Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biol Chem ; 300(4): 105778, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38395307

RESUMEN

The mechanistic target of rapamycin (mTOR) signaling is influenced by multiple regulatory proteins and post-translational modifications; however, underlying mechanisms remain unclear. Here, we report a novel role of small ubiquitin-like modifier (SUMO) in mTOR complex assembly and activity. By investigating the SUMOylation status of core mTOR components, we observed that the regulatory subunit, GßL (G protein ß-subunit-like protein, also known as mLST8), is modified by SUMO1, 2, and 3 isoforms. Using mutagenesis and mass spectrometry, we identified that GßL is SUMOylated at lysine sites K86, K215, K245, K261, and K305. We found that SUMO depletion reduces mTOR-Raptor (regulatory protein associated with mTOR) and mTOR-Rictor (rapamycin-insensitive companion of mTOR) complex formation and diminishes nutrient-induced mTOR signaling. Reconstitution with WT GßL but not SUMOylation-defective KR mutant GßL promotes mTOR signaling in GßL-depleted cells. Taken together, we report for the very first time that SUMO modifies GßL, influences the assembly of mTOR protein complexes, and regulates mTOR activity.


Asunto(s)
Transducción de Señal , Sumoilación , Serina-Treonina Quinasas TOR , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/genética , Células HEK293 , Proteína SUMO-1/metabolismo , Proteína SUMO-1/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Ubiquitinas/metabolismo , Ubiquitinas/genética , Lisina/metabolismo
2.
Cell Oncol (Dordr) ; 45(3): 355-365, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35579750

RESUMEN

PURPOSE: N6-methyladenosine (m6A), the most prevalent mRNA modification, plays an essential role in tumorigenesis. Notably, increasing interest has been directed to bioactive peptides (BPs) with antitumor activities. Here, we set out to investigate the potential of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis on prevention and treatment of acute myeloid leukemia (AML). METHODS: The biological effects of BP on AML cells were detected by MTT and ApoLive-Glo™ multiplex assays. The role of BP in tumor growth was determined by a subcutaneous xenograft model. The ALKBH5/MLST8/EIF4EBP1 axis was identified as a potential BP target in AML via methylated RNA immunoprecipitation sequencing (MeRIP-seq) combined with RNA sequencing (RNA-seq). Western blot, RT-qPCR, MeRIP-qPCR, dual-luciferase reporter and RNA stability assays were performed to validate the function and mode of action of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis. The clinical relevance of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis in AML was confirmed by TCGA data analysis. RESULTS: We found that BP can inhibit AML cell proliferation and promote apoptosis in vitro, and repress AML tumor growth in vivo. Mechanistically, we found that BP downregulated ALKBH5 expression, which in turn repressed m6A demethylation of MLST8 and EIF4EBP1 mRNAs. Reduction of the m6A levels of MLST8 and EIF4EBP1 facilitated MLST8 and EIF4EBP1 mRNA decay, resulting in inhibition of AML cell proliferation. Furthermore, we found that the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis closely correlates with AML patient prognosis. CONCLUSIONS: Our data indicate that BP can inhibit acute myeloid leukemia cell proliferation by downregulating ALKBH5-mediated m6A demethylation of EIF4EBP1 and MLST8 mRNAs, which may have potential to prevent and treat this disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Desmetilasa de ARN, Homólogo 5 de AlkB , Proteínas de Ciclo Celular , Leucemia Mieloide Aguda , Péptidos , Homóloga LST8 de la Proteína Asociada al mTOR , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Desmetilación/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Péptidos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
3.
J Clin Invest ; 132(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35239512

RESUMEN

Y-box-binding protein 1 (YB-1) is a multifunctional RNA binding protein involved in virtually every step of RNA metabolism. However, the functions and mechanisms of YB-1 in one of the most aggressive cancers, glioblastoma, are not well understood. In this study, we found that YB-1 protein was markedly overexpressed in glioblastoma and acted as a critical activator of both mTORC1 and mTORC2 signaling. Mechanistically, YB-1 bound the 5'UTR of CCT4 mRNA to promote the translation of CCT4, a component of the CCT chaperone complex, that in turn activated the mTOR signaling pathway by promoting mLST8 folding. In addition, YB-1 autoregulated its own translation by binding to its 5'UTR, leading to sustained activation of mTOR signaling. In patients with glioblastoma, high protein expression of YB-1 correlated with increased expression of CCT4 and mLST8 and activated mTOR signaling. Importantly, the administration of RNA decoys specifically targeting YB-1 in a mouse xenograft model resulted in slower tumor growth and better survival. Taken together, these findings uncover a disrupted proteostasis pathway involving a YB-1/CCT4/mLST8/mTOR axis in promoting glioblastoma growth, suggesting that YB-1 is a potential therapeutic target for the treatment of glioblastoma.


Asunto(s)
Glioblastoma , Proteína 1 de Unión a la Caja Y , Regiones no Traducidas 5' , Animales , Línea Celular Tumoral , Chaperonina con TCP-1 , Glioblastoma/genética , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética , Proteína 1 de Unión a la Caja Y/genética , Proteína 1 de Unión a la Caja Y/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
4.
Cancer Sci ; 113(1): 91-108, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34741373

RESUMEN

Recent studies have reported that MLST8 is upregulated in many malignant tumors. Nevertheless, the underlying molecular mechanism is still unclear. The aim of this work was to investigate how MLST8 contributes to the development and progression of clear cell renal cell carcinoma (ccRCC). MLST8 is an oncogenic protein in the TCGA database and ccRCC clinical specimens. We also ascertain that MLST8 interacts with FBXW7, which was universally regarded as an E3 ubiquitin ligase. MLST8 can be degraded and ubiquitinated by tumor suppressor FBXW7. FBXW7 recognizes a consensus motif (T/S) PXX (S/T/D/E) of MLST8 and triggers MLST8 degradation via the ubiquitin-proteasome pathway. Strikingly, the activated cyclin dependent kinase 1 (CDK1) kinase engages in the MLST8 phosphorylation required for FBXW7-mediated degradation. In vitro, we further prove that MLST8 is an essential mediator of FBXW7 inactivation-induced tumor growth, migration, and invasion. Furthermore, the MLST8 and FBXW7 proteins are negatively correlated in human renal cancer specimens. Our findings suggest that MLST8 is a putative oncogene that functions via interaction with FBXW7, and inhibition MLST8 could be a potential future target in ccRCC treatment.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Carcinoma de Células Renales/patología , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Neoplasias Renales/patología , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo , Secuencias de Aminoácidos , Animales , Biomarcadores de Tumor/química , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Fosforilación , Proteolisis , Ubiquitinación , Regulación hacia Arriba , Homóloga LST8 de la Proteína Asociada al mTOR/química
5.
PLoS Comput Biol ; 17(9): e1008513, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34529665

RESUMEN

The PI3K/MTOR signalling network regulates a broad array of critical cellular processes, including cell growth, metabolism and autophagy. The mechanistic target of rapamycin (MTOR) kinase functions as a core catalytic subunit in two physically and functionally distinct complexes mTORC1 and mTORC2, which also share other common components including MLST8 (also known as GßL) and DEPTOR. Despite intensive research, how mTORC1 and 2 assembly and activity are coordinated, and how they are functionally linked remain to be fully characterized. This is due in part to the complex network wiring, featuring multiple feedback loops and intricate post-translational modifications. Here, we integrate predictive network modelling, in vitro experiments and -omics data analysis to elucidate the emergent dynamic behaviour of the PI3K/MTOR network. We construct new mechanistic models that encapsulate critical mechanistic details, including mTORC1/2 coordination by MLST8 (de)ubiquitination and the Akt-to-mTORC2 positive feedback loop. Model simulations validated by experimental studies revealed a previously unknown biphasic, threshold-gated dependence of mTORC1 activity on the key mTORC2 subunit SIN1, which is robust against cell-to-cell variation in protein expression. In addition, our integrative analysis demonstrates that ubiquitination of MLST8, which is reversed by OTUD7B, is regulated by IRS1/2. Our results further support the essential role of MLST8 in enabling both mTORC1 and 2's activity and suggest MLST8 as a viable therapeutic target in breast cancer. Overall, our study reports a new mechanistic model of PI3K/MTOR signalling incorporating MLST8-mediated mTORC1/2 formation and unveils a novel regulatory linkage between mTORC1 and mTORC2.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular , Péptidos y Proteínas de Señalización Intracelular , Diana Mecanicista del Complejo 2 de la Rapamicina/química , Reproducibilidad de los Resultados , Transducción de Señal , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
6.
Neuropharmacology ; 193: 108619, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34023336

RESUMEN

The newfound antidepressant efficacy of ketamine has provided opportunities for the development of new-generation, rapid-acting, glutamate-based antidepressants. We previously identified that methoxetamine (MXE), a ketamine analog, and an N-Methyl-d-aspartate (NMDA) receptor antagonist, produced rapid and sustained antidepressant effects in mice. MXE (R, S (±)-MXE) is a racemic mixture containing equal parts of S (+)-MXE and R (-)-MXE. However, studies have yet to investigate the antidepressant effects of its enantiomers. Here, we examined the potential antidepressant properties and behavioral side effects of S- and R-MXE in mice. Both S- and R-MXE showed significant NMDA receptor affinity and appreciable inhibitory activity on serotonin transporter. Also, S- and R-MXE (10 mg kg-1) exerted antidepressant effects and increased gamma waves (electroencephalography) but were inhibited by NBQX (an AMPA receptor antagonist). Subsequently, they increased mammalian target of rapamycin phosphorylation and AMPA receptor subunits GluA1 and GluA2 protein levels in the hippocampus or prefrontal cortex. Furthermore, they increased 5HT2a and 5HT2c receptor mRNA levels in the prefrontal cortex, with their antidepressant effects inhibited by ketanserin (a 5HT2a/c receptor antagonist). Taken together, S-MXE and R-MXE elicit antidepressant effects that are probably mediated via glutamatergic and serotonergic mechanisms. Unlike S-MXE, R-MXE did not induce prepulse inhibition deficits, hyperlocomotion, conditioned place preference, and locomotor sensitization, although it acutely altered motor coordination. This suggests that R-MXE induces fewer behavioral side effects and is a safer antidepressant than S-MXE. Overall, this study provides significant implications for future research on the next generation of rapid-acting, glutamate-based antidepressant drugs.


Asunto(s)
Antidepresivos/efectos adversos , Antidepresivos/farmacología , Ciclohexanonas/farmacología , Ciclohexilaminas/farmacología , Depresión/tratamiento farmacológico , Depresión/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ciclohexanonas/efectos adversos , Ciclohexilaminas/efectos adversos , Prueba de Laberinto Elevado , Células HEK293 , Suspensión Trasera , Humanos , Ketamina , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Receptores AMPA/metabolismo , Receptores de Serotonina/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
7.
Mol Oncol ; 14(8): 1850-1867, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32495998

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) regulates cell growth, cell survival, angiogenesis, metastasis of cancer cells, and cancer immune evasion by regulating gene expression as a transcription factor. However, the effect of STAT3 on translation is almost unknown. We demonstrated that STAT3 acts as a trans-acting factor for MLST8 gene expression and the protein level of mLST8, a core component of mechanistic target of rapamycin complex 1 and 2 (mTORC1/2), positively regulates the mTORC1/2 downstream pathways. Suppression of STAT3 by siRNA attenuated 4E-BP1 phosphorylation, cap-dependent translation, and cell proliferation in a variety of cancer cells. In HCT116 cells, STAT3 knockdown-induced decreases in 4E-BP1 and AKT phosphorylation levels were further attenuated by MLST8 knockdown or recovered by mLST8 overexpression. STAT3 knockdown-induced G2/M phase arrest was partially restored by co-knockdown of 4EBP1, and the attenuation of cell proliferation was enhanced by the expression of an mTORC1-mediated phosphorylation-defective mutant of 4E-BP1. ChIP and promoter mapping using a luciferase reporter assay showed that the -951 to -894 bp of MLST8 promoter seems to include STAT3-binding site. Overall, these results suggest that STAT3-driven MLST8 gene expression regulates cap-dependent translation through 4E-BP1 phosphorylation in cancer cells.


Asunto(s)
Neoplasias/genética , Biosíntesis de Proteínas/genética , Caperuzas de ARN/metabolismo , Factor de Transcripción STAT3/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Fosforilación , Regiones Promotoras Genéticas/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
8.
EBioMedicine ; 51: 102609, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31915116

RESUMEN

BACKGROUND: Endometrial carcinoma (EC) is one of the most common gynecological malignancies among women. Maternal embryonic leucine Zipper Kinase (MELK) is upregulated in a variety of human tumors, where it contributes to malignant phenotype and correlates with a poor prognosis. However, the biological function of MELK in EC progression remains largely unknown. METHODS: We explored the MELK expression in EC using TCGA and GEO databases and verified it using clinical samples by IHC methods. CCK-8 assay, colony formation assay, cell cycle assay, wound healing assay and subcutaneous xenograft mouse model were generated to estimate the functions of MELK and its inhibitor OTSSP167. qRT-PCR, western blotting, co-immunoprecipitation, chromatin immunoprecipitation and luciferase reporter assay were performed to uncover the underlying mechanism concerning MELK during the progression of EC. FINDINGS: MELK was significantly elevated in patients with EC, and high expression of MELK was associated with serous EC, high histological grade, advanced clinical stage and reduced overall survival and disease-free survival. MELK knockdown decreased the ability of cell proliferation and migration in vitro and subcutaneous tumorigenesis in vivo. In addition, high expression of MELK could be regulated by transcription factor E2F1. Moreover, we found that MELK had a direct interaction with MLST8 and then activated mTORC1 and mTORC2 signaling pathway for EC progression. Furthermore, OTSSP167, an effective inhibitor, could inhibit cell proliferation driven by MELK in vivo and vitro assays. INTERPRETATION: We have explored the crucial role of the E2F1/MELK/mTORC1/2 axis in the progression of EC, which could be served as potential therapeutic targets for treatment of EC. FUNDING: This research was supported by National Natural Science Foundation of China (No:81672565), the Natural Science Foundation of Shanghai (Grant NO:17ZR1421400 to Dr. Zhihong Ai) and the fundamental research funds for central universities (No: 22120180595).


Asunto(s)
Progresión de la Enfermedad , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estudios de Cohortes , Factor de Transcripción E2F1/metabolismo , Neoplasias Endometriales/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Modelos Biológicos , Análisis Multivariante , Naftiridinas/farmacología , Pronóstico , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
9.
Immunobiology ; 225(2): 151880, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31836304

RESUMEN

INTRODUCTION: AIM2 inflammasome activation leads to the release of IL-ß, which plays an important role in rheumatoid arthritis pathogenesis. In this work, we evaluated AIM2 expression and activity in RA patients and healthy controls. METHODS: AIM2 and RANKL expression were evaluated by flow cytometry. Inflammasome activity was determined in monocyte cultures stimulated with synthetic DNA by measuring IL-1ß levels in supernatants using an ELISA assay. The caspase-1 expression in monocytes was measured by western blot, the POP3 expression was analysed by qPCR, and serum levels of IFN-γ were evaluated using ELISA assay. RESULTS: We observed a diminution of CD14+AIM2+ cells in RA patients, associated with disease activity and evolution. Likewise, the levels of IL-1ß were increased in monocyte cultures un-stimulated and stimulated with LPS from RA patients with DAS28 ≥ 4. The Caspase-1 activity and RANKL + monocytes in RA patients were slightly increased. Finally, augmented POP3 expression and diminished IFN-γ serum levels were detected in RA patients. CONCLUSION: Our results showed that the monocytes from RA patients were prone to release IL-1ß in the absence of the AIM2 inflammasome signal. The down-regulation of AIM2 to a systemic level in RA patients might be a consequence of augmented POP3 expression and might imply the survival of pro-inflammatory cells contributing to the inflammation process.


Asunto(s)
Artritis Reumatoide/metabolismo , Proteínas de Unión al ADN/metabolismo , Inflamasomas/metabolismo , Adulto , Caspasa 1/metabolismo , Células Cultivadas , Femenino , Humanos , Inflamación/metabolismo , Interferón gamma/metabolismo , Interleucina-1beta/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Masculino , Monocitos/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
10.
Clin Transl Oncol ; 22(7): 1040-1048, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31630355

RESUMEN

OBJECTIVE: To investigate the gene expression profile of CSCs and to explore the key pathways and specific molecular signatures involved in the characteristic of CSCs. MATERIALS AND METHODS: CD133+ /CD44+ CSCs and bulk population (non-CSCs) were isolated from DU-145 cells using fluorescence-activated cell sorting (FACS). We used Illumina HumanHT-12 v4 Expression to investigate gene expression profiling of CSCs and non-CSCs. Protein-protein interaction (PPI) network analysis was performed using the STRING database. Biomarkers selected based on gene expression profiling were visually analyzed using immunofluorescence staining method. An image analysis program, ImageJ®, was used for the analysis of fluorescence intensity. RESULTS: In microarray analysis, we found that many ribosomal proteins and translation initiation factors that constitute the mTOR complex were highly expressed. PPI analysis using the 33 genes demonstrated that there was a close interaction between ribosome biogenesis, translation, and mTOR signaling. The fluorescence amount of mTOR and MLST8 were higher in CSCs compared to non-CSCs. CONCLUSIONS: The increase in a number of genes associated with ribosome biogenesis, translation, and mTOR signaling may be important to evaluate prognosis and determine treatment approach for prostate cancer (PCa). A better understanding of the molecular pathways associated with CSCs may be promising to develop targeted therapies to prolong survival in PCa.


Asunto(s)
Factores Eucarióticos de Iniciación/genética , Células Madre Neoplásicas/metabolismo , Biogénesis de Organelos , Neoplasias de la Próstata/genética , Ribosomas/genética , Serina-Treonina Quinasas TOR/genética , Transcriptoma , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Antígeno AC133/metabolismo , Línea Celular Tumoral , Factores Eucarióticos de Iniciación/metabolismo , Citometría de Flujo , Humanos , Receptores de Hialuranos/metabolismo , Masculino , Neoplasias de la Próstata/metabolismo , Mapas de Interacción de Proteínas , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Esferoides Celulares , Serina-Treonina Quinasas TOR/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
11.
Nat Commun ; 10(1): 2865, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31253771

RESUMEN

The mechanistic target of rapamycin (mTOR) kinase forms two multi-protein signaling complexes, mTORC1 and mTORC2, which are master regulators of cell growth, metabolism, survival and autophagy. Two of the subunits of these complexes are mLST8 and Raptor, ß-propeller proteins that stabilize the mTOR kinase and recruit substrates, respectively. Here we report that the eukaryotic chaperonin CCT plays a key role in mTORC assembly and signaling by folding both mLST8 and Raptor. A high resolution (4.0 Å) cryo-EM structure of the human mLST8-CCT intermediate isolated directly from cells shows mLST8 in a near-native state bound to CCT deep within the folding chamber between the two CCT rings, and interacting mainly with the disordered N- and C-termini of specific CCT subunits of both rings. These findings describe a unique function of CCT in mTORC assembly and a distinct binding site in CCT for mLST8, far from those found for similar ß-propeller proteins.


Asunto(s)
Chaperonina con TCP-1/fisiología , Proteína Reguladora Asociada a mTOR/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Microscopía por Crioelectrón , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Células Hep G2 , Humanos , Espectrometría de Masas , Modelos Moleculares , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Proteína Reguladora Asociada a mTOR/genética , Serina-Treonina Quinasas TOR/genética , Homóloga LST8 de la Proteína Asociada al mTOR/genética
12.
Aging (Albany NY) ; 11(9): 2681-2698, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31076559

RESUMEN

The aim of this study was to identify the function of the Mg2+ transporter protein solute carrier family 41 member 1 SLC41A1 in pancreatic ductal adenocarcinoma and the underlying mechanisms. A total of 27 solute carrier proteins were differentially expressed in pancreatic ductal adenocarcinoma. Three of these proteins were correlated with clinical outcomes in patients, among which SLC41A1 was downregulated in tumour. Overexpression of SLC41A1 suppressed orthotopic tumour growth in a mouse model and reduced the cell proliferation, colony formation, and invasiveness of KP3 and Panc-1 cells, which may have been associated with the increased population of apoptotic-prone cells. Overexpression of SLC41A1 reduced the mitochondrial membrane potential, induced Bax while suppressed Bcl-2 expression. Suppression of Bax abrogated the tumour-suppressive effects of SLC41A1. Furthermore, overexpression of SLC41A1 promoted Mg2+ efflux and suppressed Akt/mTOR activity, which is the upstream regulator of Bax and Bcl-2. An increase in Akt activity and supplementation with Mg2+ abolished SLC41A1-induced tumour suppression. The results of this study suggest that SLC41A1 may be a potential target for the treatment of pancreatic ductal adenocarcinoma.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas de Transporte de Catión/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/antagonistas & inhibidores , Animales , Apoproteínas , Carcinoma Ductal Pancreático/genética , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias/metabolismo , Estadificación de Neoplasias , Neoplasias Pancreáticas/genética , Embarazo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína X Asociada a bcl-2/genética , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
13.
Cancer Res ; 79(13): 3178-3184, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31085701

RESUMEN

mTOR is a serine/threonine kinase that acts in two distinct complexes, mTORC1 and mTORC2, and is dysregulated in many diseases including cancer. mLST8 is a shared component of both mTORC1 and mTORC2, yet little is known regarding how mLST8 contributes to assembly and activity of the mTOR complexes. Here we assessed mLST8 loss in a panel of normal and cancer cells and observed little to no impact on assembly or activity of mTORC1. However, mLST8 loss blocked mTOR association with mTORC2 cofactors RICTOR and SIN1, thus abrogating mTORC2 activity. Similarly, a single pair of mutations on mLST8 with a corresponding mutation on mTOR interfered with mTORC2 assembly and activity without affecting mTORC1. We also discovered a direct interaction between mLST8 and the NH2-terminal domain of the mTORC2 cofactor SIN1. In PTEN-null prostate cancer xenografts, mLST8 mutations disrupting the mTOR interaction motif inhibited AKT S473 phosphorylation and decreased tumor cell proliferation and tumor growth in vivo. Together, these data suggest that the scaffolding function of mLST8 is critical for assembly and activity of mTORC2, but not mTORC1, an observation that could enable therapeutic mTORC2-selective inhibition as a therapeutic strategy. SIGNIFICANCE: These findings show that mLST8 functions as a scaffold to maintain mTORC2 integrity and kinase activity, unveiling a new avenue for development of mTORC2-specific inhibitors.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Neoplasias de la Próstata/patología , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Ratones , Ratones Desnudos , Fosforilación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Homóloga LST8 de la Proteína Asociada al mTOR/genética
14.
Aging (Albany NY) ; 10(7): 1649-1665, 2018 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-30036188

RESUMEN

Fluoride is known to impair organism's development and function via adverse effects, and autophagy plays a regulation role in human or animal health and disease. At present, there are no reports focused on fluoride-induced autophagy in the animal and human spleen. The objective of this study was to investigate sodium fluoride (NaF)-induced splenocyte autophagy and the potential mechanism via regulation of p-mTOR in growing mice by using the methods of transmission electron microscopy (TEM), immunohistochemistry (IHC), quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. A total of 240 ICR mice were equally allocated into four groups with intragastric administration of distilled water in the control group and 12, 24, 48 mg/kg NaF solution in the experimental groups for 42 days. Results revealed that NaF increased autophagosomes or autolysosomes in spleen. Simultaneously, the autophagy marker LC3 brown punctate staining was increased with NaF dosage increase. On the other hand, NaF caused inhibition of mTOR activity, which was characterized by down-regulation of PI3K, Akt and mTOR mRNA and protein expression levels. And the suppression of mTOR activity in turn resulted in the significantly increased of ULK1 and Atg13 expression levels. Concurrently, NaF increased the levels of mRNA and protein expression of autophagy markers LC3, Beclin1, Atg16L1, Atg12, Atg5 and decreased the mRNA and protein expression levels of p62. The above-mentioned findings verify that NaF induces autophagy via mTOR signaling pathway. The inhibition of mTOR activity and alteration of autophagy-related genes and proteins are the potential molecular mechanism of NaF-induced splenocyte autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Fluoruro de Sodio/farmacología , Bazo/citología , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Beclina-1/genética , Beclina-1/metabolismo , Biomarcadores , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación , ARN Mensajero , Transducción de Señal , Bazo/ultraestructura , Homóloga LST8 de la Proteína Asociada al mTOR/genética
15.
Innate Immun ; 23(7): 615-624, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28836874

RESUMEN

Hirsutella sinensis, cultured in vitro, is an attractive substitute for Cordyceps sinensis as health supplement. The aim of this study was to demonstrate whether H. sinensis mycelium (HSM) attenuates murine pulmonary fibrosis induced by bleomycin and to explore the underlying molecular mechanisms. Using lung fibrosis modle induced by intratracheal instillation of bleomycin (BLM; 4 mg/kg), we observed that the administration of HSM reduced HYP, TGF-ß1 and the production of several pro-fibrosis cytokines (α-smooth muscle actin, fibronectin and vimentin) in fibrotic mice lung sections. Histopathological examination of lung tissues also demonstrated that HSM improved BLM-induced pathological damage. Concurrently, HSM supplementation markedly reduced the chemotaxis of alveolar macrophages and potently suppressed the expression of inflammatory cytokines. Also, HSM influenced Th1/Th2 and Th17/Treg imbalance and blocked the phosphorylation of mTOR pathway in vivo. Alveolar epithelial A549 cells acquired a mesenchymal phenotype and an increased expression of myofibroblast markers of differentiation (vimentin and fibronectin) after treatment with TGF-ß1. HSM suppressed these markers and blocked the phosphorylation of mTOR pathway in vitro. The results provide evidence supporting the use of HSM in the intervention of pulmonary fibrosis and suggest that HSM is a potential therapeutic agent for lung fibrosis.


Asunto(s)
Cordyceps/inmunología , Pulmón/fisiología , Macrófagos Alveolares/inmunología , Micelio/metabolismo , Fibrosis Pulmonar/terapia , Células A549 , Actinas/metabolismo , Animales , Bleomicina , Movimiento Celular , Suplementos Dietéticos , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Fibronectinas/metabolismo , Humanos , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
16.
J Cell Mol Med ; 21(12): 3204-3213, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28643459

RESUMEN

Increasing epidemic of type 2 diabetes mellitus (T2DM) and its comorbidities makes it urgent to understand the pathogenesis and regulatory mechanism. However, little is known about the regulatory role of lncRNAs in diabetes. Here, we constructed a T2DM-related competitive endogenous RNA (ceRNA) network (DMCN) to explore biological function of lncRNAs during the development of diabetes mellitus. This network contained 351 nodes including 98 mRNAs, 86 microRNAs and 167 lncRNAs. Functional analysis showed that the mRNAs in DMCN were annotated into some diabetes-related pathways. Furthermore, mTOR-centred subnetwork was extracted and ncRNA-involved mTOR pathway was established. Finally, we validated that NEAT1 was potentially communicated with mTOR signalling target protein mLST8 via the association with miR-181b. These findings provide significant insight into lncRNA regulatory network in T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Redes Reguladoras de Genes , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Mensajero/genética , Emparejamiento Base , Secuencia de Bases , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica , Ontología de Genes , Hepatocitos , Humanos , MicroARNs/metabolismo , Anotación de Secuencia Molecular , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
17.
Cell Signal ; 33: 98-106, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28214587

RESUMEN

Amino acids are the key activators of the mTOR complex 1 (mTORC1, mainly composed of mTOR, Raptor and mLST8) required for cell growth and proliferation. On the other hand, deprivation of amino acids induces autophagy via inhibition of mTORC1 signaling. We report here that amino acid-induced mTORC1 activity and amino acid deprivation-induced autophagy are regulated by PAQR3, a newly found tumor suppressor. At the cellular level, PAQR3 negatively regulates amino acid-induced activation of mTORC1. The N-terminal end of PAQR3 interacts with the WD domains of Raptor and mLST8 directly. PAQR3 reduces the interaction of mTOR with Raptor and mLST8, thus disrupts formation of intact mTORC1 complex. PAQR3 modulates leucine-induced alteration in cell size. In addition, PAQR3 knockdown reduces amino acid deprivation-induced autophagy. The inhibitory effect of PAQR3 knockdown on autophagy is abrogated by rapamycin treatment, indicating that PAQR3 modulates autophagy via its regulation on mTORC1 signaling. In conclusion, our finding reveals a new mode of regulation of mTORC1 signaling and autophagy by PAQR3 in response to alterations of amino acids.


Asunto(s)
Aminoácidos/deficiencia , Autofagia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Línea Celular , Tamaño de la Célula , Humanos , Ratones , Unión Proteica , Proteína Reguladora Asociada a mTOR/metabolismo , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA