Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28.490
Filtrar
1.
Nat Commun ; 15(1): 5515, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951495

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 8 , Proteínas Inmediatas-Precoces , Transactivadores , Ubiquitinación , Replicación Viral , Herpesvirus Humano 8/fisiología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/genética , Células HEK293 , Transactivadores/metabolismo , Transactivadores/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Activación Viral , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Núcleo Celular/metabolismo
2.
Vet Res ; 55(1): 84, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965634

RESUMEN

Pseudorabies virus (PRV) has evolved multiple strategies to evade host antiviral responses to benefit virus replication and establish persistent infection. Recently, tripartite motif 26 (TRIM26), a TRIM family protein, has been shown to be involved in a broad range of biological processes involved in innate immunity, especially in regulating viral infection. Herein, we found that the expression of TRIM26 was significantly induced after PRV infection. Surprisingly, the overexpression of TRIM26 promoted PRV production, while the depletion of this protein inhibited virus replication, suggesting that TRIM26 could positively regulate PRV infection. Further analysis revealed that TRIM26 negatively regulates the innate immune response by targeting the RIG-I-triggered type I interferon signalling pathway. TRIM26 was physically associated with MAVS independent of viral infection and reduced MAVS expression. Mechanistically, we found that NDP52 interacted with both TRIM26 and MAVS and that TRIM26-induced MAVS degradation was almost entirely blocked in NDP52-knockdown cells, demonstrating that TRIM26 degrades MAVS through NDP52-mediated selective autophagy. Our results reveal a novel mechanism by which PRV escapes host antiviral innate immunity and provide insights into the crosstalk among virus infection, autophagy, and the innate immune response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Autofagia , Inmunidad Innata , Animales , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Porcinos , Replicación Viral , Humanos , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética
3.
Arch Insect Biochem Physiol ; 116(3): e22127, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38976652

RESUMEN

Ubiquitin-fold modifier 1 (UFM1) is attached to protein substrates through the sequential activity of an E1 (UBA5)-E2 (UFC1)-E3 (UFL1) cascade. UFL1 is the E3 ligase for UFMylation in vertebrates. However, there have been no studies on UFL1 in silkworm to date. In this study, we identified a UFL1 ortholog in Bombyx mori genome. Spatio-temporal expression profiles showed that BmUFL1 expression was high in the midgut, epidermis, and testis and in the pupa-adult stage. BmUFL1 knockdown inhibited B. mori nucleopolyhedrovirus (BmNPV) proliferation, while BmUFL1 overexpression promoted BmNPV proliferation. Mechanically, protein kinase RNA-like endoplasmic reticulum kinase (PERK) signaling and cell apoptosis are involved in BmUFL1-regulated BmNPV proliferation. Overall, these results suggest that BmUFL1 facilitates BmNPV proliferation in silkworm.


Asunto(s)
Apoptosis , Bombyx , Proteínas de Insectos , Nucleopoliedrovirus , eIF-2 Quinasa , Animales , Bombyx/virología , Bombyx/genética , Bombyx/crecimiento & desarrollo , Nucleopoliedrovirus/fisiología , Proteínas de Insectos/metabolismo , Proteínas de Insectos/genética , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Replicación Viral , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Larva/virología , Larva/crecimiento & desarrollo , Larva/metabolismo , Larva/genética
4.
Int J Biol Sci ; 20(9): 3656-3674, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38993561

RESUMEN

Ubiquitination plays a pivotal regulatory role in tumor progression. Among the components of the ubiquitin-proteasome system (UPS), ubiquitin-protein ligase E3 has emerged as a key molecule. Nevertheless, the biological functions of E3 ubiquitin ligases and their potential mechanisms orchestrating glycolysis in gastric cancer (GC) remain to be elucidated. In this study, we conducted a comprehensive transcriptomic analysis to identify the core E3 ubiquitin ligases in GC, followed by extensive validation of the expression patterns and clinical significance of Tripartite motif-containing 50 (TRIM50) both in vitro and in vivo. Remarkably, we found that TRIM50 was downregulated in GC tissues, associated with malignant progression and poor patient survival. Functionally, overexpression of TRIM50 suppressed GC cell proliferation and indirectly mitigated the invasion and migration of GC cells by inhibiting the M2 polarization of tumor-associated macrophages (TAMs). Mechanistically, TRIM50 inhibited the glycolytic pathway by ubiquitinating Phosphoglycerate Kinase 1 (PGK1), thereby directly suppressing GC cell proliferation. Simultaneously, the reduction in lactate led to diminished M2 polarization of TAMs, indirectly inhibiting the invasion and migration of GC cells. Notably, the downregulation of TRIM50 in GC was mediated by the METTL3/YTHDF2 axis in an m6A-dependent manner. In our study, we definitively identified TRIM50 as a tumor suppressor gene (TSG) that effectively inhibits glycolysis and the malignant progression of GC by ubiquitinating PGK1, thus offering novel insights and promising targets for the diagnosis and treatment of GC.


Asunto(s)
Glucólisis , Fosfoglicerato Quinasa , Neoplasias Gástricas , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Ubiquitinación , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Humanos , Fosfoglicerato Quinasa/metabolismo , Fosfoglicerato Quinasa/genética , Línea Celular Tumoral , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Proliferación Celular/genética , Animales , Ratones , Ratones Desnudos , Progresión de la Enfermedad , Movimiento Celular/genética , Ratones Endogámicos BALB C , Regulación Neoplásica de la Expresión Génica , Metiltransferasas/metabolismo , Metiltransferasas/genética
5.
Sci Adv ; 10(28): eadg1421, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996019

RESUMEN

Genomic mechanisms enhancing risk in males may contribute to sex bias in autism. The ubiquitin protein ligase E3A gene (Ube3a) affects cellular homeostasis via control of protein turnover and by acting as transcriptional coactivator with steroid hormone receptors. Overdosage of Ube3a via duplication or triplication of chromosomal region 15q11-13 causes 1 to 2% of autistic cases. Here, we test the hypothesis that increased dosage of Ube3a may influence autism-relevant phenotypes in a sex-biased manner. We show that mice with extra copies of Ube3a exhibit sex-biasing effects on brain connectomics and autism-relevant behaviors. These effects are associated with transcriptional dysregulation of autism-associated genes, as well as genes differentially expressed in 15q duplication and in autistic people. Increased Ube3a dosage also affects expression of genes on the X chromosome, genes influenced by sex steroid hormone, and genes sex-differentially regulated by transcription factors. These results suggest that Ube3a overdosage can contribute to sex bias in neurodevelopmental conditions via influence on sex-differential mechanisms.


Asunto(s)
Trastorno Autístico , Transcriptoma , Ubiquitina-Proteína Ligasas , Animales , Masculino , Femenino , Trastorno Autístico/genética , Ratones , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Humanos , Conducta Animal , Caracteres Sexuales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad
7.
Methods Mol Biol ; 2780: 345-359, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38987477

RESUMEN

Chemical protein knockdown technology using proteolysis-targeting chimeras (PROTACs) to hijack the endogenous ubiquitin-proteasome system is a powerful strategy to degrade disease-related proteins. This chapter describes in silico design of a hematopoietic prostaglandin D synthase (H-PGDS) degrader, PROTAC(H-PGDS), using a docking simulation of the ternary complex of H-PGDS/PROTAC/E3 ligase as well as the synthesis of the designed PROTAC(H-PGDS)s and evaluation of their H-PGDS degradation activity.


Asunto(s)
Oxidorreductasas Intramoleculares , Lipocalinas , Simulación del Acoplamiento Molecular , Proteolisis , Oxidorreductasas Intramoleculares/metabolismo , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Humanos , Lipocalinas/metabolismo , Lipocalinas/química , Simulación por Computador , Diseño de Fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/química
8.
J Med Virol ; 96(7): e29789, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38988206

RESUMEN

Merkel cell carcinoma (MCC) is a highly aggressive skin cancer associated with integration of Merkel cell polyomavirus (MCPyV). MCPyV-encoded T-antigens (TAs) are pivotal for sustaining MCC's oncogenic phenotype, i.e., repression of TAs results in reactivation of the RB pathway and subsequent cell cycle arrest. However, the MCC cell line LoKe, characterized by a homozygous loss of the RB1 gene, exhibits uninterrupted cell cycle progression after shRNA-mediated TA repression. This unique feature allows an in-depth analysis of the effects of TAs beyond inhibition of the RB pathway, revealing the decrease in expression of stem cell-related genes upon panTA-knockdown. Analysis of gene regulatory networks identified members of the E2F family (E2F1, E2F8, TFDP1) as key transcriptional regulators that maintain stem cell properties in TA-expressing MCC cells. Furthermore, minichromosome maintenance (MCM) genes, which encodes DNA-binding licensing proteins essential for stem cell maintenance, were suppressed upon panTA-knockdown. The decline in stemness occurred simultaneously with neural differentiation, marked by the increased expression of neurogenesis-related genes such as neurexins, BTG2, and MYT1L. This upregulation can be attributed to heightened activity of PBX1 and BPTF, crucial regulators of neurogenesis pathways. The observations in LoKe were confirmed in an additional MCPyV-positive MCC cell line in which RB1 was silenced before panTA-knockdown. Moreover, spatially resolved transcriptomics demonstrated reduced TA expression in situ in a part of a MCC tumor characterized by neural differentiation. In summary, TAs are critical for maintaining stemness of MCC cells and suppressing neural differentiation, irrespective of their impact on the RB-signaling pathway.


Asunto(s)
Antígenos Virales de Tumores , Diferenciación Celular , Técnicas de Silenciamiento del Gen , Poliomavirus de Células de Merkel , Proteínas de Unión a Retinoblastoma , Humanos , Poliomavirus de Células de Merkel/genética , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Línea Celular Tumoral , Diferenciación Celular/genética , Proteínas de Unión a Retinoblastoma/genética , Proteínas de Unión a Retinoblastoma/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Carcinoma de Células de Merkel/virología , Carcinoma de Células de Merkel/genética , Células Madre Neoplásicas/virología , Células Madre Neoplásicas/metabolismo , Neoplasias Cutáneas/virología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Redes Reguladoras de Genes , Neuronas/virología
9.
Cell Host Microbe ; 32(7): 1114-1128.e10, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38955187

RESUMEN

Plant immune homeostasis is achieved through a balanced immune activation and suppression, enabling effective defense while averting autoimmunity. In Arabidopsis, disrupting a mitogen-activated protein (MAP) kinase cascade triggers nucleotide-binding leucine-rich-repeat (NLR) SUPPRESSOR OF mkk1/2 2 (SUMM2)-mediated autoimmunity. Through an RNAi screen, we identify PUB5, a putative plant U-box E3 ligase, as a critical regulator of SUMM2-mediated autoimmunity. In contrast to typical E3 ligases, PUB5 stabilizes CRCK3, a calmodulin-binding receptor-like cytoplasmic kinase involved in SUMM2 activation. A closely related E3 ligase, PUB44, functions oppositely with PUB5 to degrade CRCK3 through monoubiquitylation and internalization. Furthermore, CRCK3, highly expressed in roots and conserved across plant species, confers resistance to Fusarium oxysporum, a devastating soil-borne fungal pathogen, in both Arabidopsis and cotton. These findings demonstrate the antagonistic role of an E3 ligase pair in fine-tuning kinase proteostasis for the regulation of NLR-mediated autoimmunity and highlight the function of autoimmune activators in governing plant root immunity against fungal pathogens.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Autoinmunidad , Resistencia a la Enfermedad , Fusarium , Enfermedades de las Plantas , Inmunidad de la Planta , Ubiquitina-Proteína Ligasas , Arabidopsis/inmunología , Arabidopsis/microbiología , Arabidopsis/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/inmunología , Fusarium/inmunología , Proteínas NLR/metabolismo , Proteínas NLR/genética , Regulación de la Expresión Génica de las Plantas , Ubiquitinación , Proteínas Portadoras
10.
Sci Rep ; 14(1): 15848, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38982159

RESUMEN

E3 ubiquitin protein ligase encoded by ARIH2 gene catalyses the ubiquitination of target proteins and plays a crucial role in posttranslational modifications across various cellular processes. As prior documented, mutations in genes involved in the ubiquitination process are often associated with autism spectrum disorder (ASD) and/or intellectual disability (ID). In the current study, a de novo heterozygous mutation was identified in the splicing intronic region adjacent to the last exon of the ARIH2 gene using whole exome sequencing (WES). We hypothesize that this mutation, found in an ASD/ID patient, disrupts the protein Ariadne domain which is involved in the autoinhibition of ARIH2 enzyme. Predictive analyses elucidated the implications of the novel mutation in the splicing process and confirmed its autosomal dominant inheritance model. Nevertheless, we cannot exclude the possibility that other genetic factors, undetectable by WES, such as mutations in non-coding regions and polygenic risk in inter-allelic complementation, may contribute to the patient's phenotype. This work aims to suggest potential relationship between the detected mutation in ARIH2 gene and both ASD and ID, even though functional studies combined with new sequencing approaches will be necessary to validate this hypothesis.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Mutación , Ubiquitina-Proteína Ligasas , Humanos , Trastorno del Espectro Autista/genética , Discapacidad Intelectual/genética , Ubiquitina-Proteína Ligasas/genética , Masculino , Secuenciación del Exoma , Femenino , Predisposición Genética a la Enfermedad , Niño
11.
Int J Mol Sci ; 25(13)2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-39000606

RESUMEN

Sarcopenia refers to an age-related decrease in muscle mass and strength. The gut-muscle axis has been proposed as a promising target to alleviate muscle atrophy. The effect of KL-Biome-a postbiotic preparation comprising heat-killed Lactiplantibacillus plantarum KM-2, its metabolites, and an excipient (soybean powder)-on muscle atrophy was evaluated using dexamethasone (DEX)-induced atrophic C2C12 myoblasts and C57BL/6J mice. KL-Biome significantly downregulated the expression of genes (Atrogin-1 and MuRF1) associated with skeletal muscle degradation but increased the anabolic phosphorylation of FoxO3a, Akt, and mTOR in C2C12 cells. Oral administration of KL-Biome (900 mg/kg) for 8 weeks significantly improved muscle mass, muscle function, and serum lactate dehydrogenase levels in DEX-treated mice. KL-Biome administration increased gut microbiome diversity and reversed DEX-mediated gut microbiota alterations. Furthermore, it significantly increased the relative abundances of the genera Subdologranulum, Alistipes, and Faecalibacterium prausnitzii, which are substantially involved in short-chain fatty acid production. These findings suggest that KL-Biome exerts beneficial effects on muscle atrophy by regulating gut microbiota.


Asunto(s)
Dexametasona , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Músculo Esquelético , Atrofia Muscular , Animales , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/metabolismo , Atrofia Muscular/inducido químicamente , Ratones , Dexametasona/farmacología , Dexametasona/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Masculino , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Proteína Forkhead Box O3/metabolismo , Proteína Forkhead Box O3/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Probióticos/administración & dosificación , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Sarcopenia/tratamiento farmacológico , Sarcopenia/metabolismo , Sarcopenia/patología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Línea Celular , Lactobacillus plantarum
12.
J Cell Biol ; 223(10)2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-38967608

RESUMEN

Peroxisomes are membrane-bound organelles harboring metabolic enzymes. In humans, peroxisomes are required for normal development, yet the genes regulating peroxisome function remain unclear. We performed a genome-wide CRISPRi screen to identify novel factors involved in peroxisomal homeostasis. We found that inhibition of RNF146, an E3 ligase activated by poly(ADP-ribose), reduced the import of proteins into peroxisomes. RNF146-mediated loss of peroxisome import depended on the stabilization and activity of the poly(ADP-ribose) polymerases TNKS and TNKS2, which bind the peroxisomal membrane protein PEX14. We propose that RNF146 and TNKS/2 regulate peroxisome import efficiency by PARsylation of proteins at the peroxisome membrane. Interestingly, we found that the loss of peroxisomes increased TNKS/2 and RNF146-dependent degradation of non-peroxisomal substrates, including the ß-catenin destruction complex component AXIN1, which was sufficient to alter the amplitude of ß-catenin transcription. Together, these observations not only suggest previously undescribed roles for RNF146 in peroxisomal regulation but also a novel role in bridging peroxisome function with Wnt/ß-catenin signaling during development.


Asunto(s)
Proteína Axina , Peroxisomas , Ubiquitina-Proteína Ligasas , Vía de Señalización Wnt , Peroxisomas/metabolismo , Peroxisomas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Humanos , Proteína Axina/metabolismo , Proteína Axina/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , beta Catenina/metabolismo , beta Catenina/genética , Células HEK293 , Transporte de Proteínas , Sistemas CRISPR-Cas
13.
Nat Commun ; 15(1): 5961, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39013878

RESUMEN

Autoreactive CD4+ T helper cells are critical players that orchestrate the immune response both in multiple sclerosis (MS) and in other neuroinflammatory autoimmune diseases. Ubiquitination is a posttranslational protein modification involved in regulating a variety of cellular processes, including CD4+ T cell differentiation and function. However, only a limited number of E3 ubiquitin ligases have been characterized in terms of their biological functions, particularly in CD4+ T cell differentiation and function. In this study, we found that the RING finger protein 213 (RNF213) specifically promoted regulatory T (Treg) cell differentiation in CD4+ T cells and attenuated autoimmune disease development in an FOXO1-dependent manner. Mechanistically, RNF213 interacts with Forkhead Box Protein O1 (FOXO1) and promotes nuclear translocation of FOXO1 by K63-linked ubiquitination. Notably, RNF213 expression in CD4+ T cells was induced by IFN-ß and exerts a crucial role in the therapeutic efficacy of IFN-ß for MS. Together, our study findings collectively emphasize the pivotal role of RNF213 in modulating adaptive immune responses. RNF213 holds potential as a promising therapeutic target for addressing disorders associated with Treg cells.


Asunto(s)
Diferenciación Celular , Proteína Forkhead Box O1 , Interferón beta , Linfocitos T Reguladores , Ubiquitina-Proteína Ligasas , Ubiquitinación , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Animales , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Ratones , Humanos , Interferón beta/metabolismo , Ratones Endogámicos C57BL , Núcleo Celular/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Transporte Activo de Núcleo Celular , Femenino , Ratones Noqueados , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Células HEK293
14.
Nat Commun ; 15(1): 6177, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39039081

RESUMEN

The ankyrin (ANK) SOCS box (ASB) family, encompassing ASB1-18, is the largest group of substrate receptors of cullin 5 Ring E3 ubiquitin ligase. Nonetheless, the mechanism of substrate recognition by ASB family proteins has remained largely elusive. Here we present the crystal structure of ASB7-Elongin B-Elongin C ternary complex bound to a conserved helical degron. ASB7 employs its ANK3-6 to form an extended groove, effectively interacting with the internal α-helix-degron through a network of side-chain-mediated electrostatic and hydrophobic interactions. Our structural findings, combined with biochemical and cellular analyses, identify the key residues of the degron motif and ASB7 required for their recognition. This will facilitate the identification of additional physiological substrates of ASB7 by providing a defined degron motif for screening. Furthermore, the structural insights provide a basis for the rational design of compounds that can specifically target ASB7 by disrupting its interaction with its cognate degron.


Asunto(s)
Unión Proteica , Proteínas Supresoras de la Señalización de Citocinas , Humanos , Cristalografía por Rayos X , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ancirinas/metabolismo , Ancirinas/química , Ancirinas/genética , Modelos Moleculares , Elonguina/metabolismo , Elonguina/genética , Elonguina/química , Células HEK293 , Secuencias de Aminoácidos , Degrones
15.
Nat Commun ; 15(1): 6172, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39039092

RESUMEN

The severity of bacterial pneumonia can be worsened by impaired innate immunity resulting in ineffective pathogen clearance. We describe a mitochondrial protein, aspartyl-tRNA synthetase (DARS2), which is released in circulation during bacterial pneumonia in humans and displays intrinsic innate immune properties and cellular repair properties. DARS2 interacts with a bacterial-induced ubiquitin E3 ligase subunit, FBXO24, which targets the synthetase for ubiquitylation and degradation, a process that is inhibited by DARS2 acetylation. During experimental pneumonia, Fbxo24 knockout mice exhibit elevated DARS2 levels with an increase in pulmonary cellular and cytokine levels. In silico modeling identified an FBXO24 inhibitory compound with immunostimulatory properties which extended DARS2 lifespan in cells. Here, we show a unique biological role for an extracellular, mitochondrially derived enzyme and its molecular control by the ubiquitin apparatus, which may serve as a mechanistic platform to enhance protective host immunity through small molecule discovery.


Asunto(s)
Aspartato-ARNt Ligasa , Inmunidad Innata , Ratones Noqueados , Mitocondrias , Ubiquitinación , Animales , Aspartato-ARNt Ligasa/metabolismo , Aspartato-ARNt Ligasa/genética , Humanos , Ratones , Mitocondrias/metabolismo , Proteínas F-Box/metabolismo , Proteínas F-Box/genética , Ratones Endogámicos C57BL , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteolisis , Femenino , Masculino , Citocinas/metabolismo , Células HEK293 , Acetilación , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética
16.
Nat Commun ; 15(1): 6197, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043663

RESUMEN

Replication stress compromises genomic integrity. Fork blocking lesions such as those induced by cisplatin and other chemotherapeutic agents arrest replication forks. Repriming downstream of these lesions represents an important mechanism of replication restart, however the single stranded DNA (ssDNA) gaps left behind, unless efficiently filled, can serve as entry point for nucleases. Nascent strand gaps can be repaired by BRCA-mediated homology repair. Alternatively, gaps can also be filled by translesion synthesis (TLS) polymerases. How these events are regulated is still not clear. Here, we show that PARP10, a poorly-characterized mono-ADP-ribosyltransferase, is recruited to nascent strand gaps to promote their repair. PARP10 interacts with the ubiquitin ligase RAD18 and recruits it to these structures, resulting in the ubiquitination of the replication factor PCNA. PCNA ubiquitination, in turn, recruits the TLS polymerase REV1 for gap filling. We show that PARP10 recruitment to gaps and the subsequent REV1-mediated gap filling requires both the catalytic activity of PARP10, and its ability to interact with PCNA. We moreover show that PARP10 is hyperactive in BRCA-deficient cells, and its inactivation potentiates gap accumulations and cytotoxicity in these cells. Our work uncovers PARP10 as a regulator of ssDNA gap filling, which promotes genomic stability in BRCA-deficient cells.


Asunto(s)
Reparación del ADN , Replicación del ADN , ADN de Cadena Simple , Proteínas de Unión al ADN , Poli(ADP-Ribosa) Polimerasas , Antígeno Nuclear de Célula en Proliferación , Ubiquitina-Proteína Ligasas , Ubiquitinación , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Daño del ADN , Proteína BRCA2/metabolismo , Proteína BRCA2/genética , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Línea Celular Tumoral , Células HEK293 , Síntesis Translesional de ADN , ADN Polimerasa Dirigida por ADN , Proteínas Proto-Oncogénicas
17.
Acta Biochim Biophys Sin (Shanghai) ; 56(7): 963-972, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38993132

RESUMEN

Endothelial dysfunction (ED) serves as the pathological basis for various cardiovascular diseases. Guanosine triphosphate cyclopyrrolone 1 (GCH1) emerges as a pivotal protein in sustaining nitric oxide (NO) production within endothelial cells, yet it undergoes degradation under oxidative stress, contributing to endothelial cell dysfunction. Citronellal (CT), a monoterpenoid, has been shown to ameliorate endothelial dysfunction induced by in atherosclerosis rats. However, whether CT can inhibit the degradation of GCH1 protein is not clear. It has been reported that ubiquitination may play a crucial role in regulating GCH1 protein levels and activities. However, the specific E3 ligase for GCH1 and the molecular mechanism of GCH1 ubiquitination remain unclear. Using data-base exploration analysis, we find that the levels of the E3 ligase Smad-ubiquitination regulatory factor 2 (Smurf2) negatively correlate with those of GCH1 in vascular tissues and HUVECs. We observe that Smurf2 interacts with GCH1 and promotes its degradation via the proteasome pathway. Interestingly, ectopic Smurf2 expression not only decreases GCH1 levels but also reduces cell proliferation and reactive oxygen species (ROS) levels, mostly because of increased GCH1 accumulation. Furthermore, we identify BH 4/eNOS as downstream of GCH1. Taken together, our results indicate that CT can obviously improve vascular endothelial injury in Type 1 diabetes mellitus (T1DM) rats and reverse the expressions of GCH1 and Smurf2 proteins in aorta of T1DM rats. Smurf2 promotes ubiquitination and degradation of GCH1 through proteasome pathway in HUVECs. We conclude that the Smurf2-GCH1 interaction might represent a potential target for improving endothelial injury.


Asunto(s)
Monoterpenos Acíclicos , Células Endoteliales de la Vena Umbilical Humana , Ubiquitina-Proteína Ligasas , Animales , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Humanos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Monoterpenos Acíclicos/farmacología , Monoterpenos Acíclicos/metabolismo , Ratas , Ubiquitinación , Aldehídos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Masculino , Ratas Sprague-Dawley , Óxido Nítrico/metabolismo , Proliferación Celular , Estabilidad Proteica , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Estrés Oxidativo
18.
J Physiol Pharmacol ; 75(3)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39042393

RESUMEN

Circular E3 ubiquitin-protein ligase (circ-ITCH), a novel circRNA, is generated from several exons of itchy E3 ubiquitin protein ligase. Reports on circ-ITCH have discussed its pathogenic performance in human diseases. Based on this, this study determines whether and how circ-ITCH is involved in the pathogenesis of chronic glomerulonephritis (CGN). First, a rat model of CGN induced by cationic bovine serum albumin was established. Then, CGN rats were injected with lentiviruses interfering with the expression of circ-ITCH, miR-146a-5p or tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein gamma (YWHAG). Then, blood urea nitrogen and serum creatinine levels were measured to evaluate renal function; inflammatory factor content and fibrosis marker expression in kidney tissue were detected; renal pathological damage was analyzed by hematoxylin-eosin staining and periodic acid-Schiff staining. Finally, the binding relationship between miR-146a-5p and circ-ITCH or YWHAG was verified. Elevating circ-ITCH or depleting miR-146a-5p improved renal function (both P<0.05), reduced inflammatory factor content and fibrosis marker expression (all P<0.05) and alleviated renal pathological damage in CGN rats. Circ-ITCH negatively regulated miR-146a-5p expression by adsorbing miR-146a-5p (P<0.05), and miR-146a-5p inhibited YWHAG expression by binding to the 3'-UTR of YWHAG (P<0.05). Loss of YWHAG reversed the protective effect of upregulated circ-ITCH in CGN rats (all P<0.05). We conclude that circ-ITCH improves renal function and attenuates inflammation and renal injury in rats with CGN via the miR-146a-5p/YWHAG axis.


Asunto(s)
Glomerulonefritis , Inflamación , Riñón , MicroARNs , Ratas Sprague-Dawley , Ubiquitina-Proteína Ligasas , Animales , MicroARNs/genética , MicroARNs/metabolismo , Ratas , Masculino , Riñón/patología , Riñón/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Glomerulonefritis/metabolismo , Glomerulonefritis/genética , Inflamación/metabolismo , Inflamación/genética , ARN Circular/genética , ARN Circular/metabolismo , Enfermedad Crónica
19.
Int J Mol Sci ; 25(13)2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-39000117

RESUMEN

Diabetic cardiomyopathy (DCM) is a major determinant of mortality in diabetic populations, and the potential strategies are insufficient. Canagliflozin has emerged as a potential cardioprotective agent in diabetes, yet its underlying molecular mechanisms remain unclear. We employed a high-glucose challenge (60 mM for 48 h) in vitro to rat cardiomyocytes (H9C2), with or without canagliflozin treatment (20 µM). In vivo, male C57BL/6J mice were subjected to streptozotocin and a high-fat diet to induce diabetes, followed by canagliflozin administration (10, 30 mg·kg-1·d-1) for 12 weeks. Proteomics and echocardiography were used to assess the heart. Histopathological alterations were assessed by the use of Oil Red O and Masson's trichrome staining. Additionally, mitochondrial morphology and mitophagy were analyzed through biochemical and imaging techniques. A proteomic analysis highlighted alterations in mitochondrial and autophagy-related proteins after the treatment with canagliflozin. Diabetic conditions impaired mitochondrial respiration and ATP production, alongside decreasing the related expression of the PINK1-Parkin pathway. High-glucose conditions also reduced PGC-1α-TFAM signaling, which is responsible for mitochondrial biogenesis. Canagliflozin significantly alleviated cardiac dysfunction and improved mitochondrial function both in vitro and in vivo. Specifically, canagliflozin suppressed mitochondrial oxidative stress, enhancing ATP levels and sustaining mitochondrial respiratory capacity. It activated PINK1-Parkin-dependent mitophagy and improved mitochondrial function via increased phosphorylation of adenosine monophosphate-activated protein kinase (AMPK). Notably, PINK1 knockdown negated the beneficial effects of canagliflozin on mitochondrial integrity, underscoring the critical role of PINK1 in mediating these protective effects. Canagliflozin fosters PINK1-Parkin mitophagy and mitochondrial function, highlighting its potential as an effective treatment for DCM.


Asunto(s)
Canagliflozina , Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Ratones Endogámicos C57BL , Mitofagia , Proteínas Quinasas , Ubiquitina-Proteína Ligasas , Animales , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Mitofagia/efectos de los fármacos , Masculino , Ratones , Proteínas Quinasas/metabolismo , Proteínas Quinasas/genética , Ratas , Canagliflozina/farmacología , Canagliflozina/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Línea Celular , Transducción de Señal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos
20.
Life Sci Alliance ; 7(9)2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38969364

RESUMEN

The transmembrane E3 ligases RNF43 and ZNRF3 perform key tumour suppressor roles by inducing endocytosis of members of the Frizzled (FZD) family, the primary receptors for WNT. Loss-of-function mutations in RNF43 and ZNRF3 mediate FZD stabilisation and a WNT-hypersensitive growth state in various cancer types. Strikingly, RNF43 and ZNRF3 mutations are differentially distributed across cancer types, raising questions about their functional redundancy. Here, we compare the efficacy of RNF43 and ZNRF3 of targeting different FZDs for endocytosis. We find that RNF43 preferentially down-regulates FZD1/FZD5/FZD7, whereas ZNRF3 displays a preference towards FZD6. We show that the RNF43 transmembrane domain (TMD) is a key molecular determinant for inducing FZD5 endocytosis. Furthermore, a TMD swap between RNF43 and ZNRF3 re-directs their preference for FZD5 down-regulation. We conclude that RNF43 and ZNRF3 preferentially down-regulate specific FZDs, in part by a TMD-dependent mechanism. In accordance, tissue-specific expression patterns of FZD homologues correlate with the incidence of RNF43 or ZNRF3 cancer mutations in those tissues. Consequently, our data point to druggable vulnerabilities of specific FZD receptors in RNF43- or ZNRF3-mutant human cancers.


Asunto(s)
Endocitosis , Receptores Frizzled , Ubiquitina-Proteína Ligasas , Receptores Frizzled/metabolismo , Receptores Frizzled/genética , Humanos , Endocitosis/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Células HEK293 , Mutación , Vía de Señalización Wnt/genética , Regulación hacia Abajo/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...