Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Cell Mol Life Sci ; 81(1): 232, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38780644

RESUMEN

Ubiquitin-proteasome system dysfunction triggers α-synuclein aggregation, a hallmark of neurodegenerative diseases, such as Parkinson's disease (PD). However, the crosstalk between deubiquitinating enzyme (DUBs) and α-synuclein pathology remains unclear. In this study, we observed a decrease in the level of ubiquitin-specific protease 14 (USP14), a DUB, in the cerebrospinal fluid (CSF) of PD patients, particularly females. Moreover, CSF USP14 exhibited a dual correlation with α-synuclein in male and female PD patients. To investigate the impact of USP14 deficiency, we crossed USP14 heterozygous mouse (USP14+/-) with transgenic A53T PD mouse (A53T-Tg) or injected adeno-associated virus (AAV) carrying human α-synuclein (AAV-hα-Syn) in USP14+/- mice. We found that Usp14 deficiency improved the behavioral abnormities and pathological α-synuclein deposition in female A53T-Tg or AAV-hα-Syn mice. Additionally, Usp14 inactivation attenuates the pro-inflammatory response in female AAV-hα-Syn mice, whereas Usp14 inactivation demonstrated opposite effects in male AAV-hα-Syn mice. Mechanistically, the heterodimeric protein S100A8/A9 may be the downstream target of Usp14 deficiency in female mouse models of α-synucleinopathies. Furthermore, upregulated S100A8/A9 was responsible for α-synuclein degradation by autophagy and the suppression of the pro-inflammatory response in microglia after Usp14 knockdown. Consequently, our study suggests that USP14 could serve as a novel therapeutic target in PD.


Asunto(s)
Calgranulina A , Calgranulina B , Ratones Transgénicos , Enfermedad de Parkinson , Ubiquitina Tiolesterasa , alfa-Sinucleína , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Animales , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/deficiencia , Humanos , Ratones , Femenino , Masculino , Calgranulina B/metabolismo , Calgranulina B/genética , Calgranulina A/metabolismo , Calgranulina A/genética , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
2.
J Transl Med ; 22(1): 478, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769534

RESUMEN

BACKGROUND: Miscarriage is a frustrating complication of pregnancy that is common among women of reproductive age. Insufficient decidualization which not only impairs embryo implantation but disturbs fetomaternal immune-tolerance, has been widely regarded as a major cause of miscarriage; however, the underlying mechanisms resulting in decidual impairment are largely unknown. METHODS: With informed consent, decidual tissue from patients with spontaneous abortion or normal pregnant women was collected to detect the expression profile of UCHL1. Human endometrial stromal cells (HESCs) were used to explore the roles of UCHL1 in decidualization and dNK modulation, as well as the mechanisms involved. C57/BL6 female mice (7-10 weeks old) were used to construct pregnancy model or artificially induced decidualization model to evaluate the effect of UCHL1 on mice decidualization and pregnancy outcome. RESULTS: The Ubiquitin C-terminal hydrolase L1 (UCHL1), as a deubiquitinating enzyme, was significantly downregulated in decidua from patients with miscarriage, along with impaired decidualization and decreased dNKs. Blockage of UCHL1 led to insufficient decidualization and resultant decreased expression of cytokines CXCL12, IL-15, TGF-ß which were critical for generation of decidual NK cells (dNKs), whereas UCHL1 overexpression enhanced decidualization accompanied by increase in dNKs. Mechanistically, the promotion of UCHL1 on decidualization was dependent on its deubiquitinating activity, and intervention of UCHL1 inhibited the activation of JAK2/STAT3 signaling pathway, resulting in aberrant decidualization and decreased production of cytokines associated with dNKs modulation. Furthermore, we found that inhibition of UCHL1 also disrupted the decidualization in mice and eventually caused adverse pregnancy outcome. CONCLUSIONS: UCHL1 plays significant roles in decidualization and dNKs modulation during pregnancy in both humans and mice. Its deficiency indicates a poor pregnancy outcome due to defective decidualization, making UCHL1 a potential target for the diagnosis and treatment of miscarriage.


Asunto(s)
Aborto Espontáneo , Decidua , Células Asesinas Naturales , Ratones Endogámicos C57BL , Ubiquitina Tiolesterasa , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/deficiencia , Femenino , Decidua/metabolismo , Animales , Embarazo , Aborto Espontáneo/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/inmunología , Adulto , Ratones , Células del Estroma/metabolismo , Transducción de Señal
3.
Am J Hum Genet ; 109(2): 361-372, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35051358

RESUMEN

Nuclear deubiquitinase BAP1 (BRCA1-associated protein 1) is a core component of multiprotein complexes that promote transcription by reversing the ubiquitination of histone 2A (H2A). BAP1 is a tumor suppressor whose germline loss-of-function variants predispose to cancer. To our knowledge, there are very rare examples of different germline variants in the same gene causing either a neurodevelopmental disorder (NDD) or a tumor predisposition syndrome. Here, we report a series of 11 de novo germline heterozygous missense BAP1 variants associated with a rare syndromic NDD. Functional analysis showed that most of the variants cannot rescue the consequences of BAP1 inactivation, suggesting a loss-of-function mechanism. In T cells isolated from two affected children, H2A deubiquitination was impaired. In matching peripheral blood mononuclear cells, histone H3 K27 acetylation ChIP-seq indicated that these BAP1 variants induced genome-wide chromatin state alterations, with enrichment for regulatory regions surrounding genes of the ubiquitin-proteasome system (UPS). Altogether, these results define a clinical syndrome caused by rare germline missense BAP1 variants that alter chromatin remodeling through abnormal histone ubiquitination and lead to transcriptional dysregulation of developmental genes.


Asunto(s)
Proteína BRCA1/genética , Mutación de Línea Germinal , Mutación con Pérdida de Función , Mutación Missense , Trastornos del Neurodesarrollo/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Adolescente , Proteína BRCA1/inmunología , Niño , Preescolar , Cromatina/química , Cromatina/inmunología , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/inmunología , Familia , Femenino , Regulación de la Expresión Génica , Heterocigoto , Histonas/genética , Histonas/inmunología , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/inmunología , Humanos , Lactante , Masculino , Trastornos del Neurodesarrollo/inmunología , Trastornos del Neurodesarrollo/patología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/inmunología , Ubiquitina/genética , Ubiquitina/inmunología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación
4.
FASEB J ; 35(9): e21870, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34436790

RESUMEN

COVID-19 is often characterized by dysregulated inflammatory and immune responses. It has been shown that the Traditional Chinese Medicine formulation Qing-Fei-Pai-Du decoction (QFPDD) is effective in the treatment of the disease, especially for patients in the early stage. Our network pharmacology analyses indicated that many inflammation and immune-related molecules were the targets of the active components of QFPDD, which propelled us to examine the effects of the decoction on inflammation. We found in the present study that QFPDD effectively alleviated dextran sulfate sodium-induced intestinal inflammation in mice. It inhibited the production of pro-inflammatory cytokines IL-6 and TNFα, and promoted the expression of anti-inflammatory cytokine IL-10 by macrophagic cells. Further investigations found that QFPDD and one of its active components wogonoside markedly reduced LPS-stimulated phosphorylation of transcription factor ATF2, an important regulator of multiple cytokines expression. Our data revealed that both QFPDD and wogonoside decreased the half-life of ATF2 and promoted its proteasomal degradation. Of note, QFPDD and wogonoside down-regulated deubiquitinating enzyme USP14 along with inducing ATF2 degradation. Inhibition of USP14 with the small molecular inhibitor IU1 also led to the decrease of ATF2 in the cells, indicating that QFPDD and wogonoside may act through regulating USP14 to promote ATF2 degradation. To further assess the importance of ubiquitination in regulating ATF2, we generated mice that were intestinal-specific KLHL5 deficiency, a CUL3-interacting protein participating in substrate recognition of E3s. In these mice, QFPDD mitigated inflammatory reaction in the spleen, but not intestinal inflammation, suggesting CUL3-KLHL5 may function as an E3 for ATF2 degradation.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Flavanonas/farmacología , Glucósidos/farmacología , Inflamación/tratamiento farmacológico , Proteolisis/efectos de los fármacos , Ubiquitina Tiolesterasa/deficiencia , Animales , Línea Celular , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Proteínas Cullin/metabolismo , Citocinas/metabolismo , Sulfato de Dextran/farmacología , Sulfato de Dextran/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Flavanonas/uso terapéutico , Glucósidos/uso terapéutico , Inflamación/inducido químicamente , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Pirroles/farmacología , Pirrolidinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitinación
5.
Mol Ther ; 29(6): 2108-2120, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-33601053

RESUMEN

Genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated 9 (Cas9)-mediated loss-of-function screens are powerful tools for identifying genes responsible for diverse phenotypes. Here, we perturbed genes in melanoma cells to screen for genes involved in tumor escape from T cell-mediated killing. Multiple interferon gamma (IFNγ) signaling-related genes were enriched in melanoma cells resistant to T cell killing. In addition, deletion of the deubiquitinating protease ubiquitin specific peptidase 22 (USP22) in mouse melanoma (B16-OVA) cells decreased the efficacy of T cell-mediated killing, both in vitro and in vivo, while overexpression enhanced tumor-cell sensitivity to T (OT-I) cell-mediated killing. USP22 deficiency in both mouse and human melanoma cells showed impaired sensitivity to interferon pathway and USP22 was positively correlated with key molecules of interferon pathway in clinical melanoma samples. Mechanistically, USP22 may directly interact with signal transducer and activator of transcription 1 (STAT1), deubiquitinate it, and improve its stability in both human and mouse melanoma cells. Our findings identified a previously unknown function of USP22 and linked the loss of genes in tumor cells that are essential for escaping the effector function of CD8+ T cells during immunotherapy.


Asunto(s)
Interferón gamma/metabolismo , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ubiquitina Tiolesterasa/deficiencia , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Citotoxicidad Inmunológica/genética , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Melanoma , Ratones , Estabilidad Proteica , Ubiquitinación
6.
Eur J Immunol ; 51(1): 138-150, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32686110

RESUMEN

The IFN stimulated gene 15 (ISG15) encodes a 15-kDa ubiquitin-like protein, that is induced by type I IFNs and is conjugated to the bulk of newly synthesized polypeptides at the ribosome. ISG15 functions as an antiviral molecule possibly by being covalently conjugated to viral proteins and disturbing virus particle assembly. Here, we have investigated the effect of ISGylation on degradation and antigen presentation of viral and cellular proteins. ISGylation did not induce proteasomal degradation of bulk ISG15 target proteins neither after overexpressing ISG15 nor after induction by IFN-ß. The MHC class I cell surface expression of splenocytes derived from ISG15-deficient mice or mice lacking the catalytic activity of the major de-ISGylating enzyme USP18 was unaltered as compared to WT mice. Fusion of ubiquitin or FAT10 to the long-lived nucleoprotein (NP) of lymphocytic choriomeningitis virus accelerated the proteasomal degradation of NP while fusion to ISG15 did not detectably speed up NP degradation. Nevertheless, MHC-I restricted presentation of two epitopes of NP were markedly enhanced when it was fused to ISG15 similarly to fusion with ubiquitin or FAT10. Thus, we provide evidence that ISG15 can enhance the presentation of antigens on MHC-I most likely by promoting co-translational antigen processing.


Asunto(s)
Presentación de Antígeno/inmunología , Citocinas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Ubiquitinas/inmunología , Animales , Citocinas/deficiencia , Citocinas/genética , Citocinas/metabolismo , Células HEK293 , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/inmunología , Proteínas de la Nucleocápside/metabolismo , Complejo de la Endopetidasa Proteasomal/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Modificación Traduccional de las Proteínas/inmunología , Proteolisis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/inmunología , Ubiquitinas/deficiencia , Ubiquitinas/genética , Ubiquitinas/metabolismo
7.
Cell Mol Life Sci ; 78(6): 2929-2948, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33104844

RESUMEN

Macrophages are innate immune cells that contribute to classical immune functions and tissue homeostasis. Ubiquitin-specific protease 2 (USP2) controls cytokine production in macrophages, but its organ-specific roles are still unknown. In this study, we generated myeloid-selective Usp2 knockout (msUsp2KO) mice and specifically explored the roles of testicular macrophage-derived USP2 in reproduction. The msUsp2KO mice exhibited normal macrophage characteristics in various tissues. In the testis, macrophage Usp2 deficiency negligibly affected testicular macrophage subpopulations, spermatogenesis, and testicular organogenesis. However, frozen-thawed sperm derived from msUsp2KO mice exhibited reduced motility, capacitation, and hyperactivation. In addition, macrophage Usp2 ablation led to a decrease in the sperm population exhibiting high intracellular pH, calcium influx, and mitochondrial membrane potential. Interrupted pronuclei formation in eggs was observed when using frozen-thawed sperm from msUsp2KO mice for in vitro fertilization. Administration of granulocyte macrophage-colony stimulating factor (GM-CSF), whose expression was decreased in testicular macrophages derived from msUsp2KO mice, restored mitochondrial membrane potential and total sperm motility. Our observations demonstrate a distinct role of the deubiquitinating enzyme in organ-specific macrophages that directly affect sperm function.


Asunto(s)
Macrófagos/metabolismo , Capacitación Espermática/fisiología , Motilidad Espermática/fisiología , Espermatozoides/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Calcio/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Fertilización In Vitro , Congelación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Concentración de Iones de Hidrógeno , Macrófagos/citología , Macrófagos/inmunología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Motilidad Espermática/efectos de los fármacos , Testículo/anatomía & histología , Testículo/fisiología , Testosterona/metabolismo , Tretinoina/metabolismo , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/genética
8.
Br J Cancer ; 124(4): 817-830, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33214684

RESUMEN

BACKGROUND: Interferon (IFN) signalling pathways, a key element of the innate immune response, contribute to resistance to conventional chemotherapy, radiotherapy, and immunotherapy, and are often deregulated in cancer. The deubiquitylating enzyme USP18 is a major negative regulator of the IFN signalling cascade and is the predominant human protease that cleaves ISG15, a ubiquitin-like protein tightly regulated in the context of innate immunity, from its modified substrate proteins in vivo. METHODS: In this study, using advanced proteomic techniques, we have significantly expanded the USP18-dependent ISGylome and proteome in a chronic myeloid leukaemia (CML)-derived cell line. USP18-dependent effects were explored further in CML and colorectal carcinoma cellular models. RESULTS: Novel ISGylation targets were characterised that modulate the sensing of innate ligands, antigen presentation and secretion of cytokines. Consequently, CML USP18-deficient cells are more antigenic, driving increased activation of cytotoxic T lymphocytes (CTLs) and are more susceptible to irradiation. CONCLUSIONS: Our results provide strong evidence for USP18 in regulating antigenicity and radiosensitivity, highlighting its potential as a cancer target.


Asunto(s)
Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/inmunología , Citocinas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinas/metabolismo , Variación Antigénica , Línea Celular Tumoral , Neoplasias Colorrectales/radioterapia , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/radioterapia , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/genética
9.
Nature ; 588(7838): 479-484, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33177714

RESUMEN

Cholesterol is an essential lipid and its synthesis is nutritionally and energetically costly1,2. In mammals, cholesterol biosynthesis increases after feeding and is inhibited under fasting conditions3. However, the regulatory mechanisms of cholesterol biosynthesis at the fasting-feeding transition remain poorly understood. Here we show that the deubiquitylase ubiquitin-specific peptidase 20 (USP20) stabilizes HMG-CoA reductase (HMGCR), the rate-limiting enzyme in the cholesterol biosynthetic pathway, in the feeding state. The post-prandial increase in insulin and glucose concentration stimulates mTORC1 to phosphorylate USP20 at S132 and S134; USP20 is recruited to the HMGCR complex and antagonizes its degradation. The feeding-induced stabilization of HMGCR is abolished in mice with liver-specific Usp20 deletion and in USP20(S132A/S134A) knock-in mice. Genetic deletion or pharmacological inhibition of USP20 markedly decreases diet-induced body weight gain, reduces lipid levels in the serum and liver, improves insulin sensitivity and increases energy expenditure. These metabolic changes are reversed by expression of the constitutively stable HMGCR(K248R). This study reveals an unexpected regulatory axis from mTORC1 to HMGCR via USP20 phosphorylation and suggests that inhibitors of USP20 could be used to lower cholesterol levels to treat metabolic diseases including hyperlipidaemia, liver steatosis, obesity and diabetes.


Asunto(s)
Colesterol/biosíntesis , Ingestión de Alimentos/fisiología , Hidroximetilglutaril-CoA Reductasas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Hígado/metabolismo , Masculino , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Metabolismo/genética , Ratones , Ratones Endogámicos C57BL , Fosforilación , Fosfoserina/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/deficiencia , Ubiquitinación , Aumento de Peso
10.
PLoS One ; 15(11): e0241716, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33137160

RESUMEN

Ubiquitin C-terminal Hydrolase L1 (UCHL1) is a deubiquitinating enzyme that was originally identified in neurons. Our recent study showed that UCHL1 was expressed in C2C12 myoblast cells and mouse skeletal muscle. Here we report that in mouse skeletal muscle, UCHL1 is primarily expressed in oxidative muscle fibers. Skeletal muscle specific gene knockout (smKO) of UCHL1 in mice reduced oxidative activity in skeletal muscle measured by SDH staining. The in situ muscle contraction test revealed that gastrocnemius muscle from UCHL1 smKO mice was more prone to fatigue in response to the repetitive stimulation. This data suggests that UCHL1 plays a role in maintenance of muscle oxidative metabolism. Moreover, UCHL1 smKO caused a significant reduction in key proteins that are involved in mitochondrial oxidative phosphorylation in soleus muscles, suggesting that UCHL1 may be involved in regulation of mitochondrial content and function. Immunostaining showed the co-localization of UCHL1 and mitochondrial marker VDAC in skeletal muscle. Mitochondrial fractionation assay revealed that, although UCHL1 was primarily present in the cytosolic fraction, a low level of UCHL1 protein was present in mitochondrial fraction. The level of phosphorylation of AMPKα, a master regulator of mitochondrial biogenesis, were unchanged in UCHL1 smKO muscle. On the other hand, immunoprecipitation from soleus muscle sample indicated the interaction between UCHL1 and HSP60, a chaperon protein that is involved in mitochondrial protein transport. There was a trend of downregulation of HSP60 in UCHL1 smKO muscle. Overall, our data suggests UCHL1 is a novel regulator of mitochondrial function and oxidative activity in skeletal muscle.


Asunto(s)
Músculo Esquelético/metabolismo , Estrés Oxidativo , Ubiquitina Tiolesterasa/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Chaperonina 60/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Contracción Muscular , Mioblastos/citología , Mioblastos/metabolismo , Fosforilación Oxidativa , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/genética
11.
Biochem Biophys Res Commun ; 529(4): 1025-1032, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819560

RESUMEN

Liver kinase B1 (LKB1), a tumour suppressor, participates in many cellular processes, including cell survival, growth, apoptosis, transformation, and metabolism. Upon performing yeast two-hybrid screening, co-immunoprecipitation, and GST pull-down, we identified that BRCA1-associated protein 1 (BAP1), a deubiquitinase, interacts with LKB1. Immunoblotting was performed to examine the effect of BAP1 on the activation of 5' AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR), downstream of LKB1. The relationship between BAP1 deficiency and cancer cell proliferation was examined using cell survival assay and soft agar assay. qRT-PCR and oil red O staining were performed to evaluate lipid synthesis. Our findings reveal that BAP1 deubiquitinates LKB1, inhibits its degradation, and stabilises it, thereby affecting AMPK activation and downstream mTOR activity. BAP1 deficiency may enhance cellular proliferation as well as lipid synthesis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular , Humanos , Lípidos/biosíntesis , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Estabilidad Proteica , Especificidad por Sustrato , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Ubiquitina Tiolesterasa/deficiencia
12.
Nature ; 582(7812): 416-420, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32499641

RESUMEN

Regulatory T (Treg) cells are required to control immune responses and maintain homeostasis, but are a significant barrier to antitumour immunity1. Conversely, Treg instability, characterized by loss of the master transcription factor Foxp3 and acquisition of proinflammatory properties2, can promote autoimmunity and/or facilitate more effective tumour immunity3,4. A comprehensive understanding of the pathways that regulate Foxp3 could lead to more effective Treg therapies for autoimmune disease and cancer. The availability of new functional genetic tools has enabled the possibility of systematic dissection of the gene regulatory programs that modulate Foxp3 expression. Here we developed a CRISPR-based pooled screening platform for phenotypes in primary mouse Treg cells and applied this technology to perform a targeted loss-of-function screen of around 500 nuclear factors to identify gene regulatory programs that promote or disrupt Foxp3 expression. We identified several modulators of Foxp3 expression, including ubiquitin-specific peptidase 22 (Usp22) and ring finger protein 20 (Rnf20). Usp22, a member of the deubiquitination module of the SAGA chromatin-modifying complex, was revealed to be a positive regulator that stabilized Foxp3 expression; whereas the screen suggested that Rnf20, an E3 ubiquitin ligase, can serve as a negative regulator of Foxp3. Treg-specific ablation of Usp22 in mice reduced Foxp3 protein levels and caused defects in their suppressive function that led to spontaneous autoimmunity but protected against tumour growth in multiple cancer models. Foxp3 destabilization in Usp22-deficient Treg cells could be rescued by ablation of Rnf20, revealing a reciprocal ubiquitin switch in Treg cells. These results reveal previously unknown modulators of Foxp3 and demonstrate a screening method that can be broadly applied to discover new targets for Treg immunotherapies for cancer and autoimmune disease.


Asunto(s)
Sistemas CRISPR-Cas , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Autoinmunidad/inmunología , Células Cultivadas , Factores de Transcripción Forkhead/biosíntesis , Edición Génica , Regulación de la Expresión Génica , Humanos , Inmunoterapia , Masculino , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/prevención & control , Estabilidad Proteica , Reproducibilidad de los Resultados , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/metabolismo
13.
Histochem Cell Biol ; 154(3): 275-286, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32451617

RESUMEN

UCHL1 is expressed specifically in the brain and gonads of almost all studied model organisms including Drosophila, zebrafish, amphibians, and mammals, suggesting a high degree of evolutionary conservation in its structure and function. Although UCHL1 has been involved in spermatogenesis in mice, its specific expression in mammal placenta remains elusive. Our previous work has revealed that UCHL1 is highly expressed in oocytes, and has been involved in mouse ovarian follicular development. Here, we further examined UCHL1 expression change in endometria during early natural pregnancy, with different stages of the estrous cycle and pseudopregnancy as control. The UCHL1 gene deletion model showed that UCHL1 protein is associated with endometrial development, and its deletion leads to infertility. Notably, we demonstrate evidence showing the distinct expression pattern of UCHL1: weak expression over the uterine endometria, strong expression in decidualized stromal cells at the implantation site with a peak at pregnancy D6, and a shift with primary decidualization to secondary decidualized zones. Using the delayed implantation, the delayed implantation activation, and the artificial decidualization models, we have demonstrated that strong expression of UCHL1 occurred in response to decidualization and estrogen stimulation. These observations suggest that during the early proliferation and differentiation of mouse uterine decidua, UCHL1 expression is up-regulated, and formed an unique intracellular distribution mode. Therefore, we proposed that UCHL1 is involved in decidualization, and possibly in response to estrogen regulation.


Asunto(s)
Decidua/metabolismo , Estrógenos/metabolismo , Ubiquitina Tiolesterasa/genética , Útero/metabolismo , Animales , Decidua/citología , Implantación del Embrión/genética , Femenino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Embarazo , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/metabolismo , Útero/citología
14.
J Exp Med ; 217(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32130408

RESUMEN

USP22 is a cytoplasmic and nuclear deubiquitinating enzyme, and the functions of cytoplasmic USP22 are unclear. Here, we discovered that cytoplasmic USP22 promoted nuclear translocation of IRF3 by deubiquitianting and stabilizing KPNA2 after viral infection. Viral infection induced USP22-IRF3 association in the cytoplasm in a KPNA2-depedent manner, and knockdown or knockout of USP22 or KPNA2 impaired IRF3 nuclear translocation and expression of downstream genes after viral infection. Consistently, Cre-ER Usp22fl/fl or Lyz2-Cre Usp22fl/fl mice produced decreased levels of type I IFNs after viral infection and exhibited increased susceptibility to lethal viral infection compared with the respective control littermates. Mechanistically, USP22 deubiquitinated and stabilized KPNA2 after viral infection to facilitate efficient nuclear translocation of IRF3. Reconstitution of KPNA2 into USP22 knockout cells restored virus-triggered nuclear translocation of IRF3 and cellular antiviral responses. These findings define a previously unknown function of cytoplasmic USP22 and establish a mechanistic link between USP22 and IRF3 nuclear translocation that expands potential therapeutic strategies for infectious diseases.


Asunto(s)
Antivirales/metabolismo , Núcleo Celular/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación , alfa Carioferinas/metabolismo , Animales , Línea Celular , Susceptibilidad a Enfermedades , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Interferón Tipo I/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Estabilidad Proteica , Transporte de Proteínas , Transducción de Señal , Ubiquitina Tiolesterasa/deficiencia , Virosis/metabolismo
15.
J Exp Med ; 217(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32092142

RESUMEN

Type I interferonopathies are monogenic disorders characterized by enhanced type I interferon (IFN-I) cytokine activity. Inherited USP18 and ISG15 deficiencies underlie type I interferonopathies by preventing the regulation of late responses to IFN-I. Specifically, USP18, being stabilized by ISG15, sterically hinders JAK1 from binding to the IFNAR2 subunit of the IFN-I receptor. We report an infant who died of autoinflammation due to a homozygous missense mutation (R148Q) in STAT2. The variant is a gain of function (GOF) for induction of the late, but not early, response to IFN-I. Surprisingly, the mutation does not enhance the intrinsic activity of the STAT2-containing transcriptional complex responsible for IFN-I-stimulated gene induction. Rather, the STAT2 R148Q variant is a GOF because it fails to appropriately traffic USP18 to IFNAR2, thereby preventing USP18 from negatively regulating responses to IFN-I. Homozygosity for STAT2 R148Q represents a novel molecular and clinical phenocopy of inherited USP18 deficiency, which, together with inherited ISG15 deficiency, defines a group of type I interferonopathies characterized by an impaired regulation of late cellular responses to IFN-I.


Asunto(s)
Mutación con Ganancia de Función/genética , Interferón Tipo I/metabolismo , Factor de Transcripción STAT2/genética , Ubiquitina Tiolesterasa/deficiencia , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Femenino , Regulación de la Expresión Génica , Homocigoto , Humanos , Recién Nacido , Masculino , Linaje , Fenotipo , Dominios Proteicos , Factor de Transcripción STAT2/química , Ubiquitina Tiolesterasa/genética , Secuenciación del Exoma
16.
J Exp Med ; 217(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32069354

RESUMEN

The ubiquitin pathway has been shown to regulate iNKT cell immunity, but the deubiquitinase involved in this process has not been identified. Herein we found that ubiquitin-specific peptidase 22 (USP22) is highly expressed in iNKT cells during their early developmental stage 1. USP22 deficiency blocked the transition from stage 1 to 2 during iNKT cell development in a cell-intrinsic manner. USP22 suppression also diminishes iNKT17 and iNKT1 differentiation but favors iNKT2 polarization without altering conventional T cell activation and differentiation. USP22 interacts with the Mediator complex subunit 1 (MED1), a transcription coactivator involved in iNKT cell development. Interestingly, while interacting with MED1, USP22 does not function as a deubiquitinase to suppress MED1 ubiquitination for its stabilization. Instead, USP22 enhances MED1 functions for IL-2Rß and T-bet gene expression through deubiquitinating histone H2A but not H2B monoubiquitination. Therefore, our study revealed USP22-mediated histone H2A deubiquitination fine-tunes MED1 transcriptional activation as a previously unappreciated molecular mechanism to control iNKT development and functions.


Asunto(s)
Histonas/metabolismo , Inmunidad , Subunidad 1 del Complejo Mediador/metabolismo , Células T Asesinas Naturales/inmunología , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación , Animales , Linfocitos T CD4-Positivos/citología , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Ubiquitina Tiolesterasa/deficiencia
17.
N Engl J Med ; 382(3): 256-265, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31940699

RESUMEN

Deficiency of ubiquitin-specific peptidase 18 (USP18) is a severe type I interferonopathy. USP18 down-regulates type I interferon signaling by blocking the access of Janus-associated kinase 1 (JAK1) to the type I interferon receptor. The absence of USP18 results in unmitigated interferon-mediated inflammation and is lethal during the perinatal period. We describe a neonate who presented with hydrocephalus, necrotizing cellulitis, systemic inflammation, and respiratory failure. Exome sequencing identified a homozygous mutation at an essential splice site on USP18. The encoded protein was expressed but devoid of negative regulatory ability. Treatment with ruxolitinib was followed by a prompt and sustained recovery. (Funded by King Saud University and others.).


Asunto(s)
Enfermedades Autoinflamatorias Hereditarias/tratamiento farmacológico , Interferones/metabolismo , Interleucinas/metabolismo , Janus Quinasa 1/antagonistas & inhibidores , Inhibidores de las Cinasas Janus/uso terapéutico , Mutación con Pérdida de Función , Pirazoles/uso terapéutico , Ubiquitina Tiolesterasa/deficiencia , Homocigoto , Humanos , Hidrocefalia/genética , Recién Nacido , Masculino , Nitrilos , Pirimidinas , Receptores de Interferón/metabolismo , Inducción de Remisión , Choque Séptico/genética , Transducción de Señal/genética , Ubiquitina Tiolesterasa/genética , Secuenciación del Exoma
18.
Cell Death Differ ; 27(4): 1328-1340, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31527800

RESUMEN

USP22, the deubiquitinating subunit of the SAGA transcriptional cofactor complex, is a member of an 11-gene "death-from-cancer" signature. USP22 has been considered an attractive therapeutic target since high levels of its expression were associated with distant metastasis, poor survival, and high recurrence rates in a wide variety of solid tumors, including colorectal cancer (CRC). We sought to investigate the role of Usp22 during tumorigenesis in vivo using a mouse model for intestinal carcinogenesis with a tissue-specific Usp22 ablation. In addition, we assessed the effects of USP22 depletion in human CRC cells on tumorigenic potential and identified underlying molecular mechanisms. For the first time, we report that USP22 has an unexpected tumor-suppressive function in vivo. Intriguingly, intestine-specific Usp22 deletion exacerbated the tumor phenotype caused by Apc mutation, resulting in significantly decreased survival and higher intestinal tumor incidence. Accordingly, human CRC cells showed increased tumorigenic properties upon USP22 reduction in vitro and in vivo and induced gene expression signatures associated with an unfavorable outcome in CRC patients. Notably, USP22 loss resulted in increased mTOR activity with the tumorigenic properties elicited by the loss of USP22 being reversible by mTOR inhibitor treatment in vitro and in vivo. Here, we demonstrate that USP22 can exert tumor-suppressive functions in CRC where its loss increases CRC burden by modulating mTOR activity. Importantly, our data uncover a tumor- and context-specific role of USP22, suggesting that USP22 expression could serve as a marker for therapeutic stratification of cancer patients.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/patología , Eliminación de Gen , Células HCT116 , Humanos , Ratones Endogámicos C57BL , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ubiquitina Tiolesterasa/deficiencia
19.
Cell Death Dis ; 10(12): 911, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801945

RESUMEN

As a member of the 11-gene "death-from-cancer" gene expression signature, overexpression of the Ubiquitin-Specific Protease 22 (USP22) was associated with poor prognosis in various human malignancies. To investigate the function of USP22 in cancer development and progression, we sought to detect common USP22-dependent molecular mechanisms in human colorectal and breast cancer cell lines. We performed mRNA-seq to compare gene expression profiles of various colorectal (SW837, SW480, HCT116) and mammary (HCC1954 and MCF10A) cell lines upon siRNA-mediated knockdown of USP22. Intriguingly, while USP22 depletion had highly heterogeneous effects across the cell lines, all cell lines displayed a common reduction in the expression of Heat Shock Protein 90 Alpha Family Class B Member 1 (HSP90AB1). The downregulation of HSP90AB1 was confirmed at the protein level in these cell lines as well as in colorectal and mammary tumors in mice with tissue-specific Usp22 deletions. Mechanistically, we detected a significant reduction of H3K9ac on the HSP90AB1 gene in USP22-deficient cells. Interestingly, USP22-deficient cells displayed a high dependence on HSP90AB1 expression and diminishing HSP90 activity further using the HSP90 inhibitor Ganetespib resulted in increased therapeutic vulnerability in both colorectal and breast cancer cells in vitro. Accordingly, subcutaneously transplanted CRC cells deficient in USP22 expression displayed increased sensitivity towards Ganetespib treatment in vivo. Together, we discovered that HSP90AB1 is USP22-dependent and that cooperative targeting of USP22 and HSP90 may provide an effective approach to the treatment of colorectal and breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular Tumoral , Regulación hacia Abajo/genética , Femenino , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones SCID , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Ubiquitina Tiolesterasa/deficiencia , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cell Commun Signal ; 17(1): 167, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31842906

RESUMEN

BACKGROUND: Loss of monoubiquitination of histone H2B (H2Bub1) was found to be associated with poor differentiation, cancer stemness, and enhanced malignancy of non-small cell lung cancer (NSCLC). Herein, we investigated the biological significance and therapeutic implications of ubiquitin-specific protease 22 (USP22), an H2Bub1 deubiquitinase, in non-small cell lung cancer (NSCLC). METHODS: USP22 expression and its clinical relevance were assessed in NSCLC patients. The effects of USP22 knockout on sensitivity to cisplatin and irradiation, and growth, metastasis of NSCLC xenografts, and survival of cancer-bearing mice were investigated. The underlying mechanisms of targeting USP22 were explored. RESULTS: Overexpression of USP22 was observed in 49.0% (99/202) of NSCLC tissues; higher USP22 immunostaining was found to be associated with enhanced angiogenesis and recurrence of NSCLC. Notably, USP22 knockout dramatically suppressed in vitro proliferation, colony formation; and angiogenesis, growth, metastasis of A549 and H1299 in mouse xenograft model, and significantly prolonged survival of metastatic cancer-bearing mice. Furthermore, USP22 knockout significantly impaired non-homologous DNA damage repair capacity, enhanced cisplatin and irradiation-induced apoptosis in these cells. In terms of underlying mechanisms, RNA sequencing and gene ontology enrichment analysis demonstrated that USP22 knockout significantly suppressed angiogenesis, proliferation, EMT, RAS, c-Myc pathways, concurrently enhanced oxidative phosphorylation and tight junction pathways in A549 and H1299 NSCLC cells. Immunoblot analysis confirmed that USP22 knockout upregulated E-cadherin, p16; reduced ALDH1A3, Cyclin E1, c-Myc, and attenuated activation of AKT and ERK pathways in these cells. CONCLUSIONS: Our findings suggest USP22 plays critical roles in the malignancy and progression of NSCLC and provide rationales for targeting USP22, which induces broad anti-cancer activities, as a novel therapeutic strategy for NSCLC patient.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neovascularización Patológica/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/terapia , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Reparación del ADN , ADN de Neoplasias/análisis , Modelos Animales de Enfermedad , Femenino , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/secundario , Neoplasias Pulmonares/terapia , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Metástasis de la Neoplasia , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...