1
|
Anashkin VA, Kirillova EA, Orlov VN, Baykov AA. Specific Mutations Reverse Regulatory Effects of Adenosine Phosphates and Increase Their Binding Stoichiometry in CBS Domain-Containing Pyrophosphatase. Int J Mol Sci 2024; 25:5768. [PMID: 38891956 PMCID: PMC11172384 DOI: 10.3390/ijms25115768] [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: 04/25/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Regulatory cystathionine β-synthase (CBS) domains are widespread in proteins; however, difficulty in structure determination prevents a comprehensive understanding of the underlying regulation mechanism. Tetrameric microbial inorganic pyrophosphatase containing such domains (CBS-PPase) is allosterically inhibited by AMP and ADP and activated by ATP and cell alarmones diadenosine polyphosphates. Each CBS-PPase subunit contains a pair of CBS domains but binds cooperatively to only one molecule of the mono-adenosine derivatives. We used site-directed mutagenesis of Desulfitobacterium hafniense CBS-PPase to identify the key elements determining the direction of the effect (activation or inhibition) and the "half-of-the-sites" ligand binding stoichiometry. Seven amino acid residues were selected in the CBS1 domain, based on the available X-ray structure of the regulatory domains, and substituted by alanine and other residues. The interaction of 11 CBS-PPase variants with the regulating ligands was characterized by activity measurements and isothermal titration calorimetry. Lys100 replacement reversed the effect of ADP from inhibition to activation, whereas Lys95 and Gly118 replacements made ADP an activator at low concentrations but an inhibitor at high concentrations. Replacement of these residues for alanine increased the stoichiometry of mono-adenosine phosphate binding by twofold. These findings identified several key protein residues and suggested a "two non-interacting pairs of interacting regulatory sites" concept in CBS-PPase regulation.
Collapse
Affiliation(s)
- Viktor A. Anashkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119899, Russia (V.N.O.)
| | | | | | - Alexander A. Baykov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119899, Russia (V.N.O.)
| |
Collapse
|
2
|
Aikawa A, Kozako T, Kato N, Ohsugi T, Honda SI. Anti-tumor activity of 5-aminoimidazole-4-carboxamide riboside with AMPK-independent cell death in human adult T-cell leukemia/lymphoma. Eur J Pharmacol 2023; 961:176180. [PMID: 37956732 DOI: 10.1016/j.ejphar.2023.176180] [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: 07/23/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
Adult T-cell leukemia/lymphoma (ATL) is an aggressive T cell leukemia/lymphoma caused by human T-cell lymphotropic virus type I (HTLV-1). Acadesine or 5-aminoimidazole-4-carboxamide riboside (AICAR) is an AMP-activated protein kinase (AMPK) activator that was recently shown to have tumor suppressive effects on B cell chronic lymphocytic leukemia, but not ATL. This study evaluated the cytotoxic effects of AICAR on ATL-related cell lines and its anti-tumor activity. Here, we demonstrated that AICAR induced cell death via apoptosis and the mitochondrial membrane depolarization of ATL-related cell lines (S1T, MT-1, and MT-2) but not non-HTLV-1-infected Jurkat cells. However, AICAR did not increase the phosphorylation levels of AMPKα. In addition, AICAR increased the expression of the death receptors (DR) DR4 and DR5, and necroptosis-related proteins including phosphorylated receptor-interacting protein family members and the mixed lineage kinase domain-like protein. Interestingly, HTLV-1 Tax, an HTLV-1-encoded oncogenic factor, did not affect AICAR-induced apoptosis. Furthermore, AICAR inhibited the growth of human ATL tumor xenografts in NOD/SCID/gamma mice in vivo. Together, these results suggest that AICAR induces AMPK-independent cell death in ATL-related cell lines and has anti-tumor activity, indicating that it might be a therapeutic agent for ATL.
Collapse
Affiliation(s)
- Akiyoshi Aikawa
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.
| | - Tomohiro Kozako
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.
| | - Naho Kato
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.
| | - Takeo Ohsugi
- Department of Hematology and Immunology, Rakuno Gakuen University, Hokkaido, Japan.
| | - Shin-Ichiro Honda
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.
| |
Collapse
|
3
|
Gedeon A, Ayoub N, Brûlé S, Raynal B, Karimova G, Gelin M, Mechaly A, Haouz A, Labesse G, Munier‐Lehmann H. Insight into the role of the Bateman domain at the molecular and physiological levels through engineered IMP dehydrogenases. Protein Sci 2023; 32:e4703. [PMID: 37338125 PMCID: PMC10357500 DOI: 10.1002/pro.4703] [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: 03/03/2023] [Revised: 05/15/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Inosine 5'-monophosphate (IMP) dehydrogenase (IMPDH) is an ubiquitous enzyme that catalyzes the NAD+ -dependent oxidation of inosine 5'-monophosphate into xanthosine 5'-monophosphate. This enzyme is formed of two distinct domains, a core domain where the catalytic reaction occurs, and a less-conserved Bateman domain. Our previous studies gave rise to the classification of bacterial IMPDHs into two classes, according to their oligomeric and kinetic properties. MgATP is a common effector but cause to different effects when it binds within the Bateman domain: it is either an allosteric activator for Class I IMPDHs or a modulator of the oligomeric state for Class II IMPDHs. To get insight into the role of the Bateman domain in the dissimilar properties of the two classes, deleted variants of the Bateman domain and chimeras issued from the interchange of the Bateman domain between the three selected IMPDHs have been generated and characterized using an integrative structural biology approach. Biochemical, biophysical, structural, and physiological studies of these variants unveil the Bateman domain as being the carrier of the molecular behaviors of both classes.
Collapse
Affiliation(s)
- Antoine Gedeon
- Institut Pasteur, Université Paris Cité, Unité de Chimie et Biocatalyse, CNRS UMR3523ParisFrance
- Present address:
Institut Pasteur, Université Paris Cité, Unité de Microbiologie Structurale, CNRS UMR3525ParisFrance
| | - Nour Ayoub
- Institut Pasteur, Université Paris Cité, Unité de Chimie et Biocatalyse, CNRS UMR3523ParisFrance
- Present address:
Institut Pasteur, Université Paris Cité, Plate‐Forme de Criblage Chémogénomique et Biologique, CNRS UMR3523ParisFrance
| | - Sébastien Brûlé
- Institut Pasteur, Université Paris Cité, Plate‐Forme de Biophysique Moléculaire, C2RT, CNRS UMR3528ParisFrance
| | - Bertrand Raynal
- Institut Pasteur, Université Paris Cité, Plate‐Forme de Biophysique Moléculaire, C2RT, CNRS UMR3528ParisFrance
| | - Gouzel Karimova
- Institut Pasteur, Université Paris Cité, Unité de Biochimie des Interactions Macromoléculaires, CNRS UMR3528ParisFrance
| | - Muriel Gelin
- Centre de Biologie StructuraleUniversité Montpellier, INSERM, CNRSMontpellierFrance
| | - Ariel Mechaly
- Institut Pasteur, Université Paris Cité, Plate‐Forme de Cristallographie, C2RT, CNRS UMR3528ParisFrance
| | - Ahmed Haouz
- Institut Pasteur, Université Paris Cité, Plate‐Forme de Cristallographie, C2RT, CNRS UMR3528ParisFrance
| | - Gilles Labesse
- Centre de Biologie StructuraleUniversité Montpellier, INSERM, CNRSMontpellierFrance
| | - Hélène Munier‐Lehmann
- Institut Pasteur, Université Paris Cité, Unité de Chimie et Biocatalyse, CNRS UMR3523ParisFrance
- Present address:
Institut Pasteur, Université Paris Cité, Plate‐Forme de Criblage Chémogénomique et Biologique, CNRS UMR3523ParisFrance
| |
Collapse
|
4
|
Pyla R, Hartney TJ, Segar L. AICAR promotes endothelium-independent vasorelaxation by activating AMP-activated protein kinase via increased ZMP and decreased ATP/ADP ratio in aortic smooth muscle. J Basic Clin Physiol Pharmacol 2022; 33:759-768. [PMID: 35503763 DOI: 10.1515/jbcpp-2021-0308] [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: 10/14/2021] [Accepted: 04/05/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES AICAR, an adenosine analog, has been shown to exhibit vascular protective effects through activation of AMP-activated protein kinase (AMPK). However, it remains unclear as to whether adenosine kinase-mediated ZMP formation or adenosine receptor activation contributes to AICAR-mediated AMPK activation and/or vasorelaxant response in vascular smooth muscle. METHODS AND RESULTS In the present study using endothelium-denuded rat aortic ring preparations, isometric tension measurements revealed that exposure to 1 mM AICAR for 30 min resulted in inhibition of phenylephrine (1 μM)-induced smooth muscle contractility by ∼35%. Importantly, this vasorelaxant response by AICAR was prevented after pretreatment of aortic rings with an AMPK inhibitor (compound C, 40 µM) and adenosine kinase inhibitor (5-iodotubercidin, 1 µM), but not with an adenosine receptor blocker (8-sulfophenyltheophylline, 100 µM). Immunoblot analysis of respective aortic tissues showed that AMPK activation seen during vasorelaxant response by AICAR was abolished by compound C and 5-iodotubercidin, but not by 8-sulfophenyltheophylline, suggesting ZMP involvement in AMPK activation. Furthermore, LC-MS/MS MRM analysis revealed that exposure of aortic smooth muscle cells to 1 mM AICAR for 30 min enhanced ZMP level to 2014.9 ± 179.4 picomoles/mg protein (vs. control value of 8.5 ± 0.6; p<0.01), which was accompanied by a significant decrease in ATP/ADP ratio (1.08 ± 0.02 vs. 2.08 ± 0.06; p<0.01). CONCLUSIONS Together, the present findings demonstrate that AICAR-mediated ZMP elevation and the resultant AMPK activation in vascular smooth muscle contribute to vasorelaxation.
Collapse
Affiliation(s)
- Rajkumar Pyla
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA
| | | | - Lakshman Segar
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA.,Vascular Biology Center, Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA.,Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
5
|
Antonioli L, Pellegrini C, Fornai M, Benvenuti L, D’Antongiovanni V, Colucci R, Bertani L, Di Salvo C, Semeghini G, La Motta C, Giusti L, Zallocco L, Ronci M, Quattrini L, Angelucci F, Coviello V, Oh WK, Ha QTK, Németh ZH, Haskó G, Blandizzi C. Preclinical Development of FA5, a Novel AMP-Activated Protein Kinase (AMPK) Activator as an Innovative Drug for the Management of Bowel Inflammation. Int J Mol Sci 2021; 22:6325. [PMID: 34199160 PMCID: PMC8231528 DOI: 10.3390/ijms22126325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
Acadesine (ACA), a pharmacological activator of AMP-activated protein kinase (AMPK), showed a promising beneficial effect in a mouse model of colitis, indicating this drug as an alternative tool to manage IBDs. However, ACA displays some pharmacodynamic limitations precluding its therapeutical applications. Our study was aimed at evaluating the in vitro and in vivo effects of FA-5 (a novel direct AMPK activator synthesized in our laboratories) in an experimental model of colitis in rats. A set of experiments evaluated the ability of FA5 to activate AMPK and to compare the efficacy of FA5 with ACA in an experimental model of colitis. The effects of FA-5, ACA, or dexamethasone were tested in rats with 2,4-dinitrobenzenesulfonic acid (DNBS)-induced colitis to assess systemic and tissue inflammatory parameters. In in vitro experiments, FA5 induced phosphorylation, and thus the activation, of AMPK, contextually to the activation of SIRT-1. In vivo, FA5 counteracted the increase in spleen weight, improved the colon length, ameliorated macroscopic damage score, and reduced TNF and MDA tissue levels in DNBS-treated rats. Of note, FA-5 displayed an increased anti-inflammatory efficacy as compared with ACA. The novel AMPK activator FA-5 displays an improved anti-inflammatory efficacy representing a promising pharmacological tool against bowel inflammation.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Carolina Pellegrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Vanessa D’Antongiovanni
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy;
| | - Lorenzo Bertani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Clelia Di Salvo
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Giorgia Semeghini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Concettina La Motta
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Laura Giusti
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Lorenzo Zallocco
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Maurizio Ronci
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy;
| | - Luca Quattrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Francesco Angelucci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Vito Coviello
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Won-Keun Oh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Quy Thi Kim Ha
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Zoltan H. Németh
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
- Department of Surgery, Morristown Medical Center, Morristown, NJ 07960, USA
| | - Gyorgy Haskó
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| |
Collapse
|
6
|
Yang Y, Wu Y, Zou J, Wang YH, Xu MX, Huang W, Yu DJ, Zhang L, Zhang YY, Sun XD. Naringenin Attenuates Non-Alcoholic Fatty Liver Disease by Enhancing Energy Expenditure and Regulating Autophagy via AMPK. Front Pharmacol 2021; 12:687095. [PMID: 34163366 PMCID: PMC8215389 DOI: 10.3389/fphar.2021.687095] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background: The prevalence of non-alcoholic fatty liver disease (NAFLD) keeps growing recently. Purpose: To investigate the effects and mechanisms of naringenin (NAR) on NAFLD. Methods: High-fat diet (HFD)-induced NAFLD rats were orally administered with NAR at 10, 30, and 90 mg/kg for 2 weeks. The serum level of triglyceride (TG), total cholesterol (TC), glutamic-oxaloacetic transaminase (AST), and glutamic-pyruvic transaminase (ALT) was measured. The hepatic histology was detected by H&E and oil red O staining. L02 and Huh-7 cells were induced by sodium oleate to establish a NAFLD cell model. The effects of NAR on lipid accumulation were detected by oil red O staining. The glucose uptake and ATP content of 3T3-L1 adipocytes and C2C12 myotubes were measured. The expression of proteins of the AMPK signaling pathway in 3T3-L1 adipocytes and C2C12 myotubes was assessed by Western blotting. The mitochondrial biogenesis of 3T3-L1 adipocytes and C2C12 myotubes was measured by mitotracker orange staining and Western blotting. The biomarkers of autophagy were detected by Western blotting and immunofluorescence. The binding of NAR to AMPKγ1 was analyzed by molecular docking. Chloroquine and compound C were employed to block autophagic flux and AMPK, respectively. Results: NAR alleviated HFD-induced NAFLD in rats at 10, 30, and 90 mg/kg. NAR attenuated lipid accumulation in L02 and Huh-7 cells at 0.7, 2.2, 6.7, and 20 μM. NAR increased glucose uptake, decreased the ATP content, activated the CaMKKβ/AMPK/ACC pathway, and enhanced the mitochondrial biogenesis in 3T3-L1 adipocytes and C2C12 myotubes. NAR increased autophagy and promoted the initiation of autophagic flux in 3T3-L1 preadipocytes and C2C12 myoblasts, while it inhibited autophagy in NAFLD rats, 3T3-L1 adipocytes, and C2C12 myotubes. Molecular docking showed that NAR binds to AMPKγ1. Compound C blocked effects of NAR on lipid accumulation and autophagy in L02 cells. Conclusion: NAR alleviates NAFLD by increasing energy expenditure and regulating autophagy via activating AMPK directly and indirectly. The direct binding of NAR and AMPKγ1 needs further validation.
Collapse
Affiliation(s)
- Ying Yang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yue Wu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jie Zou
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yu-Hao Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Meng-Xia Xu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Wei Huang
- Department of Endocrinology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Dao-Jiang Yu
- Department of Plastic Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Li Zhang
- Analytical and Testing Center, Sichuan University, Chengdu, China
| | - Yuan-Yuan Zhang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.,Department of Endocrinology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiao-Dong Sun
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.,Department of Plastic Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| |
Collapse
|
7
|
Alghamdi F, Alshuweishi Y, Salt IP. Regulation of nutrient uptake by AMP-activated protein kinase. Cell Signal 2020; 76:109807. [DOI: 10.1016/j.cellsig.2020.109807] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
|
8
|
Chatterjee A, Singh S, Rai R, Rai S, Rai L. Functional Characterization of Alr0765, A Hypothetical Protein from Anabaena PCC 7120 Involved in Cellular Energy Status Sensing, Iron Acquisition and Abiotic Stress Management in E. coli Using Molecular, Biochemical and Computational Approaches. Curr Genomics 2020; 21:295-310. [PMID: 33071622 PMCID: PMC7521041 DOI: 10.2174/1389202921999200424181239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Cyanobacteria are excellent model to understand the basic metabolic processes taking place in response to abiotic stress. The present study involves the characterization of a hypothetical protein Alr0765 of Anabaena PCC7120 comprising the CBS-CP12 domain and deciphering its role in abiotic stress tolerance. METHODS Molecular cloning, heterologous expression and protein purification using affinity chromatography were performed to obtain native purified protein Alr0765. The energy sensing property of Alr0765 was inferred from its binding affinity with different ligand molecules as analyzed by FTIR and TNP-ATP binding assay. AAS and real time-PCR were applied to evaluate the iron acquisition property and cyclic voltammetry was employed to check the redox sensitivity of the target protein. Transcript levels under different abiotic stresses, as well as spot assay, CFU count, ROS level and cellular H2O2 level, were used to show the potential role of Alr0765 in abiotic stress tolerance. In-silico analysis of Alr0765 included molecular function probability analysis, multiple sequence analysis, protein domain and motif finding, secondary structure analysis, protein-ligand interaction, homologous modeling, model refinement and verification and molecular docking was performed with COFACTOR, PROMALS-3D, InterProScan, MEME, TheaDomEx, COACH, Swiss modeller, Modrefiner, PROCHECK, ERRAT, MolProbity, ProSA, TM-align, and Discovery studio, respectively. RESULTS Transcript levels of alr0765 significantly increased by 20, 13, 15, 14.8, 12, 7, 6 and 2.5 fold when Anabaena PCC7120 treated with LC50 dose of heat, arsenic, cadmium, butachlor, salt, mannitol (drought), UV-B, and methyl viologen respectively, with respect to control (untreated). Heterologous expression resulted in 23KDa protein observed on the SDS-PAGE. Immunoblotting and MALDI-TOF-MS/MS, followed by MASCOT search analysis, confirmed the identity of the protein and ESI/MS revealed that the purified protein was a dimer. Binding possibility of Alr0765 with ATP was observed with an almost 6-fold increment in relative fluorescence during TNP-ATP binding assay with a λ max of 538 nm. FTIR spectra revealed modification in protein confirmation upon binding of Alr0765 with ATP, ADP, AMP and NADH. A 10-fold higher accumulation of iron was observed in digests of E. coli with recombinant vector after induction as compared to control, which affirms the iron acquisition property of the protein. Moreover, the generation of the redox potential of 146 mV by Alr0765 suggested its probable role in maintaining the redox status of the cell under environmental constraints. As per CFU count recombinant, E. coli BL21 cells showed about 14.7, 7.3, 6.9, 1.9, 3 and 4.9 fold higher number of colonies under heat, cadmium (CdCl2), arsenic (Na3AsO4), salt (NaCl), UV-B and drought (mannitol) respectively compared to pET21a harboring E. coli BL21 cells. Deterioration in the cellular ROS level and total cellular H2O2 concentration validated the stress tolerance ability of Alr0765. In-silico analysis unraveled novel findings and attested experimental findings in determining the role of Alr0765. CONCLUSION Alr0765 is a novel CBS-CP12 domain protein that maintains cellular energy level and iron homeostasis which provides tolerance against multiple abiotic stresses.
Collapse
Affiliation(s)
- Antra Chatterjee
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Shilpi Singh
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Ruchi Rai
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Shweta Rai
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - L.C. Rai
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| |
Collapse
|
9
|
Novel antioxidant astaxanthin-s-allyl cysteine biconjugate diminished oxidative stress and mitochondrial dysfunction to triumph diabetes in rat model. Life Sci 2020; 245:117367. [PMID: 32001265 DOI: 10.1016/j.lfs.2020.117367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/14/2020] [Accepted: 01/26/2020] [Indexed: 01/14/2023]
Abstract
AIMS The present study determines the effect of administration of novel antioxidant astaxanthin-s-allyl cysteine biconjugate (AST-SAC) against streptozotocin-induced diabetes mellitus (DM) in rats. MAIN METHODS AST-SAC (1 mg/kg/day) was treated against DM in rats for 45 days. The oxidative stress, antioxidants level, insulin secretion, activities of various carbohydrate metabolizing enzymes were studied. The glucose uptake in L6 myotubes was studied. In addition, in silico analysis of interaction of AST-SAC with proteins such as insulin receptor (IR) and 5'-adenosine monophosphate-activated protein kinase (AMPK) were carried out. KEY FINDINGS Administration of AST-SAC in DM rats has protected the mitochondrial function (decreased oxidative stress and normalized oxidative phosphorylation activities) and antioxidant capacity of the pancreas which has resulted in beta cells rejuvenation and insulin secretion restoration. AST-SAC decreased the alpha-glucosidases activities to bring glycemic control in DM rats. Due to these effects the glycoprotein components and lipids were restored to near normalcy in DM rats. AST-SAC protected the antioxidant status of liver, kidney and plasma; and curbed the progression of secondary complications of DM. AST-SAC treatment stimulated glucose uptake in L6 myotubes in in vitro. To support this observation, AST-SAC interacted with proteins such as IR and AMPK in silico. SIGNIFICANCE AST-SAC can be considered as "multi-target-directed ligand", that is, through these manifold effects, AST-SAC has been able to prevail over DM in rats.
Collapse
|
10
|
Shen Z, Chen Q, Jin T, Wang M, Ying H, Lu J, Wang M, Zhang W, Qiu F, Jin C, Zhao Y, Fu G. Theaflavin 3,3'-digallate reverses the downregulation of connexin 43 and autophagy induced by high glucose via AMPK activation in cardiomyocytes. J Cell Physiol 2019; 234:17999-18016. [PMID: 30847932 DOI: 10.1002/jcp.28432] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/03/2019] [Accepted: 02/14/2019] [Indexed: 12/16/2022]
Abstract
Theaflavin 3,3'-digallate (TF3), is reported to protect cardiomyocytes from lipotoxicity and reperfusion injury. However, the role of TF3 in the protection of high-glucose injury is still poorly understood. This study investigated the protective effects of TF3 on gap junctions and autophagy in neonatal cardiomyocytes (NRCMs). NRCMs preincubated with high glucose were coincubated with TF3. The expression of connexins and autophagy-related proteins was determined. The functioning of gap-junctional intercellular communication (GJIC) was measured by a dye transfer assay. Adenosine monophosphate-activated protein kinase (AMPK) activity was determined by western blot. Moreover, AMPK was activated with aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) or inhibited by AMPKα small interfering RNA (siRNA) to explore the role of AMPK in the modulation of connexin 43 (Cx43) and autophagy. Meanwhile, autophagy was activated or blocked to observe the change in Cx43 expression. It was found that the protein expression of Cx43 and autophagy-related proteins was increased in a TF3 dose- and time-dependent manner under high glucose. TF3 also recovered the reduced GJIC function induced by high glucose concentrations. TF3 activated phosphorylated AMPK in a time-dependent way. AMPKα siRNA abrogated the protection of TF3, while AICAR showed similar results compared to the TF3 treatment. Meanwhile, autophagy activation caused decreased Cx43, while cotreatment with baf A1 enhanced Cx43 expression further compared with the TF3 treatment alone under high glucose. We concluded that TF3 partly reversed the inhibition of Cx43 expression and autophagy induced by high glucose in NRCMs, partly by restoring AMPK activity. Inhibition of autophagy might be protective by preserving Cx43 expression in NRCMs stimulated by high glucose.
Collapse
Affiliation(s)
- Zhida Shen
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qi Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tingting Jin
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meihui Wang
- Department of Cardiology Basic Research, Biomedical Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hangying Ying
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiangting Lu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ming Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fuyu Qiu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chongying Jin
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanbo Zhao
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Shen S, Liao Q, Feng Y, Liu J, Pan R, Lee SMY, Lin L. Myricanol mitigates lipid accumulation in 3T3-L1 adipocytes and high fat diet-fed zebrafish via activating AMP-activated protein kinase. Food Chem 2019; 270:305-314. [DOI: 10.1016/j.foodchem.2018.07.117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
|
12
|
Douillet DC, Pinson B, Ceschin J, Hürlimann HC, Saint-Marc C, Laporte D, Claverol S, Konrad M, Bonneu M, Daignan-Fornier B. Metabolomics and proteomics identify the toxic form and the associated cellular binding targets of the anti-proliferative drug AICAR. J Biol Chem 2018; 294:805-815. [PMID: 30478173 DOI: 10.1074/jbc.ra118.004964] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
5-Aminoimidazole-4-carboxamide 1-β-d-ribofuranoside (AICAR, or acadesine) is a precursor of the monophosphate derivative 5-amino-4-imidazole carboxamide ribonucleoside 5'-phosphate (ZMP), an intermediate in de novo purine biosynthesis. AICAR proved to have promising anti-proliferative properties, although the molecular basis of its toxicity is poorly understood. To exert cytotoxicity, AICAR needs to be metabolized, but the AICAR-derived toxic metabolite was not identified. Here, we show that ZMP is the major toxic derivative of AICAR in yeast and establish that its metabolization to succinyl-ZMP, ZDP, or ZTP (di- and triphosphate derivatives of AICAR) strongly reduced its toxicity. Affinity chromatography identified 74 ZMP-binding proteins, including 41 that were found neither as AMP nor as AICAR or succinyl-ZMP binders. Overexpression of karyopherin-β Kap123, one of the ZMP-specific binders, partially rescued AICAR toxicity. Quantitative proteomic analyses revealed 57 proteins significantly less abundant on nuclei-enriched fractions from AICAR-fed cells, this effect being compensated by overexpression of KAP123 for 15 of them. These results reveal nuclear protein trafficking as a function affected by AICAR.
Collapse
Affiliation(s)
- Delphine C Douillet
- From the Université de Bordeaux, IBGC UMR 5095, F-33077 Bordeaux, France.,the Centre National de la Recherche Scientifique, IBGC UMR 5095, F-33077 Bordeaux, France
| | - Benoît Pinson
- From the Université de Bordeaux, IBGC UMR 5095, F-33077 Bordeaux, France.,the Centre National de la Recherche Scientifique, IBGC UMR 5095, F-33077 Bordeaux, France
| | - Johanna Ceschin
- From the Université de Bordeaux, IBGC UMR 5095, F-33077 Bordeaux, France.,the Centre National de la Recherche Scientifique, IBGC UMR 5095, F-33077 Bordeaux, France
| | - Hans C Hürlimann
- From the Université de Bordeaux, IBGC UMR 5095, F-33077 Bordeaux, France.,the Centre National de la Recherche Scientifique, IBGC UMR 5095, F-33077 Bordeaux, France
| | - Christelle Saint-Marc
- From the Université de Bordeaux, IBGC UMR 5095, F-33077 Bordeaux, France.,the Centre National de la Recherche Scientifique, IBGC UMR 5095, F-33077 Bordeaux, France
| | - Damien Laporte
- From the Université de Bordeaux, IBGC UMR 5095, F-33077 Bordeaux, France.,the Centre National de la Recherche Scientifique, IBGC UMR 5095, F-33077 Bordeaux, France
| | - Stéphane Claverol
- the University of Bordeaux, Bordeaux INP, Plateforme Proteome, F-33076 Bordeaux, France, and
| | - Manfred Konrad
- the Max-Planck-Institute for Biophysical Chemistry, D-37077 Goettingen, Germany
| | - Marc Bonneu
- the University of Bordeaux, Bordeaux INP, Plateforme Proteome, F-33076 Bordeaux, France, and
| | - Bertrand Daignan-Fornier
- From the Université de Bordeaux, IBGC UMR 5095, F-33077 Bordeaux, France, .,the Centre National de la Recherche Scientifique, IBGC UMR 5095, F-33077 Bordeaux, France
| |
Collapse
|
13
|
The conserved mosaic prophage protein paratox inhibits the natural competence regulator ComR in Streptococcus. Sci Rep 2018; 8:16535. [PMID: 30409983 PMCID: PMC6224593 DOI: 10.1038/s41598-018-34816-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 10/25/2018] [Indexed: 01/05/2023] Open
Abstract
Horizontal gene transfer is an important means of bacterial evolution. This includes natural genetic transformation, where bacterial cells become “competent” and DNA is acquired from the extracellular environment. Natural competence in many species of Streptococcus, is regulated by quorum sensing via the ComRS receptor-signal pair. The ComR-XIP (mature ComS peptide) complex induces expression of the alternative sigma factor SigX, which targets RNA polymerase to CIN-box promoters to activate genes involved in DNA uptake and recombination. In addition, the widely distributed Streptococcus prophage gene paratox (prx) also contains a CIN-box, and here we demonstrate it to be transcriptionally activated by XIP. In vitro experiments demonstrate that Prx binds ComR directly and prevents the ComR-XIP complex from interacting with DNA. Mutations of prx in vivo caused increased expression of the late competence gene ssb when induced with XIP as compared to wild-type, and Prx orthologues are able to inhibit ComR activation by XIP in a reporter strain which lacks an endogenous prx. Additionally, an X-ray crystal structure of Prx reveals a unique fold that implies a novel molecular mechanism to inhibit ComR. Overall, our results suggest Prx functions to inhibit the acquisition of new DNA by Streptococcus.
Collapse
|
14
|
Dolinar K, Jan V, Pavlin M, Chibalin AV, Pirkmajer S. Nucleosides block AICAR-stimulated activation of AMPK in skeletal muscle and cancer cells. Am J Physiol Cell Physiol 2018; 315:C803-C817. [PMID: 30230919 DOI: 10.1152/ajpcell.00311.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
AMP-activated kinase (AMPK) is a major regulator of energy metabolism and a promising target for development of new treatments for type 2 diabetes and cancer. 5-Aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR), an adenosine analog, is a standard positive control for AMPK activation in cell-based assays. Some broadly used cell culture media, such as minimal essential medium α (MEMα), contain high concentrations of adenosine and other nucleosides. We determined whether such media alter AICAR action in skeletal muscle and cancer cells. In nucleoside-free media, AICAR stimulated AMPK activation, increased glucose uptake, and suppressed cell proliferation. Conversely, these effects were blunted or completely blocked in MEMα that contains nucleosides. Addition of adenosine or 2'-deoxyadenosine to nucleoside-free media also suppressed AICAR action. MEMα with nucleosides blocked AICAR-stimulated AMPK activation even in the presence of methotrexate, which normally markedly enhances AICAR action by reducing its intracellular clearance. Other common media components, such as vitamin B-12, vitamin C, and α-lipoic acid, had a minor modulatory effect on AICAR action. Our findings show that nucleoside-containing media, commonly used in AMPK research, block action of the most widely used pharmacological AMPK activator AICAR. Results of cell-based assays in which AICAR is used for AMPK activation therefore critically depend on media formulation. Furthermore, our findings highlight a role for extracellular nucleosides and nucleoside transporters in regulation of AMPK activation.
Collapse
Affiliation(s)
- Klemen Dolinar
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana , Ljubljana , Slovenia.,Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana , Ljubljana , Slovenia
| | - Vid Jan
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana , Ljubljana , Slovenia
| | - Mojca Pavlin
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana , Ljubljana , Slovenia.,Institute of Biophysics, Faculty of Medicine, University of Ljubljana , Ljubljana , Slovenia
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet , Stockholm , Sweden.,National Research Tomsk State University , Tomsk , Russia
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana , Ljubljana , Slovenia
| |
Collapse
|
15
|
Philippe C, Pinson B, Dompierre J, Pantesco V, Viollet B, Daignan-Fornier B, Moenner M. AICAR Antiproliferative Properties Involve the AMPK-Independent Activation of the Tumor Suppressors LATS 1 and 2. Neoplasia 2018; 20:555-562. [PMID: 29730476 PMCID: PMC5994775 DOI: 10.1016/j.neo.2018.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/14/2018] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
AICAR (Acadesine) is a pharmacological precursor of purine nucleotide biosynthesis with anti-tumoral properties. Although recognized as an AMP mimetic activator of the protein kinase AMPK, the AICAR monophosphate derivative ZMP was also shown to mediate AMPK-independent effects. In order to unveil these AMPK-independent functions, we performed a transcriptomic analysis in AMPKα1/α2 double knockout murine embryonic cells. Kinetic analysis of the cellular response to AICAR revealed the up-regulation of the large tumor suppressor kinases (Lats) 1 and 2 transcripts, followed by the repression of numerous genes downstream of the transcriptional regulators Yap1 and Taz. This transcriptional signature, together with the observation of increased levels in phosphorylation of Lats1 and Yap1 proteins, suggested that the Hippo signaling pathway was activated by AICAR. This effect was observed in both fibroblasts and epithelial cells. Knockdown of Lats1/2 prevented the cytoplasmic delocalization of Yap1/Taz proteins in response to AICAR and conferred a higher resistance to the drug. These results indicate that activation of the most downstream steps of the Hippo cascade participates to the antiproliferative effects of AICAR.
Collapse
Affiliation(s)
- Chloé Philippe
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France
| | - Benoît Pinson
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France
| | - Jim Dompierre
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France
| | | | - Benoît Viollet
- INSERM U1016, Institut Cochin, Paris, France; CNRS (UMR 8104), Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertrand Daignan-Fornier
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France.
| | - Michel Moenner
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France.
| |
Collapse
|
16
|
Shen Y, Sherman JW, Chen X, Wang R. Phosphorylation of CDC25C by AMP-activated protein kinase mediates a metabolic checkpoint during cell-cycle G 2/M-phase transition. J Biol Chem 2018; 293:5185-5199. [PMID: 29467227 PMCID: PMC5892595 DOI: 10.1074/jbc.ra117.001379] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/01/2018] [Indexed: 12/30/2022] Open
Abstract
From unicellular to multicellular organisms, cell-cycle progression is tightly coupled to biosynthetic and bioenergetic demands. Accumulating evidence has demonstrated the G1/S-phase transition as a key checkpoint where cells respond to their metabolic status and commit to replicating the genome. However, the mechanism underlying the coordination of metabolism and the G2/M-phase transition in mammalian cells remains unclear. Here, we show that the activation of AMP-activated protein kinase (AMPK), a highly conserved cellular energy sensor, significantly delays mitosis entry. The cell-cycle G2/M-phase transition is controlled by mitotic cyclin-dependent kinase complex (CDC2-cyclin B), which is inactivated by WEE1 family protein kinases and activated by the opposing phosphatase CDC25C. AMPK directly phosphorylates CDC25C on serine 216, a well-conserved inhibitory phosphorylation event, which has been shown to mediate DNA damage–induced G2-phase arrest. The acute induction of CDC25C or suppression of WEE1 partially restores mitosis entry in the context of AMPK activation. These findings suggest that AMPK-dependent phosphorylation of CDC25C orchestrates a metabolic checkpoint for the cell-cycle G2/M-phase transition.
Collapse
Affiliation(s)
- Yuqing Shen
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and.,the Department of Microbiology and Immunology, Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Medical School, Southeast University, Nanjing 210009, China
| | - John William Sherman
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and
| | - Xuyong Chen
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and
| | - Ruoning Wang
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and
| |
Collapse
|
17
|
Kjøbsted R, Hingst JR, Fentz J, Foretz M, Sanz MN, Pehmøller C, Shum M, Marette A, Mounier R, Treebak JT, Wojtaszewski JFP, Viollet B, Lantier L. AMPK in skeletal muscle function and metabolism. FASEB J 2018; 32:1741-1777. [PMID: 29242278 PMCID: PMC5945561 DOI: 10.1096/fj.201700442r] [Citation(s) in RCA: 275] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle possesses a remarkable ability to adapt to various physiologic conditions. AMPK is a sensor of intracellular energy status that maintains energy stores by fine-tuning anabolic and catabolic pathways. AMPK’s role as an energy sensor is particularly critical in tissues displaying highly changeable energy turnover. Due to the drastic changes in energy demand that occur between the resting and exercising state, skeletal muscle is one such tissue. Here, we review the complex regulation of AMPK in skeletal muscle and its consequences on metabolism (e.g., substrate uptake, oxidation, and storage as well as mitochondrial function of skeletal muscle fibers). We focus on the role of AMPK in skeletal muscle during exercise and in exercise recovery. We also address adaptations to exercise training, including skeletal muscle plasticity, highlighting novel concepts and future perspectives that need to be investigated. Furthermore, we discuss the possible role of AMPK as a therapeutic target as well as different AMPK activators and their potential for future drug development.—Kjøbsted, R., Hingst, J. R., Fentz, J., Foretz, M., Sanz, M.-N., Pehmøller, C., Shum, M., Marette, A., Mounier, R., Treebak, J. T., Wojtaszewski, J. F. P., Viollet, B., Lantier, L. AMPK in skeletal muscle function and metabolism.
Collapse
Affiliation(s)
- Rasmus Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Janne R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Joachim Fentz
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Marc Foretz
- INSERM, Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria-Nieves Sanz
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland, and.,Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Christian Pehmøller
- Internal Medicine Research Unit, Pfizer Global Research and Development, Cambridge, Massachusetts, USA
| | - Michael Shum
- Axe Cardiologie, Quebec Heart and Lung Research Institute, Laval University, Québec, Canada.,Institute for Nutrition and Functional Foods, Laval University, Québec, Canada
| | - André Marette
- Axe Cardiologie, Quebec Heart and Lung Research Institute, Laval University, Québec, Canada.,Institute for Nutrition and Functional Foods, Laval University, Québec, Canada
| | - Remi Mounier
- Institute NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM Unité 1217, CNRS UMR, Villeurbanne, France
| | - Jonas T Treebak
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Benoit Viollet
- INSERM, Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA.,Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
18
|
Cao Y, Bojjireddy N, Kim M, Li T, Zhai P, Nagarajan N, Sadoshima J, Palmiter RD, Tian R. Activation of γ2-AMPK Suppresses Ribosome Biogenesis and Protects Against Myocardial Ischemia/Reperfusion Injury. Circ Res 2017; 121:1182-1191. [PMID: 28835357 DOI: 10.1161/circresaha.117.311159] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/17/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE AMPK (AMP-activated protein kinase) is a heterotrimeric protein that plays an important role in energy homeostasis and cardioprotection. Two isoforms of each subunit are expressed in the heart, but the isoform-specific function of AMPK remains unclear. OBJECTIVE We sought to determine the role of γ2-AMPK in cardiac stress response using bioengineered cell lines and mouse models containing either isoform of the γ-subunit in the heart. METHODS AND RESULTS We found that γ2 but not γ1 or γ3 subunit translocated into nucleus on AMPK activation. Nuclear accumulation of AMPK complexes containing γ2-subunit phosphorylated and inactivated RNA Pol I (polymerase I)-associated transcription factor TIF-IA at Ser-635, precluding the assembly of transcription initiation complexes for rDNA. The subsequent downregulation of pre-rRNA level led to attenuated endoplasmic reticulum (ER) stress and cell death. Deleting γ2-AMPK led to increases in pre-rRNA level, ER stress markers, and cell death during glucose deprivation, which could be rescued by inhibition of rRNA processing or ER stress. To study the function of γ2-AMPK in the heart, we generated a mouse model with cardiac-specific deletion of γ2-AMPK (cardiac knockout [cKO]). Although the total AMPK activity was unaltered in cKO hearts because of upregulation of γ1-AMPK, the lack of γ2-AMPK sensitizes the heart to myocardial ischemia/reperfusion injury. The cKO failed to suppress pre-rRNA level during ischemia/reperfusion and showed a greater infarct size. Conversely, cardiac-specific overexpression of γ2-AMPK decreased ribosome biosynthesis and ER stress during ischemia/reperfusion insult, and the infarct size was reduced. CONCLUSIONS The γ2-AMPK translocates into the nucleus to suppress pre-rRNA transcription and ribosome biosynthesis during stress, thus ameliorating ER stress and cell death. Increased γ2-AMPK activity is required to protect against ischemia/reperfusion injury. Our study reveals an isoform-specific function of γ2-AMPK in modulating ribosome biosynthesis, cell survival, and cardioprotection.
Collapse
Affiliation(s)
- Yang Cao
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Naveen Bojjireddy
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Maengjo Kim
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Tao Li
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Peiyong Zhai
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Narayani Nagarajan
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Junichi Sadoshima
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Richard D Palmiter
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Rong Tian
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.).
| |
Collapse
|
19
|
Kjøbsted R, Wojtaszewski JFP, Treebak JT. Role of AMP-Activated Protein Kinase for Regulating Post-exercise Insulin Sensitivity. ACTA ACUST UNITED AC 2017; 107:81-126. [PMID: 27812978 DOI: 10.1007/978-3-319-43589-3_5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Skeletal muscle insulin resistance precedes development of type 2 diabetes (T2D). As skeletal muscle is a major sink for glucose disposal, understanding the molecular mechanisms involved in maintaining insulin sensitivity of this tissue could potentially benefit millions of people that are diagnosed with insulin resistance. Regular physical activity in both healthy and insulin-resistant individuals is recognized as the single most effective intervention to increase whole-body insulin sensitivity and thereby positively affect glucose homeostasis. A single bout of exercise has long been known to increase glucose disposal in skeletal muscle in response to physiological insulin concentrations. While this effect is identified to be restricted to the previously exercised muscle, the molecular basis for an apparent convergence between exercise- and insulin-induced signaling pathways is incompletely known. In recent years, we and others have identified the Rab GTPase-activating protein, TBC1 domain family member 4 (TBC1D4) as a target of key protein kinases in the insulin- and exercise-activated signaling pathways. Our working hypothesis is that the AMP-activated protein kinase (AMPK) is important for the ability of exercise to insulin sensitize skeletal muscle through TBC1D4. Here, we aim to provide an overview of the current available evidence linking AMPK to post-exercise insulin sensitivity.
Collapse
Affiliation(s)
- Rasmus Kjøbsted
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200, Copenhagen, Denmark
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200, Copenhagen, Denmark.
| |
Collapse
|
20
|
Abstract
AMP-activated protein kinase is a family of heterotrimeric serine/threonine protein kinases that come in twelve different flavors. They serve an essential function in all eukaryotes of conserving cellular energy levels. AMPK complexes are regulated by changes in cellular AMP:ATP or ADP:ATP ratios and by a number of neutraceuticals and some of the widely-used diabetes medications such as metformin and thiazolinonediones. Moreover, biochemical activities of AMPK are tightly regulated by phosphorylation or dephosphorylation by upstream kinases and phosphatases respectively. Efforts are underway in many pharmaceutical companies to discover direct AMPK activators for the treatment of cardiovascular and metabolic diseases such as diabetes, non-alcoholic steatohepatitis (NASH) and diabetic nephropathy. Many advances have been made in the AMPK structural biology arena over the last few years that are beginning to provide detailed molecular insights into the overall topology of these fascinating enzymes and how binding of small molecules elicit subtle conformational changes leading to their activation and protection from dephosphorylation. In the brief review below on AMPK structure and function, we have focused on the recent crystallographic results especially on specific molecular interactions of direct synthetic AMPK activators which lead to biased activation of a sub-family of AMPK isoforms.
Collapse
Affiliation(s)
- Ravi G Kurumbail
- Pfizer Worldwide Research and Development, Pfizer Inc, Eastern Point Road, Groton, CT, 06340, USA.
| | - Matthew F Calabrese
- Pfizer Worldwide Research and Development, Pfizer Inc, Eastern Point Road, Groton, CT, 06340, USA
| |
Collapse
|
21
|
Broeckx T, Hulsmans S, Rolland F. The plant energy sensor: evolutionary conservation and divergence of SnRK1 structure, regulation, and function. JOURNAL OF EXPERIMENTAL BOTANY 2016; 67:6215-6252. [PMID: 27856705 DOI: 10.1093/jxb/erw416] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The SnRK1 (SNF1-related kinase 1) kinases are the plant cellular fuel gauges, activated in response to energy-depleting stress conditions to maintain energy homeostasis while also gatekeeping important developmental transitions for optimal growth and survival. Similar to their opisthokont counterparts (animal AMP-activated kinase, AMPK, and yeast Sucrose Non-Fermenting 1, SNF), they function as heterotrimeric complexes with a catalytic (kinase) α subunit and regulatory β and γ subunits. Although the overall configuration of the kinase complexes is well conserved, plant-specific structural modifications (including a unique hybrid βγ subunit) and associated differences in regulation reflect evolutionary divergence in response to fundamentally different lifestyles. While AMP is the key metabolic signal activating AMPK in animals, the plant kinases appear to be allosterically inhibited by sugar-phosphates. Their function is further fine-tuned by differential subunit expression, localization, and diverse post-translational modifications. The SnRK1 kinases act by direct phosphorylation of key metabolic enzymes and regulatory proteins, extensive transcriptional regulation (e.g. through bZIP transcription factors), and down-regulation of TOR (target of rapamycin) kinase signaling. Significant progress has been made in recent years. New tools and more directed approaches will help answer important fundamental questions regarding their structure, regulation, and function, as well as explore their potential as targets for selection and modification for improved plant performance in a changing environment.
Collapse
Affiliation(s)
- Tom Broeckx
- Laboratory for Molecular Plant Biology, Biology Department, University of Leuven-KU Leuven, Kasteelpark Arenberg 31, 3001 Heverlee-Leuven, Belgium
| | - Sander Hulsmans
- Laboratory for Molecular Plant Biology, Biology Department, University of Leuven-KU Leuven, Kasteelpark Arenberg 31, 3001 Heverlee-Leuven, Belgium
| | - Filip Rolland
- Laboratory for Molecular Plant Biology, Biology Department, University of Leuven-KU Leuven, Kasteelpark Arenberg 31, 3001 Heverlee-Leuven, Belgium
| |
Collapse
|
22
|
Mohanta TK, Park YH, Bae H. Novel Genomic and Evolutionary Insight of WRKY Transcription Factors in Plant Lineage. Sci Rep 2016; 6:37309. [PMID: 27853303 PMCID: PMC5112548 DOI: 10.1038/srep37309] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/27/2016] [Indexed: 11/22/2022] Open
Abstract
The evolutionarily conserved WRKY transcription factor (TF) regulates different aspects of gene expression in plants, and modulates growth, development, as well as biotic and abiotic stress responses. Therefore, understanding the details regarding WRKY TFs is very important. In this study, large-scale genomic analyses of the WRKY TF gene family from 43 plant species were conducted. The results of our study revealed that WRKY TFs could be grouped and specifically classified as those belonging to the monocot or dicot plant lineage. In this study, we identified several novel WRKY TFs. To our knowledge, this is the first report on a revised grouping system of the WRKY TF gene family in plants. The different forms of novel chimeric forms of WRKY TFs in the plant genome might play a crucial role in their evolution. Tissue-specific gene expression analyses in Glycine max and Phaseolus vulgaris showed that WRKY11-1, WRKY11-2 and WRKY11-3 were ubiquitously expressed in all tissue types, and WRKY15-2 was highly expressed in the stem, root, nodule and pod tissues in G. max and P. vulgaris.
Collapse
Affiliation(s)
- Tapan Kumar Mohanta
- Free Major of Natural Sciences, College of Basic Studies, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Yong-Hwan Park
- School of Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hanhong Bae
- School of Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| |
Collapse
|
23
|
Kim J, Yang G, Ha J. Targeting of AMP-activated protein kinase: prospects for computer-aided drug design. Expert Opin Drug Discov 2016; 12:47-59. [PMID: 27797589 DOI: 10.1080/17460441.2017.1255194] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Dysregulation of energy homeostasis has been implicated in a number of human chronic diseases including diabetes, obesity, cancer, and inflammation. Given the functional attributes as a central regulator of energy homeostasis, AMP-activated protein kinase (AMPK) is emerging as a therapeutic target for these diseases, and lines of evidence have highlighted the need for rational and robust screening systems for identifying specific AMPK modulators with a therapeutic potential for preventing and/or curing these diseases. Areas covered: Here, the authors review the recent advances in the understanding of three-dimensional structures of AMPK in relationship with the regulatory mechanisms, potentials of AMPK as a therapeutic target in human chronic diseases, and prospects of computer-based drug design for AMPK. Expert opinion: Accumulating information of AMPK structure has provided us with deep insight into the molecular basis underlying the regulatory mechanisms, and further discloses several structural domains, which can be served for a target site for computer-based drug design. Molecular docking and simulations provides useful information about the binding sites between potent drugs and AMPK as well as a rational screening format to discover isoform-specific AMPK modulators. For these reasons, the authors suggest that computer-aided virtual screening methods hold promise as a rational approach for discovering more specific AMPK modulators.
Collapse
Affiliation(s)
- Joungmok Kim
- a Department of Oral Biochemistry and Molecular Biology, School of Dentistry , Kyung Hee University , Dongdaemun-gu , Republic of Korea
| | - Goowon Yang
- b Department of Biochemistry and Molecular Biology, Graduate School , Kyung Hee University , Seoul , Republic of Korea
| | - Joohun Ha
- b Department of Biochemistry and Molecular Biology, Graduate School , Kyung Hee University , Seoul , Republic of Korea
| |
Collapse
|
24
|
Janda E, Lascala A, Martino C, Ragusa S, Nucera S, Walker R, Gratteri S, Mollace V. Molecular mechanisms of lipid- and glucose-lowering activities of bergamot flavonoids. PHARMANUTRITION 2016. [DOI: 10.1016/j.phanu.2016.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
25
|
Small-molecule activators of AMP-activated protein kinase as modulators of energy metabolism. Russ Chem Bull 2016. [DOI: 10.1007/s11172-015-1036-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
26
|
Nakabayashi M, Shibata N, Ishido-Nakai E, Kanagawa M, Iio Y, Komori H, Ueda Y, Nakagawa N, Kuramitsu S, Higuchi Y. Crystal structure of a hypothetical protein, TTHA0829 from Thermus thermophilus HB8, composed of cystathionine-β-synthase (CBS) and aspartate-kinase chorismate-mutase tyrA (ACT) domains. Extremophiles 2016; 20:275-82. [DOI: 10.1007/s00792-016-0817-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/14/2016] [Indexed: 01/01/2023]
|
27
|
Abstract
The AMP-protein kinase (AMPK) pathway is very versatile as it regulates cellular energetic homeostasis in many different tissue types. An appreciation for the importance of AMPK signalling and regulation in cardiovascular and tumor biology is increasing. Recently, a link has been established between anti-cancer therapy and susceptibility to cardiac disease. It has been shown that some anti-cancer drugs lead to an increased risk of cardiac disease, underlined by de-regulation of AMPK signalling. This review explores the AMPK signalling axis in both cardiac and tumor metabolism. We then examine off-target AMPK inhibition by cancer drugs and how this may translate into increased risk of cardiovascular disease. Finally, we discuss the implication of deregulated AMPK signalling during different stages of cardiac hypertrophy. Better understanding of the molecular pathways behind pathological processes will lead to the development of more effective therapeutics for cancer and cardiovascular diseases.
Collapse
Affiliation(s)
- Yulia Lipovka
- Department of Physiology, University of Arizona, Sarver Molecular Cardiovascular Research Program, USA
| | - John P Konhilas
- Department of Physiology, University of Arizona, Sarver Molecular Cardiovascular Research Program, USA
| |
Collapse
|
28
|
Grahame Hardie D. Regulation of AMP-activated protein kinase by natural and synthetic activators. Acta Pharm Sin B 2016; 6:1-19. [PMID: 26904394 PMCID: PMC4724661 DOI: 10.1016/j.apsb.2015.06.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/28/2015] [Indexed: 12/11/2022] Open
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that is almost universally expressed in eukaryotic cells. While it appears to have evolved in single-celled eukaryotes to regulate energy balance in a cell-autonomous manner, during the evolution of multicellular animals its role has become adapted so that it also regulates energy balance at the whole body level, by responding to hormones that act primarily on the hypothalamus. AMPK monitors energy balance at the cellular level by sensing the ratios of AMP/ATP and ADP/ATP, and recent structural analyses of the AMPK heterotrimer that have provided insight into the complex mechanisms for these effects will be discussed. Given the central importance of energy balance in diseases that are major causes of morbidity or death in humans, such as type 2 diabetes, cancer and inflammatory disorders, there has been a major drive to develop pharmacological activators of AMPK. Many such activators have been described, and the various mechanisms by which these activate AMPK will be discussed. A particularly large class of AMPK activators are natural products of plants derived from traditional herbal medicines. While the mechanism by which most of these activate AMPK has not yet been addressed, I will argue that many of them may be defensive compounds produced by plants to deter infection by pathogens or grazing by insects or herbivores, and that many of them will turn out to be inhibitors of mitochondrial function.
Collapse
Affiliation(s)
- David Grahame Hardie
- Division of Cell Signaling & Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| |
Collapse
|
29
|
Ramesh M, Vepuri SB, Oosthuizen F, Soliman ME. Adenosine Monophosphate-Activated Protein Kinase (AMPK) as a Diverse Therapeutic Target: A Computational Perspective. Appl Biochem Biotechnol 2015; 178:810-30. [DOI: 10.1007/s12010-015-1911-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 10/26/2015] [Indexed: 12/12/2022]
|
30
|
Adenosine Monophosphate-activated Protein Kinase Regulates Interleukin-1β Expression and Glial Glutamate Transporter Function in Rodents with Neuropathic Pain. Anesthesiology 2015; 122:1401-13. [PMID: 25710409 DOI: 10.1097/aln.0000000000000619] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Neuroinflammation and dysfunctional glial glutamate transporters (GTs) in the spinal dorsal horn are implicated in the genesis of neuropathic pain. The authors determined whether adenosine monophosphate-activated protein kinase (AMPK) in the spinal dorsal horn regulates these processes in rodents with neuropathic pain. METHODS Hind paw withdrawal responses to radiant heat and mechanical stimuli were used to assess nociceptive behaviors. Spinal markers related to neuroinflammation and glial GTs were determined by Western blotting. AMPK activities were manipulated pharmacologically and genetically. Regulation of glial GTs was determined by measuring protein expression and activities of glial GTs. RESULTS AMPK activities were reduced in the spinal dorsal horn of rats (n = 5) with thermal hyperalgesia induced by nerve injury, which were accompanied with the activation of astrocytes, increased production of interleukin-1β and activities of glycogen synthase kinase 3β, and suppressed protein expression of glial glutamate transporter-1. Thermal hyperalgesia was reversed by spinal activation of AMPK in neuropathic rats (n = 10) and induced by inhibiting spinal AMPK in naive rats (n = 7 to 8). Spinal AMPKα knockdown (n = 6) and AMPKα1 conditional knockout (n = 6) induced thermal hyperalgesia and mechanical allodynia. These genetic alterations mimicked the changes of molecular markers induced by nerve injury. Pharmacological activation of AMPK enhanced glial GT activity in mice with neuropathic pain (n = 8) and attenuated glial glutamate transporter-1 internalization induced by interleukin-1β (n = 4). CONCLUSIONS These findings suggest that enhancing spinal AMPK activities could be an effective approach for the treatment of neuropathic pain.
Collapse
|
31
|
Pirkmajer S, Kulkarni SS, Tom RZ, Ross FA, Hawley SA, Hardie DG, Zierath JR, Chibalin AV. Methotrexate promotes glucose uptake and lipid oxidation in skeletal muscle via AMPK activation. Diabetes 2015; 64:360-9. [PMID: 25338814 PMCID: PMC5703413 DOI: 10.2337/db14-0508] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Methotrexate (MTX) is a widely used anticancer and antirheumatic drug that has been postulated to protect against metabolic risk factors associated with type 2 diabetes, although the mechanism remains unknown. MTX inhibits 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase (ATIC) and thereby slows the metabolism of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranosyl-5'-monophosphate (ZMP) and its precursor AICAR, which is a pharmacological AMPK activator. We explored whether MTX promotes AMPK activation in cultured myotubes and isolated skeletal muscle. We found MTX markedly reduced the threshold for AICAR-induced AMPK activation and potentiated glucose uptake and lipid oxidation. Gene silencing of the MTX target ATIC activated AMPK and stimulated lipid oxidation in cultured myotubes. Furthermore, MTX activated AMPK in wild-type HEK-293 cells. These effects were abolished in skeletal muscle lacking the muscle-specific, ZMP-sensitive AMPK-γ3 subunit and in HEK-293 cells expressing a ZMP-insensitive mutant AMPK-γ2 subunit. Collectively, our findings underscore a role for AMPK as a direct molecular link between MTX and energy metabolism in skeletal muscle. Cotherapy with AICAR and MTX could represent a novel strategy to treat metabolic disorders and overcome current limitations of AICAR monotherapy.
Collapse
Affiliation(s)
- Sergej Pirkmajer
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Sameer S Kulkarni
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Robby Z Tom
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Fiona A Ross
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dundee, Scotland, U.K
| | - Simon A Hawley
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dundee, Scotland, U.K
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dundee, Scotland, U.K
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
32
|
Rana S, Blowers EC, Natarajan A. Small molecule adenosine 5'-monophosphate activated protein kinase (AMPK) modulators and human diseases. J Med Chem 2014; 58:2-29. [PMID: 25122135 DOI: 10.1021/jm401994c] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Adenosine 5'-monophosphate activated protein kinase (AMPK) is a master sensor of cellular energy status that plays a key role in the regulation of whole-body energy homeostasis. AMPK is a serine/threonine kinase that is activated by upstream kinases LKB1, CaMKKβ, and Tak1, among others. AMPK exists as αβγ trimeric complexes that are allosterically regulated by AMP, ADP, and ATP. Dysregulation of AMPK has been implicated in a number of metabolic diseases including type 2 diabetes mellitus and obesity. Recent studies have associated roles of AMPK with the development of cancer and neurological disorders, making it a potential therapeutic target to treat human diseases. This review focuses on the structure and function of AMPK, its role in human diseases, and its direct substrates and provides a brief synopsis of key AMPK modulators and their relevance in human diseases.
Collapse
Affiliation(s)
- Sandeep Rana
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha, Nebraska 68198-6805, United States
| | | | | |
Collapse
|
33
|
Abstract
Recent discoveries of AMPK activators point to the large number of therapeutic candidates that can be transformed to successful designs of novel drugs. AMPK is a universal energy sensor and influences almost all physiological processes in the cells. Thus, regulation of the cellular energy metabolism can be achieved in selective tissues via the artificial activation of AMPK by small molecules. Recently, special attention has been given to direct activators of AMPK that are regulated by several nonspecific upstream factors. The direct activation of AMPK, by definition, should lead to more specific biological activities and as a result minimize possible side effects.
Collapse
|
34
|
Ceschin J, Saint-Marc C, Laporte J, Labriet A, Philippe C, Moenner M, Daignan-Fornier B, Pinson B. Identification of yeast and human 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAr) transporters. J Biol Chem 2014; 289:16844-54. [PMID: 24778186 DOI: 10.1074/jbc.m114.551192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
5-Aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAr) is the precursor of the active monophosphate form (AICAR), a small molecule with potent anti-proliferative and low energy mimetic properties. The molecular bases for AICAR toxicity at the cellular level are poorly understood. Here, we report the isolation and characterization of several yeast AICAr-hypersensitive mutants. Identification of the cognate genes allowed us to establish that thiamine transporters Thi7 and Thi72 can efficiently take up AICAr under conditions where they are overexpressed. We establish that, under standard growth conditions, Nrt1, the nicotinamide riboside carrier, is the major AICAr transporter in yeast. A study of AICAR accumulation in human cells revealed substantial disparities among cell lines and confirmed that AICAr enters cells via purine nucleoside transporters. Together, our results point to significant differences between yeast and human cells for both AICAr uptake and AICAR accumulation.
Collapse
Affiliation(s)
- Johanna Ceschin
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| | - Christelle Saint-Marc
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| | - Jean Laporte
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| | - Adrien Labriet
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| | - Chloé Philippe
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| | - Michel Moenner
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| | - Bertrand Daignan-Fornier
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| | - Benoît Pinson
- From the Université de Bordeaux IBGC UMR 5095 1, F-33077 Bordeaux, France and the Centre National de la Recherche Scientifique IBGC UMR 5095 1, F-33077 Bordeaux, France
| |
Collapse
|
35
|
Wolf NM, Abad-Zapatero C, Johnson ME, Fung LWM. Structures of SAICAR synthetase (PurC) from Streptococcus pneumoniae with ADP, Mg2+, AIR and Asp. ACTA ACUST UNITED AC 2014; 70:841-50. [PMID: 24598753 DOI: 10.1107/s139900471303366x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/12/2013] [Indexed: 11/10/2022]
Abstract
Streptococcus pneumoniae is a multidrug-resistant pathogen that is a target of considerable interest for antibacterial drug development. One strategy for drug discovery is to inhibit an essential metabolic enzyme. The seventh step of the de novo purine-biosynthesis pathway converts carboxyaminoimidazoleribonucleotide (CAIR) and L-aspartic acid (Asp) to 4-(N-succino)-5-aminoimidazole-4-carboxamide ribonucleotide (SAICAR) in the presence of adenosine 5'-triphosphate (ATP) using the enzyme PurC. PurC has been shown to be conditionally essential for bacterial replication. Two crystal structures of this essential enzyme from Streptococcus pneumoniae (spPurC) in the presence of adenosine 5'-diphosphate (ADP), Mg(2+), aminoimidazoleribonucleotide (AIR) and/or Asp have been obtained. This is the first structural study of spPurC, as well as the first of PurC from any species with Asp in the active site. Based on these findings, two model structures are proposed for the active site with all of the essential ligands (ATP, Mg(2+), Asp and CAIR) present, and a relay mechanism for the formation of the product SAICAR is suggested.
Collapse
Affiliation(s)
- Nina M Wolf
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Celerino Abad-Zapatero
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Michael E Johnson
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Leslie W-M Fung
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
36
|
Crozet P, Margalha L, Confraria A, Rodrigues A, Martinho C, Adamo M, Elias CA, Baena-González E. Mechanisms of regulation of SNF1/AMPK/SnRK1 protein kinases. FRONTIERS IN PLANT SCIENCE 2014; 5:190. [PMID: 24904600 PMCID: PMC4033248 DOI: 10.3389/fpls.2014.00190] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/22/2014] [Indexed: 05/17/2023]
Abstract
The SNF1 (sucrose non-fermenting 1)-related protein kinases 1 (SnRKs1) are the plant orthologs of the budding yeast SNF1 and mammalian AMPK (AMP-activated protein kinase). These evolutionarily conserved kinases are metabolic sensors that undergo activation in response to declining energy levels. Upon activation, SNF1/AMPK/SnRK1 kinases trigger a vast transcriptional and metabolic reprograming that restores energy homeostasis and promotes tolerance to adverse conditions, partly through an induction of catabolic processes and a general repression of anabolism. These kinases typically function as a heterotrimeric complex composed of two regulatory subunits, β and γ, and an α-catalytic subunit, which requires phosphorylation of a conserved activation loop residue for activity. Additionally, SNF1/AMPK/SnRK1 kinases are controlled by multiple mechanisms that have an impact on kinase activity, stability, and/or subcellular localization. Here we will review current knowledge on the regulation of SNF1/AMPK/SnRK1 by upstream components, post-translational modifications, various metabolites, hormones, and others, in an attempt to highlight both the commonalities of these essential eukaryotic kinases and the divergences that have evolved to cope with the particularities of each one of these systems.
Collapse
Affiliation(s)
| | | | | | - Américo Rodrigues
- Instituto Gulbenkian de CiênciaOeiras, Portugal
- Escola Superior de Turismo e Tecnologia do Mar de Peniche, Instituto Politécnico de LeiriaPeniche, Portugal
| | | | | | | | - Elena Baena-González
- Instituto Gulbenkian de CiênciaOeiras, Portugal
- *Correspondence: Elena Baena-González, Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal e-mail:
| |
Collapse
|
37
|
Ereño-Orbea J, Oyenarte I, Martínez-Cruz LA. CBS domains: Ligand binding sites and conformational variability. Arch Biochem Biophys 2013; 540:70-81. [DOI: 10.1016/j.abb.2013.10.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 02/07/2023]
|
38
|
Jeong BC, Park SH, Yoo KS, Shin JS, Song HK. Change in single cystathionine β-synthase domain-containing protein from a bent to flat conformation upon adenosine monophosphate binding. J Struct Biol 2013; 183:40-6. [PMID: 23664870 DOI: 10.1016/j.jsb.2013.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 04/25/2013] [Accepted: 04/28/2013] [Indexed: 12/25/2022]
Abstract
Cystathionine β-synthase (CBS) domains are small intracellular modules that can act as binding domains for adenosine derivatives, and they may regulate the activity of associated enzymes or other functional domains. Among these, the single CBS domain-containing proteins, CBSXs, from Arabidopsis thaliana, have recently been identified as redox regulators of the thioredoxin system. Here, the crystal structure of CBSX2 in complex with adenosine monophosphate (AMP) is reported at 2.2Å resolution. The structure of dimeric CBSX2 with bound-AMP is shown to be approximately flat, which is in stark contrast to the bent form of apo-CBSXs. This conformational change in quaternary structure is triggered by a local structural change of the unique α5 helix, and by moving each loop P into an open conformation to accommodate incoming ligands. Furthermore, subtle rearrangement of the dimer interface triggers movement of all subunits, and consequently, the bent structure of the CBSX2 dimer becomes a flat structure. This reshaping of the structure upon complex formation with adenosine-containing ligand provides evidence that ligand-induced conformational reorganization of antiparallel CBS domains is an important regulatory mechanism.
Collapse
Affiliation(s)
- Byung-Cheon Jeong
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, South Korea
| | | | | | | | | |
Collapse
|
39
|
Stanley DN, Raines CA, Kerfeld CA. Comparative analysis of 126 cyanobacterial genomes reveals evidence of functional diversity among homologs of the redox-regulated CP12 protein. PLANT PHYSIOLOGY 2013; 161. [PMID: 23184231 PMCID: PMC3561022 DOI: 10.1104/pp.112.210542] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
CP12 is found almost universally among photosynthetic organisms, where it plays a key role in regulation of the Calvin cycle by forming a ternary complex with glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and phosphoribulokinase. Newly available genomic sequence data for the phylum Cyanobacteria reveals a heretofore unobserved diversity in cyanobacterial CP12 proteins. Cyanobacterial CP12 proteins can be classified into eight different types based on primary structure features. Among these are CP12-CBS (for cystathionine-β-synthase) domain fusions. CBS domains are regulatory modules for a wide range of cellular activities; many of these bind adenine nucleotides through a conserved motif that is also present in the CBS domains fused to CP12. In addition, a survey of expression data sets shows that the CP12 paralogs are differentially regulated. Furthermore, modeling of the cyanobacterial CP12 protein variants based on the recently available three-dimensional structure of the canonical cyanobacterial CP12 in complex with GAPDH suggests that some of the newly identified cyanobacterial CP12 types are unlikely to bind to GAPDH. Collectively these data show that, as is becoming increasingly apparent for plant CP12 proteins, the role of CP12 in cyanobacteria is likely more complex than previously appreciated, possibly involving other signals in addition to light. Moreover, our findings substantiate the proposal that this small protein may have multiple roles in photosynthetic organisms.
Collapse
|
40
|
Thomas EC, Gunter JH, Webster JA, Schieber NL, Oorschot V, Parton RG, Whitehead JP. Different characteristics and nucleotide binding properties of inosine monophosphate dehydrogenase (IMPDH) isoforms. PLoS One 2012; 7:e51096. [PMID: 23236438 PMCID: PMC3517587 DOI: 10.1371/journal.pone.0051096] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 10/29/2012] [Indexed: 11/18/2022] Open
Abstract
We recently reported that Inosine Monophosphate Dehydrogenase (IMPDH), a rate-limiting enzyme in de novo guanine nucleotide biosynthesis, clustered into macrostructures in response to decreased nucleotide levels and that there were differences between the IMPDH isoforms, IMPDH1 and IMPDH2. We hypothesised that the Bateman domains, which are present in both isoforms and serve as energy-sensing/allosteric modules in unrelated proteins, would contribute to isoform-specific differences and that mutations situated in and around this domain in IMPDH1 which give rise to retinitis pigmentosa (RP) would compromise regulation. We employed immuno-electron microscopy to investigate the ultrastructure of IMPDH macrostructures and live-cell imaging to follow clustering of an IMPDH2-GFP chimera in real-time. Using a series of IMPDH1/IMPDH2 chimera we demonstrated that the propensity to cluster was conferred by the N-terminal 244 amino acids, which includes the Bateman domain. A protease protection assay suggested isoform-specific purine nucleotide binding characteristics, with ATP protecting IMPDH1 and AMP protecting IMPDH2, via a mechanism involving conformational changes upon nucleotide binding to the Bateman domain without affecting IMPDH catalytic activity. ATP binding to IMPDH1 was confirmed in a nucleotide binding assay. The RP-causing mutation, R224P, abolished ATP binding and nucleotide protection and this correlated with an altered propensity to cluster. Collectively these data demonstrate that (i) the isoforms are differentially regulated by AMP and ATP by a mechanism involving the Bateman domain, (ii) communication occurs between the Bateman and catalytic domains and (iii) the RP-causing mutations compromise such regulation. These findings support the idea that the IMPDH isoforms are subject to distinct regulation and that regulatory defects contribute to human disease.
Collapse
Affiliation(s)
- Elaine C. Thomas
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
- * E-mail: (ECT); (JPW)
| | - Jennifer H. Gunter
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Julie A. Webster
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
- Metabolic Medicine, Mater Medical Research Institute, South Brisbane, Queensland, Australia
| | - Nicole L. Schieber
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Viola Oorschot
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Robert G. Parton
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Jonathan P. Whitehead
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
- Metabolic Medicine, Mater Medical Research Institute, South Brisbane, Queensland, Australia
- * E-mail: (ECT); (JPW)
| |
Collapse
|
41
|
Banerjee S, Ghoshal S, Porter TD. Phosphorylation of hepatic AMP-activated protein kinase and liver kinase B1 is increased after a single oral dose of green tea extract to mice. Nutr Res 2012; 32:985-90. [PMID: 23244544 DOI: 10.1016/j.nutres.2012.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 01/07/2023]
Abstract
We have previously shown that green and black tea extracts increase the phosphorylation of AMP-activated protein kinase (AMPK) and HMG-CoA reductase in rat hepatoma cells in culture, concomitant with a decrease in cholesterol synthesis. In the present study, we evaluated the ability of a single oral dose of green or black tea extract to promote the phosphorylation of AMPK, liver kinase B1 (LKB1, an AMPK-kinase), and HMG-CoA reductase in mouse liver. Green tea extract administered by gavage at 50 and 100 mg/kg caused a 2- to 3-fold increase in hepatic AMPK phosphorylation at 3 and 6 hours after dosing and a 1.5- to 2-fold increase in LKB1 phosphorylation at these same time points. The phosphorylation of HMG-CoA reductase at these and later time points was not significantly increased. Black tea administered by gavage at up to 250 mg/kg was ineffective in increasing hepatic AMPK phosphorylation. Both green and black tea extracts increased LKB1 phosphorylation in hepatoma cells in culture at 15 μg/mL, and black tea also increased the phosphorylation of protein kinase A in hepatoma cells. These results suggest that compounds in both tea extracts activate AMPK by activating its upstream kinase, LKB1, and that black tea may do so by first activating protein kinase A, a known kinase for LKB1. Only green tea, at 50 and 100 mg/kg, was able to activate AMPK and LKB1 in mouse liver after oral dosing, suggesting that the polymerized catechins present in black tea do not reach the liver in sufficient concentration to affect AMPK activity.
Collapse
Affiliation(s)
- Subhashis Banerjee
- Graduate Center for Toxicology, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | | | | |
Collapse
|
42
|
Hardie DG, Ross FA, Hawley SA. AMP-activated protein kinase: a target for drugs both ancient and modern. CHEMISTRY & BIOLOGY 2012; 19:1222-36. [PMID: 23102217 PMCID: PMC5722193 DOI: 10.1016/j.chembiol.2012.08.019] [Citation(s) in RCA: 281] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 08/28/2012] [Accepted: 08/31/2012] [Indexed: 02/07/2023]
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status. It is activated, by a mechanism requiring the tumor suppressor LKB1, by metabolic stresses that increase cellular ADP:ATP and/or AMP:ATP ratios. Once activated, it switches on catabolic pathways that generate ATP, while switching off biosynthetic pathways and cell-cycle progress. These effects suggest that AMPK activators might be useful for treatment and/or prevention of type 2 diabetes and cancer. Indeed, AMPK is activated by the drugs metformin and salicylate, the latter being the major breakdown product of aspirin. Metformin is widely used to treat diabetes, while there is epidemiological evidence that both metformin and aspirin provide protection against cancer. We review the mechanisms of AMPK activation by these and other drugs, and by natural products derived from traditional herbal medicines.
Collapse
Affiliation(s)
- D Grahame Hardie
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, Scotland, UK.
| | | | | |
Collapse
|
43
|
Transcript variant dictates Prkag2 cardiomyopathy? J Mol Cell Cardiol 2012; 53:317-9. [DOI: 10.1016/j.yjmcc.2012.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 06/24/2012] [Indexed: 12/31/2022]
|
44
|
Martelli AM, Chiarini F, Evangelisti C, Ognibene A, Bressanin D, Billi AM, Manzoli L, Cappellini A, McCubrey JA. Targeting the liver kinase B1/AMP-activated protein kinase pathway as a therapeutic strategy for hematological malignancies. Expert Opin Ther Targets 2012; 16:729-42. [PMID: 22686561 DOI: 10.1517/14728222.2012.694869] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Despite considerable advances, several hematological malignancies remain incurable with standard treatments. Therefore, there is a need for novel targeted and less toxic therapies, particularly for patients who develop resistance to traditional chemotherapeutic drugs. The liver kinase B1 (LKB1)/AMP-activated protein kinase (AMPK) signaling pathway has recently emerged as a tumor suppressor axis. A critical point is that the LKB1/AMPK network remains functional in a wide range of cancers and could be stimulated by drugs, such as N,N-dimethylimidodicarbonimidic diamide (metformin) or 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside (AICAR). AREAS COVERED The literature data show that drugs activating LKB1/AMPK signaling induced cell cycle arrest, caspase-dependent apoptosis or autophagy in hematopoietic tumors. Moreover, metformin effectively inhibited mammalian target of rapamycin complex 1 (mTORC1)-controlled oncogenetic protein translation, which does not occur with allosteric mTORC1 inhibitors, such as rapamycin and its derivatives. Metformin was also capable of targeting leukemic stem cells, the most relevant target for leukemia eradication. EXPERT OPINION Data emerging from preclinical settings suggest that the LKB1/AMPK pathway is critically involved in regulating proliferation and survival of malignant hematopoietic cells. Thus, it is proposed that drugs activating the LKB1/AMPK axis may offer a novel and less toxic treatment option for some types of hematological malignancies.
Collapse
Affiliation(s)
- Alberto M Martelli
- University of Bologna, Human Anatomy, via Irnerio 48, Bologna, 40126, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lee JW, Choe SS, Jang H, Kim J, Jeong HW, Jo H, Jeong KH, Tadi S, Park MG, Kwak TH, Man Kim J, Hyun DH, Kim JB. AMPK activation with glabridin ameliorates adiposity and lipid dysregulation in obesity. J Lipid Res 2012; 53:1277-86. [PMID: 22493094 DOI: 10.1194/jlr.m022897] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we demonstrate that activation of AMP-activated protein kinase (AMPK) with glabridin alleviates adiposity and hyperlipidemia in obesity. In several obese rodent models, glabridin decreased body weight and adiposity with a concomitant reduction in fat cell size. Further, glabridin ameliorated fatty liver and plasma levels of triglyceride and cholesterol. In accordance with these findings, glabridin suppressed the expression of lipogenic genes such as sterol regulatory element binding transcription factor (SREBP)-1c, fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and stearoyl-CoA desaturase (SCD)-1 in white adipose tissues and liver, whereas it elevated the expression of fatty acid oxidation genes such as carnitine palmitoyl transferase (CPT)1, acyl-CoA oxidase (ACO), and peroxisome proliferator-activated receptor (PPAR)α in muscle. Moreover, glabridin enhanced phosphorylation of AMPK in muscle and liver and promoted fatty acid oxidation by modulating mitochondrial activity. Together, these data suggest that glabridin is a novel AMPK activator that would exert therapeutic effects in obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Joo-Won Lee
- Department of Biophysics and Chemical Biology, School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
5-Aminoimidazole-4-carboxamide-1-beta-D-ribofuranosyl 5'-Monophosphate (AICAR), a Highly Conserved Purine Intermediate with Multiple Effects. Metabolites 2012; 2:292-302. [PMID: 24957512 PMCID: PMC3901205 DOI: 10.3390/metabo2020292] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 03/15/2012] [Accepted: 03/16/2012] [Indexed: 12/25/2022] Open
Abstract
AICAR (5-Aminoimidazole-4-carboxamide-1-beta-D-ribofuranosyl 5'-monophosphate) is a natural metabolic intermediate of purine biosynthesis that is present in all organisms. In yeast, AICAR plays important regulatory roles under physiological conditions, notably through its direct interactions with transcription factors. In humans, AICAR accumulates in several metabolic diseases, but its contribution to the symptoms has not yet been elucidated. Further, AICAR has highly promising properties which have been recently revealed. Indeed, it enhances endurance of sedentary mice. In addition, it has antiproliferative effects notably by specifically inducing apoptosis of aneuploid cells. Some of the effects of AICAR are due to its ability to stimulate the AMP-activated protein kinase but some others are not. It is consequently clear that AICAR affects multiple targets although only few of them have been identified so far. This review proposes an overview of the field and suggests future directions.
Collapse
|
47
|
Baykov AA, Tuominen HK, Lahti R. The CBS domain: a protein module with an emerging prominent role in regulation. ACS Chem Biol 2011; 6:1156-63. [PMID: 21958115 DOI: 10.1021/cb200231c] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Regulatory CBS (cystathionine β-synthase) domains exist as two or four tandem copies in thousands of cytosolic and membrane-associated proteins from all kingdoms of life. Mutations in the CBS domains of human enzymes and membrane channels are associated with an array of hereditary diseases. Four CBS domains encoded within a single polypeptide or two identical polypeptides (each having a pair of CBS domains at the subunit interface) form a highly conserved disk-like structure. CBS domains act as autoinhibitory regulatory units in some proteins and activate or further inhibit protein function upon binding to adenosine nucleotides (AMP, ADP, ATP, S-adenosyl methionine, NAD, diadenosine polyphosphates). As a result of the differential effects of the nucleotides, CBS domain-containing proteins can sense cell energy levels. Significant conformational changes are induced in CBS domains by bound ligands, highlighting the structural basis for their effects.
Collapse
Affiliation(s)
- Alexander A. Baykov
- A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119899, Russia
| | - Heidi K. Tuominen
- Department of Biochemistry and Food Chemistry, University of Turku, Turku FIN-20014, Finland
| | - Reijo Lahti
- Department of Biochemistry and Food Chemistry, University of Turku, Turku FIN-20014, Finland
| |
Collapse
|
48
|
Increased transsulfuration mediates longevity and dietary restriction in Drosophila. Proc Natl Acad Sci U S A 2011; 108:16831-6. [PMID: 21930912 DOI: 10.1073/pnas.1102008108] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanisms through which dietary restriction enhances health and longevity in diverse species are unclear. The transsulfuration pathway (TSP) is a highly conserved mechanism for metabolizing the sulfur-containing amino acids, methionine and cysteine. Here we show that Drosophila cystathionine β-synthase (dCBS), which catalyzes the rate-determining step in the TSP, is a positive regulator of lifespan in Drosophila and that the pathway is required for the effects of diet restriction on animal physiology and lifespan. dCBS activity was up-regulated in flies exposed to reduced nutrient conditions, and ubiquitous or neuron-specific transgenic overexpression of dCBS enhanced longevity in fully fed animals. Inhibition of the TSP abrogated the changes in lifespan, adiposity, and protein content that normally accompany diet restriction. RNAi-mediated knockdown of dCBS also limited lifespan extension by diet. Diet restriction reduced levels of protein translation in Drosophila, and we show that this is largely caused by increased metabolic commitment of methionine cycle intermediates to transsulfuration. However, dietary supplementation of methionine restored normal levels of protein synthesis to restricted animals without affecting lifespan, indicating that global reductions in translation alone are not required for diet-restriction longevity. Our results indicate a mechanism by which dietary restriction influences physiology and aging.
Collapse
|
49
|
Oyenarte I, Lucas M, Gómez García I, Martínez-Cruz LA. Purification, crystallization and preliminary crystallographic analysis of the CBS-domain protein MJ1004 from Methanocaldococcus jannaschii. Acta Crystallogr Sect F Struct Biol Cryst Commun 2011; 67:318-24. [PMID: 21393835 PMCID: PMC3053155 DOI: 10.1107/s1744309110053479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Accepted: 12/20/2010] [Indexed: 11/10/2022]
Abstract
The purification and preliminary crystallographic analysis of the archaeal CBS-domain protein MJ1004 from Methanocaldococcus jannaschii are described. The native protein was overexpressed, purified and crystallized in the monoclinic space group P2(1), with unit-cell parameters a=54.4, b=53.8, c=82.6 Å, β=106.1°. The crystals diffracted X-rays to 2.7 Å resolution using synchrotron radiation. Matthews-volume calculations suggested the presence of two molecules in the asymmetric unit that are likely to correspond to a dimeric species, which is also observed in solution.
Collapse
Affiliation(s)
- Iker Oyenarte
- Structural Biology Unit, CIC bioGUNE, Parque Tecnológico de Bizkaia, Edificio 800, 48160 Derio, Bizkaia, Spain
| | - María Lucas
- Center for Integrated Protein Sciences and Munich Center for Advanced Photonics at the Gene Center, Department of Biochemistry, Ludwig-Maximilians-University Munich, Feodor-Lynen-Strasse 25, 81377 Munich, Germany
| | - Inmaculada Gómez García
- Structural Biology Unit, CIC bioGUNE, Parque Tecnológico de Bizkaia, Edificio 800, 48160 Derio, Bizkaia, Spain
| | - Luis Alfonso Martínez-Cruz
- Structural Biology Unit, CIC bioGUNE, Parque Tecnológico de Bizkaia, Edificio 800, 48160 Derio, Bizkaia, Spain
| |
Collapse
|
50
|
Mutational analysis of residues in the regulatory CBS domains of Moorella thermoacetica pyrophosphatase corresponding to disease-related residues of human proteins. Biochem J 2011; 433:497-504. [PMID: 21067517 DOI: 10.1042/bj20101204] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
mtCBS-PPase [CBS (cystathionine β-synthase) domain-containing pyrophosphatase from Moorella thermoacetica] contains a pair of CBS domains that strongly bind adenine nucleotides, thereby regulating enzyme activity. Eight residues associated with the CBS domains of mtCBS-PPase were screened to explore possible associations with regulation of enzyme activity. The majority of the substitutions (V99A, R168A, Y169A, Y169F, Y188A and H189A) enhanced the catalytic activity of mtCBS-PPase, two substitutions (R170A and R187G) decreased activity, and one substitution (K100G) had no effect. AMP-binding affinity was markedly decreased in the V99A, R168A and Y169A mutant proteins, and elevated in the R187G and H189A mutant proteins. Remarkably, the R168A and Y169A substitutions changed the effect of AMP from inhibition to activation. The stoichiometry of AMP binding increased from one to two AMP molecules per CBS domain pair in the Y169F, R170A, R187G and Y188A variants. The ADP-binding affinity decreased in three and increased in four mutant proteins. These findings identify residues determining the strength and selectivity of nucleotide binding, as well as the direction (inhibition or activation) of the subsequent effect. The data suggest that mutations in human CBS domain-containing proteins can be translated into a bacterial context. Furthermore, our data support the hypothesis that the CBS domains act as an 'internal inhibitor' of mtCBS-PPase.
Collapse
|