Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Mol Biol Rep ; 51(1): 587, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683225

RESUMEN

BACKGROUND: Patients with multiple myeloma exhibit malignant osteolytic bone disease due to excessive osteoclast formation and function. We recently identified that osteoclastogenic stimulator selenoprotein W (SELENOW) is upregulated via ERK signaling and downregulated via p38 signaling during receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation. In the intrinsic physiological process, RANKL-induced downregulation of SELENOW maintains proper osteoclast differentiation; in contrast, forced overexpression of SELENOW leads to overactive osteoclast formation and function. METHODS AND RESULTS: We observed that SELENOW is highly expressed in multiple myeloma-derived peripheral blood mononuclear cells (PBMCs) and mature osteoclasts when compared to healthy controls. Also, the level of tumor necrosis factor alpha (TNFα), a pathological osteoclastogenic factor, is increased in the PBMCs and serum of patients with multiple myeloma. ERK activation by TNFα was more marked and sustained than that by RANKL, allowing SELENOW upregulation. Excessive expression of SELENOW in osteoclast progenitors and mature osteoclasts derived from multiple myeloma facilitated efficient nuclear translocation of osteoclastogenic transcription factors NF-κB and NFATc1, which are favorable for osteoclast formation. CONCLUSION: Our findings suggest a possibility that feedforward signaling of osteoclastogenic SELENOW by TNFα derived from multiple myeloma induces overactive osteoclast differentiation, leading to bone loss during multiple myeloma.


Asunto(s)
Diferenciación Celular , Mieloma Múltiple , Osteoclastos , Selenoproteína W , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Diferenciación Celular/genética , Leucocitos Mononucleares/metabolismo , Sistema de Señalización de MAP Quinasas , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Mieloma Múltiple/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Selenoproteína W/metabolismo , Selenoproteína W/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
2.
Biometals ; 35(6): 1359-1370, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36261677

RESUMEN

Selenium (Se) plays an essential role in the growth of fish and performs its physiological functions mainly through incorporation into selenoproteins. Our previous studies suggested that the selenoprotein W gene (selenow) is sensitive to changes in dietary Se in rainbow trout. However, the molecular characterization and tissue expression pattern of selenow are still unknown. Here, we revealed the molecular characterization, the tissue expression pattern of rainbow trout selenow and analyzed its response to dietary Se. The open reading frame (ORF) of the selenow gene was composed of 393 base pairs (bp) and encodes a 130-amino-acid protein. The 3' untranslated region (UTR) was 372 bp with a selenocysteine insertion sequence (SECIS) element. Remarkably, the rainbow trout selenow gene sequence was longer than those reported for mammals and most other fish. A ß1-α1-ß2-ß3-ß4-α2 pattern made up the secondary structure of SELENOW. Furthermore, multiple sequence alignment revealed that rainbow trout SELENOW showed a high level of identity with SELENOW from Salmo salar. In addition, the selenow gene was ubiquitously distributed in 13 tissues with various abundances and was predominantly expressed in muscle and brain. Interestingly, dietary Se significantly increased selenow mRNA expression in muscle. Our results highlight the vital role of selenow in rainbow trout muscle response to dietary Se levels and provide a theoretical basis for studies of selenow.


Asunto(s)
Oncorhynchus mykiss , Selenio , Animales , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/metabolismo , Selenoproteína W/genética , Selenoproteína W/metabolismo , Selenio/metabolismo , Selenocisteína/genética , Selenocisteína/metabolismo , Selenoproteínas/genética , Selenoproteínas/metabolismo , Clonación Molecular , Mamíferos/genética
3.
Blood ; 131(23): 2568-2580, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29615406

RESUMEN

Micronutrient selenium (Se) plays a key role in redox regulation through its incorporation into selenoproteins as the 21st amino acid selenocysteine (Sec). Because Se deficiency appears to be a cofactor in the anemia associated with chronic inflammatory diseases, we reasoned that selenoproteins may contribute to erythropoietic recovery from anemia, referred to as stress erythropoiesis. Here, we report that loss of selenoproteins through Se deficiency or by mutation of the Sec tRNA (tRNA[Sec]) gene (Trsp) severely impairs stress erythropoiesis at 2 stages. Early stress erythroid progenitors failed to expand and properly differentiate into burst-forming unit-erythroid cells , whereas late-stage erythroid progenitors exhibited a maturation defect that affected the transition of proerythroblasts to basophilic erythroblasts. These defects were, in part, a result of the loss of selenoprotein W (SelenoW), whose expression was reduced at both transcript and protein levels in Se-deficient erythroblasts. Mutation of SelenoW in the bone marrow cells significantly decreased the expansion of stress burst-forming unit-erythroid cell colonies, which recapitulated the phenotypes induced by Se deficiency or mutation of Trsp Similarly, mutation of SelenoW in murine erythroblast (G1E) cell line led to defects in terminal differentiation. In addition to the erythroid defects, the spleens of Se-deficient mice contained fewer red pulp macrophages and exhibited impaired development of erythroblastic island macrophages, which make up the niche supporting erythroblast development. Taken together, these data reveal a critical role of selenoproteins in the expansion and development of stress erythroid progenitors, as well as the erythroid niche during acute anemia recovery.


Asunto(s)
Anemia/metabolismo , Células Precursoras Eritroides/citología , Eritropoyesis , Selenio/deficiencia , Selenoproteínas/metabolismo , Anemia/genética , Animales , Regulación hacia Abajo , Eritroblastos/citología , Eritroblastos/metabolismo , Células Precursoras Eritroides/metabolismo , Ratones Endogámicos C57BL , Mutación , Selenio/metabolismo , Selenoproteína W/genética , Selenoproteína W/metabolismo , Selenoproteínas/genética , Bazo/citología , Bazo/metabolismo
4.
Am J Physiol Cell Physiol ; 317(6): C1172-C1182, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31509445

RESUMEN

Selenoprotein W (SelW) is a selenium-containing protein with a redox motif found abundantly in the skeletal muscle of rodents. Previous in vitro studies suggest that SelW plays an antioxidant role; however, relatively few in vivo studies have addressed the antioxidant role of SelW. Since oxidative stress is a causative factor for the development of insulin resistance in obese subjects, we hypothesized that if SelW plays a role as an antioxidant, SelW deficiency could aggravate the oxidative stress and insulin resistance caused by a high-fat diet. SelW deficiency did not affect insulin sensitivity and H2O2 levels in the skeletal muscle of control diet-fed mice. SelW levels in the skeletal muscle were decreased by high-fat diet feeding for 12 wk. High-fat diet induced obesity and insulin resistance and increased the levels of H2O2 and oxidative stress makers, which were not affected by SelW deficiency. High-fat diet feeding increased the expression of antioxidant enzymes; however, SelW deficiency did not affect the expression levels of antioxidants. These results suggest that SelW does not play a protective role against oxidative stress and insulin resistance in the skeletal muscle of high-fat diet-fed obese mice.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Músculo Esquelético/metabolismo , Obesidad/genética , Estrés Oxidativo , Selenoproteína W/genética , Animales , Catalasa/genética , Catalasa/metabolismo , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Humanos , Peróxido de Hidrógeno/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Selenoproteína W/deficiencia , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
5.
Mol Cancer ; 18(1): 9, 2019 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-30636640

RESUMEN

BACKGROUND: Breast cancer is one of the most common malignancies and the major cause of cancer-related death in women. Although the importance of PIWI-interacting RNAs (piRNAs) in cancer has been increasingly recognized, few studies have been explored the functional mechanism of piRNAs in breast cancer development and progression. METHODS: We examined the top 20 highly expressed piRNAs based on the analysis of TCGA breast cancer data in two patient cohorts to test the roles of piRNAs in breast cancer. The effects of piRNA-36,712 on the malignant phenotypes and chemosensitivity of breast cancer cells were detected in vitro and in vivo. MS2-RIP and reporter gene assays were conducted to identify the interaction and regulation among piRNA-36,712, miRNAs and SEPW1P. Kaplan-Meier estimate with log-rank test was used to compare patient survival by different piRNA-36,712 expression levels. RESULTS: We found piRNA-36,712 level was significantly lower in breast cancer than in normal breast tissues and low level was correlated with poor clinical outcome in patients. Functional studies demonstrated that piRNA-36,712 interacts with RNAs produced by SEPW1P, a retroprocessed pseudogene of SEPW1, and subsequently inhibits SEPW1 expression through competition of SEPW1 mRNA with SEPW1P RNA for microRNA-7 and microRNA-324. We also found that higher SEPW1 expression due to downregulation of piRNA-36,712 in breast cancer may suppress P53, leading to the upregulated Slug but decreased P21 and E-cadherin levels, thus promoting cancer cell proliferation, invasion and migration. Furthermore, we found that piRNA-36,712 had synergistic anticancer effects with the paclitaxel and doxorubicin, two chemotherapeutic agents for breast cancer. CONCLUSIONS: These findings suggest that piRNA-36,712 is a novel tumor suppressor and may serve as a potential predictor for the prognosis of breast cancer patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , ARN Interferente Pequeño/genética , Selenoproteína W/genética , Animales , Mama/efectos de los fármacos , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación hacia Abajo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Femenino , Humanos , MicroARNs/genética , Paclitaxel/farmacología , Pronóstico , Seudogenes , ARN Mensajero/genética , ARN Interferente Pequeño/biosíntesis , Regulación hacia Arriba
6.
Biochem Biophys Res Commun ; 512(1): 137-143, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30876693

RESUMEN

Selenoprotien W (SelW) plays a key role in brain development, although the exact biological function and mechanisms remain unclear. We performed a yeast two-hybrid screen on a human fetal brain cDNA library and identified FAM96B as a novel binding partner of SelW. FRET analyses confirmed the interaction between SelW' and FAM96B. The mutated SelW' construct was cloned and overexpressed in E. coli, and a pull-down assay verified a direct interaction between SelW' and FAM96B. Finally, Co-Immunoprecipitation on murine brain tissue proteins demonstrated an endogenous interaction between the two proteins in the brain. Taken together, our findings prove a direct interaction between SelW and FAM96B, which may provide new insights into the role of SelW in brain development and neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Metaloproteínas/metabolismo , Proteínas Nucleares/metabolismo , Selenoproteína W/metabolismo , Animales , Femenino , Feto/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Biblioteca de Genes , Células HEK293 , Humanos , Metaloproteínas/genética , Ratones , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Selenoproteína W/genética , Técnicas del Sistema de Dos Híbridos
7.
Biochim Biophys Acta ; 1863(1): 10-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26474786

RESUMEN

Selenoprotein W (SelW) contains a selenocysteine (Sec, U) in a conserved CXXU motif corresponding to the CXXC redox motif of thioredoxin, suggesting a putative redox function of SelW. We have previously reported that the binding of 14-3-3 protein to its target proteins, including CDC25B, Rictor and TAZ, is inhibited by the interaction of 14-3-3 protein with SelW. However, the binding mechanism is unclear. In this study, we sought to determine the binding site of SelW to understand the regulatory mechanism of the interaction between SelW and 14-3-3 and its biological effects. Phosphorylated Ser(pS) or Thr(pT) residues in RSXpSXP or RXXXp(S/T)XP motifs are well-known common 14-3-3-binding sites, but Thr41, Ser59, and T69 of SelW, which are computationally predicted to serve are phosphorylation sites, were neither phosphorylation sites nor sites involved in the interaction. A mutant SelW in which Sec13 is changed to Ser (U13S) was unable to interact with 14-3-3 protein and thus did not inhibit the interaction of 14-3-3 to other target proteins. However, other Cys mutants of SelW(C10S, C33S and C37S) normally interacted with 14-3-3 protein. The interaction of SelW to 14-3-3 protein was enhanced by diamide or H2O2 and decreased by dithiothreitol (DTT). Taken together, these findings demonstrate that the Sec of SelW is involved in its interaction with 14-3-3 protein and that this interaction is increased under oxidative stress conditions. Thus, SelW may have a regulatory function in redox cell signaling by interacting with 14-3-3 protein.


Asunto(s)
Proteínas 14-3-3/metabolismo , Estrés Oxidativo/fisiología , Selenoproteína W/metabolismo , Proteínas 14-3-3/genética , Secuencias de Aminoácidos , Ditiotreitol/farmacología , Femenino , Humanos , Peróxido de Hidrógeno/farmacología , Células MCF-7 , Mutación Missense , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Selenoproteína W/genética
8.
Fish Physiol Biochem ; 43(1): 115-126, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27506211

RESUMEN

In this study, a selenoprotein W cDNA was cloned from topmouth culter (Erythroculter ilishaeformis), and it was designated as EISelW. The EISelW open reading frame was composed of 261 base pairs (bp), encoding 86-amino-acid protein. The 5' untranslated region (UTR) consisted of 104 bp, and the 3'-UTR was composed of 365 bp. A selenocysteine insertion sequence (SECIS) element was found in the 3'-UTR of EISelW mRNA. The SECIS element was classified as form II because of a small additional apical loop presented in SECIS element of EISelW mRNA. Bioinformatic approaches showed that the secondary structure of EISelW was a ß1-α1-ß2-ß3-ß4-α2 pattern from amino-terminal to carboxy-terminal. Real-time PCR analysis of EISelW mRNAs expression in 17 tissues showed that the EISelW mRNA was predominantly expressed in liver, ovary, pituitary, various regions of the brain, spinal cord and head kidney. Study of intraperitoneal injection showed that the levels of EISelW mRNA in brain, liver, ovary and spleen were regulated by somatostatin 14 (SS14), 17ß-estradiol (E2), cysteamine hydrochloride (CSH) and a binary mixture of E2 and CSH, dependent on the dosage. These results suggest that E2, SS14 and CSH status may affect tissues of selenium metabolism by regulating the expression of SelW mRNA, as SelW plays a central role in selenium metabolism.


Asunto(s)
Cisteamina/farmacología , Estradiol/farmacología , Perciformes/genética , Selenoproteína W/genética , Somatostatina/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Encéfalo/metabolismo , ADN Complementario/genética , Interacciones Farmacológicas , Femenino , Hígado/metabolismo , Masculino , Ovario/metabolismo , Filogenia , ARN Mensajero/metabolismo , Bazo/metabolismo
9.
Biochim Biophys Acta ; 1843(7): 1356-64, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24726955

RESUMEN

Selenoprotein W (SelW) is expressed in various tissues, particularly in skeletal muscle. We have previously reported that SelW is up-regulated during C2C12 skeletal muscle differentiation and inhibits binding of 14-3-3 to its target proteins. 14-3-3 reduces myogenic differentiation by inhibiting nuclear translocation of transcriptional co-activator with PDZ-binding motif (TAZ). Phosphorylation of TAZ at Ser89 is required for binding to 14-3-3, leading to cytoplasmic retention of TAZ and a delay in myogenic differentiation. Here, we show that myogenic differentiation was delayed in SelW-knockdown C2C12 cells. Down-regulation of SelW also increased TAZ binding to 14-3-3, which eventually resulted in decreasing translocation of TAZ to the nucleus. However, phosphorylation of TAZ at Ser89 was not affected. Although phosphorylation of TAZ at Ser89 was sustained by the phosphatase inhibitor okadaic acid, nuclear translocation of TAZ was increased by ectopic expression of SelW. This result was due to decreased binding of TAZ to 14-3-3. We also found that the interaction between TAZ and MyoD was increased by ectopic expression of SelW. Taken together, these findings strongly demonstrate that SelW enhances C2C12 cell differentiation by inhibiting TAZ binding to 14-3-3.


Asunto(s)
Proteínas 14-3-3/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Selenoproteína W/metabolismo , Factores de Transcripción/metabolismo , Proteínas 14-3-3/genética , Aciltransferasas , Animales , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Ratones , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/citología , Mioblastos/efectos de los fármacos , Ácido Ocadaico/farmacología , Fosforilación , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Selenoproteína W/antagonistas & inhibidores , Selenoproteína W/genética , Transducción de Señal , Factores de Transcripción/genética
10.
RNA ; 19(12): 1755-66, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24152552

RESUMEN

Spinal muscular atrophy is a neuromuscular disease resulting from mutations in the SMN1 gene, which encodes the survival motor neuron (SMN) protein. SMN is part of a large complex that is essential for the biogenesis of spliceosomal small nuclear RNPs. SMN also colocalizes with mRNAs in granules that are actively transported in neuronal processes, supporting the hypothesis that SMN is involved in axonal trafficking of mRNPs. Here, we have performed a genome-wide analysis of RNAs present in complexes containing the SMN protein and identified more than 200 mRNAs associated with SMN in differentiated NSC-34 motor neuron-like cells. Remarkably, ~30% are described to localize in axons of different neuron types. In situ hybridization and immuno-fluorescence experiments performed on several candidates indicate that these mRNAs colocalize with the SMN protein in neurites and axons of differentiated NSC-34 cells. Moreover, they localize in cell processes in an SMN-dependent manner. Thus, low SMN levels might result in localization deficiencies of mRNAs required for axonogenesis.


Asunto(s)
Neuritas/metabolismo , ARN Mensajero/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Anexina A2/genética , Anexina A2/metabolismo , Línea Celular , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Técnicas de Silenciamiento del Gen , Genoma , Ratones , Neuronas Motoras/metabolismo , Unión Neuromuscular/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Transporte de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Selenoproteína W/genética , Selenoproteína W/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética
11.
Biometals ; 28(1): 75-87, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25351959

RESUMEN

Selenoprotein W (SelW) is mainly understood in terms of its antioxidant effects in the cellular defense system. Inflammation is an important indicator of animal tissue injury, and the inflammatory cells may trigger a sophisticated and well-orchestrated inflammatory cascade, resulting in exaggerated oxidative stress. To investigate the role of SelW in inflammatory injury in chicken immune tissues and cultured splenic lymphocyte, in this report, the effects of selenium (Se) on mRNA expressions of SelW and inflammatory factors (iNOS, COX-2, NF-κB, PTGEs, and TNF-α) in the chicken immune organs (spleen, thymus and bursa of Fabricius) and cultured splenic lymphocyte treated with sodium selenite and H2O2, or knocked down SelW with small interfering RNAs (siRNAs) were examined. The results showed that Se-deficient diets effectively decreased the mRNA expression of SelW (P < 0.05), and induced a significantly up-regulation of COX-2, iNOS, NF-κB, PTGEs and TNF-α mRNA levels (P < 0.05). The histopathological analysis showed that immune tissues were obviously injured in the low-Se groups. In vitro, H2O2 induced a significantly up-regulation of the mRNA levels of inflammation-related genes (iNOS, COX-2, NF-κB, PTGEs, and TNF-α) in cultured splenic lymphocyte (P < 0.05). When lymphocytes were pretreated with Se before treated with H2O2, the inflammation-related genes were significantly decreased (P < 0.05). Silencing of SelW significantly up-regulated the inflammation-related genes (iNOS, COX-2, NF-κB, PTGEs, and TNF-α) in cultured splenic lymphocyte (P < 0.05). The results suggested that the expression levels of inflammatory factors (iNOS, COX-2, NF-κB, PTGEs, and TNF-α) and SelW can be influenced by Se in birds. SelW commonly played an important role in the protection of immune organs of birds from inflammatory injury by the regulations of inflammation-related genes.


Asunto(s)
Inflamación/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Selenoproteína W/metabolismo , Animales , Células Cultivadas , Pollos , Inflamación/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Selenoproteína W/genética , Bazo/citología , Bazo/metabolismo
12.
Biochim Biophys Acta ; 1833(10): 2135-42, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23680186

RESUMEN

14-3-3 reduces cell proliferation by inhibiting the activity of proteins involved in the signaling pathway that includes Akt kinase. Activation of Akt is enhanced by activating the mammalian target of rapamycin complex 2 (mTORC2). 14-3-3 is also a negative regulator of the mTORC2/Akt pathway, by interacting with a component of mTORC2. Recently, we reported that selenoprotein W (SelW) regulated the interaction between 14-3-3 and its target protein, CDC25B. Here, we show that the binding of Rictor, a component of mTORC2, to 14-3-3, is regulated by the interaction of 14-3-3 with SelW. When SelW was down-regulated, mTORC2-dependent phosphorylation of Akt at Ser473 was decreased. However, the phosphorylation of Thr308 was not affected. The interaction of Rictor with 14-3-3 was increased in SelW-knockdown cells, as compared to control cells. SelW-knockdown cells were also more sensitive to DNA damage induced by etoposide, than control cells. This phenomenon was due to the decreased phosphorylation of Akt at Ser473. We also found that ectopic expression of SelW(U13C) reduced the interaction between Rictor and 14-3-3, leading to Akt phosphorylation at Ser473. Taken together, these findings demonstrate that SelW activates the mTORC2/Akt pathway for Akt phosphorylation at Ser473, by interrupting the binding of Rictor to 14-3-3.


Asunto(s)
Proteínas 14-3-3/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Pulmonares/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Selenoproteína W/metabolismo , Serina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas 14-3-3/antagonistas & inhibidores , Proteínas 14-3-3/genética , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Portadoras/genética , Proliferación Celular , Citometría de Flujo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/genética , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , Proteína Asociada al mTOR Insensible a la Rapamicina , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenoproteína W/genética , Serina/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre , Cicatrización de Heridas
13.
Commun Biol ; 7(1): 872, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39020075

RESUMEN

Lower selenium levels are observed in Alzheimer's disease (AD) brains, while supplementation shows multiple benefits. Selenoprotein W (SELENOW) is sensitive to selenium changes and binds to tau, reducing tau accumulation. However, whether restoration of SELENOW has any protective effect in AD models and its underlying mechanism remain unknown. Here, we confirm the association between SELENOW downregulation and tau pathology, revealing SELENOW's role in promoting tau degradation through the ubiquitin‒proteasome system. SELENOW competes with Hsp70 to interact with tau, promoting its ubiquitination and inhibiting tau acetylation at K281. SELENOW deficiency leads to synaptic defects, tau dysregulation and impaired long-term potentiation, resulting in memory deficits in mice. Conversely, SELENOW overexpression in the triple transgenic AD mice ameliorates memory impairment and tau-related pathologies, featuring decreased 4-repeat tau isoform, phosphorylation at Ser396 and Ser404, neurofibrillary tangles and neuroinflammation. Thus, SELENOW contributes to the regulation of tau homeostasis and synaptic maintenance, implicating its potential role in AD.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Homeostasis , Ratones Transgénicos , Selenoproteína W , Proteínas tau , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Ratones , Selenoproteína W/metabolismo , Selenoproteína W/genética , Masculino , Fosforilación , Humanos , Ratones Endogámicos C57BL
14.
Sci Adv ; 10(38): eadj4122, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39303039

RESUMEN

Sarcopenia is characterized by accelerated muscle mass and function loss, which burdens and challenges public health worldwide. Several studies indicated that selenium deficiency is associated with sarcopenia; however, the specific mechanism remains unclear. Here, we demonstrated that selenoprotein W (SELENOW) containing selenium in the form of selenocysteine functioned in sarcopenia. SELENOW expression is up-regulated in dexamethasone (DEX)-induced muscle atrophy and age-related sarcopenia mouse models. Knockout (KO) of SELENOW profoundly aggravated the process of muscle mass loss in the two mouse models. Mechanistically, SELENOW KO suppressed the RAC1-mTOR cascade by the interaction between SELENOW and RAC1 and induced the imbalance of protein synthesis and degradation. Consistently, overexpression of SELENOW in vivo and in vitro alleviated the muscle and myotube atrophy induced by DEX. SELENOW played a role in age-related sarcopenia and regulated the genes associated with aging. Together, our study uncovered the function of SELENOW in age-related sarcopenia and provides promising evidence for the prevention and treatment of sarcopenia.


Asunto(s)
Ratones Noqueados , Complejo de la Endopetidasa Proteasomal , Biosíntesis de Proteínas , Sarcopenia , Selenoproteína W , Ubiquitina , Animales , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratones , Sarcopenia/metabolismo , Sarcopenia/genética , Sarcopenia/patología , Ubiquitina/metabolismo , Selenoproteína W/genética , Selenoproteína W/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Dexametasona/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Modelos Animales de Enfermedad , Atrofia Muscular/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Atrofia Muscular/inducido químicamente , Envejecimiento/metabolismo , Masculino , Transducción de Señal , Neuropéptidos
15.
J Biol Chem ; 287(33): 27371-9, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22730327

RESUMEN

Selenoprotein W (SEPW1) is a ubiquitous, highly conserved thioredoxin-like protein whose depletion causes a transient p53- and p21(Cip1)-dependent G(1)-phase cell cycle arrest in breast and prostate epithelial cells. SEPW1 depletion increases phosphorylation of Ser-33 in p53, which is associated with decreased p53 ubiquitination and stabilization of p53. We report here that delayed cell cycle progression, Ser-33 phosphorylation, and p53 nuclear accumulation from SEPW1 depletion require mitogen-activated protein kinase kinase 4 (MKK4). Silencing MKK4 rescued G(1) arrest, Ser-33 phosphorylation, and nuclear accumulation of p53 induced by SEPW1 depletion, but silencing MKK3, MKK6, or MKK7 did not. SEPW1 silencing did not change the phosphorylation state of MKK4 but increased total MKK4 protein. Silencing p38γ, p38δ, or JNK2 partially rescued G(1) arrest from SEPW1 silencing, suggesting they signal downstream from MKK4. These results imply that SEPW1 silencing increases MKK4, which activates p38γ, p38δ, and JNK2 to phosphorylate p53 on Ser-33 and cause a transient G(1) arrest.


Asunto(s)
Puntos de Control del Ciclo Celular , Núcleo Celular/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Selenoproteína W/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Transporte Activo de Núcleo Celular/genética , Línea Celular Tumoral , Núcleo Celular/genética , Fase G1/genética , Silenciador del Gen , Humanos , MAP Quinasa Quinasa 4/genética , Masculino , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/genética , Fosforilación/genética , Selenoproteína W/genética , Proteína p53 Supresora de Tumor/genética
16.
Biochim Biophys Acta ; 1823(12): 2217-26, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22982242

RESUMEN

Selenoprotein W (SelW) contains a highly reactive selenocysteine (Sec; U) in the CXXU motif corresponding to the CXXC motif in thioredoxin (Trx) and thus it appears to be involved in regulating the cellular redox state. Recent reports on the interaction between SelW and 14-3-3 suggest that SelW may be redox dependently involved in the cell cycle. However, the precise function of SelW has not yet been elucidated. Here, we show that SelW is involved in the G2-M transition, especially in the recovery from G2 arrest after deoxyribonucleic acid (DNA) damage. Knockdown of SelW significantly accumulated phosphorylated cyclin-dependent kinase (Cdk1), which eventually led to a delay in recovery from G2 arrest. We also found that inactive Cdk1 is caused by the sustained inactivation of CDC25B, which removes the inhibitory phosphate from Cdk1. Our observation from this study reveals that SelW activated CDC25B by promoting the dissociation of 14-3-3 from CDC25B through the reduction of the intramolecular disulfide bond during recovery. We suggest that SelW plays an important role in the recovery from G2 arrest by determining the dissociation of 14-3-3 from CDC25B in a redox-dependent manner.


Asunto(s)
Proteínas 14-3-3/metabolismo , Apoptosis , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Selenoproteína W/metabolismo , Fosfatasas cdc25/metabolismo , Proteínas 14-3-3/genética , Animales , Western Blotting , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Humanos , Inmunoprecipitación , Ratones , Células 3T3 NIH , Proteínas Quinasas , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenoproteína W/antagonistas & inhibidores , Selenoproteína W/genética , Fosfatasas cdc25/genética
17.
Redox Biol ; 59: 102571, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36516721

RESUMEN

Macrophages play a pivotal role in mediating inflammation and subsequent resolution of inflammation. The availability of selenium as a micronutrient and the subsequent biosynthesis of selenoproteins, containing the 21st amino acid selenocysteine (Sec), are important for the physiological functions of macrophages. Selenoproteins regulate the redox tone in macrophages during inflammation, the early onset of which involves oxidative burst of reactive oxygen and nitrogen species. SELENOW is a highly expressed selenoprotein in bone marrow-derived macrophages (BMDMs). Beyond its described general role as a thiol and peroxide reductase and as an interacting partner for 14-3-3 proteins, its cellular functions, particularly in macrophages, remain largely unknown. In this study, we utilized Selenow knock-out (KO) murine bone marrow-derived macrophages (BMDMs) to address the role of SELENOW in inflammation following stimulation with bacterial endotoxin lipopolysaccharide (LPS). RNAseq-based temporal analyses of expression of selenoproteins and the Sec incorporation machinery genes suggested no major differences in the selenium utilization pathway in the Selenow KO BMDMs compared to their wild-type counterparts. However, selective enrichment of oxidative stress-related selenoproteins and increased ROS in Selenow-/- BMDMs indicated anomalies in redox homeostasis associated with hierarchical expression of selenoproteins. Selenow-/- BMDMs also exhibited reduced expression of arginase-1, a key enzyme associated with anti-inflammatory (M2) phenotype necessary to resolve inflammation, along with a significant decrease in efferocytosis of neutrophils that triggers pathways of resolution. Parallel targeted metabolomics analysis also confirmed an impairment in arginine metabolism in Selenow-/- BMDMs. Furthermore, Selenow-/- BMDMs lacked the ability to enhance characteristic glycolytic metabolism during inflammation. Instead, these macrophages atypically relied on oxidative phosphorylation for energy production when glucose was used as an energy source. These findings suggest that SELENOW expression in macrophages may have important implications on cellular redox processes and bioenergetics during inflammation and its resolution.


Asunto(s)
Selenio , Selenoproteína W , Ratones , Animales , Selenoproteína W/genética , Selenoproteína W/metabolismo , Selenio/metabolismo , Selenoproteínas/genética , Selenoproteínas/metabolismo , Macrófagos/metabolismo , Oxidación-Reducción , Inflamación/genética
18.
J Biol Chem ; 286(11): 9031-7, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21296890

RESUMEN

DNA methylation is a major epigenetic modification that regulates gene expression. Dnmt1, the maintenance DNA methylation enzyme, is abundantly expressed in the adult brain and is mainly located in the nuclear compartment, where it has access to chromatin. Hypomethylation of CpG islands at intron 1 of the SNCA gene has recently been reported to result in overexpression of α-synuclein in Parkinson disease (PD) and related disorders. We therefore investigated the mechanisms underlying altered DNA methylation in PD and dementia with Lewy bodies (DLB). We present evidence of reduction of nuclear Dnmt1 levels in human postmortem brain samples from PD and DLB patients as well as in the brains of α-synuclein transgenic mice models. Furthermore, sequestration of Dnmt1 in the cytoplasm results in global DNA hypomethylation in human and mouse brains, involving CpG islands upstream of SNCA, SEPW1, and PRKAR2A genes. We report that association of Dnmt1 and α-synuclein might mediate aberrant subcellular localization of Dnmt1. Nuclear Dnmt1 levels were partially rescued by overexpression of Dnmt1 in neuronal cell cultures and in α-synuclein transgenic mice brains. Our results underscore a novel mechanism for epigenetic dysregulation in Lewy body diseases, which might underlie the decrease in DNA methylation reported for PD and DLB.


Asunto(s)
Encéfalo/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Epigénesis Genética , Enfermedad por Cuerpos de Lewy/metabolismo , alfa-Sinucleína/metabolismo , Adulto , Animales , Islas de CpG , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Humanos , Enfermedad por Cuerpos de Lewy/genética , Ratones , Ratones Noqueados , Selenoproteína W/genética , Selenoproteína W/metabolismo , alfa-Sinucleína/genética
19.
J Cell Biochem ; 113(1): 61-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21866568

RESUMEN

The anticancer activity of selenium (Se) has been demonstrated in myriad animal and in vitro studies, yet the mechanisms remain obscure. The main form of Se in animal tissues is selenocysteine in selenoproteins, but the relative importance of selenoproteins versus smaller Se compounds in cancer protection is unresolved. Selenoprotein W (SEPW1) is a highly conserved protein ubiquitously expressed in animals, bacteria, and archaea. SEPW1 depletion causes a delay in cell cycle progression at the G1/S transition of the cell cycle in breast and prostate epithelial cells. Tumor suppressor protein p53 is a master regulator of cell cycle progression and is the most frequently mutated gene in human cancers. p53 was increased in SEPW1 silenced cells and was inversely correlated with SEPW1 mRNA in cell lines with altered SEPW1 expression. Silencing SEPW1 decreased ubiquitination of p53 and increased p53 half-life. SEPW1 silencing increased p21(Cip1/WAF1/CDKN1A), while p27 (Kip1/CDKN1B) levels were unaffected. G1-phase arrest from SEPW1 knockdown was abolished by silencing p53 or p21. Cell cycle arrest from SEPW1 silencing was not associated with activation of ATM or phosphorylation of Ser-15 in p53, suggesting the DNA damage response pathway was not involved. Silencing GPX1 had no effect on cell cycle, suggesting that G1-phase arrest from SEPW1 silencing was not due to loss of antioxidant protection. More research is required to identify the function of SEPW1 and how it affects stability of p53.


Asunto(s)
Puntos de Control del Ciclo Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/citología , Selenoproteína W/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , División Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Silenciador del Gen , Humanos , Masculino , Próstata/citología , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño , Selenio , Selenoproteína W/genética , Proteína p53 Supresora de Tumor/genética , Ubiquitinación/genética
20.
Biometals ; 25(2): 459-68, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22234495

RESUMEN

Selenium (Se) and Selenoprotein W (SelW) plays a pivotal role in the brain development, function, and degeneration and that SelW expression in the brain may be affected by Se. However, the mechanism which Se regulates the SelW gene expression in neurons remains to be unclear. To investigate the effects of the SelW gene expression and mRNA stability induced by Se, primary cultured chicken embryos neurons derived from 8-day-old chick embryo cerebral hemispheres were treated with 10(-9)-10(-5) mol/l Se as selenite for 3, 6, 12, 24 or 48 h, respectively. The morphology and viability of Neurons was detected. The SelW mRNA expression level and mRNA half-life was examined in Se-treated neurons. The relative low concentrations of Se enhanced the neurite outgrowth, increased the SelW mRNA levels and elevated the mRNA half-life of chick embryo neurons. In contrast, the high concentrations of Se presented neurotoxic to neurons, decreased the SelW mRNA levels and reduced the mRNA half-life of neuronal cells. These results suggest that the alteration of post-transcriptional stabilization of SelW mRNA is an important mechanism of Se-induced the elevation or reduction of the SelW expression level in chick embryo neurons.


Asunto(s)
Neuronas/efectos de los fármacos , Estabilidad del ARN , Selenio/farmacología , Selenoproteína W/genética , Animales , Supervivencia Celular , Embrión de Pollo , Neuronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA