1
|
Maietta I, Viscusi E, Laudati S, Iannaci G, D’Antonio A, Melillo RM, Motti ML, De Falco V. Targeting the p90RSK/MDM2/p53 Pathway Is Effective in Blocking Tumors with Oncogenic Up-Regulation of the MAPK Pathway Such as Melanoma and Lung Cancer. Cells 2024; 13:1546. [PMID: 39329730 PMCID: PMC11430938 DOI: 10.3390/cells13181546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
In most human tumors, the MAPK pathway is constitutively activated. Since p90RSK is downstream of MAPK, it is often hyperactive and capable of phosphorylating oncogenic substrates. We have previously shown that p90RSK phosphorylates MDM2 at S166, promoting p53 degradation in follicular thyroid carcinomas. Thus, the inhibition of p90RSK restores p53 expression, which in turn inhibits cell proliferation and promotes apoptosis. In the present study, we demonstrated that the p90RSK/MDM2/p53 pathway proved to be an excellent target in the therapy of tumors with MAPK hyperactivation. For this purpose, we selected p53wt melanoma, lung and medullary thyroid carcinoma cell lines with high activation of p90RSK. In these cell lines, we demonstrated that the p90RSK/MDM2/p53 pathway is implicated in the regulation of the cell cycle and apoptosis through p53-dependent transcriptional control of p21 and Bcl-2. Furthermore, with an immunohistochemical evaluation of primary melanomas and lung tumors, which exhibit highly activated p90RSK compared to corresponding normal tissue, we demonstrated that MDM2 stabilization was associated with p90RSK phosphorylation. The results indicate that p90RSK is able to control the proliferative rate and induction of apoptosis through the regulation of p53wt levels by stabilizing MDM2 in selected tumors with constitutively activated MAPKs, making p90RSK a new attractive target for anticancer therapy.
Collapse
Affiliation(s)
- Immacolata Maietta
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy; (I.M.); (R.M.M.)
| | - Eleonora Viscusi
- U.O.C. Anatomia Patologica, P.O. Pellegrini ASL NA1 Centro, 80134 Naples, Italy; (E.V.); (G.I.)
| | - Stefano Laudati
- U.O.C. Anatomia Patologica, Ospedale del Mare ASL NA1 Centro, 80147 Naples, Italy; (S.L.); (A.D.)
| | - Giuseppe Iannaci
- U.O.C. Anatomia Patologica, P.O. Pellegrini ASL NA1 Centro, 80134 Naples, Italy; (E.V.); (G.I.)
| | - Antonio D’Antonio
- U.O.C. Anatomia Patologica, Ospedale del Mare ASL NA1 Centro, 80147 Naples, Italy; (S.L.); (A.D.)
| | - Rosa Marina Melillo
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy; (I.M.); (R.M.M.)
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Letizia Motti
- Department of Medical, Movement and Wellbeing Sciences, University of Naples Parthenope, 80133 Naples, Italy
| | - Valentina De Falco
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy; (I.M.); (R.M.M.)
| |
Collapse
|
2
|
Leachman J, Creeden J, Turner M, Ahmed N, Dalmasso C, Loria AS. Sex-specific sequels of early life stress on serine/threonine kinase activity in visceral adipose tissue from obese mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587852. [PMID: 38617246 PMCID: PMC11014506 DOI: 10.1101/2024.04.03.587852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Adverse childhood experiences (ACEs) are an established independent risk factor for chronic disease including obesity and hypertension; however, only women exposed to multiple ACEs show a positive relationship with BMI. Our lab has reported that maternal separation and early weaning (MSEW), a mouse model of early life stress, induces sex-specific mechanisms underlying greater blood pressure response to a chronic high fat diet (HF). Specifically, female MSEW mice fed a HF display exacerbated perigonadal white adipose tissue (pgWAT) expansion and a metabolic syndrome-like phenotype compared to control counterparts, whereas hypertension is caused by sympathoactivation in male MSEW mice. Thus, this study aimed to determine whether there is a sex-specific serine/threonine kinase (STKA) activity in pgWAT adipose tissue associated with early life stress. Frozen pgWAT was collected from MSEW and control, male and female mice fed a HF to assess STKA activity using the Pamstation12 instrument. Overall, MSEW induces significant reduction of 7 phosphokinases (|Z| >=1.5) in females (QIK, MLK, PKCH, MST, STE7, PEK, FRAY) and 5 in males (AKT, SGK, P38, MARK, CDK), while 15 were downregulated in both sexes (DMPK, PKA, PKG, RSK, PLK, DYRK, NMO, CAMK1, JNK, PAKA, RAD53, ERK, PAKB, PKD, PIM, AMPK). This data provides new insights into the sex-specific dysregulation of the molecular network controlling cellular phosphorylation signals in visceral adipose tissue and identifies possible target phosphokinases implicated in adipocyte hypertrophy as a result of exposure to early life stress. Identifying functional metabolic signatures is critical to elucidate the underlying molecular mechanisms behind the sex-specific obesity risk associated with early life stress.
Collapse
Affiliation(s)
- Jacqueline Leachman
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY 405362
| | - Justin Creeden
- The Department of Neurosciences at the University of Toledo Medical Center
| | - Meghan Turner
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY 405362
| | - Nermin Ahmed
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY 405362
| | - Carolina Dalmasso
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY 405362
| | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY 405362
| |
Collapse
|
3
|
Lizcano-Perret B, Vertommen D, Herinckx G, Calabrese V, Gatto L, Roux PP, Michiels T. Identification of RSK substrates using an analog-sensitive kinase approach. J Biol Chem 2024; 300:105739. [PMID: 38342435 PMCID: PMC10945272 DOI: 10.1016/j.jbc.2024.105739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/28/2024] [Accepted: 02/02/2024] [Indexed: 02/13/2024] Open
Abstract
The p90 ribosomal S6 kinases (RSK) family of serine/threonine kinases comprises four isoforms (RSK1-4) that lie downstream of the ERK1/2 mitogen-activated protein kinase pathway. RSKs are implicated in fine tuning of cellular processes such as translation, transcription, proliferation, and motility. Previous work showed that pathogens such as Cardioviruses could hijack any of the four RSK isoforms to inhibit PKR activation or to disrupt cellular nucleocytoplasmic trafficking. In contrast, some reports suggest nonredundant functions for distinct RSK isoforms, whereas Coffin-Lowry syndrome has only been associated with mutations in the gene encoding RSK2. In this work, we used the analog-sensitive kinase strategy to ask whether the cellular substrates of distinct RSK isoforms differ. We compared the substrates of two of the most distant RSK isoforms: RSK1 and RSK4. We identified a series of potential substrates for both RSKs in cells and validated RanBP3, PDCD4, IRS2, and ZC3H11A as substrates of both RSK1 and RSK4, and SORBS2 as an RSK1 substrate. In addition, using mutagenesis and inhibitors, we confirmed analog-sensitive kinase data showing that endogenous RSKs phosphorylate TRIM33 at S1119. Our data thus identify a series of potential RSK substrates and suggest that the substrates of RSK1 and RSK4 largely overlap and that the specificity of the various RSK isoforms likely depends on their cell- or tissue-specific expression pattern.
Collapse
Affiliation(s)
- Belén Lizcano-Perret
- Molecular Virology Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Didier Vertommen
- MASSPROT Platform, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Gaëtan Herinckx
- MASSPROT Platform, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Viviane Calabrese
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada
| | - Laurent Gatto
- Computational Biology and Bioinformatics Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Faculty of Medicine, Department of Pathology and Cell Biology, Université de Montréal, Montreal, Quebec, Canada
| | - Thomas Michiels
- Molecular Virology Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
4
|
Kumar A, Schwab M, Laborit Labrada B, Silveira MAD, Goudreault M, Fournier É, Bellmann K, Beauchemin N, Gingras AC, Bilodeau S, Laplante M, Marette A. SHP-1 phosphatase acts as a coactivator of PCK1 transcription to control gluconeogenesis. J Biol Chem 2023; 299:105164. [PMID: 37595871 PMCID: PMC10504565 DOI: 10.1016/j.jbc.2023.105164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
We previously reported that the protein-tyrosine phosphatase SHP-1 (PTPN6) negatively regulates insulin signaling, but its impact on hepatic glucose metabolism and systemic glucose control remains poorly understood. Here, we use co-immunoprecipitation assays, chromatin immunoprecipitation sequencing, in silico methods, and gluconeogenesis assay, and found a new mechanism whereby SHP-1 acts as a coactivator for transcription of the phosphoenolpyruvate carboxykinase 1 (PCK1) gene to increase liver gluconeogenesis. SHP-1 is recruited to the regulatory regions of the PCK1 gene and interacts with RNA polymerase II. The recruitment of SHP-1 to chromatin is dependent on its association with the transcription factor signal transducer and activator of transcription 5 (STAT5). Loss of SHP-1 as well as STAT5 decrease RNA polymerase II recruitment to the PCK1 promoter and consequently PCK1 mRNA levels leading to blunted gluconeogenesis. This work highlights a novel nuclear role of SHP-1 as a key transcriptional regulator of hepatic gluconeogenesis adding a new mechanism to the repertoire of SHP-1 functions in metabolic control.
Collapse
Affiliation(s)
- Amit Kumar
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Michael Schwab
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Beisy Laborit Labrada
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Maruhen Amir Datsch Silveira
- Centre de Recherche du CHU de Québec - Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Marilyn Goudreault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Éric Fournier
- Centre de Recherche du CHU de Québec - Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada; Centre de recherche en données massives de l'Université Laval, Québec, Quebec, Canada
| | - Kerstin Bellmann
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Nicole Beauchemin
- Department of Oncology, Medicine and Biochemistry, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Steve Bilodeau
- Centre de Recherche du CHU de Québec - Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada; Centre de recherche en données massives de l'Université Laval, Québec, Quebec, Canada
| | - Mathieu Laplante
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada
| | - André Marette
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada; Institute of Nutrition and Functional Foods, Laval University, Québec, Quebec, Canada.
| |
Collapse
|
5
|
Xie XM, Cao QL, Sun YJ, Zhang J, Liu KL, Qin YF, Long WJ, Luo ZJ, Li XW, Liang XH, Yuan GD, Luo XP, Xuan XP. LRP6 Bidirectionally Regulates Insulin Sensitivity through Insulin Receptor and S6K Signaling in Rats with CG-IUGR. Curr Med Sci 2023; 43:274-283. [PMID: 36913109 DOI: 10.1007/s11596-022-2683-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 10/27/2022] [Indexed: 03/14/2023]
Abstract
OBJECTIVE Intrauterine growth restriction followed by postnatal catch-up growth (CG-IUGR) increases the risk of insulin resistance-related diseases. Low-density lipoprotein receptor-related protein 6 (LRP6) plays a substantial role in glucose metabolism. However, whether LRP6 is involved in the insulin resistance of CG-IUGR is unclear. This study aimed to explore the role of LRP6 in insulin signaling in response to CG-IUGR. METHODS The CG-IUGR rat model was established via a maternal gestational nutritional restriction followed by postnatal litter size reduction. The mRNA and protein expression of the components in the insulin pathway, LRP6/β-catenin and mammalian target of rapamycin (mTOR)/S6 kinase (S6K) signaling, was determined. Liver tissues were immunostained for the expression of LRP6 and β-catenin. LRP6 was overexpressed or silenced in primary hepatocytes to explore its role in insulin signaling. RESULTS Compared with the control rats, CG-IUGR rats showed higher homeostasis model assessment for insulin resistance (HOMA-IR) index and fasting insulin level, decreased insulin signaling, reduced mTOR/S6K/ insulin receptor substrate-1 (IRS-1) serine307 activity, and decreased LRP6/β-catenin in the liver tissue. The knockdown of LRP6 in hepatocytes from appropriate-for-gestational-age (AGA) rats led to reductions in insulin receptor (IR) signaling and mTOR/S6K/IRS-1 serine307 activity. In contrast, LRP6 overexpression in hepatocytes of CG-IUGR rats resulted in elevated IR signaling and mTOR/S6K/IRS-1 serine307 activity. CONCLUSION LRP6 regulated the insulin signaling in the CG-IUGR rats via two distinct pathways, IR and mTOR-S6K signaling. LRP6 may be a potential therapeutic target for insulin resistance in CG-IUGR individuals.
Collapse
Affiliation(s)
- Xue-Mei Xie
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Qiu-Li Cao
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yu-Jie Sun
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Jie Zhang
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Kai-Li Liu
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Ying-Fen Qin
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Wen-Jun Long
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zuo-Jie Luo
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiao-Wei Li
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xing-Huan Liang
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Guan-Dou Yuan
- Division of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiao-Ping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiu-Ping Xuan
- Department of Endocrinology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
6
|
Fricke AL, Mühlhäuser WWD, Reimann L, Zimmermann JP, Reichenbach C, Knapp B, Peikert CD, Heberle AM, Faessler E, Schäuble S, Hahn U, Thedieck K, Radziwill G, Warscheid B. Phosphoproteomics Profiling Defines a Target Landscape of the Basophilic Protein Kinases AKT, S6K, and RSK in Skeletal Myotubes. J Proteome Res 2023; 22:768-789. [PMID: 36763541 DOI: 10.1021/acs.jproteome.2c00505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Phosphorylation-dependent signal transduction plays an important role in regulating the functions and fate of skeletal muscle cells. Central players in the phospho-signaling network are the protein kinases AKT, S6K, and RSK as part of the PI3K-AKT-mTOR-S6K and RAF-MEK-ERK-RSK pathways. However, despite their functional importance, knowledge about their specific targets is incomplete because these kinases share the same basophilic substrate motif RxRxxp[ST]. To address this, we performed a multifaceted quantitative phosphoproteomics study of skeletal myotubes following kinase inhibition. Our data corroborate a cross talk between AKT and RAF, a negative feedback loop of RSK on ERK, and a putative connection between RSK and PI3K signaling. Altogether, we report a kinase target landscape containing 49 so far unknown target sites. AKT, S6K, and RSK phosphorylate numerous proteins involved in muscle development, integrity, and functions, and signaling converges on factors that are central for the skeletal muscle cytoskeleton. Whereas AKT controls insulin signaling and impinges on GTPase signaling, nuclear signaling is characteristic for RSK. Our data further support a role of RSK in glucose metabolism. Shared targets have functions in RNA maturation, stability, and translation, which suggests that these basophilic kinases establish an intricate signaling network to orchestrate and regulate processes involved in translation.
Collapse
Affiliation(s)
- Anna L Fricke
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,Biochemistry II, Theodor Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Wignand W D Mühlhäuser
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Lena Reimann
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Johannes P Zimmermann
- Biochemistry II, Theodor Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Christa Reichenbach
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Bettina Knapp
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Christian D Peikert
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Alexander M Heberle
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020 Innsbruck, Austria
| | - Erik Faessler
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Sascha Schäuble
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, 07743 Jena, Germany.,Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology─Leibniz-HKI, 07745 Jena, Germany
| | - Udo Hahn
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020 Innsbruck, Austria.,Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands.,Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
| | - Gerald Radziwill
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Bettina Warscheid
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,Biochemistry II, Theodor Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
7
|
Park JE, Han JS. A bioactive component of Portulaca Oleracea L., HM-chromanone, improves palmitate-induced insulin resistance by inhibiting mTOR/S6K1 through activation of the AMPK pathway in L6 skeletal muscle cells. Toxicol Res (Camb) 2022; 11:774-783. [PMID: 36337245 PMCID: PMC9618117 DOI: 10.1093/toxres/tfac055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 08/28/2023] Open
Abstract
Increased free fatty acid levels in the blood are common in obesity and cause insulin resistance associated with type 2 diabetes in the muscles. Previous studies have confirmed the antidiabetic and anti-obesity potential of (E)-5-hydroxy-7-methoxy-3-(2-hydroxybenzyl)-4-chromanone (HM-chromanone). However, it is unknown how HM-chromanone alleviates obesity-related insulin resistance in L6 skeletal muscle cells. Palmitate induced insulin resistance and reduced glucose uptake, whereas HM-chromanone significantly increased glucose uptake. In palmitate-treated L6 skeletal muscle cells, HM-chromanone stimulated liver kinase B1 (LKB1) and 5'-adenosine monophosphate-activated protein kinase (AMPK) phosphorylation. The AMPK inhibitor compound C, and the LKB1 inhibitor radicicol blocked the effects of HM-chromanone. Furthermore, HM-chromanone significantly inhibited mammalian target of rapamycin (mTOR) and ribosomal protein S6 kinase 1 (S6K1) activation, but there was no change in protein kinase C θ (PKC θ) expression. When pAMPK was inhibited with compound C, the effect of HM-chromanone on the inhibition of mTOR and S6K1 was significantly diminished. This indicates that HM-chromanone inhibits mTOR and S6K1 activation through pAMPK activation. Inhibition of mTOR and S6K1 by HM-chromanone significantly reduced IRS-1Ser307 and IRS-1Ser632 phosphorylation, leading to insulin resistance. This resulted in an increase in PM-GLUT4 (glucose transporter 4) expression, thereby stimulating glucose uptake in insulin-resistant muscle cells. HM-chromanone can improve palmitate-induced insulin resistance by inhibiting mTOR and S6K1 through activation of the AMPK pathway in L6 skeletal muscle cells. These results show the therapeutic potential of HM-chromanone for improving insulin resistance in type 2 diabetes.
Collapse
Affiliation(s)
- Jae Eun Park
- Department of Food Science and Nutrition, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumgeong-gu, Busan 46241, Republic of South Korea
| | - Ji Sook Han
- Department of Food Science and Nutrition, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumgeong-gu, Busan 46241, Republic of South Korea
| |
Collapse
|
8
|
Free fatty acid impairs myogenic differentiation through AMPKα-miR-206 pathway. Mol Cell Biol 2021; 42:e0032721. [PMID: 34694913 DOI: 10.1128/mcb.00327-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The activity of AMPKα is reduced in type-2 diabetes, and type-2 diabetes is associated with muscular atrophy. To date, there is little known about the mechanism by which FFA participates in muscular impairment. The purpose of the present study was to explore whether FFA damages myogenesis through AMPKα-HDAC4-miR-206 pathway. The results showed that 1mM FFA produced lipid accumulation, significantly impaired insulin signaling pathway and decreased myogenic differentiation of C2C12 myoblast cells. FFA reduced LKB1-AMPKα pathway; activation of AMPKα rescued the myogenic impairment caused by FFA (P < 0.05). AMPKα promoted myogenesis by regulating the expression of miR-206 through HDAC4 (P < 0.05); AMPKα affected cell cycle and cell proliferation to promote myogenesis by regulating miR-206 and miR-206's target gene - cyclin D1. In addition, AICAR and HDAC4 siRNA promoted myogenic differentiation compared with FFA group; however, this positive effect was significantly down-regulated after transfection of miR-206 inhibitor. In summary, AMPKα plays positive roles in myogenic differentiation and myogenesis, and FFA decreased myogenic differentiation and myotubes formation through AMPKα-HDAC4-miR-206 pathway.
Collapse
|
9
|
Alboushi L, Hackett AP, Naeli P, Bakhti M, Jafarnejad SM. Multifaceted control of mRNA translation machinery in cancer. Cell Signal 2021; 84:110037. [PMID: 33975011 DOI: 10.1016/j.cellsig.2021.110037] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/06/2021] [Indexed: 12/15/2022]
Abstract
The mRNA translation machinery is tightly regulated through several, at times overlapping, mechanisms that modulate its efficiency and accuracy. Due to their fast rate of growth and metabolism, cancer cells require an excessive amount of mRNA translation and protein synthesis. However, unfavorable conditions, such as hypoxia, amino acid starvation, and oxidative stress, which are abundant in cancer, as well as many anti-cancer treatments inhibit mRNA translation. Cancer cells adapt to the various internal and environmental stresses by employing specialised transcript-specific translation to survive and gain a proliferative advantage. We will highlight the major signaling pathways and mechanisms of translation that regulate the global or mRNA-specific translation in response to the intra- or extra-cellular signals and stresses that are key components in the process of tumourigenesis.
Collapse
Affiliation(s)
- Lilas Alboushi
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Angela P Hackett
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Parisa Naeli
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Seyed Mehdi Jafarnejad
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
10
|
Liu YC, Tsai FJ, Chen CJ. A rapid, multiplexed kinase activity assay using 8-plex iTRAQ labeling, SPE, and MALDI-TOF/TOF MS. Analyst 2020; 145:992-1000. [PMID: 31829320 DOI: 10.1039/c9an01810g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Synthesized peptide substrates have been used for in vitro phosphorylation using purified kinases or cell lysates. For screening assays, a direct readout and comparison among different experimental conditions without using an internal standard would be preferred. In this study, we developed a rapid, quantitative measurement method of multikinase activity based on MALDI-TOF/TOF MS. We combined 8-plex iTRAQ-labeled peptide substrates, solid phase extraction (SPE), and a phosphorylated peptide purification plate to rapidly determine multikinase activity in cell lysates. To enable our platform to be applicable in insulin stimulation and cancer drug inhibition, a list of peptide substrates was designed. By labeling peptide substrates with 8-plex iTRAQ reagents, protein kinase activity in 8 samples could be directly compared using the mass tags on their fragmented ion spectra. The protein amount and incubation time for multikinase activity assays were optimized, and the effect of insulin stimulation and an inhibitory drug on the cellular protein kinase activity was evaluated.
Collapse
Affiliation(s)
- Yu-Ching Liu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40402, Taiwan.
| | | | | |
Collapse
|
11
|
Genome-wide association study identifies novel risk variants from RPS6KA1, CADPS, VARS, and DHX58 for fasting plasma glucose in Arab population. Sci Rep 2020; 10:152. [PMID: 31932636 PMCID: PMC6957513 DOI: 10.1038/s41598-019-57072-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
Consanguineous populations of the Arabian Peninsula, which has seen an uncontrolled rise in type 2 diabetes incidence, are underrepresented in global studies on diabetes genetics. We performed a genome-wide association study on the quantitative trait of fasting plasma glucose (FPG) in unrelated Arab individuals from Kuwait (discovery-cohort:n = 1,353; replication-cohort:n = 1,196). Genome-wide genotyping in discovery phase was performed for 632,375 markers from Illumina HumanOmniExpress Beadchip; and top-associating markers were replicated using candidate genotyping. Genetic models based on additive and recessive transmission modes were used in statistical tests for associations in discovery phase, replication phase, and meta-analysis that combines data from both the phases. A genome-wide significant association with high FPG was found at rs1002487 (RPS6KA1) (p-discovery = 1.64E-08, p-replication = 3.71E-04, p-combined = 5.72E-11; β-discovery = 8.315; β-replication = 3.442; β-combined = 6.551). Further, three suggestive associations (p-values < 8.2E-06) with high FPG were observed at rs487321 (CADPS), rs707927 (VARS and 2Kb upstream of VWA7), and rs12600570 (DHX58); the first two markers reached genome-wide significance in the combined analysis (p-combined = 1.83E-12 and 3.07E-09, respectively). Significant interactions of diabetes traits (serum triglycerides, FPG, and glycated hemoglobin) with homeostatic model assessment of insulin resistance were identified for genotypes heterozygous or homozygous for the risk allele. Literature reports support the involvement of these gene loci in type 2 diabetes etiology.
Collapse
|
12
|
Zuo ML, Wang AP, Tian Y, Mao L, Song GL, Yang ZB. Oxymatrine ameliorates insulin resistance in rats with type 2 diabetes by regulating the expression of KSRP, PETN, and AKT in the liver. J Cell Biochem 2019; 120:16185-16194. [PMID: 31087709 DOI: 10.1002/jcb.28898] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/03/2019] [Accepted: 03/15/2019] [Indexed: 12/23/2022]
Abstract
Insulin resistance plays a key role in the development and progression of type 2 diabetes mellitus (T2DM). Recent studies found that insulin resistance was associated with the dysfunction of KH-type splicing regulatory protein (KSRP) expression and AKT pathway, and that oxymatrine possesses an antidiabetic effect. The aim of the present study was to investigate whether the protection of oxymatrine against T2DM was associated with the modulation of the KSRP expression and AKT pathway. Sprague-Dawley rats were fed a high-fat diet and injected with streptozotocin intraperitoneally to induce T2DM, which led to an increase in blood glucose levels and insulin resistance, and a decrease in insulin sensitivity and glycogen synthesis concomitant with KSRP downregulation, PTEN upregulation, and AKT phosphorylation deficiency. The administration of oxymatrine decreased blood glucose levels and insulin resistance, increased insulin sensitivity, and improved glycogen synthesis in the liver of T2DM rats, through a reversal in the expression of KSRP, PTEN, and AKT. On the basis of these observations, we concluded that oxymatrine can protect T2DM rats from insulin resistance through the regulation of the KSRP, PETN, and AKT expression in the liver.
Collapse
Affiliation(s)
- Mei-Ling Zuo
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Ying Tian
- Institute of Clinical Research, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Li Mao
- Department of Basic Medicine, Changsha Health Vocational College, Changsha, Hunan, China
| | - Gui-Lin Song
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China.,Institute of Emergency and Critical Care Medicine of Changsha, Changsha, China
| | - Zhong-Bao Yang
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China.,Institute of Emergency and Critical Care Medicine of Changsha, Changsha, China
| |
Collapse
|
13
|
Abdulghani M, Song G, Kaur H, Walley JW, Tuteja G. Comparative Analysis of the Transcriptome and Proteome during Mouse Placental Development. J Proteome Res 2019; 18:2088-2099. [PMID: 30986076 DOI: 10.1021/acs.jproteome.8b00970] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The condition of the placenta is a determinant of the short- and long-term health of the mother and the fetus. However, critical processes occurring in early placental development, such as trophoblast invasion and establishment of placental metabolism, remain poorly understood. To gain a better understanding of the genes involved in regulating these processes, we utilized a multiomics approach, incorporating transcriptome, proteome, and phosphoproteome data generated from mouse placental tissue collected at two critical developmental time points. We found that incorporating information from both the transcriptome and proteome identifies genes associated with time point-specific biological processes, unlike using the proteome alone. We further inferred genes upregulated on the basis of the proteome data but not the transcriptome data at each time point, leading us to identify 27 genes that we predict to have a role in trophoblast migration or placental metabolism. Finally, using the phosphoproteome data set, we discovered novel phosphosites that may play crucial roles in the regulation of placental transcription factors. By generating the largest proteome and phosphoproteome data sets in the developing placenta, and integrating transcriptome analysis, we uncovered novel aspects of placental gene regulation.
Collapse
Affiliation(s)
- Majd Abdulghani
- Interdepartmental Genetics and Genomics , Iowa State University , Ames , Iowa 50011-1079 , United States.,Department of Genetics, Development, and Cell Biology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Gaoyuan Song
- Department of Plant Pathology and Microbiology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Haninder Kaur
- Department of Genetics, Development, and Cell Biology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Justin W Walley
- Interdepartmental Genetics and Genomics , Iowa State University , Ames , Iowa 50011-1079 , United States.,Department of Plant Pathology and Microbiology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Geetu Tuteja
- Interdepartmental Genetics and Genomics , Iowa State University , Ames , Iowa 50011-1079 , United States.,Department of Genetics, Development, and Cell Biology , Iowa State University , Ames , Iowa 50011-1079 , United States
| |
Collapse
|
14
|
Hebbar P, Nizam R, Melhem M, Alkayal F, Elkum N, John SE, Tuomilehto J, Alsmadi O, Thanaraj TA. Genome-wide association study identifies novel recessive genetic variants for high TGs in an Arab population. J Lipid Res 2018; 59:1951-1966. [PMID: 30108155 DOI: 10.1194/jlr.p080218] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Abnormal blood lipid levels are influenced by genetic and lifestyle/dietary factors. Although many genetic variants associated with blood lipid traits have been identified in Europeans, similar data in Middle Eastern populations are limited. We performed a genome-wide association study with Arab individuals (discovery cohort: 1,353; replication cohort: 1,176) from Kuwait to identify possible associations of genetic variants with high lipid levels. We used Illumina HumanOmniExpress BeadChip and candidate SNP genotyping in the discovery and replication phases, respectively. For association tests, we used genetic models that were based on additive and recessive modes of inheritance. High triglycerides (TGs) were recessively associated with six risk variants (rs1002487/RPS6KA1, rs11805972/LAD1) rs7761746/Or5v1, rs39745/CTTNBP2-LSM8, rs2934952/PGAP3, and rs9626773/RP11-191L9.4-CERK) at genome-wide significance (P 6.12E-09), and another six variants (rs10873925/ST6GALNAC5, rs4663379/SPP2-ARL4C, rs10033119/NPY1R, rs17709449/LINC00911-FLRT2, rs11654954/CDK12-NEUROD2, and rs9972882/STARD3) were associated at borderline significance (P 5.0E-08). High TG was also additively associated with rs11654954. All of the 12 identified markers are novel and are harbored in runs of homozygosity. Literature evidence supports the involvement of these gene loci in lipid-related processes. This study in an Arab population augments international efforts to identify genetic regulation of lipid traits.
Collapse
Affiliation(s)
- Prashantha Hebbar
- Dasman Diabetes Institute, Dasman 15462, Kuwait.,Faculty of Medicine, Univerisity of Helsinki, Helsinki, Finland
| | | | | | | | - Naser Elkum
- Dasman Diabetes Institute, Dasman 15462, Kuwait
| | | | | | | | | |
Collapse
|
15
|
Houles T, Gravel SP, Lavoie G, Shin S, Savall M, Méant A, Grondin B, Gaboury L, Yoon SO, St-Pierre J, Roux PP. RSK Regulates PFK-2 Activity to Promote Metabolic Rewiring in Melanoma. Cancer Res 2018; 78:2191-2204. [DOI: 10.1158/0008-5472.can-17-2215] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/03/2017] [Accepted: 02/06/2018] [Indexed: 11/16/2022]
|
16
|
Mitochondrial AKAP1 supports mTOR pathway and tumor growth. Cell Death Dis 2017; 8:e2842. [PMID: 28569781 PMCID: PMC5520900 DOI: 10.1038/cddis.2017.241] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/06/2017] [Accepted: 04/28/2017] [Indexed: 12/23/2022]
Abstract
Mitochondria are the powerhouses of energy production and the sites where metabolic pathway and survival signals integrate and focus, promoting adaptive responses to hormone stimulation and nutrient availability. Increasing evidence suggests that mitochondrial bioenergetics, metabolism and signaling are linked to tumorigenesis. AKAP1 scaffolding protein integrates cAMP and src signaling on mitochondria, regulating organelle biogenesis, oxidative metabolism and cell survival. Here, we provide evidence that AKAP1 is a transcriptional target of Myc and supports the growth of cancer cells. We identify Sestrin2, a leucine sensor and inhibitor of the mammalian target of rapamycin (mTOR), as a novel component of the complex assembled by AKAP1 on mitochondria. Downregulation of AKAP1 impaired mTOR pathway and inhibited glioblastoma growth. Both effects were reversed by concomitant depletion of AKAP1 and sestrin2. High levels of AKAP1 were found in a wide variety of high-grade cancer tissues. In lung cancer, AKAP1 expression correlates with high levels of Myc, mTOR phosphorylation and reduced patient survival. Collectively, these data disclose a previously unrecognized role of AKAP1 in mTOR pathway regulation and cancer growth. AKAP1/mTOR signal integration on mitochondria may provide a new target for cancer therapy.
Collapse
|
17
|
Guo J, Dou L, Meng X, Chen Z, Yang W, Fang W, Yang C, Huang X, Tang W, Yang J, Li J. Hepatic MiR-291b-3p Mediated Glucose Metabolism by Directly Targeting p65 to Upregulate PTEN Expression. Sci Rep 2017; 7:39899. [PMID: 28054586 PMCID: PMC5214750 DOI: 10.1038/srep39899] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 11/29/2016] [Indexed: 12/14/2022] Open
Abstract
Several studies have suggested an important role of miR-291b-3p in the development of embryonic stem cells. In previous study, we found that the expression of miR-291b-3p was significantly upregulated in the liver of db/db mice. However, the role of miR-291b-3p in glucose metabolism and its underlying mechanisms remain unknown. In the present study, we demonstrated that miR-291b-3p was abundantly expressed in the liver. Of note, hepatic miR-291b-3p expression was upregulated in HFD-fed mice and induced by fasting in C57BL/6 J normal mice. Importantly, hepatic inhibition miR-291b-3p expression ameliorated hyperglycemia and insulin resistance in HFD-fed mice, whereas hepatic overexpression of miR-291b-3p led to hyperglycemia and insulin resistance in C57BL/6 J normal mice. Further study revealed that miR-291b-3p suppressed insulin-stimulated AKT/GSK signaling and increased the expression of gluconeogenic genes in hepatocytes. Moreover, we identified that p65, a subunit of nuclear factor-κB (NF-κB), is a target of miR-291b-3p by bioinformatics analysis and luciferase reporter assay. Silencing of p65 significantly augmented the expression of PTEN and impaired AKT activation. In conclusion, we found novel evidence suggesting that hepatic miR-291b-3p mediated glycogen synthesis and gluconeogenesis through targeting p65 to regulate PTEN expression. Our findings indicate the therapeutic potential of miR-291b-3p inhibitor in hyperglycemia and insulin resistance.
Collapse
Affiliation(s)
- Jun Guo
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| | - Lin Dou
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| | - Xiangyu Meng
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| | - Zhenzhen Chen
- Department of physiology and pathophysiology, key laboratory of molecular cardiovascular science of the ministry of education, Peking University Health Science Center, Beijing 100191, China
| | - Weili Yang
- Department of physiology and pathophysiology, key laboratory of molecular cardiovascular science of the ministry of education, Peking University Health Science Center, Beijing 100191, China
| | - Weiwei Fang
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| | - Chunxiao Yang
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| | - Xiuqing Huang
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| | - Weiqing Tang
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| | - Jichun Yang
- Department of physiology and pathophysiology, key laboratory of molecular cardiovascular science of the ministry of education, Peking University Health Science Center, Beijing 100191, China
| | - Jian Li
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, P. R. China
| |
Collapse
|
18
|
Kim HS, Kim SJ, Bae J, Wang Y, Park SY, Min YS, Je HD, Sohn UD. The p90rsk-mediated signaling of ethanol-induced cell proliferation in HepG2 cell line. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:595-603. [PMID: 27847436 PMCID: PMC5106393 DOI: 10.4196/kjpp.2016.20.6.595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/23/2016] [Accepted: 07/28/2016] [Indexed: 01/17/2023]
Abstract
Ribosomal S6 kinase is a family of serine/threonine protein kinases involved in the regulation of cell viability. There are two subfamilies of ribosomal s6 kinase, (p90rsk, p70rsk). Especially, p90rsk is known to be an important downstream kinase of p44/42 MAPK. We investigated the role of p90rsk on ethanol-induced cell proliferation of HepG2 cells. HepG2 cells were treated with 10~50 mM of ethanol with or without ERK and p90rsk inhibitors. Cell viability was measured by MTT assay. The expression of pERK1, NHE1 was measured by Western blots. The phosphorylation of p90rsk was measured by ELISA kits. The expression of Bcl-2 was measured by qRT-PCR. When the cells were treated with 10~30 mM of ethanol for 24 hour, it showed significant increase in cell viability versus control group. Besides, 10~30 mM of ethanol induced increased expression of pERK1, p-p90rsk, NHE1 and Bcl-2. Moreover treatment of p90rsk inhibitor attenuated the ethanol-induced increase in cell viability and NHE1 and Bcl-2 expression. In summary, these results suggest that p90rsk, a downstream kinase of ERK, plays a stimulatory role on ethanol-induced hepatocellular carcinoma progression by activating anti-apoptotic factor Bcl-2 and NHE1 known to regulate cell survival.
Collapse
Affiliation(s)
- Han Sang Kim
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Su-Jin Kim
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Jinhyung Bae
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Yiyi Wang
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Sun Young Park
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Young Sil Min
- Department of Medicinal Plant Science, College of Science and Engineering, Jungwon University, Chungbuk 28024, Korea
| | - Hyun Dong Je
- Department of Pharmacology, College of Pharmacy, Catholic University of Daegu, Daegu 38430, Korea
| | - Uy Dong Sohn
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
19
|
Shum M, Bellmann K, St-Pierre P, Marette A. Pharmacological inhibition of S6K1 increases glucose metabolism and Akt signalling in vitro and in diet-induced obese mice. Diabetologia 2016; 59:592-603. [PMID: 26733005 DOI: 10.1007/s00125-015-3839-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/13/2015] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS The mammalian target of rapamycin complex 1 (mTORC1)/p70 ribosomal S6 kinase (S6K)1 pathway is overactivated in obesity, leading to inhibition of phosphoinositide 3-kinase (PI3K)/Akt signalling and insulin resistance. However, chronic mTORC1 inhibition by rapamycin impairs glucose homeostasis because of robust induction of liver gluconeogenesis. Here, we compared the effect of rapamycin with that of the selective S6K1 inhibitor, PF-4708671, on glucose metabolism in vitro and in vivo. METHODS We used L6 myocytes and FAO hepatocytes to explore the effect of PF-4708671 on the regulation of glucose uptake, glucose production and insulin signalling. We also treated high-fat (HF)-fed obese mice for 7 days with PF-4708671 in comparison with rapamycin to assess glucose tolerance, insulin resistance and insulin signalling in vivo. RESULTS Chronic rapamycin treatment induced insulin resistance and impaired glucose metabolism in hepatic and muscle cells. Conversely, chronic S6K1 inhibition with PF-4708671 reduced glucose production in hepatocytes and enhanced glucose uptake in myocytes. Whereas rapamycin treatment inhibited Akt phosphorylation, PF-4708671 increased Akt phosphorylation in both cell lines. These opposite effects of the mTORC1 and S6K1 inhibitors were also observed in vivo. Indeed, while rapamycin treatment induced glucose intolerance and failed to improve Akt phosphorylation in liver and muscle of HF-fed mice, PF-4708671 treatment improved glucose tolerance and increased Akt phosphorylation in metabolic tissues of these obese mice. CONCLUSIONS/INTERPRETATION Chronic S6K1 inhibition by PF-4708671 improves glucose homeostasis in obese mice through enhanced Akt activation in liver and muscle. Our results suggest that specific S6K1 blockade is a valid pharmacological approach to improve glucose disposal in obese diabetic individuals.
Collapse
Affiliation(s)
- Michael Shum
- Department of Medicine, Quebec Heart and Lung Institute, Hôpital Laval, Pavillon Marguerite d'Youville, Room Y4308, 2705 Chemin Ste-Foy, Québec, Canada, G1V 4G5
| | - Kerstin Bellmann
- Department of Medicine, Quebec Heart and Lung Institute, Hôpital Laval, Pavillon Marguerite d'Youville, Room Y4308, 2705 Chemin Ste-Foy, Québec, Canada, G1V 4G5
| | - Philippe St-Pierre
- Department of Medicine, Quebec Heart and Lung Institute, Hôpital Laval, Pavillon Marguerite d'Youville, Room Y4308, 2705 Chemin Ste-Foy, Québec, Canada, G1V 4G5
| | - André Marette
- Department of Medicine, Quebec Heart and Lung Institute, Hôpital Laval, Pavillon Marguerite d'Youville, Room Y4308, 2705 Chemin Ste-Foy, Québec, Canada, G1V 4G5.
| |
Collapse
|
20
|
Rozengurt E, Soares HP, Sinnet-Smith J. Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol Cancer Ther 2014; 13:2477-88. [PMID: 25323681 PMCID: PMC4222988 DOI: 10.1158/1535-7163.mct-14-0330] [Citation(s) in RCA: 219] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The development of drug resistance by cancer cells is recognized as a major cause for drug failure and disease progression. The PI3K/AKT/mTOR pathway is aberrantly stimulated in many cancer cells and thus it has emerged as a target for therapy. However, mTORC1 and S6K also mediate potent negative feedback loops that attenuate signaling via insulin/insulin growth factor receptor and other tyrosine kinase receptors. Suppression of these feedback loops causes overactivation of upstream pathways, including PI3K, AKT, and ERK that potentially oppose the antiproliferative effects of mTOR inhibitors and lead to drug resistance. A corollary of this concept is that release of negative feedback loops and consequent compensatory overactivation of promitogenic pathways in response to signal inhibitors can circumvent the mitogenic block imposed by targeting only one pathway. Consequently, the elucidation of the negative feedback loops that regulate the outputs of signaling networks has emerged as an area of fundamental importance for the rational design of effective anticancer combinations of inhibitors. Here, we review pathways that undergo compensatory overactivation in response to inhibitors that suppress feedback inhibition of upstream signaling and underscore the importance of unintended pathway activation in the development of drug resistance to clinically relevant inhibitors of mTOR, AKT, PI3K, or PI3K/mTOR.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California. CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, California. Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California.
| | - Heloisa P Soares
- Division of Hematology-Oncology and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - James Sinnet-Smith
- Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California. CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
21
|
Zordoky BNM, Nagendran J, Pulinilkunnil T, Kienesberger PC, Masson G, Waller TJ, Kemp BE, Steinberg GR, Dyck JRB. AMPK-dependent inhibitory phosphorylation of ACC is not essential for maintaining myocardial fatty acid oxidation. Circ Res 2014; 115:518-24. [PMID: 25001074 DOI: 10.1161/circresaha.115.304538] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The energy sensor AMP-activated protein kinases (AMPK) is thought to play an important role in regulating myocardial fatty acid oxidation (FAO) via its phosphorylation and inactivation of acetyl coenzyme A carboxylase (ACC). However, studies supporting this have not directly assessed whether the maintenance of FAO rates and subsequent cardiac function requires AMPK-dependent inhibitory phosphorylation of ACC. OBJECTIVE To determine whether preventing AMPK-mediated inactivation of ACC influences myocardial FAO or function. METHODS AND RESULTS A double knock-in (DKI) mouse (ACC-DKI) model was generated in which the AMPK phosphorylation sites Ser79 on ACC1 and Ser221 (Ser212 mouse) on ACC2 were mutated to prevent AMPK-dependent inhibitory phosphorylation of ACC. Hearts from ACC-DKI mice displayed a complete loss of ACC phosphorylation at the AMPK phosphorylation sites. Despite the inability of AMPK to regulate ACC activity, hearts from ACC-DKI mice displayed normal basal AMPK activation and cardiac function at both standard and elevated workloads. In agreement with the inability of AMPK in hearts from ACC-DKI mice to phosphorylate and inhibit ACC, there was a significant increase in cardiac malonyl-CoA content compared with wild-type mice. However, cardiac FAO rates were comparable between wild-type and ACC-DKI mice at baseline, during elevated workloads, and after a more stressful condition of myocardial ischemia that is known to robustly activate AMPK. CONCLUSIONS Our findings show AMPK-dependent inactivation of ACC is not essential for the control of myocardial FAO and subsequent cardiac function during a variety of conditions involving AMPK-independent and AMPK-dependent metabolic adaptations.
Collapse
Affiliation(s)
- Beshay N M Zordoky
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Jeevan Nagendran
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Thomas Pulinilkunnil
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Petra C Kienesberger
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Grant Masson
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Terri J Waller
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Bruce E Kemp
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Gregory R Steinberg
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.)
| | - Jason R B Dyck
- From the Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (B.N.M.Z., J.N., T.P., P.C.K., G.M., T.J.W., J.R.B.D.); Department of Medicine, St. Vincent's Institute of Medical Research, University of Melbourne, Melbourne, Victoria, Australia (B.E.K.); and Division of Endocrinology and Metabolism, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada (G.R.S.).
| |
Collapse
|
22
|
Leontieva OV, Demidenko ZN, Blagosklonny MV. Rapamycin reverses insulin resistance (IR) in high-glucose medium without causing IR in normoglycemic medium. Cell Death Dis 2014; 5:e1214. [PMID: 24810050 PMCID: PMC4047870 DOI: 10.1038/cddis.2014.178] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/17/2014] [Accepted: 03/26/2014] [Indexed: 02/08/2023]
Abstract
Mammalian target of rapamycin (mTOR) is involved in insulin resistance (IR) and diabetic retinopathy. In retinal pigment epithelial (RPE) cells, insulin activates the mTOR pathway, inducing hypoxia-inducible factor-1α (HIF-1α) and HIF-dependent transcription in serum-free minimum essential medium Eagle (MEM). Serendipitously, we found that insulin failed to induce the HIF-1α-dependent response, when RPE cells were cultured in Dulbecco's modification of Eagle's medium (DMEM). Whereas concentration of glucose in MEM corresponds to normal glucose levels in blood (5.5 mM), its concentration in DMEM corresponds to severe diabetic hyperglycemia (25 mM). Addition of glucose to MEM also caused IR. Glucose-mediated IR was characterized by basal activation of mTORC1 and its poor inducibility by insulin. Basal levels of phosphorylated S6 kinase (S6K), S6 and insulin receptor substrate 1 (IRS1) S635/639 were high, whereas their inducibilities were decreased. Insulin-induced Akt phosphorylation was decreased and restored by rapamycin and an inhibitor of S6K. IR was associated with de-phosphorylation of IRS1 at S1011, which was reversed by rapamycin. Both short (16–40 h) and chronic (2 weeks) treatment with rapamycin reversed IR. Furthermore, rapamycin did not impair Akt activation in RPE cells cultured in normoglycemic media. In contrast, Torin 1 blocked Akt activation by insulin. We conclude that by activating mTOR/S6K glucose causes feedback IR, preventable by rapamycin. Rapamycin does not cause IR in RPE cells regardless of the duration of treatment. We confirmed that rapamycin also did not impair phosphorylation of Akt at T308 and S473 in normal myoblast C2C12 cells. Our work provides insights in glucose-induced IR and suggests therapeutic approaches to treat patients with IR and severe hyperglycemia and to prevent diabetic complications such as retinopathy. Also our results prompt to reconsider physiological relevance of numerous data and paradigms on IR given that most cell lines are cultured with grossly super-physiological levels of glucose.
Collapse
Affiliation(s)
- O V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA
| | - Z N Demidenko
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA
| | - M V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA
| |
Collapse
|