threonine kinase (ATM)-dependent DNA damage response in host cells that suppresses viral replication. Viruses evolved different strategies to counteract this antiviral surveillance system. Here, we report that human herpesvirus 6B (HHV-6B) infection causes genomic instability by suppressing ATM signaling in host cells. Expression of immediate-early protein 1 (IE1) phenocopies this phenotype and blocks homology-directed double-strand break repair. Mechanistically, IE1 interacts with NBS1, and inhibits ATM signaling through two distinct domains. HHV-6B seems to efficiently inhibit ATM signaling as further depletion of either NBS1 or ATM do not significantly boost viral replication in infected cells. Interestingly, viral integration of HHV-6B into the host's telomeres is not strictly dependent on NBS1, challenging current models where integration occurs through homology-directed repair. Given that spontaneous IE1 expression has been detected in cells of subjects with inherited chromosomally-integrated form of HHV-6B (iciHHV-6B), a condition associated with several health conditions, our results raise the possibility of a link between genomic instability and the development of iciHHV-6-associated diseases.


Asunto(s)
Herpesvirus Humano 6 , Proteínas Inmediatas-Precoces , Infecciones por Roseolovirus , Humanos , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Infecciones por Roseolovirus/genética , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Integración Viral , Inestabilidad Genómica , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo
15.
Nat Commun ; 15(1): 881, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38286811

RESUMEN

Excessive host immune responses contribute to severe malaria with high mortality. Here, we show that PRL2 in innate immune cells is highly related to experimental malaria disease progression, especially the development of murine severe malaria. In the absence of PRL2 in myeloid cells, Plasmodium berghei infection results in augmented lung injury, leading to significantly increased mortality. Intravital imaging revealed greater neutrophilic inflammation and NET formation in the lungs of PRL2 myeloid conditional knockout mice. Depletion of neutrophils prior to the onset of severe disease protected mice from NETs associated lung injury, and eliminated the difference between WT and PRL2 CKO mice. PRL2 regulates neutrophil activation and NET accumulation via the Rac-ROS pathway, thus contributing to NETs associated ALI. Hydroxychloroquine, an inhibitor of PRL2 degradation alleviates NETs associated tissue damage in vivo. Our findings suggest that PRL2 serves as an indicator of progression to severe malaria and ALI. In addition, our study indicated the importance of PRL2 in NET formation and tissue injury. It might open a promising path for adjunctive treatment of NET-associated disease.


Asunto(s)
Lesión Pulmonar Aguda , Trampas Extracelulares , Proteínas Inmediatas-Precoces , Malaria , Proteínas Tirosina Fosfatasas , Animales , Ratones , Lesión Pulmonar Aguda/metabolismo , Trampas Extracelulares/metabolismo , Pulmón/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Inmediatas-Precoces/metabolismo
16.
J Virol ; 98(2): e0138623, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38240593

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV) genome consists of an approximately 140-kb unique coding region flanked by 30-40 copies of a 0.8-kb terminal repeat (TR) sequence. A gene enhancer recruits transcription-related enzymes by having arrays of transcription factor binding sites. Here, we show that KSHV TR possesses transcription regulatory function with latency-associated nuclear antigen (LANA). Cleavage under targets and release using nuclease demonstrated that TR fragments were occupied by LANA-interacting histone-modifying enzymes in naturally infected cells. The TR was enriched with histone H3K27 acetylation (H3K27Ac) and H3K4 tri-methylation (H3K4me3) modifications and also expressed nascent RNAs. The sites of H3K27Ac and H3K4me3 modifications were also conserved in the KSHV unique region among naturally infected primary effusion lymphoma cells. KSHV origin of lytic replication (Ori-Lyt) showed similar protein and histone modification occupancies with that of TR. In the Ori-Lyt region, the LANA and LANA-interacting proteins colocalized with an H3K27Ac-modified nucleosome along with paused RNA polymerase II. The KSHV transactivator KSHV replication and transcription activator (K-Rta) recruitment sites franked the LANA-bound nucleosome, and reactivation evicted the LANA-bound nucleosome. Including TR fragments in reporter plasmid enhanced inducible viral gene promoter activities independent of the orientations. In the presence of TR in reporter plasmids, K-Rta transactivation was drastically increased, while LANA acquired the promoter repression function. KSHV TR, therefore, functions as an enhancer for KSHV inducible genes. However, in contrast to cellular enhancers bound by multiple transcription factors, perhaps the KSHV enhancer is predominantly regulated by the LANA nuclear body.IMPORTANCEEnhancers are a crucial regulator of differential gene expression programs. Enhancers are the cis-regulatory sequences determining target genes' spatiotemporal and quantitative expression. Here, we show that Kaposi's sarcoma-associated herpesvirus (KSHV) terminal repeats fulfill the enhancer definition for KSHV inducible gene promoters. The KSHV enhancer is occupied by latency-associated nuclear antigen (LANA) and its interacting proteins, such as CHD4. Neighboring terminal repeat (TR) fragments to lytic gene promoters drastically enhanced KSHV replication and transcription activator and LANA transcription regulatory functions. This study, thus, proposes a new latency-lytic switch model in which TR accessibility to the KSHV gene promoters regulates viral inducible gene expression.


Asunto(s)
Herpesvirus Humano 8 , Proteínas Inmediatas-Precoces , Sarcoma de Kaposi , Humanos , Herpesvirus Humano 8/fisiología , Histonas/genética , Histonas/metabolismo , Nucleosomas , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Latencia del Virus/genética , Antígenos Virales/genética , Antígenos Virales/metabolismo , Secuencias Repetidas Terminales/genética , Regulación Viral de la Expresión Génica
17.
mBio ; 15(2): e0262123, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38165154

RESUMEN

Human cytomegalovirus (HCMV) requires the robust expression of two immediate early proteins, IE1 and IE2, immediately upon infection to suppress the antiviral response and promote viral gene expression. While transcriptional control of IE1 and IE2 has been extensively studied, the role of post-transcriptional regulation of IE1 and IE2 expression is relatively unexplored. We previously found that the shared major immediate early 5' untranslated region (MIE 5' UTR) of the mature IE1 and IE2 transcripts plays a critical role in facilitating the translation of the IE1 and IE2 mRNAs. As RNA secondary structure in 5' UTRs can regulate mRNA translation efficiency, we used selective 2'-hydroxyl acylation analyzed by primer extension and mutational profiling (SHAPE-MaP) to identify RNA structures in the shared MIE 5' UTR. We found that the MIE 5' UTR contains three stable stem loop structures. Using a series of recombinant viruses to investigate the role of each stem loop in IE1 and IE2 protein synthesis, we found that the stem loop closest to the 5' end of the MIE 5' UTR (SL1) is both necessary and sufficient for efficient IE1 and IE2 mRNA translation and HCMV replication. The positive effect of SL1 on mRNA translation and virus replication was dependent on its location within the 5' UTR. Surprisingly, a synthetic stem loop with the same free energy as SL1 in its native location also supported wild type levels of IE1 and IE2 mRNA translation and virus replication, suggesting that the presence of RNA structure at a specific location in the 5' UTR, rather than the primary sequence of the RNA, is critical for efficient IE1 and IE2 protein synthesis. These data reveal a novel post-transcriptional regulatory mechanism controlling IE1 and IE2 expression and reinforce the critical role of RNA structure in regulating HCMV protein synthesis and replication.IMPORTANCEThese results reveal a new aspect of immediate early gene regulation controlled by non-coding RNA structures in viral mRNAs. Previous studies have largely focused on understanding viral gene expression at the level of transcriptional control. Our results show that a complete understanding of the control of viral gene expression must include an understanding of viral mRNA translation, which is driven in part by RNA structure(s) in the 5' UTR of viral mRNAs. Our results illustrate the importance of these additional layers of regulation by defining specific 5' UTR RNA structures regulating immediate early gene expression in the context of infection and identify important features of RNA structure that govern viral mRNA translation efficiency. These results may therefore broadly impact current thinking on how viral gene expression is regulated for human cytomegalovirus and other DNA viruses.


Asunto(s)
Citomegalovirus , Proteínas Inmediatas-Precoces , Humanos , Regiones no Traducidas 5' , Citomegalovirus/fisiología , Proteínas Virales/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Replicación Viral , ARN Mensajero/genética , ARN Mensajero/metabolismo
18.
PLoS Pathog ; 20(1): e1011943, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38215174

RESUMEN

Deubiquitinases (DUBs) remove ubiquitin from substrates and play crucial roles in diverse biological processes. However, our understanding of deubiquitination in viral replication remains limited. Employing an oncogenic human herpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV) to probe the role of protein deubiquitination, we found that Ovarian tumor family deubiquitinase 4 (OTUD4) promotes KSHV reactivation. OTUD4 interacts with the replication and transcription activator (K-RTA), a key transcription factor that controls KSHV reactivation, and enhances K-RTA stability by promoting its deubiquitination. Notably, the DUB activity of OTUD4 is not required for K-RTA stabilization; instead, OTUD4 functions as an adaptor protein to recruit another DUB, USP7, to deubiquitinate K-RTA and facilitate KSHV lytic reactivation. Our study has revealed a novel mechanism whereby KSHV hijacks OTUD4-USP7 deubiquitinases to promote lytic reactivation, which could be potentially harnessed for the development of new antiviral therapies.


Asunto(s)
Herpesvirus Humano 8 , Proteínas Inmediatas-Precoces , Sarcoma de Kaposi , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Peptidasa Específica de Ubiquitina 7/genética , Peptidasa Específica de Ubiquitina 7/metabolismo , Transactivadores/genética , Herpesvirus Humano 8/genética , Replicación Viral , Regulación Viral de la Expresión Génica , Activación Viral , Proteasas Ubiquitina-Específicas/metabolismo
19.
J Virol ; 97(11): e0110723, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37902400

RESUMEN

IMPORTANCE: Immune evasion and latency are key mechanisms that underlie the success of herpesviruses. In each case, interactions between viral and host proteins are required and due to co-evolution, not all mechanisms are preserved across host species, even if infection is possible. This is highlighted by the herpes simplex virus (HSV) protein immediate early-infected cell protein (ICP)47, which inhibits the detection of infected cells by killer T cells and acts with high efficiency in humans, but poorly, if at all in mouse cells. Here, we show that ICP47 retains modest but detectable function in mouse cells, but in an in vivo model we found no role during acute infection or latency. We also explored the activity of the ICP47 promoter, finding that it could be active during latency, but this was dependent on genome location. These results are important to interpret HSV pathogenesis work done in mice.


Asunto(s)
Herpes Simple , Proteínas Inmediatas-Precoces , Simplexvirus , Animales , Ratones , Herpes Simple/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Evasión Inmune , Regiones Promotoras Genéticas , Simplexvirus/genética , Simplexvirus/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Latencia del Virus
20.
J Virol ; 97(11): e0138923, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37888983

RESUMEN

IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is a cancer-causing human herpesvirus that establishes a persistent infection in humans. The lytic viral cycle plays a crucial part in lifelong infection as it is involved in the viral dissemination. The master regulator of the KSHV lytic replication cycle is the viral replication and transcription activator (RTA) protein, which is necessary and sufficient to push the virus from latency into the lytic phase. Thus, the identification of host factors utilized by RTA for controlling the lytic cycle can help to find novel targets that could be used for the development of antiviral therapies against KSHV. Using a proteomics approach, we have identified a novel interaction between RTA and the cellular E3 ubiquitin ligase complex RNF20/40, which we have shown to be necessary for promoting RTA-induced KSHV lytic cycle.


Asunto(s)
Herpesvirus Humano 8 , Interacciones Microbiota-Huesped , Proteínas Inmediatas-Precoces , Ubiquitina-Proteína Ligasas , Proteínas Virales , Activación Viral , Latencia del Virus , Replicación Viral , Humanos , Herpesvirus Humano 8/crecimiento & desarrollo , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Unión Proteica , Proteómica , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA