Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
1.
MedComm (2020) ; 4(6): e439, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38045832

RESUMEN

Ubiquitin-specific protease 22 (USP22) has been identified as a potential marker for cancer stem cells in hepatocellular carcinoma (HCC). It can promote HCC stemness, which is considered a driver of tumorigenesis. Here, we sought to determine the role of USP22 in tumorigenesis, elucidate its underlying mechanism, and explore its therapeutic significance in HCC. As a result, we found that tissue-specific Usp22 overexpression accelerated tumorigenesis, whereas Usp22 ablation decelerated it in a c-Myc/NRasGV12-induced HCC mouse model and that the mammalian target of rapamycin complex 1 (mTORC1) pathway was activated downstream. USP22 overexpression resulted in increased tumorigenic properties that were reversed by rapamycin in vitro and in vivo. In addition, USP22 activated mTORC1 by deubiquitinating FK506-binding protein 12 (FKBP12) and activated mTORC1, in turn, further stabilizing USP22 by inhibiting autophagic degradation. Clinically, HCC patients with high USP22 expression tend to benefit from mTOR inhibitors after liver transplantation (LT). Our results revealed that USP22 promoted tumorigenesis and progression via an FKBP12/mTORC1/autophagy positive feedback loop in HCC. Clinically, USP22 may be an effective biomarker for selecting eligible recipients with HCC for anti-mTOR-based therapy after LT.

2.
J Vet Med Sci ; 85(8): 820-827, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37407446

RESUMEN

Canine lymphoma is the most common cancer in dogs and has a poor prognosis. We recently found that the endocytosis inhibitor dynasore suppresses the viability of human cancer cell lines, especially hematopoietic cancers, by inducing apoptosis. In the present study, we examined the anticancer effects of dynasore on five previously established canine lymphoma cell lines (CLBL-1, Ema, Nody-1, CLC, and GL-1). Dynasore suppressed cell viability in these canine lymphoma cell lines more effectively than in human cancer cell lines. It also induced apoptosis in CLBL-1 and Ema cells but not in peripheral blood mononuclear cells in healthy dogs or in Madin-Darby canine kidney (MDCK) cells, suggesting that the ability of dynasore to induce apoptosis is cancer-specific. Furthermore, dynasore induced a DNA damage response in CLBL-1 and Ema cells, suggesting that it acts as a genotoxic agent in canine lymphoma cell lines. These findings suggest that endocytosis inhibitors may provide a new anticancer treatment for canine lymphoma.


Asunto(s)
Enfermedades de los Perros , Linfoma , Animales , Perros , Humanos , Leucocitos Mononucleares/metabolismo , Línea Celular Tumoral , Linfoma/tratamiento farmacológico , Linfoma/veterinaria , Linfoma/genética , Apoptosis , Endocitosis , Enfermedades de los Perros/genética
3.
J Vet Med Sci ; 85(7): 772-780, 2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37225457

RESUMEN

Mechanistic/mammalian target of rapamycin complex 1 (mTORC1) is a serine/threonine kinase that plays a major role in cell metabolism. Although mTORC1 inhibitors are known to exert immunosuppressive effects, their effects on immune cells are not fully understood. In the present study, we examined the involvement of mTORC1 in the differentiation and functions of macrophages using THP-1 cells, which are derived from human monocytic leukemia and differentiate into macrophage-like cells upon treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA). We also examined the effects of two mTOR inhibitors, Torin 1 and rapamycin, on TPA-stimulated THP-1 cells. Although mTORC1 activation was observed upon TPA stimulation, mTOR inhibitors did not affect TPA-induced morphological changes or expression of the general macrophage marker, CD11b. In contrast, phagocytosis and fluid endocytosis were significantly impaired by the mTOR inhibitors. Endocytosis suppression was observed when mTOR inhibitors were added during differentiation, but not before or after differentiation, suggesting that endocytosis suppression altered the direction of differentiation. Furthermore, mTOR inhibitors altered the expression of M1/M2 polarization markers. These results suggest that the immunosuppressive effects of mTOR inhibitors may involve the suppression of macrophage endocytosis caused by abnormal cell differentiation.


Asunto(s)
Inhibidores mTOR , Serina-Treonina Quinasas TOR , Humanos , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células THP-1 , Diferenciación Celular , Fagocitosis , Mamíferos
4.
Chemosphere ; 320: 138071, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36754296

RESUMEN

Aflatoxin B1 (AFB1) is a common contaminant in many foodstuffs and is considered a public health concern worldwide due to its hepatotoxicity caused by lipid metabolism disorders. However, the molecular mechanism underlying AFB1-induced lipotoxicity-dependent liver injury via regulating cholesterol metabolism remains unclear. We established a cholesterol trafficking disorder-mediated hepatic lipotoxicity model with AFB1 mixture exposure in vitro (HepaRG and HepG2 cells, 1.6 µM for 36 h) and in vivo (C57BL/6 mice, 3 mg kg-1, i.g., every other day for 6 weeks). In vitro, the interaction between lysosomal Niemann-Pick type C1 (NPC1) protein and mitochondrial translocator protein (TSPO) regulated lipotoxicity induced by AFB1 mixture exposure, including lysosomal membrane permeabilization and mitochondria-dependent necroptosis. Moreover, the downregulation of lysosomal Ras-associated protein 7a (Rab7a) enhanced the mammalian target of rapamycin complex 1 (mTORC1)-mediated disorders of cholesterol trafficking from the lysosome to mitochondria. Furthermore, cholesterol trafficking disorder-mediated hepatic lipotoxicity induced by the low-dose level of AFB1 exposure was relieved by genetic or pharmaceutic activation of Rab7a to inhibit mTORC1 in vitro and ex vivo. In vivo, mTORC1 inhibitor (Torin1, 4 mg kg-1, i.p., every other day for 3 weeks) alleviated the cholesterol trafficking disorder-mediated hepatic lipotoxicity via upregulating the molecular machinery of lysosomes and mitochondria contact mediated by NPC1 and TSPO interaction in the low dose of AFB1 exposure. Altogether, our data suggested a novel mechanism that lysosomal Rab7a-mTORC1 signaling determined the cholesterol trafficking regulated by NPC1-TSPO from the lysosome to mitochondria, which promoted hepatic lipotoxicity via lysosomal quality control and mitochondria-dependent necroptosis signaling pathways in chemical mixture exposure.


Asunto(s)
Aflatoxina B1 , Hígado , Animales , Ratones , Aflatoxina B1/metabolismo , Colesterol/metabolismo , Hígado/metabolismo , Lisosomas/metabolismo , Mamíferos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteínas de Unión a GTP rab7/metabolismo
5.
J Agric Food Chem ; 70(8): 2589-2599, 2022 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-35180345

RESUMEN

Autophagy, an evolutionarily conserved process, is intricately involved in many aspects of human health and a variety of human diseases, including cancer. Discovery of small-molecule autophagy modulators with potent anticancer effect would be of great significance. To this end, a natural product library consisting of 170 natural compounds were screened as autophagy modulators with potent cytotoxicity in our present study. Among these compounds, gossypol acetate (GAA), the mostly used medicinal form of gossypol, was identified. GAA effectively increased the number of autophagic puncta in GFP-LC3B-labeled 293T cells and significantly decreased cell viability in different cancer cells. In A549 cells, GAA at concentrations below 10 µM triggered caspase-independent cell death via targeting autophagy, as evidenced by elevated LC3 conversion and decreased p62/SQSTM1 levels. Knocking down of LC3 significantly attenuated GAA-induced cell death. Mechanistically, GAA at low concentrations induced autophagy through targeting AMPK-mTORC1-ULK1 signaling. Interestingly, high concentrations of GAA induced LC3 conversion, p62 accumulation, and yellow autophagosome formation, indicating that GAA at high concentrations blocked autophagic flux. Mechanistically, GAA decreased intracellular ATP level and suppressed lysosome activity. Exogenous ATP partially reversed the inhibitory effect of GAA on autophagy, suggesting that decreased ATP level and lysosome activity might be involved in the blocking of autophagy flux by GAA. Collectively, our present study reveals the mechanisms by which GAA modulates autophagy and illustrates whether autophagy regulation by GAA is functionally involved in GAA-induced cancer cell death.


Asunto(s)
Gosipol , Neoplasias , Proteínas Quinasas Activadas por AMP/metabolismo , Acetatos/farmacología , Apoptosis , Autofagia , Gosipol/farmacología , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética
6.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-940383

RESUMEN

ObjectiveBy observing the effect of Xiaoluowan on phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin complex 1 (mTORC1) pathway in experimental goiter rats, this study aims to explore its therapeutic effect on experimental goiter rats. MethodSixty 5-month-old SD rats of SPF grade were purchased, half males and half females, of which 10 were used as a normal group, and the remaining rats were administrated with propylthiouracil (PTU) solution to induce nodular goiter. After successful modeling, rats were randomly divided into a model group, levothyroxine sodium tablets group, Xiaoluowan low-dose group, medium-dose group, and high-dose group, ten rats each. The levothyroxine sodium tablets group was given 15 μg·kg-1 levothyroxine sodium tablets by gavage. The Xiaoluowan low-, medium-, and high-dose groups were given (ig) Xiaoluowan low-dose (10 g·kg-1), medium-dose (20 g·kg-1), and high-dose (30 g·kg-1) Xiaoluowan, and the normal group and model group were administered (ig) with the same volume of 0.9% sodium chloride solution. Four weeks after the intervention, rats were sacrificed by routine intraperitoneal anesthesia using 5% phenobarbital. Subsequently, the histopathology was observed under a microscope, and serum thyroid hormone levels were measured using a Roche electrochemiluminescence immunoassay analyzer. Serum cytokines were detected by enzyme-linked immunosorbent assay (ELISA), and neurotransmitters were measured using a high-performance liquid chromatograph. The protein level of PI3K/Akt/mTORC1 pathway was determined by Western blot. ResultAs compared with the normal group, the levels of basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF-1), 5-hydroxytryptamine (5-HT), and thyroid stimulating hormone (TSH) were increased, and PI3K, Akt, and mTORC1 protein levels were up-regulated in the model group, while the levels of norepinephrine (NE), triiodothyronine (T3), tetraiodothyronine (T4), free triiodothyronine (FT3), and free thyroid hormone (FT4) were decreased (P<0.05). Compared with the model group, the levothyroxine sodium tablets group, and Xiaoluowan low-, medium-, and high-dose groups exhibited reduced levels of bFGF, VEGF, IGF-1, 5-HT, and TSH, and down-regulated PI3K, Akt, and mTORC1 protein levels, and increased NE, T3, T4, FT3, and FT4 levels (P<0.05). ConclusionXiaoluowan may act on the PI3K/Akt/mTORC1 signaling pathway to play its role in the treatment of nodular goiter, and it is dose-dependent.

7.
Pharmaceutics ; 13(12)2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34959437

RESUMEN

The inhibition of the mammalian target of rapamycin complex 1 (mTORC1) by everolimus (RAD001) was recently shown to enhance the tumor uptake of radiolabeled minigastrin. In this paper, we investigate if this finding can improve the in vivo therapeutic response to [177Lu]Lu-PP-F11N treatment. The N-terminal DOTA-conjugated gastrin analogue PP-F11N (DOTA-(DGlu)6-Ala-Tyr-Gly-Trp-Nle-Asp-Phe) was used to evaluate treatment efficacy in the human A431/CCKBR xenograft nude mouse model in combination with RAD001. Both RAD001 and [177Lu]Lu-PP-F11N single treatments as well as their combination inhibited tumor growth and increased survival. In concomitantly treated mice, the average tumor size and median survival time were significantly reduced and extended, respectively, as compared to the monotherapies. The histological analysis of kidney and stomach dissected after treatment with RAD001 and [177Lu]Lu-PP-F11N did not indicate significant adverse effects. In conclusion, our study data demonstrate the potential of mTORC1 inhibition to substantially improve the therapeutic efficacy of radiolabeled minigastrin analogues in CCKBR-positive cancers.

8.
Adv Exp Med Biol ; 1275: 35-69, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33539011

RESUMEN

Recently, aging has been tried to be explained with large numbers of theories, but none of them can elucidate the changes occurring in the aging process alone. A unified theory encompassing the mechanisms of genetic factors and repair systems in aging is becoming increasingly required. Almost 37 protein kinases contribute to all processes of aging and senescence. Furthermore, these kinases not only regulate the large number of metabolic pathways related to aging processes, but also control these pathways through 12 checkpoints. Thus, in this chapter, the metabolic targets of protein kinases signal transduction pathways were discussed in terms of the aging perspective under five headings, which are the indispensable stages of the aging process. Although the most popular classical aging theories have been stated as DNA damage theory, mitochondrial theory, free radical theory, and telomere theory, it was concluded that the aging process is controlled by protein kinases regardless of the different theories.


Asunto(s)
Daño del ADN , Proteínas Quinasas , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de Ciclo Celular/metabolismo , Reparación del ADN , Proteínas Quinasas/genética , Transducción de Señal
9.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-906508

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a common, lethal interstitial lung disease characterized by airway remodeling, inflammation, alveolar destruction, and fibrosis. The mammalian target of rapamycin complex 1/4E binding protein 1 (mTORC1/4E-BP1) axis is closely related to the expression of collagen by fibroblasts, and its role in pulmonary fibrosis remains to be further elucidated. Traditional Chinese medicine (TCM) has shown promising efficacy in improving the lung function, exercise capacity, and quality of life in patients with IPF. The theory of "same treatment for different diseases" provides a TCM theoretical basis for the treatment of pulmonary fibrosis with Bupleuri Radix, while the research in western medicine has preliminarily shown that both the formulation and single herb as well as the active ingredients of Bupleuri Radix have good therapeutic effects on pulmonary fibrosis. Therefore, this review will elaborate on the role of the mTORC1/4E-BP1 axis in the pathomechanism of IPF, as well as the research results of the active components of Bupleuri Radix on the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin protein(PI3K/AKT/mTOR) pathway, so as to provide a reference for the treatment and drug development of IPF.

10.
Biol Pharm Bull ; 43(12): 1983-1986, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33268720

RESUMEN

The mechanistic/mammalian target of rapamycin complex-1 (mTORC1) integrates multiple signaling pathways and regulates various cellular processes. Tuberous sclerosis complex 1 (Tsc1) and complex 2 (Tsc2) are critical negative regulators of mTORC1. Mouse genetic studies, including ours, have revealed that inactivation of mTORC1 in undifferentiated mesenchymal cells and chondrocytes leads to severe skeletal abnormalities, indicating a pivotal role for mTORC1 in skeletogenesis. Here, we show that hyperactivation of mTORC1 influences skeletal development through its expression in undifferentiated mesenchymal cells at the embryonic stage. Inactivation of Tsc1 in undifferentiated mesenchymal cells by paired-related homeobox 1 (Prx1)-Cre-mediated recombination led to skeletal abnormalities in appendicular skeletons. In contrast, Tsc1 deletion in chondrocytes using collagen type II α1 (Col2a1)-Cre or in osteoprogenitors using Osterix (Osx)-Cre did not result in skeletal defects in either appendicular or axial skeletons. These findings indicate that Tsc complex-mediated chronic overactivation of mTORC1 influences skeletal development at the embryonic stage through its expression in undifferentiated mesenchymal cells but not in chondrocytes or osteoprogenitors.


Asunto(s)
Desarrollo Óseo/fisiología , Condrocitos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/deficiencia , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética
11.
J Cell Physiol ; 235(12): 9958-9973, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32474911

RESUMEN

Nephron loss stimulates residual functioning nephrons to undergo compensatory growth. Excessive nephron growth may be a maladaptive response that sets the stage for progressive nephron damage, leading to kidney failure. To date, however, the mechanism of nephron growth remains incompletely understood. Our previous study revealed that class III phosphatidylinositol-3-kinase (Pik3c3) is activated in the remaining kidney after unilateral nephrectomy (UNX)-induced nephron loss, but previous studies failed to generate a Pik3c3 gene knockout animal model. Global Pik3c3 deletion results in embryonic lethality. Given that renal proximal tubule cells make up the bulk of the kidney and undergo the most prominent hypertrophic growth after UNX, in this study we used Cre-loxP-based approaches to demonstrate for the first time that tamoxifen-inducible SLC34a1 promoter-driven CreERT2 recombinase-mediated downregulation of Pik3c3 expression in renal proximal tubule cells alone is sufficient to inhibit UNX- or amino acid-induced hypertrophic nephron growth. Furthermore, our mechanistic studies unveiled that the SLC34a1-CreERT2 recombinase-mediated Pik3c3 downregulation inhibited UNX- or amino acid-stimulated lysosomal localization and signaling activation of mechanistic target of rapamycin complex 1 (mTORC1) in the renal proximal tubules. Moreover, our additional cell culture experiments using RNAi confirmed that knocking down Pik3c3 expression inhibited amino acid-stimulated mTORC1 signaling and blunted cellular growth in primary cultures of renal proximal tubule cells. Together, both our in vivo and in vitro experimental results indicate that Pik3c3 is a major mechanistic mediator responsible for sensing amino acid availability and initiating hypertrophic growth of renal proximal tubule cells by activation of the mTORC1-S6K1-rpS6 signaling pathway.


Asunto(s)
Fosfatidilinositol 3-Quinasas Clase III/genética , Túbulos Renales Proximales/crecimiento & desarrollo , Riñón/efectos de los fármacos , Nefronas/crecimiento & desarrollo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Animales , Fosfatidilinositol 3-Quinasas Clase III/antagonistas & inhibidores , Proteínas de la Matriz Extracelular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Integrasas/genética , Riñón/crecimiento & desarrollo , Riñón/patología , Riñón/cirugía , Túbulos Renales Proximales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Nefrectomía , Nefronas/metabolismo , Fosforilación/genética , Proteína-Lisina 6-Oxidasa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología
12.
Biosci Rep ; 40(4)2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32285908

RESUMEN

Autophagy is dependent upon lysosomes, which fuse with the autophagosome to complete the autophagic process and whose acidic interior permits the activity of their intraluminal degradative enzymes. Chloroquine (CQ) and bafilomycin A1 (BafA) each cause alkalinisation of the lumen and thus impair lysosomal function, although by distinct mechanisms. CQ diffuses into lysosomes and undergoes protonation, while BafA inhibits the ability of the vacuolar type H+-ATPase (v-ATPase) to transfer protons into the lysosome. In the present study, we examine the impact of CQ and BafA on the activity of mammalian target of rapamycin complex 1 (mTORC1), inhibition of which is an early step in promoting autophagy. We find each compound inhibits mTORC1 signalling, without affecting levels of protein components of the mTORC1 signalling pathway. Furthermore, these effects are not related to these agents' capacity to inhibit autophagy or the reduction in amino acid supply from lysosomal proteolysis. Instead, our data indicate that the reduction in mTORC1 signalling appears to be due to the accumulation of lysosomal storage material. However, there are differences in responses to these agents, for instance, in their abilities to up-regulate direct targets of transcription factor EB (TFEB), a substrate of mTORC1 that drives transcription of many lysosomal and autophagy-related genes. Nonetheless, our data imply that widely used agents that alkalinise intralysosomal pH are mimetics of acute lysosomal storage disorders (LSDs) and emphasise the importance of considering the result of CQ and BafA on mTORC1 signalling when interpreting the effects of these agents on cellular physiology.


Asunto(s)
Cloroquina/farmacología , Lisosomas/efectos de los fármacos , Macrólidos/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Células A549 , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagia/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Enfermedades por Almacenamiento Lisosomal/patología , Lisosomas/química , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores , ATPasas de Translocación de Protón Vacuolares/metabolismo
13.
J Biol Chem ; 295(21): 7418-7430, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32312748

RESUMEN

Autophagy and lysosomal activities play a key role in the cell by initiating and carrying out the degradation of misfolded proteins. Transcription factor EB (TFEB) functions as a master controller of lysosomal biogenesis and function during lysosomal stress, controlling most but, importantly, not all lysosomal genes. Here, we sought to better understand the regulation of lysosomal genes whose expression does not appear to be controlled by TFEB. Sixteen of these genes were screened for transactivation in response to diverse cellular insults. mRNA levels for lysosomal-associated membrane protein 3 (LAMP3), a gene that is highly up-regulated in many forms of cancer, including breast and cervical cancers, were significantly increased during the integrated stress response, which occurs in eukaryotic cells in response to accumulation of unfolded and misfolded proteins. Of note, results from siRNA-mediated knockdown of activating transcription factor 4 (ATF4) and overexpression of exogenous ATF4 cDNA indicated that ATF4 up-regulates LAMP3 mRNA levels. Finally, ChIP assays verified an ATF4-binding site in the LAMP3 gene promoter, and a dual-luciferase assay confirmed that this ATF4-binding site is indeed required for transcriptional up-regulation of LAMP3 These results reveal that ATF4 directly regulates LAMP3, representing the first identification of a gene for a lysosomal component whose expression is directly controlled by ATF4. This finding may provide a key link between stresses such as accumulation of unfolded proteins and modulation of autophagy, which removes them.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Proteínas de Membrana de los Lisosomas/biosíntesis , Proteínas de Neoplasias/biosíntesis , ARN Mensajero/biosíntesis , Elementos de Respuesta , Transcripción Genética , Regulación hacia Arriba , Células A549 , Factor de Transcripción Activador 4/genética , Humanos , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Neoplasias/genética , ARN Mensajero/genética
14.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-872783

RESUMEN

Objective::To observe the effect of Shenling Baizhusan(SBS)on the mammalian target of rapamycin complex 1 (mTORC1)/signal transducers and activators of transcription 3 (STAT3) pathway in liver hepatocyte of nonalcoholic fatty liver disease(NAFLD)rats induced by high fat diet, in order to reveal the mechanism of SBS against rat NAFLD from the perspective of inflammation. Method::Totally 80 SD rats were randomly divided into 4 groups, normal control group, model group, high-dose SBP group(30 g·kg-1), and low-dose SBS group(10 g·kg-1), with 20 rats in each group. The rats of NAFLD model were established by being fed with high-fat diets for 8 weeks, and the treatment groups were fed with high or low dose of SBS respectively. After treatment for 8 weeks, blood and liver samples of rats were collected. Alanine aminotransferase (ALT), aspartate aminotransferase(AST), total cholesterol (TC), triglyceride(TG), high-density lipoprotein cholesterol(HDL-C)and low-density lipoprotein cholesterol(LDL-C)levels in blood serum were detected with automatic biochemical analyzer. The liver tissues were observed by oil red O and hematoxylin-eosin (HE) staining. Hepatocytes were isolated by type Ⅳ collagenase perfusion in vitro. Tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-5 and IL-6 in hepatocytes were detected by enzyme-linked immunosorbent assay (ELISA), and the relevant gene and proteins expressions of mTORC1 and STAT3 in hepatocytes were detected by Real-time fluorescent quantitative polymerase chain reaction (Real-time PCR) and Western blot detection respectively. Result::Compared with the normal control group, the serum levels of TG, TC, AST, ALT and LDL-C were increased significantly, the levels of TNF-α, IL-1β, IL-5 and IL-6 in hepatocytes were increased significantly, and the expression levels of mTORC1, STAT3 mRNA and proteins in hepatocytes were increased significantly(P<0.01). Compared with the model group, the hepatic lipid accumulation of the medicine intervention group was relieved significantly, the serum levels of AST, ALT, TG and LDL-C were decreased significantly, the expression levels of TNF-α, IL-1β, IL-5 and IL-6 of hepatocytes were decreased significantly, and the expressions of mTORC1, STAT3 mRNA and proteins in hepatocytes were decreased significantly(P<0.05, P<0.01). In the high-dose SBS group, the effects in improving the lipid accumulation and inhibiting the inflammatory reaction were better than those of the low-dose SBS group, and the expressions of mTORC1 and STAT3 genes and proteins in hepatocytes were significantly decreased (P<0.05, P<0.01). Conclusion::SBS can improve the fat metabolism disorder and reduce liver lipid accumulation and inflammatory reaction in NAFLD rats induced by high-fat diet. The mechanism may be correlated with the inhibition of mTORC1/STAT3 pathway relating to genes and protein expression in hepatocytes.

15.
J Cell Biochem ; 120(3): 2886-2896, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29266342

RESUMEN

Neurofibromatosis type I (NF1), which is caused by mutations in the NF1 gene, is a common autosomal dominant genetic disease leading to skeletal abnormalities. Both NF1 gene and mammalian target of rapamycin complex 1 (mTORC1) signaling are associated with the osteogenic differentiation of bone marrow stem cells (BMSCs). In this study, we hypothesized that mTORC1 signaling is involved in NF1-modulated osteoblast differentiation of BMSCs. Human BMSCs were cultured in an osteogenic induction medium. The expression of NF1 was either inhibited or overexpressed by transfecting NF1 with a specific small interfering RNA (siRNA) or pcDNA3.0 plasmid, respectively. In addition, an mTORC1 signaling inhibitor and agonist were used to investigate the effects of mTORC1 on NF1-modulated osteogenic differentiation of BMSCs. The results indicated that inhibiting the expression of NF1 with siRNA significantly decreased the mRNA levels of NF1, whereas overexpressing the expression of NF1 with pcDNA3.0 plasmid significantly increased the mRNA levels of NF1 at days 3, 7, 14 and 21 after culture. We observed reduced osteogenic differentiation and cell proliferation in the NF1-siRNA group and enhanced osteogenic differentiation and cell proliferation of BMSCs in the NF1-pcDNA3.0 group. The activity of mTORC1 signaling (p-mTORC1, p-S6K1, and p-4EBP1) was significantly upregulated in the NF1-siRNA group and significantly inhibited in the NF1-pcDNA3.0 group, 7 and 14 days after culture. The effects of NF1-siRNA and NF1-pcDNA3.0 on osteogenic differentiation of BMSCs and cell proliferation were reversed by mTORC1 inhibitor and agonist, respectively. In conclusion, NF1 modulates osteogenic differentiation and cell proliferation of human BMSCs and mTORC1 signaling is essential for this process.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células Madre Mesenquimatosas/citología , Neurofibromina 1/genética , Osteogénesis , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/metabolismo , Morfolinas/farmacología , Neurofibromina 1/antagonistas & inhibidores , Neurofibromina 1/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos
16.
Neurochem Res ; 43(2): 465-476, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29238892

RESUMEN

Epilepsy is among the most common neurological disorders. Recurrent seizures result in neuronal death, cognitive deficits and intellectual disabilities in children. Currently, recombinant human erythropoietin (rhEPO) is considered to play a neuroprotective role in nervous system disorders. However, the precise mechanisms through which rhEPO modulates epilepsy remain unknown. Based on results from numerous studies, we hypothesized that rhEPO protects against hippocampal damage in developing rats with seizures probably by modulating autophagy via the ribosomal protein S6 (S6) in a time-dependent manner. First, we observed that rats with recurrent seizures displayed neuronal loss in the hippocampal CA1 region. Second, rhEPO injection reduced neuronal loss and decreased the number of apoptotic cells in the hippocampal CA1 region. Moreover, rhEPO increased the Bcl-2 protein expression levels and decreased the ratio of cleaved caspase-3/caspase-3 in the hippocampus. Finally, rhEPO modulated autophagy in the hippocampus in a time-dependent manner, probably via the S6 protein. In summary, rhEPO protects against hippocampal damage in developing rats with seizures by modulating autophagy in a time-dependent manner, probably via the S6 protein. Consequently, rhEPO is a likely drug candidate that is capable of attenuating brain injury.


Asunto(s)
Autofagia/efectos de los fármacos , Eritropoyetina/farmacología , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Epilepsia/tratamiento farmacológico , Epilepsia/metabolismo , Hipocampo/metabolismo , Humanos , Masculino , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Factores de Tiempo
17.
Adv Exp Med Biol ; 960: 443-467, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28585211

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is in parallel with the obesity epidemic and it is the most common cause of liver diseases. The development of hepatic steatosis in majority of patients is linked to dietary fat ingestion. NAFLD is characterized by excess accumulation of triglyceride in the hepatocyte due to both increased inflow of free fatty acids and de novo hepatic lipogenesis. Insulin resistance with the deficiency of insulin receptor substrate-2 (IRS-2)-associated phosphatidylinositol 3-kinase (PI3K) activity causes an increase in intracellular fatty acid-derived metabolites such as diacylglycerol, fatty acyl CoA or ceramides. Lipotoxicity-related mechanism of NAFLD could be explained still best by the "double-hit" hypothesis. Insulin resistance is the major mechanism in the development and progression of NAFLD/Non-alcoholic steatohepatitis (NASH). Metabolic oxidative stress, autophagy, and inflammation induce NASH progression. In the "first hit" the hepatic concentrations of diacylglycerol increase with rising saturated liver fat content in human NAFLD. Activities of mitochondrial respiratory chain complexes are decreased in liver tissue of patients with NASH. Furthermore, hepatocyte lipoapoptosis is a critical feature of NASH. In "second hit" reduced glutathione levels due to oxidative stress lead to overactivation of c-Jun N-terminal kinase (JNK)/c-Jun signaling that induces cell death in the steatotic liver. Accumulation of toxic levels of reactive oxygen species (ROS) is caused by the ineffectual cycling of the endoplasmic reticulum (ER) oxidoreductin (Ero1)-protein disulfide isomerase oxidation cycle through the downstream of the inner membrane mitochondrial oxidative metabolism and Kelch like-ECH-associated protein 1 (Keap1)- Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/fisiología , Transducción de Señal/fisiología , Triglicéridos/metabolismo
18.
J Dairy Sci ; 100(7): 5909-5921, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28478014

RESUMEN

The phosphorylation of mammalian target of rapamycin complex 1 (mTORC1) components and integrated stress response networks in the mammary glands of lactating cows have not accounted for the stimulation of milk protein yield by chronic supplementation with AA or glucose. Faster milk protein synthesis could be a consequence of increased milk protein mRNA per cell, the number of ribosomes per cell, the secretory capacity of cells, or the mammary cell number. To investigate these 4 possibilities using a translational and transcriptional approach, we performed protein and gene expression analyses of mammary and longissimus dorsi tissue collected from lactating dairy cows after 5 d of abomasal infusion with saline or 844 or 1,126 g/d of an essential AA (EAA) mixture, with and without 1,000 g/d glucose. Infusion with EAA increased milk protein yield but did not affect the phosphorylation of mTORC1-related proteins in the mammary gland. In skeletal muscle, phosphorylation of 4EBP1 (eIF4E-binding protein 1) increased in response to both EAA and glucose, and phosphorylated S6K1 (70-kDa ribosomal protein S6 kinase) increased with glucose. In response to EAA, mammary mRNA expression of the marker genes for milk proteins, ribosome biogenesis, and cell proliferation were not upregulated. Instead, reciprocal regulation of 2 arms of the unfolded protein response occurred. Infusion of EAA for 5 d activated XBP1 (X-box binding protein 1) mRNA, encoding a transcription factor for endoplasmic reticulum biogenesis, and it decreased the mRNA expression of genes encoding pro-apoptotic protein CHOP (C/EBP homologous protein) and downstream GADD34 (growth arrest and DNA damage-inducible 34). These findings implicate non-stress-related, adaptive capabilities of the unfolded protein response in the long-term nutritional regulation of milk protein yield in lactating dairy cows.


Asunto(s)
Aminoácidos Esenciales/farmacología , Bovinos , Glándulas Mamarias Animales/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Aminoácidos , Animales , Femenino , Lactancia , Leche , Proteínas de la Leche , Transducción de Señal , Serina-Treonina Quinasas TOR
19.
Cell Signal ; 32: 24-35, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28089905

RESUMEN

Lysosomal localization of mammalian target of rapamycin complex 1 (mTORC1) is a critical step for activation of the molecule. Rag GTPases are essential for this translocation. Here, we demonstrate that Nudix-type motif 2 (NUDT2) is a novel positive regulator of mTORC1 activation. Activation of mTORC1 is impaired in NUDT2-silenced cells. Mechanistically, NUDT2 binds to Rag GTPase and controls mTORC1 translocation to the lysosomal membrane. Furthermore, NUDT2-dependent mTORC1 regulation is critical for proliferation of breast cancer cells, as NUDT2-silenced cells arrest in G0/G1 phases. Taken together, these results show that NUDT2 is a novel complex formation enhancing factor regulating mTORC1-Rag GTPase signaling that is crucial for cell growth control.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Monoéster Fosfórico Hidrolasas/metabolismo , Aminoácidos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fase G1/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Insulina/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ensayo de Tumor de Célula Madre
20.
Endocrine ; 54(1): 217-224, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27503318

RESUMEN

Luteinizing hormone/human chorionic gonadotropin stimulates progesterone biosynthesis in the corpus luteum by activating cyclic adenosine monophosphate/protein kinase A cascade. Recent studies have shown that cyclic adenosine monophosphate-mediated activation of protein kinase A interacts with the mammalian target of rapamycin signaling pathways. Furthermore, the use of mammalian target of rapamycin inhibitors for immunosuppression in transplant patients has shown adverse effects in reproductive functions. This study examined whether the mammalian target of rapamycin pathway plays any role in luteinizing hormone-mediated regulation of progesterone production. Human granulosa lutein cells were isolated from follicular aspirates of women undergoing in vitro fertilization. Cells were cultured for 72 h and treated with human chorionic gonadotropin (50 ng/ml) for different time periods with or without pretreatment with mammalian target of rapamycin complex 1 inhibitor, rapamycin, (20 nM) for 1 h. Expression of steroidogenic enzymes, including steroidogenic acute regulatory protein, cholesterol side chain cleavage enzyme, and 3ß-hydroxysteroid dehydrogenase type 1 messenger RNA, were examined by real-time polymerase chain reaction after 6 h of human chorionic gonadotropin treatment. Expressions of phospho-ribosomal protein S6 kinase and cholesterol side chain cleavage enzyme were analyzed after 15 min and 24 h of human chorionic gonadotropin treatment, respectively. Progesterone production was analyzed by an enzyme immunoassay kit after human chorionic gonadotropin (50 ng/ml) or forskolin (10 µM) treatment for 24 h. Treatment with human chorionic gonadotropin increased the expression of downstream targets of mammalian target of rapamycin complex 1, as well as cholesterol side chain cleavage enzyme, 3ß-hydroxysteroid dehydrogenase type 1 and steroidogenic acute regulatory protein messenger RNAs. These increases were inhibited by rapamycin pretreatment. Increased progesterone production in response to treatment with human chorionic gonadotropin or forskolin was also blocked by rapamycin pretreatment. Our findings support a role for mammalian target of rapamycin complex 1 in regulating steroidogenesis in human granulosa lutein cells.


Asunto(s)
Gonadotropina Coriónica/farmacología , Células Lúteas/efectos de los fármacos , Células Lúteas/metabolismo , Complejos Multiproteicos/metabolismo , Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Femenino , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA