141 results on '"PRAS40"'
Search Results
2. mTOR/miR-142-3p/PRAS40 signaling cascade is critical for tuberous sclerosis complex-associated renal cystogenesis
- Author
-
Shuyun Zhao, Shuai Hao, Jiasheng Zhou, Xinran Chen, Tianhua Zhang, Zhaolai Qi, Ting Zhang, Sajid Jalal, Chuanxin Zhai, Lu Yin, Yufei Bo, Hongming Teng, Yue Wang, Dongyan Gao, Hongbing Zhang, and Lin Huang
- Subjects
Tuberous sclerosis complex ,mTOR ,PRAS40 ,Renal cystogenesis ,miRNA ,Cytology ,QH573-671 - Abstract
Abstract Background Patients with tuberous sclerosis complex (TSC) develop renal cysts and/or angiomyolipomas (AMLs) due to inactive mutations of either TSC1 or TSC2 and consequential mTOR hyperactivation. The molecular events between activated mTOR and renal cysts/AMLs are still largely unknown. Methods The mouse model of TSC-associated renal cysts were constructed by knocking out Tsc2 specifically in renal tubules (Tsc2 f/f; ksp-Cre). We further globally deleted PRAS40 in these mice to investigate the role of PRAS40. Tsc2 −/− cells were used as mTOR activation model cells. Inhibition of DNA methylation was used to increase miR-142-3p expression to examine the effects of miR-142-3p on PRAS40 expression and TSC-associated renal cysts. Results PRAS40, a component of mTOR complex 1, was overexpressed in Tsc2-deleted cell lines and mouse kidneys (Tsc2 f/f; ksp-Cre), which was decreased by mTOR inhibition. mTOR stimulated PRAS40 expression through suppression of miR-142-3p expression. Unleashed PRAS40 was critical to the proliferation of Tsc2 −/− cells and the renal cystogenesis of Tsc2 f/f; ksp-Cre mice. In contrast, inhibition of DNA methylation increased miR-142-3p expression, decreased PRAS40 expression, and hindered cell proliferation and renal cystogenesis. Conclusions Our data suggest that mTOR activation caused by TSC2 deletion increases PRAS40 expression through miR-142-3p repression. PRAS40 depletion or the pharmacological induction of miR-142-3p expression impaired TSC2 deficiency-associated renal cystogenesis. Therefore, harnessing mTOR/miR-142-3p/PRAS40 signaling cascade may mitigate hyperactivated mTOR-related diseases.
- Published
- 2024
- Full Text
- View/download PDF
3. mTOR/miR-142-3p/PRAS40 signaling cascade is critical for tuberous sclerosis complex-associated renal cystogenesis.
- Author
-
Zhao, Shuyun, Hao, Shuai, Zhou, Jiasheng, Chen, Xinran, Zhang, Tianhua, Qi, Zhaolai, Zhang, Ting, Jalal, Sajid, Zhai, Chuanxin, Yin, Lu, Bo, Yufei, Teng, Hongming, Wang, Yue, Gao, Dongyan, Zhang, Hongbing, and Huang, Lin
- Abstract
Background: Patients with tuberous sclerosis complex (TSC) develop renal cysts and/or angiomyolipomas (AMLs) due to inactive mutations of either TSC1 or TSC2 and consequential mTOR hyperactivation. The molecular events between activated mTOR and renal cysts/AMLs are still largely unknown. Methods: The mouse model of TSC-associated renal cysts were constructed by knocking out Tsc2 specifically in renal tubules (Tsc2
f/f ; ksp-Cre). We further globally deleted PRAS40 in these mice to investigate the role of PRAS40. Tsc2−/− cells were used as mTOR activation model cells. Inhibition of DNA methylation was used to increase miR-142-3p expression to examine the effects of miR-142-3p on PRAS40 expression and TSC-associated renal cysts. Results: PRAS40, a component of mTOR complex 1, was overexpressed in Tsc2-deleted cell lines and mouse kidneys (Tsc2f/f ; ksp-Cre), which was decreased by mTOR inhibition. mTOR stimulated PRAS40 expression through suppression of miR-142-3p expression. Unleashed PRAS40 was critical to the proliferation of Tsc2−/− cells and the renal cystogenesis of Tsc2f/f ; ksp-Cre mice. In contrast, inhibition of DNA methylation increased miR-142-3p expression, decreased PRAS40 expression, and hindered cell proliferation and renal cystogenesis. Conclusions: Our data suggest that mTOR activation caused by TSC2 deletion increases PRAS40 expression through miR-142-3p repression. PRAS40 depletion or the pharmacological induction of miR-142-3p expression impaired TSC2 deficiency-associated renal cystogenesis. Therefore, harnessing mTOR/miR-142-3p/PRAS40 signaling cascade may mitigate hyperactivated mTOR-related diseases. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. اثر تعاملی شش هفته تمرین مقاومتی و مکمل یاری پروتئین وی بر بیان ژن PRAS40 و میزان هایپرتروفی عضله دوقلو در رتهای نر نژاد ویستار.
- Author
-
حمیده احمدی, فرشته شهیدی, and مجید کاشف
- Subjects
RESISTANCE training ,WHEY proteins ,SKELETAL muscle ,MUSCLE growth ,LABORATORY rats - Abstract
PRAS40 is expressed in all types of body tissues and is present in the muscle growth pathway and has multiple phosphorylation sites. Studies have shown that PRAS40 plays a role in regulating cell growth. The aim of this study was to evaluate the effect of six weeks of resistance training and whey protein supplementation on PRAS40 and biceps hypertrophy in male Wistar rats Method: This developmental experimental study was carried out in 1402 year. 23 eight-week-old male Wistar rats (250 grams) were randomly divided into three groups: resistance training, resistance training+ supplement, and control. After two weeks of orientation, the training groups completed six weeks of resistance training, climbing the ladder five days a week. The training +supplement group received whey protein (2.05 g/kg dose) through gavage. 48 hours after the last training, a blood sample was taken and the gastrocnemius muscle was removed, frizzed, and saved to evaluate the expression of the PRAS40 gene. Findings: Six weeks of resistance training with whey supplement caused a significant decrease in PRAS40 gene expression and an increase in body and muscle weight in the exercise and supplement + exercise groups. Conclusion: The resistance training protocol along with her supplement can lead to muscle hypertrophy and activate the anabolic pathways in the muscle through the suppression of the PRAS40 pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
5. Exogenous PRAS40 reduces KLF4 expression and alleviates hypertrophic scar fibrosis and collagen deposition through inhibiting mTORC1.
- Author
-
Wang, Chao and Jiang, Duyin
- Subjects
- *
HYPERTROPHIC scars , *FIBROSIS , *RAPAMYCIN , *COLLAGEN , *SCARS , *PATHOLOGICAL physiology - Abstract
To identify the anti-fibrosis effect of PRAS40 in scar, and its potential mechanism. We constructed a rat model of hypertrophic scarthat was locally injected the PRAS40 overexpression adenoviruses, mTORC1 inhibitor MHY1485 and activator rapamycin, and further observed the pathological changes of skin tissue and the severity of fibrosis by HE, Masson and sirius red staining, and analyzed the deposition of a-SMA and collagen I by western blot and immunofluorescence test. Meanwhile, the co-localization of KLF4 with a-SMA and type I collagen was analyzed, as well as the regulatory effect of PRAS40 on KLF4. In addition, we also verified whether the inhibition of scar fibrosis by PRAS40 is related to mTORC1, and whether the upregulation of KLF4 is related to mTORC1. The results showed that the expression of PRAS40 was low and p-PRAS40 was high in scar skin tissue. After local injection of PRAS40 overexpression adenovirus, the expression of PRAS40 in skin tissue was increased. The overexpression of PRAS40 can inhibit scar skin fibrosis and reduce the content of a-SMA and collagen I. Further mechanism analysis confirms that the inhibitory effect of PRAS40 on skin fibrosis is related to mTORC1, and PRAS40 inhibits the activation of mTORC1. The expression of KLF4 is relatively low in scar tissue. PRAS40 administration upregulated the expression of KLF4, which is related to mTORC1 PRAS40 significantly improves fibrosis of scar skin tissue and increases the expression of KLF4 in scars. The anti-fibrotic effect of PRAS40 depends on mTORC1. • Exogenous PRAS40 improved the scar fibrosis in the injured tissues of HS rats. • mTORC1 activation accelerates fibrosis degree in the injured skin. PRAS40 is also an inhibitor of mTORC1 inactivation, so we believe that the effect of PRAS40 on scar fibrosis is related to mTORC1. • KLF4 is an important regulator in the development of fibrosis. • Consistent with the previous report, we found a high expression of KLF4 in skin tissues , while after HS modeling, KLF4 was downregulated. Interestingly, PRAS40 could modulate the KLF4 expression, and this effect was also related to mTORC1. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Vitexicarpin Induces Apoptosis and Inhibits Metastatic Properties via the AKT-PRAS40 Pathway in Human Osteosarcoma.
- Author
-
Yun, Hyung-Mun, Kwon, Hyun Sook, Lee, Joon Yeop, and Park, Kyung-Ran
- Subjects
- *
OSTEOSARCOMA , *APOPTOSIS , *VITEX , *BONE cancer , *CELL death - Abstract
Osteosarcoma, which has poor prognosis after metastasis, is the most common type of bone cancer in children and adolescents. Therefore, plant-derived bioactive compounds are being actively developed for cancer therapy. Artemisia apiacea Hance ex Walp. is a traditional medicinal plant native to Eastern Asia, including China, Japan, and Korea. Vitexicarpin (Vitex), derived from A. apiacea, has demonstrated analgesic, anti-inflammatory, antitumour, and immunoregulatory properties; however, there are no published studies on Vitex isolated from the aerial parts of A. apiacea. Thus, this study aimed to evaluate the antitumour activity of Vitex against human osteosarcoma cells. In the present study, Vitex (>99% purity) isolated from A. apiacea induced significant cell death in human osteosarcoma MG63 cells in a dose- and time-dependent manner; cell death was mediated by apoptosis, as evidenced by the appearance of cleaved-PARP, cleaved-caspase 3, anti-apoptotic proteins (Survivin and Bcl-2), pro-apoptotic proteins (Bax), and cell cycle-related proteins (Cyclin D1, Cdk4, and Cdk6). Additionally, a human phosphokinase array proteome profiler revealed that Vitex suppressed AKT-dependent downstream kinases. Further, Vitex reduced the phosphorylation of PRAS40, which is associated with autophagy and metastasis, induced autophagosome formation, and suppressed programmed cell death and necroptosis. Furthermore, Vitex induced antimetastatic activity by suppressing the migration and invasion of MMP13, which is the primary protease that degrades type I collagen for tumour-induced osteolysis in bone tissues and preferential metastasis sites. Taken together, our results suggest that Vitex is an attractive target for treating human osteosarcoma. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Screening and evaluation of metabolites binding PRAS40 from Erxian decoction used to treat spinal cord injury.
- Author
-
Li Lin, Jingchuan Yan, Jin Sun, Jianfeng Zhang, and Bo Liao
- Subjects
SPINAL cord injuries ,ISOTHERMAL titration calorimetry ,WESTERN immunoblotting ,HEMATOXYLIN & eosin staining ,MOLECULAR docking - Abstract
Objective: The PRAS40 is an essential inhibitory subunit of the mTORC1 complex, which regulates autophagy. It has been suggested that Erxian Decoction (EXD) could treat spinal cord injury (SCI) via the autophagy pathway. However, the mechanism of whether EXD acts through PRAS40 remains unclear. Methods: With the help of immobilized PRAS40, isothermal titration calorimetry (ITC) and molecular docking, the bioactive metabolites in the EXD were screened. To establish in vitro SCI models, PC12 cells were exposed to hydrogen peroxide (H2O2) and then treated with the identified EXD substances. Furthermore, Western blot assay was carried out to identify potential molecular mechanisms involved. For assessing the effect of metabolites in vivo, the SCI model rats were first pretreated with or without the metabolite and then subjected to the immunohistochemistry (IHC) staining, Basso, Beattie & Bresnahan (BBB) locomotor rating scale, and H&E staining. Results: The immobilized PRAS40 isolated indole, 4-nitrophenol, terephthalic acid, palmatine, sinapinaldehyde, and 3-chloroaniline as the potential ligands binding to PRAS40. Furthermore, the association constants of palmatine and indole as 2.84 × 10
6 M-1 and 3.82 × 105 M-1 were elucidated via ITC due to the drug-like properties of these two metabolites. Molecular docking results also further demonstrated the mechanism of palmatine binding to PRAS40. Western blot analysis of PC12 cells demonstrated that palmatine inhibited the expression of p-mTOR by binding to PRAS40, activating the autophagic flux by markedly increasing LC3. The injection of palmatine (10 μM and 20 μM) indicated notably increased BBB scores in the SCI rat model. Additionally, a dose-dependent increase in LC3 was observed by IHC staining. Conclusion: This research proved that EXD comprises PRAS40 antagonists, and the identified metabolite, palmatine, could potentially treat SCI by activating the autophagic flux. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
8. Oncolytic Avian Reovirus p17-Modulated Inhibition of mTORC1 by Enhancement of Endogenous mTORC1 Inhibitors Binding to mTORC1 To Disrupt Its Assembly and Accumulation on Lysosomes.
- Author
-
Jyun-Yi Li, Wei-Ru Huang, Tsai-Ling Liao, Nielsen, Brent L., and Hung-Jen Liu
- Subjects
- *
VIRAL replication , *AUTOPHAGY , *CELL lines , *LYSOSOMES , *CANCER cells , *PHOSPHATIDYLINOSITOL 3-kinases - Abstract
The mechanism by which avian reovirus (ARV)-modulated suppression of mTORC1 triggers autophagy remains largely unknown. In this work, we determined that p17 functions as a negative regulator of mTORC1. This study suggest novel mechanisms whereby p17-modulated inhibition of mTORC1 occurs via upregulation of p53, inactivation of Akt, and enhancement of binding of the endogenous mTORC1 inhibitors (PRAS40, FKBP38, and FKPP12) to mTORC1 to disrupt its assembly and accumulation on lysosomes. p17-modulated inhibition of Akt leads to activation of the downstream targets PRAS40 and TSC2, which results in mTORC1 inhibition, thereby triggering autophagy and translation shutoff, which is favorable for virus replication. p17 impairs the interaction of mTORC1 with its activator Rheb, which promotes FKBP38 interaction with mTORC1. It is worth noting that p17 activates ULK1 and Beclin1 and increases the formation of the Beclin 1/class III PI3K complex. These effects could be reversed in the presence of insulin or depletion of p53. Furthermore, we found that p17 induces autophagy in cancer cell lines by upregulating the p53/PTEN pathway, which inactivates Akt and mTORC1. This study highlights p17-modulated inhibition of Akt and mTORC1, which triggers autophagy and translation shutoff by positively modulating the tumor suppressors p53 and TSC2 and endogenous mTORC1 inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. TIFA promotes colorectal cancer cell proliferation in an RSK‐ and PRAS40‐dependent manner.
- Author
-
Shen, Wenzhi, Du, Wenfei, Li, Yanping, Huang, Yongming, Jiang, Xinyu, Yang, Chenglong, Tang, Jiaping, Liu, Huan, Luo, Na, Zhang, Xiaoyuan, and Zhang, Zhixin
- Abstract
Previous studies have reported that TIFA plays different roles in various tumor types. However, the function of TIFA in colorectal cancer (CRC) remains unclear. Here, we showed that the expression of TIFA was markedly increased in CRC versus normal tissue, and positively correlated with CRC TNM stages. In agreement, we found that the CRC cell lines show increased TIFA expression levels versus normal control. The knockdown of TIFA inhibited cell proliferation but had no effect on cell apoptosis in vitro or in vivo. Moreover, the ectopic expression of TIFA enhanced cell proliferation ability in vitro and in vivo. In contrast, the expression of mutant TIFA (T9A, oligomerization site mutation; D6, TRAF6 binding site deletion) abolished TIFA‐mediated cell proliferation enhancement. Exploration of the underlying mechanism revealed that the protein synthesis‐associated kinase RSK and PRAS40 activation were responsible for TIFA‐mediated CRC progression. In summary, these findings suggest that TIFA plays a role in mediating CRC progression. This could provide a promising target for CRC therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. Activation of the PI3K/AKT/MTOR/P70S6K1 signaling cascade in peripheral blood mononuclear cells in patients with type 2 diabetes
- Author
-
L. K. Sokolova, V. M. Pushkarev, O. I. Kovzun, B. B. Guda, V. V. Pushkarev, M. D. Tronko, N. V. Skrypnyk, and L. M. Zaiats
- Subjects
peripheral blood mononuclear cells ,akt ,pras40 ,p70s6k ,insulin ,insulin-like growth factor ,type 2 diabetes ,Biochemistry ,QD415-436 ,Medicine ,Biology (General) ,QH301-705.5 - Abstract
Modern research shows that patients with diabetes mellitus have an increased risk of cancer. PI3K/Akt/mTOR/p70S6K1 signaling pathway plays an important role in the pathogenesis of cancer and diabetes. The aim of this study was to determine the state of РІ3K/Akt/mTORC1/p70S6K signaling cascade activity in peripheral mononuclear blood cells (PBMC) of patients with type 2 diabetes (T2D) relatively to the insulin and insulin-like growth factor (IGF-1) concentrations in blood plasma. Enzyme-linked immunosorbent assay was used to examine the levels of insulin and IGF-1 in blood plasma as well as the content of phosphorylated forms of Akt (Ser473), PRAS40 (Thr246), and p70S6K (Thr389) in PMBC. It was shown that in the blood plasma of patients with T2D the levels of insulin and IGF-1 were increased. Phosphorylation and activation of Akt by the mTORC2 protein kinase complex was not observed. At the same time, the relative degree of phosphorylation of mTORC1 inhibitor, PRAS40, and its substrate, p70S6K, was higher in PMBC of T2D patients in comparison with control values. These data suggest that phosphoinositide-dependent protein kinase 1 (PDK1) and, possibly, mitogen-activated protein kinase (MAPK) could mediate the effects of IGF-1 on Akt activation under type 2 diabetes.
- Published
- 2020
- Full Text
- View/download PDF
11. TGFβ signaling-induced miRNA participates in autophagic regulation by targeting PRAS40 in mesenchymal subtype of glioblastoma
- Author
-
Yingbin Xie, Luyue Chen, Junhu Zhou, Qixue Wang, Chao Yang, Can Xu, Xiangyu Fan, Yanli Tan, Yanan Wang, Chunsheng Kang, and Chuan Fang
- Subjects
autophagy ,glioblastoma ,microrna ,pras40 ,tgfβ signalling ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Objective: Mesenchymal subtype of glioblastoma (mesGBM) is a refractory disease condition characterized by therapeutic failure and tumor recurrence. Hyperactive transforming growth factor-β (TGF-β) signaling could be a signature event in mesGBM, which leads to dysregulation of downstream targets and contribute to malignant transformation. In this study we aimed to investigate the hyperactive TGFβ signaling-mediated pathogenesis and possible downstream targets for the development of novel therapeutic interventions for mesGBM. Methods: GBM-BioDP is an online resource for accessing and displaying interactive views of the TCGA GBM data set. Transcriptomic sequencing followed by bioinformatic analysis was performed to identify dysregulated microRNAs. Target prediction by MR-microT and dual luciferase reporter assay were utilized to confirm the predicted target of novel_miR56. CCK-8 assays was used to assesse cell viability. The miRNA manipulation was proceeded by cell transfection and lentivirus delivery. A plasmid expressing GFP-LC3 was introduced to visualize the formation of autophagosomes. Orthotopic GBM model was constructed for in vivo study. Results: TGFβ1 and TGFβ receptor type II (TβRII) were exclusively upregulated in mesGBM (P < 0.01). Dysregulated miRNAs were identified after LY2109761 (a TβRI/II inhibitor) treatment in a mesGBM-derived cell line, and novel_miR56 was selected as a promising candidate for further functional verification. Novel_miR56 was found to potentially bind to PRAS40 via seed region complementarity in the 3′ untranslated region, and we also confirmed that PRAS40 is a direct target of novel_miR56 in glioma cells. In vitro, over expression of novel_miR56 in tumor cells significantly promoted proliferation and inhibited autophagy (P < 0.05). The expression levels of P62/SQSTM was significantly increased accompanied by the decrease of BECN1 and LC3B-II/I, which indicated that autophagic activity was reduced after novel_miR56 treatment. In addition, over expression of novel_miR56 also promoted tumor growth and inhibited autophagy in vivo, which is associated with worse prognosis (P < 0.05). Conclusions: In summary, we provide novel insight into TGFβ signaling-mediated pathogenesis in mesGBM and TGFβ signaling-induced novel_miR56 may be a novel target for mesGBM management.
- Published
- 2020
- Full Text
- View/download PDF
12. Interaction Between CD34+ Fibrocytes and Airway Smooth Muscle Promotes IL-8 Production and Akt/PRAS40/mTOR Signaling in Asthma
- Author
-
Ting-Yu Lin, Po-Jui Chang, Chun-Yu Lo, Yu-Lun Lo, Chih-Teng Yu, Shu-Min Lin, Chih-His Scott Kuo, and Horng-Chyuan Lin
- Subjects
fibrocytes ,airway smooth muscle cells ,interleukin-8 ,interleukin-33 ,PRAS40 ,mTOR ,Medicine (General) ,R5-920 - Abstract
BackgroundThe circulating progenitor cells of fibroblasts (fibrocytes) have been shown to infiltrate the airway smooth muscle compartment of asthma patients; however, the pathological significance of this discovery has yet to be elucidated. This study established a co-culture model of airway smooth muscle cells (ASMCs) and fibrocytes from asthmatic or normal subjects to evaluate innate cytokine production, corticosteroid responses, and signaling in ASMCs.MethodsCD34+ fibrocytes were purified from peripheral blood of asthmatic (Global Initiative for Asthma treatment step 4–5) and normal subjects and cultured for 5∼7 days. In a transwell plate, ASMCs were co-cultured with fibrocytes at a ratio of 2:1, ASMCs were cultured alone (control condition), and fibrocytes were cultured alone for 48 h. Measurements were obtained of interleukin-8 (IL-8), IL-6, IL-17, thymic stromal lymphopoietin, and IL-33 levels in the supernatant and IL-33 levels in the cell lysate of the co-culture. Screening for intracellular signaling in the ASMCs after stimulation was performed using condition medium from the patients’ co-culture (PtCM) or IL-8. mRNA and western blot analysis were used to analyze AKT/mTOR signaling in ASMCs stimulated via treatment with PtCM or IL-8.ResultsCompared with ASMCs cultured alone, IL-8 levels in the supernatant and IL-33 levels in the ASMCs lysate were significantly higher in samples co-cultured from asthmatics, but not in those co-cultured from normal subjects. Corticosteroid-induced suppression of IL-8 production was less pronounced in ASMCs co-cultured with fibrocytes from asthma patients than in ASMCs co-cultured from normal subjects. ASMCs stimulated using PtCM and IL-8 presented elevating activated AKT substrate PRAS40. Treatment with IL-8 and PtCM increased mRNA expression of mTOR and P70S6 kinases in ASMCs. Treatment with IL-8 and PtCM also significantly increased phosphorylation of AKT and mTOR subtract S6 ribosomal protein in ASMCs.ConclusionThe interaction between ASMCs and fibrocytes from asthmatic patients was shown to increase IL-8 and IL-33 production and promote AKT/mTOR signaling in ASMCs. IL-8 production in the co-culture from asthmatic patients was less affected by corticosteroid than was that in the co-culture from normal subjects. Our results elucidate the novel role of fibrocytes and ASMCs in the pathogenesis of asthma.
- Published
- 2022
- Full Text
- View/download PDF
13. Rapamycin alleviates memory deficit against pentylenetetrazole-induced neural toxicity in Wistar male rats.
- Author
-
Aghaie, Fatemeh, Shemshaki, Afsaneh, Rajabi, Mojgan, Khatami, Parisa, and Hosseini, Abdolkarim
- Abstract
Numerous studies have reported that epilepsy causes memory deficits. The present study was aimed at studying the effect of rapamycin against the memory deficiency of the pentylenetetrazole (PTZ)-kindled animal model of epilepsy. In the present experiment, we randomly chose thirty male rats from the species of Wistar and categorized them in groups of control and experiment (6 for each group). The groups of experiment received the injection of rapamycin (0.5, 1 and 2 mg/kg) intraperitoneally (i.p.) and the group of control received normal saline (0.9%) treatment. Through the PTZ's sub-threshold dose (35 mg kg
−1 , i.p.), all groups were kindled 12 times. Passive avoidance test (PAT) was used for gauging the memory function and the seizure behaviors after the kindling procedure. The rodents were sacrificed at the end of the trial and their brains were scooped for measuring the expression of Gabra1 and Pras40 genes. Statistical analysis unveiled that rapamycin delayed the kindling development and the onset of seizures which are tonic-clonic. Moreover, the administration of rapamycin significantly prevented memory dysfunction in epileptic rats. Finally, it was shown that rapamycin resulted in an increase in the expression levels of Gabra1 and Pras40 genes at the brain tissues. The current research design indicated that rapamycin has beneficial effects for the prevention of memory impairment against PTZ-kindling epilepsy in rats. Such promising outcomes could be attributed to its impact on the Gabra1 and Pras40 genes. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
14. mTORC1 silencing during intestinal epithelial Caco-2 cell differentiation is mediated by the activation of the AMPK/TSC2 pathway.
- Author
-
Kaur, Harleen and Moreau, Régis
- Subjects
- *
ENTEROENDOCRINE cells , *EPITHELIAL cells , *CELL differentiation , *STEM cells , *PROTEIN synthesis , *ENTEROCYTES - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) signaling is the prototypical pathway regulating protein synthesis and cell proliferation. The level of mTORC1 activity is high in intestinal stem cells located at the base of the crypts and thought to gradually decrease as transit-amplifying cells migrate out of the crypts and differentiate into enterocytes, goblet cells or enteroendocrine cells along the epithelium. The unknown mechanism responsible for the silencing of intestinal epithelium mTORC1 during cell differentiation was investigated in Caco-2 cells, which spontaneously differentiate into enterocytes in standard growth medium. The results show that TSC2, an upstream negative regulator of mTORC1 was central to mTORC1 silencing in differentiated Caco-2 cells. AMPK-mediated activation of TSC2 (Ser1387) and repression of Raptor (Ser792), an essential component of mTORC1, were stimulated in differentiated Caco-2 cells. ERK1/2-mediated repression of TSC2 (Ser664) seen in undifferentiated Caco-2 cells was lifted in differentiated cells. IRS-1-mediated activation of AKT (Thr308) phosphorylation was stimulated in differentiated Caco-2 cells and may be involved in cross-pathway repression of ERK1/2. Additionally, PRAS40 (Thr246) phosphorylation was decreased in differentiated Caco-2 cells compared to undifferentiated cells allowing dephosphorylated PRAS40 to displace Raptor thereby repressing mTORC1 kinase activity. • AMPK-mediated activation of TSC2 (Ser1387) and repression of Raptor (Ser792) are stimulated in differentiated Caco-2 cells. • ERK1/2-mediated repression of TSC2 (Ser664) is lifted in differentiated Caco-2 cells. • IRS-1-mediated activation of AKT (Thr308) phosphorylation is stimulated in differentiated Caco-2 cells. • PRAS40 (Thr246) phosphorylation is decreased in differentiated Caco-2 cells compared to undifferentiated cells. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
15. Activation of PI3K/mTOR pathway occurs in most adult low-grade gliomas and predicts patient survival
- Author
-
McBride, Sean M., Perez, Daniel A., Polley, Mei-Yin, Vandenberg, Scott R., Smith, Justin S., Zheng, Shichun, Lamborn, Kathleen R., Wiencke, John K., Chang, Susan M., Prados, Michael D., Berger, Mitchel S., Stokoe, David, and Haas-Kogan, Daphne A.
- Subjects
Medicine & Public Health ,Neurology ,Oncology ,Low grade glioma ,mTOR ,Rapamycin ,PTEN ,PRAS40 - Abstract
Recent evidence suggests the Akt-mTOR pathway may play a role in development of low-grade gliomas (LGG). We sought to evaluate whether activation of this pathway correlates with survival in LGG by examining expression patterns of proteins within this pathway. Forty-five LGG tumor specimens from newly diagnosed patients were analyzed for methylation of the putative 5′-promoter region of PTEN using methylation-specific PCR as well as phosphorylation of S6 and PRAS40 and expression of PTEN protein using immunohistochemistry. Relationships between molecular markers and overall survival (OS) were assessed using Kaplan-Meier methods and exact log-rank test. Correlation between molecular markers was determined using the Mann-Whitney U and Spearman Rank Correlation tests. Eight of the 26 patients with methylated PTEN died, as compared to 1 of 19 without methylation. There was a trend towards statistical significance, with PTEN methylated patients having decreased survival (P = 0.128). Eight of 29 patients that expressed phospho-S6 died, whereas all 9 patients lacking p-S6 expression were alive at last follow-up. There was an inverse relationship between expression of phospho-S6 and survival (P = 0.029). There was a trend towards decreased survival in patients expressing phospho-PRAS40 (P = 0.077). Analyses of relationships between molecular markers demonstrated a statistically significant positive correlation between expression of p-S6(235) and p-PRAS40 (P = 0.04); expression of p-S6(240) correlated positively with PTEN methylation (P = 0.04) and negatively with PTEN expression (P = 0.03). Survival of LGG patients correlates with phosphorylation of S6 protein. This relationship supports the use of selective mTOR inhibitors in the treatment of low grade glioma.
- Published
- 2010
16. Gene expression and regulatory factors of the mechanistic target of rapamycin (mTOR) complex 1 predict mammalian longevity.
- Author
-
Mota-Martorell, Natalia, Jove, Mariona, Pradas, Irene, Berdún, Rebeca, Sanchez, Isabel, Naudi, Alba, Gari, Eloi, Barja, Gustavo, and Pamplona, Reinald
- Subjects
GENE expression ,REGULATOR genes ,LONGEVITY ,GENE ontology ,PROTEIN expression ,MASS spectrometry - Abstract
Species longevity varies significantly across animal species, but the underlying molecular mechanisms remain poorly understood. Recent studies and omics approaches suggest that phenotypic traits of longevity could converge in the mammalian target of rapamycin (mTOR) signalling pathway. The present study focuses on the comparative approach in heart tissue from 8 mammalian species with a ML ranging from 3.5 to 46 years. Gene expression, protein content, and concentration of regulatory metabolites of the mTOR complex 1 (mTORC1) were measured using droplet digital PCR, western blot, and mass spectrometry, respectively. Our results demonstrate (1) the existence of differences in species-specific gene expression and protein content of mTORC1, (2) that the achievement of a high longevity phenotype correlates with decreased and inhibited mTORC1, (3) a decreased content of mTORC1 activators in long-lived animals, and (4) that these differences are independent of phylogeny. Our findings, taken together, support an important role for mTORC1 downregulation in the evolution of long-lived mammals. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
17. Cryo-EM insight into the structure of MTOR complex 1 and its interactions with Rheb and substrates [version 1; referees: 3 approved]
- Author
-
Luke H. Chao and Joseph Avruch
- Subjects
Review ,Articles ,MTOR ,Raptor ,mLst8 ,PRAS40 ,AKT1S1 ,Rheb ,p70 S6 kinase ,S6K1B ,EIF4EBP1 ,4E-BP - Abstract
The mechanistic target of rapamycin (MTOR) is a giant protein kinase that, together with the accessory proteins Raptor and mLst8, forms a complex of over 1 MDa known as MTOR complex 1 (MTORC1). MTORC1, through its protein kinase activity, controls the accretion of cell mass through the regulation of gene transcription, mRNA translation, and protein turnover. MTORC1 is activated in an interdependent manner by insulin/growth factors and nutrients, especially amino acids, and is inhibited by stressors such as hypoxia and by the drug rapamycin. The action of insulin/growth factors converges on the small GTPase Rheb, which binds directly to the MTOR polypeptide in MTORC1 and, in its GTP-bound state, initiates kinase activation. Biochemical studies established that MTORC1 exists as a dimer of the MTOR/Raptor/mLst8 trimer, and progressive refinements in cryo-electron microscopy (cryo-EM) have enabled an increasingly clear picture of the architecture of MTORC1, culminating in a deep understanding of how MTORC1 interacts with and phosphorylates its best-known substrates—the eIF-4E binding protein/4E-BP, the p70 S6 kinase/S6K1B, and PRAS40/AKT1S1—and how this is inhibited by rapamycin. Most recently, Rheb-GTP has been shown to bind to MTORC1 in a cooperative manner at an allosteric site remote from the kinase domain that twists the latter into a catalytically competent configuration. Herein, we review the recent cryo-EM and associated biochemical studies of MTORC1 and seek to integrate these new results with the known physiology of MTORC1 regulation and signaling.
- Published
- 2019
- Full Text
- View/download PDF
18. Crystal Structure of Human Dual-Specificity Tyrosine-Regulated Kinase 3 Reveals New Structural Features and Insights into its Auto-phosphorylation.
- Author
-
Kim, Kuglae, Cha, Jeong Seok, Cho, Yong-Soon, Kim, Hoyoung, Chang, Nienping, Kim, Hye-Jung, and Cho, Hyun-Soo
- Subjects
- *
PHOSPHORYLATION , *PROTEIN-tyrosine kinases , *CRYSTAL structure , *ANEMIA treatment , *PROTEIN domains - Abstract
Dual-specificity tyrosine-regulated kinases (DYRKs) auto-phosphorylate a critical tyrosine residue in their activation loop and phosphorylate their substrate on serine and threonine residues. The auto-phosphorylation occurs intramolecularly and is a one-off event. DYRK3 is selectively expressed at a high level in hematopoietic cells and attenuates erythroblast development, leading to anemia. In the present study, we determined the crystal structure of the mature form of human DYRK3 in complex with harmine, an ATP competitive inhibitor. The crystal structure revealed a phosphorylation site, residue S350, whose phosphorylation increases the stability of DYRK3 and enhances its kinase activity. In addition, our structural and biochemical assays suggest that the N-terminal auto-phosphorylation accessory domain stabilizes the DYRK3 protein, followed by auto-phosphorylation of the tyrosine of the activation loop, which is important for kinase activity. Finally, our docking analysis provides information for the design of novel and potent therapeutics to treat anemia. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
19. PRAS40 plays a pivotal role in protecting against stroke by linking the Akt and mTOR pathways
- Author
-
Xiaoxing Xiong, Rong Xie, Hongfei Zhang, Lijuan Gu, Weiying Xie, Michelle Cheng, Zhihong Jian, Kristina Kovacina, and Heng Zhao
- Subjects
Focal cerebral ischemia ,Akt ,Stroke ,mTOR ,PRAS40 ,PTEN ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The proline-rich Akt substrate of 40 kDa (PRAS40) protein is not only a substrate of the protein kinase Akt but also a component of the mTOR complex 1 (mTORC1), thus it links the Akt and the mTOR pathways. We investigated the potential protective role of PRAS40 in cerebral ischemia and its underlying mechanisms by using rats with lentiviral over-expression of PRAS40 and mice with PRAS40 gene knockout (PRAS40 KO). Our results show that gene transfer of PRAS40 reduced infarction size in rats by promoting phosphorylation of Akt, FKHR (FOXO1), PRAS40, and mTOR. In contrast, PRAS40 KO increased infarction size. Although the PRAS40 KO under normal condition did not alter baseline levels of phosphorylated proteins in the Akt and mTOR pathways, PRAS40 KO that underwent stroke exhibited reduced protein levels of p-S6K and p-S6 in the mTOR pathway but not p-Akt, or p-PTEN in the Akt pathway. Furthermore, co-immunoprecipitation suggests that there were less interactive effects between Akt and mTOR in the PRAS40 KO. In conclusion, PRAS40 appears to reduce brain injury by converting cell signaling from Akt to mTOR.
- Published
- 2014
- Full Text
- View/download PDF
20. PRAS40 prevents development of diabetic cardiomyopathy and improves hepatic insulin sensitivity in obesity
- Author
-
Mirko Völkers, Shirin Doroudgar, Nathalie Nguyen, Mathias H Konstandin, Pearl Quijada, Shabana Din, Luis Ornelas, Donna J Thuerauf, Natalie Gude, Kilian Friedrich, Stephan Herzig, Christopher C Glembotski, and Mark A Sussman
- Subjects
diabetes ,PRAS40 ,mTOR ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Diabetes is a multi‐organ disease and diabetic cardiomyopathy can result in heart failure, which is a leading cause of morbidity and mortality in diabetic patients. In the liver, insulin resistance contributes to hyperglycaemia and hyperlipidaemia, which further worsens the metabolic profile. Defects in mTOR signalling are believed to contribute to metabolic dysfunctions in diabetic liver and hearts, but evidence is missing that mTOR activation is causal to the development of diabetic cardiomyopathy. This study shows that specific mTORC1 inhibition by PRAS40 prevents the development of diabetic cardiomyopathy. This phenotype was associated with improved metabolic function, blunted hypertrophic growth and preserved cardiac function. In addition PRAS40 treatment improves hepatic insulin sensitivity and reduces systemic hyperglycaemia in obese mice. Thus, unlike rapamycin, mTORC1 inhibition with PRAS40 improves metabolic profile in diabetic mice. These findings may open novel avenues for therapeutic strategies using PRAS40 directed against diabetic‐related diseases.
- Published
- 2013
- Full Text
- View/download PDF
21. Pim1 inhibition as a novel therapeutic strategy for Alzheimer's disease.
- Author
-
Velazquez, Ramon, Shaw, Darren M., Caccamo, Antonella, and Oddo, Salvatore
- Subjects
- *
ALZHEIMER'S disease treatment , *PROTEASOME inhibitors , *ANIMAL models of Alzheimer's disease , *SHORT-term memory , *ANIMAL models for aging , *LABORATORY mice , *HOMEOSTASIS , *RAPAMYCIN , *MAMMALS - Abstract
Background: Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder worldwide. Clinically, AD is characterized by impairments of memory and cognitive functions. Accumulation of amyloid-β (Aβ) and neurofibrillary tangles are the prominent neuropathologies in patients with AD. Strong evidence indicates that an imbalance between production and degradation of key proteins contributes to the pathogenesis of AD. The mammalian target of rapamycin (mTOR) plays a key role in maintaining protein homeostasis as it regulates both protein synthesis and degradation. A key regulator of mTOR activity is the proline-rich AKT substrate 40 kDa (PRAS40), which directly binds to mTOR and reduces its activity. Notably, AD patients have elevated levels of phosphorylated PRAS40, which correlate with Aβ and tau pathologies as well as cognitive deficits. Physiologically, PRAS40 phosphorylation is regulated by Pim1, a protein kinase of the protoconcogene family. Here, we tested the effects of a selective Pim1 inhibitor (Pim1i), on spatial reference and working memory and AD-like pathology in 3xTg-AD mice. Results: We have identified a Pim1i that crosses the blood brain barrier and reduces PRAS40 phosphorylation. Pim1i-treated 3xTg-AD mice performed significantly better than their vehicle treated counterparts as well as non-transgenic mice. Additionally, 3xTg-AD Pim1i-treated mice showed a reduction in soluble and insoluble Aβ40 and Aβ42 levels, as well as a 45.2 % reduction in Aβ42 plaques within the hippocampus. Furthermore, phosphorylated tau immunoreactivity was reduced in the hippocampus of Pim1i-treated 3xTg-AD mice by 38 %. Mechanistically, these changes were linked to a significant increase in proteasome activity. Conclusion: These results suggest that reductions in phosphorylated PRAS40 levels via Pim1 inhibition reduce Aβ and Tau pathology and rescue cognitive deficits by increasing proteasome function. Given that Pim1 inhibitors are already being tested in ongoing human clinical trials for cancer, the results presented here may open a new venue of drug discovery for AD by developing more Pim1 inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
22. Ischemia-Induced Changes of PRAS40 and p-PRAS40 Immunoreactivities in the Gerbil Hippocampal CA1 Region After Transient Cerebral Ischemia.
- Author
-
Park, Joon, Shin, Bich, Ahn, Ji, Cho, Jeong-Hwi, Kim, In, Kim, Dae, Won, Moo-Ho, Hong, Seongkweon, Cho, Jun, and Lee, Choong-Hyun
- Subjects
- *
CEREBRAL ischemia , *HIPPOCAMPUS (Brain) , *PYRAMIDAL neurons , *ASTROCYTES , *BRAIN damage - Abstract
Proline-rich Akt substrate of 40-kDa (PRAS40) is one of the important interactive linkers between Akt and mTOR signaling pathways. The increase of PRAS40 is related with the reduction of brain damage induced by cerebral ischemia. In the present study, we investigated time-dependent changes in PRAS40 and phospho-PRAS40 (p-PRAS40) immunoreactivities in the hippocampal CA1 region of the gerbil after 5 min of transient cerebral ischemia. PRAS40 immunoreactivity in the CA1 region was decreased in pyramidal neurons from 12 h after ischemic insult in a time-dependent manner, and, at 5 days post-ischemia, PRAS40 immunoreactivity was newly expressed in astrocytes. p-PRAS40 immunoreactivity in the CA1 pyramidal neurons was hardly found 12 h and apparently detected again 1 and 2 days after ischemic insult. At 5 days post-ischemia, p-PRAS40 immunoreactivity in the CA1 pyramidal neurons was not found. These results indicate that ischemia-induced changes in PRAS40 and p-PRAS40 immunoreactivities in CA1 pyramidal neurons and astrocytes may be closely associated with delayed neuronal death in the hippocampal CA1 region following transient cerebral ischemia. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
23. Targeting the mTOR pathway using novel ATP-competitive inhibitors, Torin1, Torin2 and XL388, in the treatment of glioblastoma
- Author
-
John L. Gillick, Raj Murali, Meena Jhanwar‑Uniyal, Chirag D. Gandhi, Tolga Sursal, Anubhav G. Amin, and Seung Won Jeong
- Subjects
Cancer Research ,mTOR inhibitors ,PRAS40 ,P70-S6 Kinase 1 ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,mTORC2 ,Cell Movement ,Cell Line, Tumor ,PTEN ,Humans ,Sulfones ,Naphthyridines ,Protein kinase B ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Torin2 ,Torin1 ,biology ,Brain Neoplasms ,glioblastoma ,Articles ,Oncology ,biology.protein ,Cancer research ,mTOR ,Phosphorylation - Abstract
Mechanistic target of rapamycin (mTOR), which functions via two multiprotein complexes termed mTORC1 and mTORC2, is positioned in the canonical phosphoinositide 3‑kinase‑related kinase (PI3K)/AKT (PI3K/AKT) pathways. These complexes exert their actions by regulating other important kinases, such as 40S ribosomal S6 kinases (S6K), eukaryotic translation initiation factor 4E (elF4E)‑binding protein 1 (4E‑BP1) and AKT, to control cell growth, proliferation, migration and survival in response to nutrients and growth factors. Glioblastoma (GB) is a devastating form of brain cancer, where the mTOR pathway is deregulated due to frequent upregulation of the Receptor Tyrosine Kinase/PI3K pathways and loss of the tumor suppressor phosphatase and tensin homologue (PTEN). Rapamycin and its analogs were less successful in clinical trials for patients with GB due to their incomplete inhibition of mTORC1 and the activation of mitogenic pathways via negative feedback loops. Here, the effects of selective ATP‑competitive dual inhibitors of mTORC1 and mTORC2, Torin1, Torin2 and XL388, are reported. Torin2 exhibited concentration‑dependent pharmacodynamic effects on inhibition of phosphorylation of the mTORC1 substrates S6KSer235/236 and 4E‑BP1Thr37/46 as well as the mTORC2 substrate AKTSer473 resulting in suppression of tumor cell migration, proliferation and S‑phase entry. Torin1 demonstrated similar effects, but only at higher doses. XL388 suppressed cell proliferation at a higher dose, but failed to inhibit cell migration. Treatment with Torin1 suppressed phosphorylation of proline rich AKT substrate of 40 kDa (PRAS40) at Threonine 246 (PRAS40Thr246) whereas Torin2 completely abolished it. XL388 treatment suppressed the phosphorylation of PRAS40Thr246 only at higher doses. Drug resistance analysis revealed that treatment of GB cells with XL388 rendered partial drug resistance, which was also seen to a lesser extent with rapamycin and Torin1 treatments. However, treatment with Torin2 completely eradicated the tumor cell population. These results strongly suggest that Torin2, compared to Torin1 or XL388, is more effective in suppressing mTORC1 and mTORC2, and therefore in the inhibition of the GB cell proliferation, dissemination and in overcoming resistance to therapy. These findings underscore the significance of Torin2 in the treatment of GB.
- Published
- 2021
24. THE STUDY OF THE ACTIVATION OF MTORC1 AND ITS SUBSTRATE P70S6K INVOLVED IN TYPE 2 DIABETES MELLITUS AND ONCOGENETIC PROCESSES
- Author
-
L.K. Sokolova, Vladimir Pushkarev, M.D. Tronko, O.I. Kovzun, V.V. Pushkarev, and T.S. Vatseba
- Subjects
signaling pathway ,0301 basic medicine ,PRAS40 ,Type 2 diabetes ,mTORC1 ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,рак ,Diabetes mellitus ,сигнальний шлях ,medicine ,cancer ,p70S6K ,PI3K/AKT/mTOR pathway ,Kinase ,business.industry ,Cancer ,Type 2 Diabetes Mellitus ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,діабет другого типу ,mTOR ,Cancer research ,type 2 diabetes ,Carcinogenesis ,business - Abstract
Вступ. Новітні дослідження довели вплив патогенетичних факторів цукрового діабету на активність внутрішньоклітинних сигнальних шляхів регуляції онкогенезу іметаболізму, одним з яких є PI3K/Akt/mTORC1. Макрофаги та лімфоцити беруть участь у патогенезі діабету та раку. Надмірна активація компонентів та субстратів PI3K/Akt/mTORC1 в цих клітинах може вказувати на необхідність додаткової корекції метаболічних процесів у хворих на діабет 2 типу з точки зору профілактики онкологічних захворювань. Мета: вивчити активацію mTORC1 шляхом визначення фосфорилювання PRAS40 та p70S6K1 у лейкоцитах хворих на діабет 2-го типу та рак. Матеріали та методи.Жінки, залучені у дослідженні, були поділені на групи: контрольна група, пацієнти з діабетом 2 типу, хворі на рак та пацієнти з поєднанням раку і діабету. Вміст фосфорильованих PRAS40 (фосфо-T246) та p70S6K1 (фосфо-T389) визначали імуноферментним методом, використовуючи лабораторні набори ELISA КНО0421 таELISA 85-86053 фірми Invitrogen (США). Концентрацію білка в лізаті визначали за допомогою набору для аналізу протеїнів BCA Novagen (США). Вимірювання проводилися намікропланшетному зчитувачі (Bio-tek Instruments, США) при довжині хвилі 450 нм.Результати. Виявлений достовірно підвищений вміст фосфорильованих PRAS40 та p70S6K1 у лейкоцитах хворих на цукровий діабет 2-го типу та рак. Кількість позитивних фосфо-PRAS40 проб у хворих на діабет становила 83,3%, а у хворих на рак -66,7%. Виявлений знижений вміст фосфо-PRAS40 в лейкоцитах пацієнтів з поєднанням діабету і раку. Висновки.Підвищений вміст фосфорильованих PRAS40 та p70S6K1 доводить активацію досліджуваного сигнального шляху при цукровому діабеті 2 типу. Зниження їх активаціїпри поєднанні раку та діабету можна пояснити можливими конкуруючими ефектами білків, які впливають на регулятори цих кіназ або на них безпосередньо. ntroduction.Pathogenetic factors of diabetes may affect the activity of intracellular systems of oncogenesis andmetabolismregulation, one of which is PI3K/Akt/mTORC1. Macrophages and lymphocytes are involved in the pathogenesis of diabetes and cancer. Detection of excessive activation of PI3K/Akt/mTORC1 components and substrates in these cells may indicate the need for additional correction of metabolic processes in patients with type 2 diabetes from the point of prevention of cancer. The aim:to study the activation of mTORC1 by determining the phosphorylation of PRAS40 and p70S6K1 in the leukocytes of patients with type 2 diabetes and cancer.Materials and methods. The study included women from the following groups: control group, patients with type 2 diabetes, cancer patients, patients with both diseases. The content of phosphorylated PRAS40 (phospho-T246) and p70S6K1 (phospho-T389) was determined using laboratory kits ELISA KNO0421 and ELISA 85-86053 of Invitrogen (USA). The protein concentration in the lysate was determined using a BCA Novagen protein assay kit (USA). Measurements were performed on a microplate reader (Bio-tek Instruments, USA) at a wavelength of 450 nm. Results.Significantly increased content of phosphorylated PRAS40 and p70S6K1 inleukocytes of patients with type 2 diabetes mellitus and cancer was detected. The number of positive phospho-PRAS40 tests in patients with diabetes was 83.3%, and in cancer patients -66.7%. Was revealedthe reduced content of phospho-PRAS40 in leukocytesof patients with a combination of diabetes and cancer.Conclusions.The increased amount of phosphorylated PRAS40 and p70S6K1 proves the activation of the studied signaling pathway by diabetes mellitus type 2. Its decrease by cancer and diabetes can be explained by the possible competing effects of the proteins that affect upstream regulators of these kinases or them directly.
- Published
- 2020
- Full Text
- View/download PDF
25. Proline-rich AKT substrate of 40-kDa (PRAS40) in the pathophysiology of cancer.
- Author
-
Malla, Ritu, Jr.Ashby, Charles R., Narayanan, Narayanan K., Narayanan, Bhagavathi, Faridi, Jesika S., and Tiwari, Amit K.
- Subjects
- *
PROLINE , *PHOSPHATIDYLINOSITOL 3-kinases , *INSULIN resistance , *CELLULAR signal transduction , *MICROARRAY technology ,CANCER pathophysiology - Abstract
Dysregulation of PI3K-AKT-mTOR pathway has been reported in various pathologies, such as cancer and insulin resistance. The proline-rich AKT substrate of 40-kDa (PRAS40), also known as AKT substrate 1 (AKT1S1), lies at the crossroads of these cascades and inhibits the activity of the mTOR complex 1 (mTORC1) kinase. This review discusses the role of PRAS40 and possible feedback mechanisms, and alterations in AKT/PRAS40/mTOR signaling that have been implicated in the pathogenesis of tumor progression. Additionally, we probed new datasets extracted from Oncomine, a cancer microarray database containing datasets derived from patient samples, to further understand the role of PRAS40 (AKT1S1). These data strongly supports the hypothesis that PRAS40 may serve as a potential therapeutic target for various cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
26. Genetic ablation of PRAS40 improves glucose homeostasis via linking the AKT and mTOR pathways.
- Author
-
Malla, Ritu, Wang, Yu, Chan, William K., Tiwari, Amit K., and Faridi, Jesika S.
- Subjects
- *
ABLATION techniques , *PROTEIN kinase B , *MTOR protein , *PROTEIN-protein interactions , *DIABETES , *HYPERGLYCEMIA , *STREPTOZOTOCIN - Abstract
Alterations in PI3K–AKT–mTOR signaling have been implicated in diabetes. This study assessed whether disruption of PRAS40, a substrate of AKT and component of mTORC1, would alter glucose homeostasis and prevent hyperglycemia in the streptozotocin (STZ)-induced diabetes mouse model. PRAS40 ablation resulted in a mild lowering of blood glucose levels and glycated hemoglobin (HbA1C), a lowered insulin requirement, and improved glucose tolerance in untreated PRAS40 gene knockout (PRAS40 −/− ) as compared to wild-type (PRAS40 +/+ ) mice. Diabetes was then induced in these mice using STZ at 50 mg/kg/day over five days. Following STZ-treatment, PRAS40 −/− mice exhibited significantly lower blood glucose and HbA1C levels than PRAS40 +/+ mice. Liver tissue of PRAS40 −/− mice and shPRAS40 Hep3B cells showed increased activation of AKT (p-AKT T308) and mTORC1 (p-p70S6K) signaling as well as decreased p-AKT (S473) and increased p-IRS1 (S612) protein levels. Altered tissue gene expression of several glucose transporters (GLUT) and increased hepatic GLUT4 protein levels were observed in PRAS40 −/− as compared to PRAS40 +/+ mice. In summary, PRAS40 deletion significantly attenuates hyperglycemia in STZ-induced PRAS40 −/− mice through increased hepatic AKT and mTORC1 signaling, a lowered serum insulin requirement, and altered hepatic GLUT4 levels. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
27. Sepsis-induced changes in amino acid transporters and leucine signaling via mTOR in skeletal muscle.
- Author
-
Laufenberg, Lacee, Pruznak, Anne, Navaratnarajah, Maithili, and Lang, Charles
- Subjects
- *
SEPSIS , *AMINO acid transport , *LEUCINE , *CELLULAR signal transduction , *MTOR protein , *SKELETAL muscle - Abstract
The present study tested the hypothesis that sepsis-induced leucine (Leu) resistance in skeletal muscle is associated with a down-regulation of amino acid transporters important in regulating Leu flux or an impairment in the formation of the Leu-sensitive mTOR-Ragulator complex. Sepsis in adult male rats decreased basal protein synthesis in gastrocnemius, associated with a reduction in mTOR activation as indicated by decreased 4E-BP1 and S6K1 phosphorylation. The ability of oral Leu to increase protein synthesis and mTOR kinase after 1 h was largely prevented in sepsis. Sepsis increased CAT1, LAT2 and SNAT2 mRNA content two- to fourfold, but only the protein content for CAT1 (20 % decrease) differed significantly. Conversely, sepsis decreased the proton-assisted amino acid transporter (PAT)-2 mRNA by 60 %, but without a coordinate change in PAT2 protein. There was no sepsis or Leu effect on the protein content for RagA-D, LAMTOR-1 and -2, raptor, Rheb or mTOR in muscle. The binding of mTOR, PRAS40 and RagC to raptor did not differ for control and septic muscle in the basal condition; however, the Leu-induced decrease in PRAS40·raptor and increase in RagC·raptor seen in control muscle was absent in sepsis. The intracellular Leu concentration was increased in septic muscle, compared to basal control conditions, and oral Leu further increased the intracellular Leu concentration similarly in both control and septic rats. Hence, while alterations in select amino acid transporters are not associated with development of sepsis-induced Leu resistance, the Leu-stimulated binding of raptor with RagC and the recruitment of mTOR/raptor to the endosome-lysosomal compartment may partially explain the inability of Leu to fully activate mTOR and muscle protein synthesis. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
28. Over-expression of PRAS40 enhances insulin sensitivity in skeletal muscle.
- Author
-
Wiza, Claudia, Chadt, Alexandra, Blumensatt, Marcel, Kanzleiter, Timo, Herzfeld De Wiza, Daniella, Horrighs, Angelika, Mueller, Heidi, Nascimento, Emmani B.M., Schürmann, Annette, Al-Hasani, Hadi, and Ouwens, D. Margriet
- Subjects
- *
PROLINE , *PROTEIN kinase B , *SKELETAL muscle , *INSULIN resistance , *GENE silencing , *MUSCLE cells , *PHOSPHORYLATION - Abstract
Context: Silencing proline-rich Akt substrate of 40-kDa (PRAS40) impairs insulin signalling in skeletal muscle. Objective: This study assessed the effects of over-expressing wild type or mutant AAA-PRAS40, in which the major phosphorylation sites and mTORC1-binding site were mutated, on insulin signalling in skeletal muscle. Results: Over-expression of WT-PRAS40, but not AAA-PRAS40, impaired the insulin-mediated activation of the mTORC1-pathway in human skeletal muscle cells (hSkMC). However, insulin-mediated Akt-phosphorylation was increased upon over-expression of WT-PRAS40 both in hSkMC and mouse skeletal muscle. Also over-expression of AAA-PRAS40 had an insulin-sensitizing effect, although to a lesser extent as WT-PRAS40. The insulin-sensitizing effect associated with increased IRS1 protein abundance and inhibition of proteasome activity. Finally, over-expression of WT-PRAS40 reversed hyperinsulinemia-induced insulin resistance. Conclusion: This study identifies PRAS40 as a regulator of insulin sensitivity in hSkMC. In contrast to the mTORC1-pathway, the insulin-sensitizing action of PRAS40 occurs independent of binding of PRAS40 to the mTORC1-complex. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
29. Dysmetabolic and intracellular mechanisms of type 2 diabetes mellitus and colorectal cancer association
- Author
-
Vatseba, T. S. and Vatseba, T. S.
- Published
- 2020
30. PRAS40 prevents development of diabetic cardiomyopathy and improves hepatic insulin sensitivity in obesity.
- Author
-
Völkers, Mirko, Doroudgar, Shirin, Nguyen, Nathalie, Konstandin, Mathias H, Quijada, Pearl, Din, Shabana, Ornelas, Luis, Thuerauf, Donna J, Gude, Natalie, Friedrich, Kilian, Herzig, Stephan, Glembotski, Christopher C, and Sussman, Mark A
- Abstract
Diabetes is a multi-organ disease and diabetic cardiomyopathy can result in heart failure, which is a leading cause of morbidity and mortality in diabetic patients. In the liver, insulin resistance contributes to hyperglycaemia and hyperlipidaemia, which further worsens the metabolic profile. Defects in mTOR signalling are believed to contribute to metabolic dysfunctions in diabetic liver and hearts, but evidence is missing that mTOR activation is causal to the development of diabetic cardiomyopathy. This study shows that specific mTORC1 inhibition by PRAS40 prevents the development of diabetic cardiomyopathy. This phenotype was associated with improved metabolic function, blunted hypertrophic growth and preserved cardiac function. In addition PRAS40 treatment improves hepatic insulin sensitivity and reduces systemic hyperglycaemia in obese mice. Thus, unlike rapamycin, mTORC1 inhibition with PRAS40 improves metabolic profile in diabetic mice. These findings may open novel avenues for therapeutic strategies using PRAS40 directed against diabetic-related diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
31. Proline-rich Akt substrate of 40-kDa contains a nuclear export signal.
- Author
-
Wiza, Claudia, Nascimento, Emmani B.M., Linssen, Margot M.L., Carlotti, Françoise, Herzfeld de Wiza, Daniella, van der Zon, Gerard C.M., Maassen, J. Antonie, Diamant, Michaela, Guigas, Bruno, and Ouwens, D. Margriet
- Subjects
- *
PROLINE , *CELLULAR signal transduction , *RAPAMYCIN , *CYTOSOL , *PHOSPHOPROTEINS , *HELA cells , *GENETIC regulation , *INSULIN receptors - Abstract
Abstract: The proline-rich Akt substrate of 40-kDa (PRAS40) has been linked to the regulation of the activity of the mammalian target of rapamycin complex 1 as well as insulin action. Despite these cytosolic functions, PRAS40 was originally identified as nuclear phosphoprotein in Hela cells. This study aimed to detail mechanisms and consequences of the nucleocytosolic trafficking of PRAS40. Sequence analysis identified a potential leucine-rich nuclear export signal (NES) within PRAS40. Incubation of A14 fibroblasts overexpressing human PRAS40 (hPRAS40) resulted in nuclear accumulation of the protein. Furthermore, mutation of the NES mimicked the effects of leptomycin B, a specific inhibitor of nuclear export, on the subcellular localization of hPRAS40. Finally, A14 cells expressing the NES-mutant showed impaired activation of components of the Akt-pathway as well as of the mTORC1 substrate p70 S6 kinase after insulin stimulation. This impaired insulin signaling could be ascribed to reduced protein levels of insulin receptor substrate 1 in cells expressing mutant NES. In conclusion, PRAS40 contains a functional nuclear export signal. Furthermore, enforced nuclear accumulation of PRAS40 impairs insulin action, thereby substantiating the function of this protein in the regulation of insulin sensitivity. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
32. Knockdown of PRAS40 inhibits insulin action via proteasome-mediated degradation of IRS1 in primary human skeletal muscle cells.
- Author
-
Wiza, C., Herzfeld de Wiza, D., Nascimento, E., Lehr, S., Al-Hasani, H., and Ouwens, D.
- Abstract
Aims/hypothesis: The proline-rich Akt substrate of 40 kDa (PRAS40) is a component of the mammalian target of rapamycin complex 1 (mTORC1) and among the most prominent Akt substrates in skeletal muscle. Yet the cellular functions of PRAS40 are incompletely defined. This study assessed the function of PRAS40 in insulin action in primary human skeletal muscle cells (hSkMC). Methods: Insulin action was examined in hSkMC following small interfering RNA-mediated silencing of PRAS40 (also known as AKT1S1) under normal conditions and following chemokine-induced insulin resistance. Results: PRAS40 knockdown (PRAS40-KD) in hSkMC decreased insulin-mediated phosphorylation of Akt by 50% ( p < 0.05) as well as of the Akt substrates glycogen synthase kinase 3 (40%) and tuberous sclerosis complex 2 (32%) (both p < 0.05). Furthermore, insulin-stimulated glucose uptake was reduced by 20% in PRAS40-KD myotubes ( p < 0.05). Exposing PRAS40-KD myotubes to chemokines caused no additional deterioration of insulin action. PRAS40-KD further reduced insulin-mediated phosphorylation of the mTORC1-regulated proteins p70S6 kinase (p70S6K) (47%), S6 (43%), and eukaryotic elongation 4E-binding protein 1 (100%), as well as protein levels of growth factor receptor bound protein 10 (35%) (all p < 0.05). The inhibition of insulin action in PRAS40-KD myotubes was associated with a reduction in IRS1 protein levels (60%) ( p < 0.05), and was reversed by pharmacological proteasome inhibition. Accordingly, expression of the genes encoding E3-ligases F-box protein 32 (also known as atrogin-1) and muscle RING-finger protein-1 and activity of the proteasome was elevated in PRAS40-KD myotubes. Conclusions/interpretation: Inhibition of insulin action in PRAS40-KD myotubes was found to associate with IRS1 degradation promoted by increased proteasome activity rather than hyperactivation of the p70S6K-negative-feedback loop. These findings identify PRAS40 as a modulator of insulin action. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
33. Insulin-like growth factor-1 induces the phosphorylation of PRAS40 via the PI3K/Akt signaling pathway in PC12 cells
- Author
-
Wang, Haitao, Zhang, Qishan, Zhang, Lang, Little, Peter J., Xie, Xiaochun, Meng, Qian, Ren, Yannan, Zhou, Lihua, Gao, Guoquan, Quirion, Remi, and Zheng, Wenhua
- Subjects
- *
SOMATOMEDIN , *PHOSPHORYLATION , *CELLULAR signal transduction , *NEURONS , *PHOSPHOINOSITIDES , *ENZYME inhibitors , *MITOGEN-activated protein kinases - Abstract
Abstract: Insulin-like growth factor-1 (IGF-1) is a polypeptide tropic factor that plays an important role in the survival and differentiation of both neuronal and non-neuronal cells. Numerous studies have demonstrated that IGF-1 promotes neuronal cell survival via the PI3K/Akt signaling pathway. Proline-rich Akt substrate of 40kDa (PRAS40) is a recently discovered downstream target of Akt. However, the relationship between IGF-1 and PRAS40 is not known. In this study, we characterized the phosphorylation of PRAS40 induced by IGF-1 in PC12 cells and explored the signaling pathway responsible for the effect of IGF-1. IGF-1 induced the phosphorylation of Akt at Thr473 and PRAS40 at Thr246 in PC12 cells. The phosphorylation of Akt and PRAS40 induced by IGF-1 (100ng/ml) was inhibited by the phosphatidylinositide 3-kinase (PI3K) specific inhibitor LY294002 (50μM), while no inhibitory effect was observed for a MAPK kinase pathway specific inhibitor PD98059 nor a p38 MAPK inhibitor PD169316, suggesting that the phosphorylation of PRAS40 induced by IGF-1 is mediated by the PI3K pathway in PC12 cells and primary cultured neurons. In further support this hypothesis, an Akt kinase specific inhibitor, Akt inhibitor VIII, attenuated IGF-1-induced phosphorylation of PRAS40 at the concentration that blocked the phosphorylation of Akt induced by IGF-1. Taken together, these data demonstrate that IGF-1 stimulates the phosphorylation of PRAS40 at Thr246 in neuronal cells and the effect of IGF-1 is mediated, at least in part, by the PI3K/Akt signaling pathway. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
34. Proline-rich Akt substrate of 40kDa (PRAS40): A novel downstream target of PI3k/Akt signaling pathway
- Author
-
Wang, Haitao, Zhang, Qishan, Wen, Qiang, Zheng, Yongxin, Philip, Lazarovici, Jiang, Hao, Lin, Jun, and Zheng, WenHua
- Subjects
- *
CELLULAR signal transduction , *PROTEIN kinases , *TYPE 2 diabetes , *PROLINE , *MELANOMA , *RAPAMYCIN , *GROWTH factors , *CARRIER proteins - Abstract
Abstract: Modifications in signaling of the proline-rich Akt substrate of 40-kDa (PRAS40) pathway is implicated in type 2 diabetes and melanoma. PRAS40 is known for its ability to regulate the mammalian target of rapamycin complex 1 (mTORC1) kinase activity, possessing a key regulatory role at the cross point of signal transduction pathways activated by growth factor receptors. Recently it has been found that PRAS40 is regulated by its upstream phosphatidylinositol 3-kinase/Akt (PI3K/Akt) which is activated by many tyrosine kinase receptors growth factors including insulin-like growth factor 1. Also, PRAS40 functions downstream of mTORC1 and upstream from its effectors ribosomal protein S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E binding protein 1 (4E-BP1). Phosphorylation of PRAS40 by Akt and mTORC1 disrupts the binding between mTORC1 and PRAS40, and relieves the inhibitory constraint of PRAS40 on mTORC1 activity. This review summarizes the signaling regulating PRAS40 phosphorylation, as well as the dual function of PRAS40 as substrate and inhibitor of mTORC1 upon growth factor stimulation and under pathophysiological conditions. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
35. Cytoplasmic polyadenylation element binding protein is a conserved target of tumor suppressor HRPT2/CDC73.
- Author
-
Zhang, J.-H., Panicker, L. M., Seigneur, E. M., Lin, L., House, C. D., Morgan, W., Chen, W. C., Mehta, H., Haj-Ali, M., Yu, Z.-X., and Simonds, W. F.
- Subjects
- *
CARRIER proteins , *HYPERPARATHYROIDISM , *MICROBIAL genetics , *APOPTOSIS , *NUCLEOPROTEINS - Abstract
Parafibromin, a tumor suppressor protein encoded by HRPT2/CDC73 and implicated in parathyroid cancer and the hyperparathyroidism-jaw tumor (HPT-JT) familial cancer syndrome, is part of the PAF1 transcriptional regulatory complex. Parafibromin has been implicated in apoptosis and growth arrest, but the mechanism by which its loss of function promotes neoplasia is poorly understood. In this study we report that a hypomorphic allele of hyrax (hyx), the Drosophila homolog of HRPT2/CDC73, rescues the loss-of-ventral-eye phenotype of lobe (Akt1s1). Such rescue is consistent with previous reports that hyx/parafibromin is required for the nuclear transduction of Wingless (Wg)/Wnt signals and that Wg signaling antagonizes lobe function. A screen using double hyx/lobe heterozygotes identified an additional interaction with orb and orb2, the homologs of mammalian cytoplasmic polyadenylation element binding protein (CPEB), a translational regulatory protein. Hyx and orb2 heterozygotes lived longer and were more resistant to starvation than controls. In mammalian cells, knockdown of parafibromin expression reduced levels of CPEB1. Chromatin immunoprecipitation (ChIP) showed occupancy of CPEB1 by endogenous parafibromin. Bioinformatic analysis revealed a significant overlap between human transcripts potentially regulated by parafibromin and CPEB. These results show that parafibromin may exert both transcriptional and, through CPEB, translational control over a subset of target genes and that loss of parafibromin (and CPEB) function may promote tumorigenesis in part by conferring resistance to nutritional stress. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
36. Regulation of Akt during torpor in the hibernating ground squirrel, Ictidomys tridecemlineatus.
- Author
-
McMullen, David C. and Hallenbeck, John M.
- Subjects
- *
GROUND squirrels , *HIBERNATION , *ANIMAL wintering , *BODY temperature , *PHOSPHORYLATION - Abstract
The 13-lined ground squirrel ( Ictidomys tridecemlineatus) is capable of entering into extended periods of torpor during winter hibernation. The state of torpor represents a hypometabolic shift wherein the rate of oxygen consuming processes are strongly repressed in an effort to maintain cellular homeostasis as the availability of food energy becomes limited. We are interested in studying hibernation/torpor because of the robust state of tolerance to constrained oxygen delivery, oligemia, and hypothermia achieved by the tissues of hibernating mammals. The role of the serine/threonine kinase Akt (also known as PKB) has been examined in torpor in previous studies. However, this is the first study that examines the level of Akt phosphorylation in the liver during the two transition phases of the hibernation cycle: entrance into torpor, and the subsequent arousal from torpor. Our results indicate that Akt is activated in the squirrel liver by phosphorylation of two key residues (Thr308 and Ser473) during entrance into torpor and arousal from torpor. Moreover, we observed increased phosphorylation of key substrates of Akt during the two transition stages of torpor. Finally, this study reports the novel finding that PRAS40, a component of the TORC1 multi-protein complex and a potentially important modulator of metabolism, is regulated during torpor. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
37. Phosphorylation of PRAS40 on Thr246 by PKB/AKT facilitates efficient phosphorylation of Ser183 by mTORC1
- Author
-
Nascimento, Emmani B.M., Snel, Marieke, Guigas, Bruno, van der Zon, Gerard C.M., Kriek, Jan, Maassen, J. Antonie, Jazet, Ingrid M., Diamant, Michaela, and Ouwens, D. Margriet
- Subjects
- *
TYPE 2 diabetes , *PHOSPHORYLATION , *PROTEIN kinases , *RAPAMYCIN , *CELLULAR signal transduction , *INSULIN , *LABORATORY rats - Abstract
Abstract: Type 2 diabetes is associated with alterations in protein kinase B (PKB/Akt) and mammalian target of rapamycin complex 1 (mTORC1) signalling. The proline-rich Akt substrate of 40-kDa (PRAS40) is a component of mTORC1, which has a regulatory function at the intersection of the PKB/Akt and mTORC1 signalling pathway. Phosphorylation of PRAS40-Thr246 by PKB/Akt, and PRAS40-Ser183 and PRAS40-Ser221 by mTORC1 results in dissociation from mTORC1, and its binding to 14-3-3 proteins. Although all phosphorylation sites within PRAS40 have been implicated in 14-3-3 binding, substitution of Thr246 by Ala alone is sufficient to abolish 14-3-3 binding under conditions of intact mTORC1 signalling. This suggests that phosphorylation of PRAS40-Thr246 may facilitate efficient phosphorylation of PRAS40 on its mTORC1-dependent sites. In the present study, we investigated the mechanism of PRAS40-Ser183 phosphorylation in response to insulin. Insulin promoted PRAS40-Ser183 phosphorylation after a euglycaemic–hyperinsulinaemic clamp in human skeletal muscle. The insulin-induced PRAS40-Ser183 phosphorylation was further evidenced in vivo in rat skeletal and cardiac muscle, and in vitro in A14 fibroblasts, 3T3L1 adipocytes and L6 myotubes. Inhibition of mTORC1 by rapamycin or amino acid deprivation partially abrogated insulin-mediated PRAS40-Ser183 phosphorylation in cultured cell lines. However, lowering insulin-induced PRAS40-Thr246 phosphorylation using wortmannin or palmitate in cell lines, or by feeding rats a high-fat diet, completely abolished insulin-mediated PRAS40-Ser183 phosphorylation. In addition, replacement of Thr246 by Ala reduced insulin-mediated PRAS40-Ser183 phosphorylation. We conclude that PRAS40-Ser183 is a component of insulin action, and that efficient phosphorylation of PRAS40-Ser183 by mTORC1 requires the phosphorylation of PRAS40-Thr246 by PKB/Akt. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
38. Key factors in mTOR regulation.
- Author
-
Xiaochun Bai and Yu Jiang
- Subjects
- *
RAPAMYCIN , *MAMMALS , *PROTEIN kinases , *CELL metabolism , *IMMUNOSUPPRESSIVE agents - Abstract
Mammalian target of rapamycin (mTOR) is a protein serine/threonine kinase that controls a wide range of growth-related cellular processes. In the past several years, many factors have been identified that are involved in controlling mTOR activity. Those factors in turn are regulated by diverse signaling cascades responsive to changes in intracellular and environmental conditions. The molecular connections between mTOR and its regulators form a complex signaling network that governs cellular metabolism, growth and proliferation. In this review, we discuss some key factors in mTOR regulation and mechanisms by which these factors control mTOR activity. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
39. Reduction of Lobe leads to TORC1 hypoactivation that induces ectopic Jak/STAT signaling to impair Drosophila eye development
- Author
-
Wang, Ying-Hsuan and Huang, Min-Lang
- Subjects
- *
FRONTAL lobe , *CELLULAR signal transduction , *DROSOPHILA physiology , *BRAIN physiology , *MAMMALS , *CELL growth , *ECTOPIC tissue , *EYE , *CELL proliferation - Abstract
Abstract: The TOR and Jak/STAT signal pathways are highly conserved from Drosophila to mammals, but it is unclear whether they interact during development. The proline-rich Akt substrate of 40kDa (PRAS40) mediates the TOR signal pathway through regulation of TORC1 activity, but its functions in TORC1 proved in cultured cells are controversial. The Drosophila gene Lobe (L) encodes the PRAS40 ortholog required for eye cell survival. L mutants exhibit apoptosis and eye-reduction phenotypes. It is unknown whether L regulates eye development via regulation of TORC1 activity. We found that reducing the L level, by hypomorphic L mutation or heterozygosity of the null L mutation, resulted in ectopic expression of unpaired (upd), which is known to act through the Jak/STAT signal pathway to promote proliferation during eye development. Unexpectedly, when L was reduced, decreasing Jak/STAT restored the eye size, whereas increasing Jak/STAT prevented eye formation. We found that ectopic Jak/STAT signaling and apoptosis are mutually dependent in L mutants, indicating that L reduction makes Jak/STAT signaling harmful to eye development. In addition, our genetic data suggest that TORC1 signaling is downregulated upon L reduction, supporting the idea that L regulates eye development through regulation of TORC1 activity. Similar to L reduction, decreasing TORC1 signaling by dTOR overexpression results in ectopic upd expression and apoptosis. A novel finding from our data is that dysregulated TORC1 signaling regulates the expression of upd and the function of the Jak/STAT signal pathway in Drosophila eye development. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
40. PRAS40: Target or modulator of mTORC1 signalling and insulin action?
- Author
-
Nascimento, Emmani B.M. and Ouwens, D. Margriet
- Subjects
- *
PROTEIN kinases , *RAPAMYCIN , *TYPE 2 diabetes , *INSULIN resistance , *PHOSPHORYLATION , *CANCER - Abstract
Alterations in signalling via protein kinase B (PKB/Akt) and the mammalian target of rapamycin (mTOR) frequently occur in type 2 diabetes and various human malignancies. Proline-rich Akt substrate of 40-kDa (PRAS40) has a regulatory function at the intersection of these pathways. The interaction of PRAS40 with the mTOR complex 1 (mTORC1) inhibits the activity of mTORC1. Phosphorylation of PRAS40 by PKB/Akt and mTORC1 disrupts the binding between mTORC1 and PRAS40, and relieves the inhibitory constraint of PRAS40 on mTORC1 activity. This review summarizes the signalling pathways regulating PRAS40 phosphorylation, as well as the dual function of PRAS40 as substrate and inhibitor of mTORC1 in the physiological situation, and under pathological conditions, such as insulin resistance and cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
41. Mammalian target of rapamycin complex 1: Signalling inputs, substrates and feedback mechanisms
- Author
-
Dunlop, E.A. and Tee, A.R.
- Subjects
- *
PROTEIN kinases , *CELLULAR signal transduction , *CARCINOGENESIS , *HAMARTOMA , *RAPAMYCIN , *REGULATION of cell growth , *CELL proliferation , *PROTEIN synthesis - Abstract
Abstract: The mammalian target of rapamycin (mTOR) signalling pathway is implicated in the pathogenesis of a number of cancers and inherited hamartoma syndromes which have led to mTOR inhibitors, such as rapamycin, being tested in clinical trials. Knowledge of the mTOR pathway is rapidly expanding. This review provides an update on the most recent additions to the mTOR pathway with particular emphasis on mTORC1 signalling. mTORC1 signalling is classically known for its role in regulating cell growth and proliferation through modulation of protein synthesis. Recent research has identified novel mTORC1 cell signalling mechanisms that modulate mitochondrial biogenesis, hypoxia signalling and cell cycle progression and uncovered novel mTORC1 targets; YY1, HIF and SGK1. It is unsurprising that regulation of mTORC1 is multifaceted with many positive and negative signalling inputs. We discuss the recent advances that have been made to determine the upstream mechanisms that control mTORC1 through hypoxia, energy sensing and nutrient signalling. Also discussed are current findings that have unravelled a series of novel mTORC1-associated proteins that directly control the activity of mTORC1 and include PRAS40, FKBP38, Rag GTPases and RalA. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
42. Hydrogen peroxide impairs insulin-stimulated assembly of mTORC1
- Author
-
Zhang, Lianqin, Kimball, Scot R., Jefferson, Leonard S., and Shenberger, Jeffrey S.
- Subjects
- *
HYDROGEN peroxide , *RAPAMYCIN , *OXIDIZING agents , *PHOSPHORYLATION , *CARRIER proteins , *PROTEIN kinases , *TUBEROUS sclerosis - Abstract
Abstract: Oxidants are well recognized for their capacity to reduce the phosphorylation of the mammalian target of rapamycin (mTOR) substrates, eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) and p70 S6 kinase 1 (S6K1), thereby hindering mRNA translation at the level of initiation. mTOR functions to regulate mRNA translation by forming the signaling complex mTORC1 (mTOR, raptor, GβL). Insulin signaling to mTORC1 is dependent upon phosphorylation of Akt/PKB and the inhibition of the tuberous sclerosis complex (TSC1/2), thereby enhancing the phosphorylation of 4E-BP1 and S6K1. In this study we report the effect of H2O2 on insulin-stimulated mTORC1 activity and assembly using A549 and bovine aortic smooth muscle cells. We show that insulin stimulated the phosphorylation of TSC2 leading to a reduction in raptor–mTOR binding and in the quantity of proline-rich Akt substrate 40 (PRAS40) precipitating with mTOR. Insulin also increased 4E-BP1 coprecipitating with mTOR and the phosphorylation of the mTORC1 substrates 4E-BP1 and S6K1. H2O2, on the other hand, opposed the effects of insulin by increasing raptor–mTOR binding and the ratio of PRAS40/raptor derived from the mTOR immunoprecipitates in both cell types. These effects occurred in conjunction with a reduction in 4E-BP1 phosphorylation and the 4E-BP1/raptor ratio. siRNA-mediated knockdown of PRAS40 in A549 cells partially reversed the effect of H2O2 on 4E-BP1 phosphorylation but not on S6K1. These findings are consistent with PRAS40 functioning as a negative regulator of insulin-stimulated mTORC1 activity during oxidant stress. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
43. Effects of contraction and insulin on protein synthesis, AMP-activated protein kinase and phosphorylation state of translation factors in rat skeletal muscle.
- Author
-
Miranda, Lisa, Horman, Sandrine, De Potter, Isabelle, Hue, Louis, Jensen, Jørgen, and Rider, Mark H.
- Subjects
- *
PHOSPHORYLATION , *MUSCLE motility , *MUSCLE contraction , *INSULIN , *PROTEIN synthesis , *CARRIER proteins - Abstract
In rat epitrochlearis skeletal muscle, contraction inhibited the basal and insulin-stimulated rates of protein synthesis by 75 and 70%, respectively, while increasing adenosine monophosphate-activated protein kinase (AMPK) activity. Insulin, on the other hand, stimulated protein synthesis (by 30%) and increased p70 ribosomal protein S6 kinase (p70S6K) Thr389, 40S ribosomal protein S6 (rpS6) Ser235/236, rpS6 Ser240/244 and eukaryotic initiation factor-4E-binding protein-1 (4E-BP1) Thr37/46 phosphorylation over basal values. Electrical stimulation had no effect on mammalian target of rapamycin complex 1 (mTORC1) signalling, as reflected by the lack of reduction in basal levels of p70S6K, rpS6 Ser235/236, rpS6 Ser240/244 and 4E-BP1 phosphorylation, but did antagonize mTORC1 signalling after stimulation of the pathway by insulin. Eukaryotic elongation factor-2 (eEF2) Thr56 phosphorylation increased rapidly on electrical stimulation reaching a maximum at 1 min, whereas AMPK Thr172 phosphorylation slowly increased to reach threefold after 30 min. Eukaryotic elongation factor-2 kinase (eEF2K) was not activated after 30 min of contraction when AMPK was activated. This could not be explained by the expression of a tissue-specific isoform of eEF2K in skeletal muscle lacking the Ser398 AMPK phosphorylation site. Therefore, in this skeletal muscle system, the contraction-induced inhibition of protein synthesis could not be attributed to a reduction in mTORC1 signalling but could be due to an increase in eEF2 phosphorylation independent of AMPK activation. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
44. Increased expression of a proline-rich Akt substrate (PRAS40) in human copper/zinc-superoxide dismutase transgenic rats protects motor neurons from death after spinal cord injury.
- Author
-
Fengshan Yu, Narasimhan, Purnima, Saito, Atsushi, Jing Liu, and Chan, Pak H.
- Subjects
- *
MOTOR neurons , *NEURONS , *SPINAL cord , *ISCHEMIA , *CEREBROVASCULAR disease , *CEREBRAL ischemia , *CENTRAL nervous system , *BLOOD flow - Abstract
The serine-threonine kinase, Akt, plays an important role in the cell survival signaling pathway. A proline-rich Akt substrate, PRAS40, has been characterized, and an increase in phospho-PRAS40 (pPRAS40) is neuroprotective after transient focal cerebral ischemia. However, the involvement of PRAS40 in the cell death/survival pathway after spinal cord injury (SCI) is unclear. Liposome-mediated PRAS40 transfection was performed to study whether overexpression of pPRAS40 is neuroprotective. We further examined the expression of pPRAS40 after SCI by immunohistochemistry and Western blot using copper/zinc-superoxide dismutase (SOD1) transgenic (Tg) rats and wild-type (Wt) littermates. We then examined the relationship between PRAS40 and Akt by injection of LY294002, a phosphatidylinositol 3-kinase (PI3K) pathway inhibitor, or Akt inhibitor IV, a compound that inhibits Akt activation after SCI. Our data demonstrated that increased pPRAS40 resulted in survival of more motor neurons compared with control complementary DNA transfection. Phosphorylated PRAS40 increased in the Wt rats after SCI, whereas there was a greater and prolonged increase in the SOD1 Tg rats. Coimmunoprecipitation showed that binding of pPRAS40 with 14-3-3 increased 1 day after SCI in the Wt rats, whereas there was a significant increase in the Tg rats. The inhibitor studies showed that phospho-Akt and pPRAS40 were decreased after injection of LY294002 or Akt inhibitor IV. We conclude that an increase in pPRAS40 by transfection after SCI results in survival of motor neurons, and overexpression of SOD1 in the Tg rats results in an increase in endogenous pPRAS40 and a decrease in motor neuron death through the PI3K/Akt pathway.Journal of Cerebral Blood Flow & Metabolism (2008) 28, 44–52; doi:10.1038/sj.jcbfm.9600501; published online 25 April 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
45. Trimedazidine alleviates pulmonary artery banding-induced acute right heart dysfunction and activates PRAS40 in rats
- Author
-
Xinli Li, Aqian Wang, Heling Fu, Xiang Li, Jiyang Song, Ying Xu, Min Zhang, Yunshan Cao, and Shutong Shen
- Subjects
0301 basic medicine ,medicine.medical_specialty ,PRAS40 ,PDK4 ,030204 cardiovascular system & hematology ,Pulmonary artery banding ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine.artery ,Intensive care ,medicine ,trimedazidine ,business.industry ,acute right dysfunction ,medicine.disease ,Brain natriuretic peptide ,Surgery ,030104 developmental biology ,Blood pressure ,Oncology ,pulmonary artery banding ,Heart failure ,Pulmonary artery ,Ventricular pressure ,Cardiology ,business ,Research Paper - Abstract
// Yunshan Cao 1, 2 , Jiyang Song 1 , Shutong Shen 3 , Heling Fu 4 , Xiang Li 5 , Ying Xu 6 , Aqian Wang 1 , Xinli Li 3 and Min Zhang 7 1 Department of Cardiology, Gansu Provincial Hospital, Lanzhou 730000, China 2 Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Research Center for Translational Medicine, Shanghai 200120, China 3 Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China 4 Animal Core Facility, Nanjing Medical University, Nanjing 210029, China 5 Department of Intensive Care, Minhang Hospital, Fudan University, Shanghai 201100, China 6 Intensive Care Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China 7 Department of Pathology, Gansu Provincial Hospital, Lanzhou 730000, China Correspondence to: Yunshan Cao, email: yunshancao@126.com Min Zhang, email: sallyzhangmin@126.com Keywords: acute right dysfunction, trimedazidine, PRAS40, pulmonary artery banding Received: May 16, 2017 Accepted: August 08, 2017 Published: September 08, 2017 ABSTRACT The molecular mechanism underlying acute right heart failure (RHF) is poorly understood. We used pulmonary artery banding (PAB) to induce acute RHF characterized by a rapid rise of right ventricular pressure, and then a decrease in right ventricular pressure along with a decrease in blood pressure right after banding. We found higher brain natriuretic peptide (BNP) and beta-myosin heavy chain (βMHC) levels and lower alpha-myosin heavy chain (αMHC) levels in RHF rats than sham-operated rats. Hemodynamic indexes in rats with acute RHF were slightly improved by trimedazidine TMZ, a key inhibitor of fatty acid (FA) oxidation. TMZ also reversed downregulation of peroxisome proliferator-activated receptor gamma coactivator 1-beta (PGC-1β) and peroxisome proliferator-activated receptor alpha (PPARα) by PAB and up-regulates peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), peroxisome proliferator-activated receptor delta (PPARδ) and pyruvate dehydrogenase kinase isoform 4 (PDK4). In addition, TMZ reversed upregulation of phosphorylated Akt by PAB and increased phosphorylated proline-rich Akt-substrate 40 (PRAS40). Autophagy and apoptosis were not modified by PAB or TMZ. An acute RHF model was established in rats through 70% constriction of the pulmonary artery. TMZ treatment alleviated PAB-induced acute RHF by activating PRAS40 and upregulatingPGC-1α, PGC-1β, PPARα, PPARδ, and PDK4.
- Published
- 2017
- Full Text
- View/download PDF
46. Expression of proline-rich Akt-substrate PRAS40 in cell survival pathway and carcinogenesis.
- Author
-
Bei Huang and Porter, Gavin
- Subjects
CANCER invasiveness ,CARCINOGENESIS ,CANCER cells ,CELLS ,PHOSPHORYLATION - Abstract
Aim: To study the expression of proline-rich Akt-substrate PRAS40 in the cell survival pathway and tumor progression. Methods: The effects of three key kinase inhibitors on PRAS40 activity in the cell survival pathway, serum withdrawal, H
2 O2 and overexpression of Akt were tested. The expression of PRAS40, Akt, Raf and 14-3-3 in normal cells and cancer cell lines was determined by Western blot. Results: The PI3K inhibitors worthmannin and Ly294002, but not rapamycin, completely inhibited the phosphorylation of Akt and PRAS40. The phosphorylation level of Akt decreased after serum withdrawal and treatment with the MEK inhibitor Uo126, but increased after treatment with H2 O2 at low concentration, whereas none of these treatments changed PRAS40 activity. 14-3-3 is a PRAS40 binding protein, nd the expression of 14-3-3, like that of PRAS40, was higher in HeLa cells than in HEK293 cells; PRAS40 had a stronger phosphorylation activity in A549 and HeLa cancer cells than in HEK293 normal cells. In the breast cancer model (MCF10A/MCF7) and lung cancer model (BEAS/H1198/H1170) we also found the same result: PRAS40 was constitutively active in H1198/H1170 and MCF7 premalignant and malignant cancer cells, but weakly expressed in MCF10A and BEAS normal cell. We also discussed PRAS40 activity in other NSCLC cell lines. Conclusion: The PI3K-Akt survival pathway is the main pathway that PRAS40 is involved in; PRAS40 is a substrate for Akt, but can also be activated by an Aktindependent mechanisms. PRAS40 activation is an early event during breast and lung carcinogenesis. [ABSTRACT FROM AUTHOR]- Published
- 2005
- Full Text
- View/download PDF
47. MELK is Upregulated in Advanced Clear Cell Renal Cell Carcinoma and Promotes Disease Progression by Phosphorylating PRAS40
- Author
-
Xingtao Han, Yue Zhang, Han Zhang, Pengtao Wei, Wenwei Lv, Jinhui Yang, and Shuaifeng Qin
- Subjects
Leucine zipper ,differentially expressed genes ,PRAS40 ,Biomedical Engineering ,Regulator ,lcsh:Medicine ,mTORC1 ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Protein Serine-Threonine Kinases ,clear cell renal cell carcinoma ,Downregulation and upregulation ,Cell Movement ,Cell Line, Tumor ,Databases, Genetic ,MELK ,medicine ,Biomarkers, Tumor ,Humans ,Phosphorylation ,Carcinoma, Renal Cell ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Neoplasm Staging ,Transplantation ,Kinase ,lcsh:R ,Computational Biology ,Cell Biology ,Regulatory-Associated Protein of mTOR ,Original Articles ,medicine.disease ,Embryonic stem cell ,Kidney Neoplasms ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Clear cell renal cell carcinoma ,Gene Ontology ,Gene Knockdown Techniques ,Cancer research ,Disease Progression ,Kidney cancer - Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer. However, stage IV ccRCC is generally incurable and its molecular mechanism has not yet been fully clarified. In this study, in order to screen differentially expressed genes (DEGs) between stage IV and stage I ccRCC specimens, we initially analyzed The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GSE73731). We found that maternal and embryonic leucine zipper kinase (MELK) is upregulated in stage IV ccRCC samples and that upregulation of MELK is significantly correlated with advanced disease status. Furthermore, both loss and gain-of-function assays strengthen the evidence that MELK enforces the malignant phenotype of ccRCC cells through over-activating the mammalian target of rapamycin complex 1 (mTORC1) pathway. Mechanistically, we verified that the oncogenic effect of MELK occurs through phosphorylating PRAS40, an inhibitory subunit of mTORC1, and through disrupting the interaction between PRAS40 and raptor. In summary, these results elucidate the important role of MELK in the progression of ccRCC and indicate that MELK may be a novel regulator of ccRCC progression by over-activating the mTORC1.
- Published
- 2019
48. TGFβ signaling-induced miRNA participates in autophagic regulation by targeting PRAS40 in mesenchymal subtype of glioblastoma
- Author
-
Xiangyu Fan, Yanli Tan, Yanan Wang, Chao Yang, Chuan Fang, Can Xu, Luyue Chen, Junhu Zhou, Chunsheng Kang, and Yingbin Xie
- Subjects
0301 basic medicine ,Untranslated region ,Cancer Research ,PRAS40 ,TGFβ signalling ,Mice, Nude ,Mice, SCID ,Biology ,lcsh:RC254-282 ,Malignant transformation ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,microRNA ,Autophagy ,Animals ,Humans ,Brain Neoplasms ,Receptor, Transforming Growth Factor-beta Type II ,glioblastoma ,BECN1 ,Transfection ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Xenograft Model Antitumor Assays ,MicroRNAs ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Original Article ,Transforming growth factor ,Signal Transduction - Abstract
Objective: Mesenchymal subtype of glioblastoma (mesGBM) is a refractory disease condition characterized by therapeutic failure and tumor recurrence. Hyperactive transforming growth factor-β (TGF-β) signaling could be a signature event in mesGBM, which leads to dysregulation of downstream targets and contribute to malignant transformation. In this study we aimed to investigate the hyperactive TGFβ signaling-mediated pathogenesis and possible downstream targets for the development of novel therapeutic interventions for mesGBM. Methods: GBM-BioDP is an online resource for accessing and displaying interactive views of the TCGA GBM data set. Transcriptomic sequencing followed by bioinformatic analysis was performed to identify dysregulated microRNAs. Target prediction by MR-microT and dual luciferase reporter assay were utilized to confirm the predicted target of novel_miR56. CCK-8 assays was used to assesse cell viability. The miRNA manipulation was proceeded by cell transfection and lentivirus delivery. A plasmid expressing GFP-LC3 was introduced to visualize the formation of autophagosomes. Orthotopic GBM model was constructed for in vivo study. Results: TGFβ1 and TGFβ receptor type II (TβRII) were exclusively upregulated in mesGBM (P < 0.01). Dysregulated miRNAs were identified after LY2109761 (a TβRI/II inhibitor) treatment in a mesGBM-derived cell line, and novel_miR56 was selected as a promising candidate for further functional verification. Novel_miR56 was found to potentially bind to PRAS40 via seed region complementarity in the 3' untranslated region, and we also confirmed that PRAS40 is a direct target of novel_miR56 in glioma cells. In vitro, over expression of novel_miR56 in tumor cells significantly promoted proliferation and inhibited autophagy (P < 0.05). The expression levels of P62/SQSTM was significantly increased accompanied by the decrease of BECN1 and LC3B-II/I, which indicated that autophagic activity was reduced after novel_miR56 treatment. In addition, over expression of novel_miR56 also promoted tumor growth and inhibited autophagy in vivo, which is associated with worse prognosis (P < 0.05). Conclusions: In summary, we provide novel insight into TGFβ signaling-mediated pathogenesis in mesGBM and TGFβ signaling-induced novel_miR56 may be a novel target for mesGBM management.
- Published
- 2019
49. P2 × 7 Receptor Inhibits Astroglial Autophagy via Regulating FAK- and PHLPP1/2-Mediated AKT-S473 Phosphorylation Following Kainic Acid-Induced Seizures
- Author
-
Duk-Shin Lee and Ji-Eun Kim
- Subjects
Male ,FAK inhibitor 14 ,LAMP1 ,lcsh:Chemistry ,Mice ,Phosphoprotein Phosphatases ,Serine ,Phosphorylation ,p70S6K ,lcsh:QH301-705.5 ,Heat-Shock Proteins ,Spectroscopy ,Mice, Knockout ,Kainic Acid ,Chemistry ,Kinase ,TOR Serine-Threonine Kinases ,General Medicine ,Raptor ,Computer Science Applications ,Cell biology ,Signal transduction ,Signal Transduction ,PRAS40 ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Catalysis ,Rictor ,Inorganic Chemistry ,Seizures ,Autophagy ,Animals ,Bif-1 ,Physical and Theoretical Chemistry ,Protein kinase A ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,PHLPP ,Organic Chemistry ,Mice, Inbred C57BL ,Rapamycin-Insensitive Companion of mTOR Protein ,lcsh:Biology (General) ,lcsh:QD1-999 ,Astrocytes ,Focal Adhesion Protein-Tyrosine Kinases ,siRNA ,Receptors, Purinergic P2X7 ,Carrier Proteins ,Proto-Oncogene Proteins c-akt - Abstract
Recently, we have reported that blockade/deletion of P2X7 receptor (P2X7R), an ATP-gated ion channel, exacerbates heat shock protein 25 (HSP25)-mediated astroglial autophagy (clasmatodendrosis) following kainic acid (KA) injection. In P2X7R knockout (KO) mice, prolonged astroglial HSP25 induction exerts 5&prime, adenosine monophosphate-activated protein kinase/unc-51 like autophagy activating kinase 1-mediated autophagic pathway independent of mammalian target of rapamycin (mTOR) activity following KA injection. Sustained HSP25 expression also enhances AKT-serine (S) 473 phosphorylation leading to astroglial autophagy via glycogen synthase kinase-3&beta, /bax interacting factor 1 signaling pathway. However, it is unanswered how P2X7R deletion induces AKT-S473 hyperphosphorylation during autophagic process in astrocytes. In the present study, we found that AKT-S473 phosphorylation was increased by enhancing activity of focal adhesion kinase (FAK), independent of mTOR complex (mTORC) 1 and 2 activities in isolated astrocytes of P2X7R knockout (KO) mice following KA injection. In addition, HSP25 overexpression in P2X7R KO mice acted as a chaperone of AKT, which retained AKT-S473 phosphorylation by inhibiting the pleckstrin homology domain and leucine-rich repeat protein phosphatase (PHLPP) 1- and 2-binding to AKT. Therefore, our findings suggest that P2X7R may be a fine-tuner of AKT-S473 activity during astroglial autophagy by regulating FAK phosphorylation and HSP25-mediated inhibition of PHLPP1/2-AKT binding following KA treatment.
- Published
- 2020
- Full Text
- View/download PDF
50. Impacto da idade em proteínas de sinalização de espermatozoides
- Author
-
Carvalho, Pedro Araújo, Fardilha, Margarida Sâncio da Cruz, and Silva, Joana Vieira da
- Subjects
P70 S6 kinase ,PRAS40 ,Signal transduction ,Male (in)fertility ,Spermatozoa - Abstract
Infertility is defined as the inability to achieve a pregnancy after twelve or more months of unprotected regular intercourse. The male factor is involved in approximately 50% of the cases of conjugal infertility, being exclusively responsible in approximately 20% of the cases. The postponement of paternity suggests that age may be a cause of reproductive problems. While it is well documented that women have a decline in fecundity with age, the data available regarding the effects of age on male fertility show a wider disparity. Despite the scientific consensus that male age is an important factor, very little is known about the molecular mechanisms underlying the connections between male age and reduced fertility. The aim of this study was to evaluate the impact of aging on human spermatozoa. To that end, the basic seminal parameters and the levels of 18 signaling proteins were analyzed in 31 men who resort to Assisted Reproductive Techniques (ART). The present study revealed that male age was associated with the percentage of midpiece defects and the presence of excess residual cytoplasm (ERC) in spermatozoa. This study also showed that the level of two phosphoproteins in human spermatozoa, PRAS40 (Thr246) and P70 S6 kinase (Thr389), presented a significant negative correlation (p
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.