Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 34(11): 14750-14767, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32910521

RESUMEN

Small ubiquitin-like modifier (SUMO1-3) conjugation (SUMOylation), a posttranslational modification, modulates almost all major cellular processes. Mounting evidence indicates that SUMOylation plays a crucial role in maintaining and regulating neural function, and importantly its dysfunction is implicated in cognitive impairment in humans. We have previously shown that simultaneously silencing SUMO1-3 expression in neurons negatively affects cognitive function. However, the roles of the individual SUMOs in modulating cognition and the mechanisms that link SUMOylation to cognitive processes remain unknown. To address these questions, in this study, we have focused on SUMO2 and generated a new conditional Sumo2 knockout mouse line. We found that conditional deletion of Sumo2 predominantly in forebrain neurons resulted in marked impairments in various cognitive tests, including episodic and fear memory. Our data further suggest that these abnormalities are attributable neither to constitutive changes in gene expression nor to alterations in neuronal morphology, but they involve impairment in dynamic SUMOylation processes associated with synaptic plasticity. Finally, we provide evidence that dysfunction on hippocampal-based cognitive tasks was associated with a significant deficit in the maintenance of hippocampal long-term potentiation in Sumo2 knockout mice. Collectively, these data demonstrate that protein conjugation by SUMO2 is critically involved in cognitive processes.


Asunto(s)
Memoria , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Cognición , Femenino , Hipocampo/metabolismo , Hipocampo/fisiología , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Prosencéfalo/metabolismo , Prosencéfalo/fisiología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética
2.
Dev Dyn ; 249(2): 237-244, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31625212

RESUMEN

Small ubiquitin-related modifier (SUMO)-specific protease 2 (SENP2) is essential for the development of healthy placenta. The loss of SENP2 causes severe placental deficiencies and leads to embryonic death that is associated with heart and brain deformities. However, tissue-specific disruption of SENP2 demonstrates its dispensable role in embryogenesis and the embryonic defects are secondary to placental insufficiency. SENP2 regulates SUMO1 modification of Mdm2, which controls p53 activities critical for trophoblast cell proliferation and differentiation. Here we use genetic analyses to examine the involvement of SUMO2 and SUMO3 for SENP2-mediated placentation. The results indicate that hyper-SUMOylation caused by SENP2 deficiency can be compensated by reducing the level of SUMO modifiers. The placental deficiencies caused by the loss of SENP2 can be alleviated by the inactivation of gene encoding SUMO2 or SUMO3. Our findings demonstrate that SENP2 genetically interacts with SUMO2 and SUMO3 pivotal for the development of three major trophoblast layers. The alleviation of placental defects in the SENP2 knockouts further leads to the proper formation of the heart structures, including atrioventricular cushion and myocardium. SUMO2 and SUMO3 modifications regulate placentation and organogenesis mediated by SENP2.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Embrión de Mamíferos/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Animales , Cisteína Endopeptidasas/genética , Femenino , Ratones , Ratones Noqueados , Insuficiencia Placentaria/genética , Insuficiencia Placentaria/metabolismo , Embarazo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Trofoblastos/metabolismo , Ubiquitinas/genética
3.
Stroke ; 51(5): 1570-1577, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32212900

RESUMEN

Background and Purpose- Ischemic stroke impairs endoplasmic reticulum (ER) function, causes ER stress, and activates the unfolded protein response. The unfolded protein response consists of 3 branches controlled by ER stress sensor proteins, which include PERK (protein kinase RNA-like ER kinase). Activated PERK phosphorylates eIF2α (eukaryotic initiation factor 2 alpha), resulting in inhibition of global protein synthesis. Here, we aimed to clarify the role of the PERK unfolded protein response branch in stroke. Methods- Neuron-specific and tamoxifen-inducible PERK conditional knockout (cKO) mice were generated by cross-breeding Camk2a-CreERT2 with Perkf/f mice. Transient middle cerebral artery occlusion was used to induce stroke. Short- and long-term stroke outcomes were evaluated. Protein synthesis in the brain was assessed using a surface-sensing-of-translation approach. Results- After tamoxifen-induced deletion of Perk in forebrain neurons was confirmed in PERK-cKO mice, PERK-cKO and control mice were subjected to transient middle cerebral artery occlusion and 3 days or 3 weeks recovery. PERK-cKO mice had larger infarcts and worse neurological outcomes compared with control mice, suggesting that PERK-induced eIF2α phosphorylation and subsequent suppression of translation protects neurons from ischemic stress. Indeed, better stroke outcomes were observed in PERK-cKO mice that received postischemic treatment with salubrinal, which can restore the ischemia-induced increase in phosphorylated eIF2α in these mice. Finally, our data showed that post-treatment with salubrinal improved functional recovery after stroke. Conclusions- Here, we presented the first evidence that postischemic suppression of translation induced by PERK activation promotes recovery of neurological function after stroke. This confirms and further extends our previous observations that recovery of ER function impaired by ischemic stress critically contributes to stroke outcome. Therefore, future research should include strategies to improve stroke outcome by targeting unfolded protein response branches to restore protein homeostasis in neurons.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Neuronas/metabolismo , Neuroprotección/genética , Respuesta de Proteína Desplegada/genética , eIF-2 Quinasa/genética , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Cinamatos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Infarto de la Arteria Cerebral Media/fisiopatología , Ratones , Ratones Noqueados , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología , Tiourea/análogos & derivados , Tiourea/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
4.
Lab Invest ; 98(6): 799-813, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29472640

RESUMEN

The intestinal epithelium constitutes a crucial defense to the potentially life-threatening effects of gut microbiota. However, due to a complex underlying vasculature, hypoperfusion and resultant tissue ischemia pose a particular risk to function and integrity of the epithelium. The small ubiquitin-like modifier (SUMO) conjugation pathway critically regulates adaptive responses to metabolic stress and is of particular significance in the gut, as inducible knockout of the SUMO-conjugating enzyme Ubc9 results in rapid intestinal epithelial disintegration. Here we analyzed the pattern of individual SUMO isoforms in intestinal epithelium and investigated their roles in intestinal ischemia/reperfusion (I/R) damage. Immunostaining revealed that epithelial SUMO2/3 expression was almost exclusively limited to crypt epithelial nuclei in unchallenged mice. However, intestinal I/R or overexpression of Ubc9 caused a remarkable enhancement of epithelial SUMO2/3 staining along the crypt-villus axis. Unexpectedly, a similar pattern was found in SUMO1 knockout mice. Ubc9 transgenic mice, but also SUMO1 knockout mice were protected from I/R injury as evidenced by better preserved barrier function and blunted inflammatory responses. PCR array analysis of microdissected villus-tip epithelia revealed a specific epithelial contribution to reduced inflammatory responses in Ubc9 transgenic mice, as key chemotactic signaling molecules such as IL17A were significantly downregulated. Together, our data indicate a critical role particularly of the SUMO2/3 isoforms in modulating responses to I/R and provide the first evidence that SUMO1 deletion activates a compensatory process that protects from ischemic damage.


Asunto(s)
Mucosa Intestinal/irrigación sanguínea , Daño por Reperfusión/prevención & control , Proteína SUMO-1/fisiología , Enzimas Ubiquitina-Conjugadoras/fisiología , Animales , Quimiocinas/análisis , Mucosa Intestinal/química , Captura por Microdisección con Láser , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína SUMO-1/deficiencia , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/análisis , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/fisiología , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitinas/análisis , Ubiquitinas/fisiología
5.
Stroke ; 48(6): 1646-1654, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28487326

RESUMEN

BACKGROUND AND PURPOSE: Impaired protein homeostasis induced by endoplasmic reticulum dysfunction is a key feature of a variety of age-related brain diseases including stroke. To restore endoplasmic reticulum function impaired by stress, the unfolded protein response is activated. A key unfolded protein response prosurvival pathway is controlled by the endoplasmic reticulum stress sensor (inositol-requiring enzyme-1), XBP1 (downstream X-box-binding protein-1), and O-GlcNAc (O-linked ß-N-acetylglucosamine) modification of proteins (O-GlcNAcylation). Stroke impairs endoplasmic reticulum function, which activates unfolded protein response. The rationale of this study was to explore the potentials of the IRE1/XBP1/O-GlcNAc axis as a target for neuroprotection in ischemic stroke. METHODS: Mice with Xbp1 loss and gain of function in neurons were generated. Stroke was induced by transient or permanent occlusion of the middle cerebral artery in young and aged mice. Thiamet-G was used to increase O-GlcNAcylation. RESULTS: Deletion of Xbp1 worsened outcome after transient and permanent middle cerebral artery occlusion. After stroke, O-GlcNAcylation was activated in neurons of the stroke penumbra in young mice, which was largely Xbp1 dependent. This activation of O-GlcNAcylation was impaired in aged mice. Pharmacological increase of O-GlcNAcylation before or after stroke improved outcome in both young and aged mice. CONCLUSIONS: Our study indicates a critical role for the IRE1/XBP1 unfolded protein response branch in stroke outcome. O-GlcNAcylation is a prosurvival pathway that is activated in the stroke penumbra in young mice but impaired in aged mice. Boosting prosurvival pathways to counterbalance the age-related decline in the brain's self-healing capacity could be a promising strategy to improve ischemic stroke outcome in aged brains.


Asunto(s)
Acetilglucosamina/metabolismo , Isquemia Encefálica/metabolismo , Proteínas de la Membrana/metabolismo , Neuroprotección/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Piranos/farmacología , Accidente Cerebrovascular/metabolismo , Tiazoles/farmacología , Respuesta de Proteína Desplegada/fisiología , Proteína 1 de Unión a la X-Box/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pliegue de Proteína
6.
EMBO Rep ; 15(8): 878-85, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24891386

RESUMEN

Small ubiquitin-like modifier (SUMO1-3) conjugation plays a critical role in embryogenesis. Embryos deficient in the SUMO-conjugating enzyme Ubc9 die at the early postimplantation stage. Sumo1(-/-) mice are viable, as SUMO2/3 can compensate for most SUMO1 functions. To uncover the role of SUMO2/3 in embryogenesis, we generated Sumo2- and Sumo3-null mutant mice. Here, we report that Sumo3(-/-) mice were viable, while Sumo2(-/-) embryos exhibited severe developmental delay and died at approximately embryonic day 10.5 (E10.5). We also provide evidence that SUMO2 is the predominantly expressed SUMO isoform. Furthermore, although Sumo2(+/-) and Sumo2(+/-);Sumo3(+/-) mice lacked any overt phenotype, only 2 Sumo2(+/-);Sumo3(-/-) mice were found at birth in 35 litters after crossing Sumo2(+/-);Sumo3(+/-) with Sumo3(-/-) mice, and these rare mice were considerably smaller than littermates of the other genotypes. Thus, our findings suggest that expression levels and not functional differences between SUMO2 and SUMO3 are critical for normal embryogenesis.


Asunto(s)
Desarrollo Embrionario , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Ubiquitinas/genética , Animales , Femenino , Expresión Génica , Genes Esenciales , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitinas/metabolismo
7.
Proteomics ; 15(5-6): 1181-91, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25236368

RESUMEN

Small ubiquitin-like modifier (SUMO1-3) conjugation is a posttranslational protein modification whereby SUMOs are conjugated to lysine residues of target proteins. SUMO conjugation can alter the activity, stability, and function of target proteins, and thereby modulate almost all major cellular pathways. Many diseases are associated with SUMO conjugation, including heart failure, arthritis, cancer, degenerative diseases, and brain ischemia/stroke. It is, therefore, of major interest to characterize the SUMO-modified proteome regulated by these disorders. SUMO proteomics analysis is hampered by low levels of SUMOylated proteins. Several strategies have, therefore, been developed to enrich SUMOylated proteins from cell/tissue extracts. These include proteomics analysis on cells expressing epitope-tagged SUMO isoforms, use of monoclonal SUMO antibodies for immunoprecipitation and epitope-specific peptides for elution, and affinity purification with peptides containing SUMO interaction motifs to specifically enrich polySUMOylated proteins. Recently, two mouse models were generated and characterized that express tagged SUMO isoforms, and allow purification of SUMOylated proteins from complex organ extracts. Ultimately, these new analytical tools will help to decipher the SUMO-modified proteome regulated by various human diseases, and thereby, identify new targets for preventive and therapeutic purposes.


Asunto(s)
Cardiopatías/metabolismo , Neoplasias/metabolismo , Proteoma , Proteómica/métodos , Proteína SUMO-1 , Sumoilación/fisiología , Animales , Biomarcadores , Humanos , Ratones , Enfermedades Neurodegenerativas/metabolismo , Proteoma/análisis , Proteoma/metabolismo , Proteoma/fisiología , Proteína SUMO-1/análisis , Proteína SUMO-1/metabolismo , Proteína SUMO-1/fisiología , Accidente Cerebrovascular/metabolismo
8.
J Proteome Res ; 13(9): 3905-18, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25072996

RESUMEN

SUMOylation is an essential posttranslational modification and regulates many cellular processes. Dysregulation of SUMOylation plays a critical role in metastasis, yet how its perturbation affects this lethal process of cancer is not well understood. We found that SUMO-2/3 modification is greatly up-regulated in metastatic breast cancer cells compared with nonmetastatic control cells. To identify proteins differentially modified by SUMO-2/3 between metastatic and nonmetastatic cells, we established a method in which endogenous SUMO-2/3 conjugates are labeled by stable isotope labeling by amino acids in cell culture (SILAC), immunopurified by SUMO-2/3 monoclonal antibodies and epitope-peptide elution, and analyzed by quantitative mass spectrometry. We identified 66 putative SUMO-2/3-conjugated proteins, of which 15 proteins show a significant increase/decrease in SUMO-2/3 modification in metastatic cells. Targets with altered SUMOylation are involved in cell cycle, migration, inflammation, glycolysis, gene expression, and SUMO/ubiquitin pathways, suggesting that perturbations of SUMO-2/3 modification might contribute to metastasis by affecting these processes. Consistent with this, up-regulation of PML SUMO-2/3 modification corresponds to an increased number of PML nuclear bodies (PML-NBs) in metastatic cells, whereas up-regulation of global SUMO-2/3 modification promotes 3D cell migration. Our findings provide a foundation for further investigating the effects of SUMOylation on breast cancer progression and metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteómica/métodos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Neoplasias de la Mama/química , Línea Celular Tumoral , Progresión de la Enfermedad , Drosophila , Femenino , Humanos , Espectrometría de Masas , Ratones , Procesos Neoplásicos , Procesamiento Proteico-Postraduccional , Alineación de Secuencia , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/análisis
9.
Stroke ; 45(4): 1115-22, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24569813

RESUMEN

BACKGROUND AND PURPOSE: Small ubiquitin-like modifier (SUMO) conjugation is a post-translational modification associated with many human diseases. Characterization of the SUMO-modified proteome is pivotal to define the mechanistic link between SUMO conjugation and such diseases. This is particularly evident for SUMO2/3 conjugation, which is massively activated after brain ischemia/stroke, and is believed to be a protective response. The purpose of this study was to perform a comprehensive analysis of the SUMO3-modified proteome regulated by brain ischemia using a novel SUMO transgenic mouse. METHODS: To enable SUMO proteomics analysis in vivo, we generated transgenic mice conditionally expressing tagged SUMO1-3 paralogues. Transgenic mice were subjected to 10 minutes forebrain ischemia and 1 hour of reperfusion. SUMO3-conjugated proteins were enriched by anti-FLAG affinity purification and analyzed by liquid chromatography-tandem mass spectrometry. RESULTS: Characterization of SUMO transgenic mice demonstrated that all 3 tagged SUMO paralogues were functionally active, and expression of exogenous SUMOs did not modify the endogenous SUMOylation machinery. Proteomics analysis identified 112 putative SUMO3 substrates of which 91 candidates were more abundant in the ischemia group than the sham group. Data analysis revealed processes/pathways with putative neuroprotective functions, including glucocorticoid receptor signaling, RNA processing, and SUMOylation-dependent ubiquitin conjugation. CONCLUSIONS: The identified proteins/pathways modulated by SUMOylation could be the key to understand the mechanisms linking SUMOylation to neuroprotection, and thus provide new promising targets for therapeutic interventions. The new transgenic mouse will be an invaluable platform for analyzing the SUMO-modified proteome in models of human disorders and thereby help to mechanistically link SUMOylation to the pathological processes.


Asunto(s)
Isquemia Encefálica/fisiopatología , Ataque Isquémico Transitorio/fisiopatología , Accidente Cerebrovascular/fisiopatología , Ubiquitinas/genética , Ubiquitinas/metabolismo , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Ataque Isquémico Transitorio/genética , Ataque Isquémico Transitorio/metabolismo , Espectrometría de Masas , Ratones , Ratones Transgénicos , Proteoma/genética , Proteoma/metabolismo , Proteómica , Procesamiento Postranscripcional del ARN , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo
10.
J Psychiatry Neurosci ; 39(4): 259-66, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24690371

RESUMEN

BACKGROUND: Growing evidence suggests that small ubiquitin-like modifier (SUMO) conjugation plays a key role in brain plasticity by modulating activity-dependent synaptic transmission. However, these observations are based largely on cell culture experiments. We hypothesized that episodic and fear memories would be affected by silencing SUMO1-3 expression. METHODS: To investigate the role of SUMO conjugation in neuronal functioning in vivo, we generated a novel Sumo transgenic mouse model in which a Thy1 promoter drives expression of 3 distinct microRNAs to silence Sumo1-3 expression, specifically in neurons. Wild-type and Sumo1-3 knockdown mice were subjected to a battery of behavioural tests to elucidate whether Sumoylation is involved in episodic and emotional memory. RESULTS: Expression of Sumo1-3 microRNAs and the corresponding silencing of Sumo expression were particularly pronounced in hippocampal, amygdala and layer V cerebral cortex neurons. The Sumo knockdown mice displayed anxiety-like responses and were impaired in episodic memory processes, contextual and cued fear conditioning and fear-potentiated startle. LIMITATIONS: Since expression of Sumo1-3 was silenced in this mouse model, we need to verify in future studies which of the SUMO paralogues play the pivotal role in episodic and emotional memory. CONCLUSION: Our results indicate that a functional SUMO conjugation pathway is essential for emotionality and cognition. This novel Sumo knockdown mouse model and the technology used in generating this mutant may help to reveal novel mechanisms that underlie a variety of neuropsychiatric conditions associated with anxiety and impairment of episodic and emotional memory.


Asunto(s)
Miedo/fisiología , Memoria/fisiología , Neuronas/fisiología , Animales , Ansiedad/fisiopatología , Encéfalo/fisiopatología , Condicionamiento Psicológico/fisiología , Señales (Psicología) , Emociones/fisiología , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Pruebas Neuropsicológicas , Reflejo de Sobresalto/fisiología
11.
Cancer Sci ; 104(1): 70-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23078246

RESUMEN

Small ubiquitin-like modifier (SUMO1-3) constitutes a group of proteins that conjugate to lysine residues of target proteins thereby modifying their activity, stability, and subcellular localization. A large number of SUMO target proteins are transcription factors and other nuclear proteins involved in gene expression. Furthermore, SUMO conjugation plays key roles in genome stability, quality control of newly synthesized proteins, proteasomal degradation of proteins, and DNA damage repair. Any marked increase in levels of SUMO-conjugated proteins is therefore expected to have a major impact on the fate of cells. We show here that SUMO conjugation is activated in human astrocytic brain tumors. Levels of both SUMO1- and SUMO2/3-conjugated proteins were markedly increased in tumor samples. The effect was least pronounced in low-grade astrocytoma (WHO Grade II) and most pronounced in glioblastoma multiforme (WHO Grade IV). We also found a marked rise in levels of Ubc9, the only SUMO conjugation enzyme identified so far. Blocking SUMO1-3 conjugation in glioblastoma cells by silencing their expression blocked DNA synthesis, cell growth, and clonogenic survival of cells. It also resulted in DNA-dependent protein kinase-induced phosphorylation of H2AX, indicative of DNA double-strand damage, and G(2) /M cell cycle arrest. Collectively, these findings highlight the pivotal role of SUMO conjugation in DNA damage repair processes and imply that the SUMO conjugation pathway could be a new target of therapeutic intervention aimed at increasing the sensitivity of glioblastomas to radiotherapy and chemotherapy.


Asunto(s)
Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación , Ubiquitinas/metabolismo , Astrocitoma/patología , Neoplasias Encefálicas/patología , Puntos de Control del Ciclo Celular , Supervivencia Celular , Reparación del ADN , Glioblastoma/patología , Histonas/metabolismo , Humanos , MicroARNs/genética , Fosforilación , Interferencia de ARN , Proteína SUMO-1/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Células Tumorales Cultivadas , Enzimas Ubiquitina-Conjugadoras/biosíntesis , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinas/genética
12.
J Proteome Res ; 11(2): 1108-17, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22082260

RESUMEN

Transient cerebral ischemia dramatically activates small ubiquitin-like modifier (SUMO2/3) conjugation. In cells exposed to 6 h of transient oxygen/glucose deprivation (OGD), a model of ischemia, SUMOylation increases profoundly between 0 and 30 min following re-oxygenation. To elucidate the effect of transient OGD on SUMO conjugation of target proteins, we exposed neuroblastoma B35 cells expressing HA-SUMO3 to transient OGD and used stable isotope labeling with amino acids in cell culture (SILAC) to quantify OGD-induced changes in levels of specific SUMOylated proteins. Lysates from control and OGD-treated cells were mixed equally, and HA-tagged proteins were immunoprecipitated and analyzed by 1D-SDS-PAGE-LC-MS/MS. We identified 188 putative SUMO3-conjugated proteins, including numerous transcription factors and coregulators, and PIAS2 and PIAS4 SUMO ligases, of which 22 were increased or decreased more than ±2-fold. In addition to SUMO3, the levels of protein-conjugated SUMO1 and SUMO2, as well as ubiquitin, were all increased. Importantly, protein ubiquitination induced by OGD was completely blocked by gene silencing of SUMO2/3. Collectively, these results suggest several mechanisms for OGD-modulated SUMOylation, point to a number of signaling pathways that may be targets of SUMO-based signaling and recovery from ischemic stress, and demonstrate a tightly controlled crosstalk between the SUMO and ubiquitin conjugation pathways.


Asunto(s)
Glucosa/metabolismo , Marcaje Isotópico/métodos , Oxígeno/metabolismo , Proteómica/métodos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Glucosa/deficiencia , Ratones , Neuroblastoma , Mapas de Interacción de Proteínas , Proteínas/análisis , Proteínas/química , Proteínas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , Estrés Fisiológico/fisiología , Ubiquitinación
13.
J Neurochem ; 123(3): 349-59, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22891650

RESUMEN

Deep hypothermia protects the brain from ischemic damage and is therefore used during major cardiovascular surgeries requiring cardiopulmonary bypass and a period of circulatory arrest. Here, we demonstrated that small ubiquitin-like modifier (SUMO1-3) conjugation is markedly activated in the brain during deep to moderate hypothermia. Animals were subjected to normothermic (37°C) or deep to moderate (18°C, 24°C, 30°C) hypothermic cardiopulmonary bypass, and the effects of hypothermia on SUMO conjugation were evaluated by Western blot and immunohistochemistry. Exposure to moderate 30°C hypothermia was sufficient to markedly increase levels and nuclear accumulation of SUMO2/3-conjugated proteins in these cells. Deep hypothermia induced nuclear translocation of the SUMO-conjugating enzyme Ubc9, suggesting that the increase in nuclear levels of SUMO2/3-conjugated proteins observed in brains of hypothermic animals is an active process. Exposure of primary neuronal cultures to deep hypothermia induced only a moderate rise in levels of SUMO2/3-conjugated proteins. This suggests that neurons in vivo have a higher capacity than neurons in vitro to activate this endogenous potentially neuroprotective pathway upon exposure to hypothermia. Identifying proteins that are SUMO2/3 conjugated during hypothermia could help to design new strategies for preventive and therapeutic interventions to make neurons more resistant to a transient interruption of blood supply.


Asunto(s)
Hipotermia Inducida/métodos , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Femenino , Masculino , Neuronas/enzimología , Neuronas/patología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/enzimología , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología , Enzimas Ubiquitina-Conjugadoras/metabolismo
14.
Exp Neurol ; 339: 113646, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33600817

RESUMEN

Spliced X-box binding protein-1 (XBP1s) together with the hexosamine biosynthetic pathway (HBP) and O-GlcNAcylation forms the XBP1s/HBP/O-GlcNAc axis. Our previous studies have provided evidence that activation of this axis is neuroprotective after ischemic stroke and critically, ischemia-induced O-GlcNAcylation is impaired in the aged brain. However, the XBP1s' neuroprotective role and its link to O-GlcNAcylation in stroke, as well as the therapeutic potential of targeting this axis in stroke, have not been well established. Moreover, the mechanisms underlying this age-related impairment of O-GlcNAcylation induction after brain ischemia remain completely unknown. In this study, using transient ischemic stroke models, we first demonstrated that neuron-specific overexpression of Xbp1s improved outcome, and pharmacologically boosting O-GlcNAcylation with thiamet-G reversed worse outcome observed in neuron-specific Xbp1 knockout mice. We further showed that thiamet-G treatment improved long-term functional recovery in both young and aged animals after transient ischemic stroke. Mechanistically, using an analytic approach developed here, we discovered that availability of UDP-GlcNAc was compromised in the aged brain, which may constitute a novel mechanism responsible for the impaired O-GlcNAcylation activation in the aged brain after ischemia. Finally, based on this new mechanistic finding, we evaluated and confirmed the therapeutic effects of glucosamine treatment in young and aged animals using both transient and permanent stroke models. Our data together support that increasing O-GlcNAcylation is a promising strategy in stroke therapy.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Encéfalo/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/prevención & control , Neuroprotección/fisiología , Factores de Edad , Animales , Encéfalo/efectos de los fármacos , Glucosamina/farmacología , Glucosamina/uso terapéutico , Glicosilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ratas , Ratas Endogámicas F344 , Proteína 1 de Unión a la X-Box/deficiencia , Proteína 1 de Unión a la X-Box/genética
15.
Biochem Biophys Res Commun ; 382(1): 215-8, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19275883

RESUMEN

Small ubiquitin-like modifier (SUMO) is a group of proteins binding to lysine residues of target proteins and thereby modifying their stability, activity and subcellular localization. Here we report that blocking SUMO2 and SUMO3 conjugation by silencing their expression markedly modifies gene expression. A microRNA-based RNAi system was used to specifically silence SUMO2 and SUMO3 expression simultaneously and stably transfected neuroblastoma B35 cells expressing dual SUMO2/3 microRNA were created. In cells stably expressing SUMO2/3 microRNA, mRNA levels of 105 and 58 known genes were significantly up- and down-regulated, respectively. About 20% of differentially regulated genes were associated with pathways involved in cell growth and differentiation. Cell division was significantly suppressed in SUMO2/3 miRNA expressing cells. Elucidating what effect the silencing of SUMO2/3 expression has on gene expression will help to identify the impact of SUMO2/3 conjugation on the various cellular pathways.


Asunto(s)
Proliferación Celular , Regulación de la Expresión Génica , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/fisiología , Animales , Línea Celular Tumoral , Silenciador del Gen , ARN Interferente Pequeño/genética , Ratas , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética
16.
J Cereb Blood Flow Metab ; 28(2): 269-79, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17565359

RESUMEN

A new group of proteins, small ubiquitin-like modifier (SUMO) proteins, has recently been identified and protein sumoylation has been shown to play a major role in various signal transduction pathways. Here, we report that transient global cerebral ischemia induces a marked increase in protein sumoylation. Mice were subjected to 10 mins severe forebrain ischemia followed by 3 or 6 h of reperfusion. Transient cerebral ischemia induced a massive increase in protein sumoylation by SUMO2/3 both in the hippocampus and cerebral cortex. SUMO2/3 conjugation was associated with a decrease in levels of free SUMO2/3. After ischemia, protein levels of the SUMO-conjugating enzyme Ubc9 were transiently decreased in the cortex but not in the hippocampus. We also exposed HT22 cells to arsenite, a respiratory poison that impairs cytoplasmic function and induces oxidative stress. Arsenite exposure induced a marked rise in protein sumoylation, implying that impairment of cytoplasmic function and oxidative stress may be involved in the massive post-ischemic activation of SUMO conjugation described here. Sumoylation of transcription factors has been shown to block their activation, with some exceptions such as the heat-shock factor and the hypoxia-responsive factor, where sumoylation blocks their degradation, and the nuclear factor-kappaB (NF-kappaB) essential modulator where sumoylation leads to an activation of NF-kappaB. Because protein sumoylation is known to be involved in the regulation of various biologic processes, the massive post-ischemic increase in protein sumoylation may play a critical role in defining the final outcome of neurons exposed to transient ischemia.


Asunto(s)
Ataque Isquémico Transitorio/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/biosíntesis , Animales , Western Blotting , Línea Celular , Células Cultivadas , Cisteína Endopeptidasas , Endopeptidasas/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/biosíntesis , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitinas/metabolismo
17.
J Cereb Blood Flow Metab ; 28(5): 892-6, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18167501

RESUMEN

This study was designed to investigate whether small ubiquitin-like modifier (SUMO) conjugation is activated after focal cerebral ischemia. Transient ischemia induced a dramatic increase in SUMO2/3 protein conjugates. The most pronounced changes were found in the parietal cortex. SUMO2/3 conjugation was particularly high in neurons located at the border of the middle cerebral artery territory where sumoylated proteins translocated to the nucleus. Considering the marked effect of SUMO conjugation on the function of target proteins, it is very likely that the postischemic activation of sumoylation has a significant effect on the fate of neurons exposed to transient ischemia.


Asunto(s)
Infarto de la Arteria Cerebral Media/metabolismo , Ataque Isquémico Transitorio/metabolismo , Lóbulo Parietal/metabolismo , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Western Blotting , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Ataque Isquémico Transitorio/patología , Masculino , Lóbulo Parietal/patología , Ratas , Ratas Wistar
18.
J Neurochem ; 106(3): 989-99, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18410505

RESUMEN

Transient cerebral ischemia/stroke activates various post-translational protein modifications such as phosphorylation and ubiquitin conjugation that are believed to play a major role in the pathological process triggered by an interruption of blood supply and culminating in cell death. A new system of post-translational protein modification has been identified, termed as small ubiquitin-like modifier (SUMO) conjugation. Like ubiquitin, SUMO is conjugated to the lysine residue of target proteins in a complex process. This review summarizes observations from recent experiments focusing on the effect of cerebral ischemia on SUMO conjugation. Transient global and focal cerebral ischemia both induced a rapid, dramatic and long-lasting rise in levels of SUMO2/3 conjugation. After transient focal cerebral ischemia, SUMO conjugation was particularly prominent in neurons located at the border of the ischemic territory where SUMO-conjugated proteins translocated to the nucleus. Many SUMO conjugation target proteins are transcription factors and sumoylation has been shown to have a major impact on the activity, stability, and cellular localization of target proteins. The rise in levels of SUMO-conjugated proteins is therefore likely to have a major effect on the fate of post-ischemic neurons. The sumoylation process could provide an exciting new target for therapeutic intervention.


Asunto(s)
Isquemia Encefálica/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Proteína SUMO-1/metabolismo , Transducción de Señal/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Humanos , Preparaciones Farmacéuticas/administración & dosificación , Proteína SUMO-1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico
19.
Biochem Biophys Res Commun ; 365(3): 521-7, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-18021742

RESUMEN

MicroRNAs (miRNAs) are endogenous non-coding small RNAs, which negatively regulate gene expression in a sequence-specific manner through the RNA interference (RNAi) pathway. Here we describe a new miRNA-based conditional RNAi expression system that relies on cellular stress-response mechanisms in mammalian cells. In our constructs, expression of miRNA mimics is tightly controlled by a heat shock-inducible promoter. This system is highly effective in silencing permanently or conditionally expressed luciferase. The stress inducible vectors also effectively deplete co-expressed pro-apoptotic protein CHOP with heat shock. Furthermore, we demonstrate cloning of a protein-coding sequence between the stress-inducible promoter and the miRNA expression cassette allows simultaneous silencing of a target gene and activation of synthesis of a protein of choice in response to stress stimulation. This new conditional gene silencing approach could be an invaluable tool for various areas of basic and applied research and for therapeutic intervention.


Asunto(s)
Silenciador del Gen , Calor , ARN Interferente Pequeño/biosíntesis , Animales , Línea Celular , Clonación Molecular , Genes Reporteros , Vectores Genéticos , Luciferasas/antagonistas & inhibidores , Luciferasas/genética , Ratones , Regiones Promotoras Genéticas
20.
Aging Dis ; 9(1): 31-39, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29392079

RESUMEN

Experimental cardiac arrest (CA) in aging research is infrequently studied in part due to the limitation of animal models. We aimed to develop an easily performed mouse CA model to meet this need. A standard mouse KCl-induced CA model using chest compressions and intravenous epinephrine for resuscitation was modified by blood withdrawal prior to CA onset, so as to decrease the requisite KCl dose to induce CA by decreasing the circulating blood volume. The modification was then compared to the standard model in young adult mice subjected to 8 min CA. 22-month old mice were then subjected to 8 min CA, resuscitated, and compared to young adult mice. Post-CA functional recovery was evaluated by measuring spontaneous locomotor activity pre-injury, and on post-CA days 1, 2, and 3. Neurological score and brain histology were examined on day 3. Brain elF2α phosphorylation levels were measured at 1 h to verify tissue stress. Compared to the standard model, the modification decreased cardiopulmonary resuscitation duration and increased 3-day survival in young mice. For aged mice, survival was 100 % at 24 h and 54% at 72 h. Neurological deficit was present 3 days post-CA, although more severe versus young mice. Mild neuronal necrosis was present in the cortex and hippocampus. The modified model markedly induced elF2α phosphorylation in both age groups. This modified procedure makes the CA model feasible in aged mice and provides a practical platform for understanding injury mechanisms and developing therapeutics for elderly patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA