1
|
Pang ZD, Wang Y, Han MZ, She G, Sun X, Bai RY, Lai BC, Zhang Y, Du XJ, Shyy JYJ, Deng XL. AMPK phosphorylation of K Ca2.3 alleviates angiotensin II-induced endothelial dysfunction. Biochem Pharmacol 2025; 236:116880. [PMID: 40112931 DOI: 10.1016/j.bcp.2025.116880] [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: 07/24/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
The endothelial small-conductance calcium-activated potassium channels (KCa2.3) are indispensable for endothelium-dependent hyperpolarization (EDH) response, mainly in resistance arteries. We recently demonstrated in diet-induced obese mice that adenosine monophosphate-activated protein kinase (AMPK) upregulates endothelial KCa2.3 expression and improves endothelial function. However, the molecular mechanism of regulation of KCa2.3 by AMPK remains less explored. Using techniques of bioinformatics, molecular biology and wire myograph system, we examined KCa2.3 phosphorylation by AMPK in human umbilical vein endothelial cells (HUVECs), human embryonic kidney 293 (HEK-293T) cells and second-order mesenteric resistance arteries from angiotensin II-induced hypertensive mice. In HUVECs, treatment with activators of AMPK (AICAR, metformin, and MK-8722) significantly increased phosphorylation of KCa2.3 Thr106 (human), which was antagonized by AMPK inhibitor compound C. In HEK-293T cells, KCa2.3 current was enhanced by AMPK activation or phosphomimetic mutant KCa2.3 (T106D), which was abolished after de-phosphomimetic mutant (T106A) or deletion of KCa2.3 of Thr106 site (T106Del). In mice with angiotensin II infusion, 2-week treatment with AICAR or overexpressing phosphomimetic mutant KCa2.3 Thr107D (mouse) restored KCa2.3-mediated EDH-dependent relaxation in mesenteric resistance arteries together with reversal of early phase hypertension. Our study demonstrates for the first time that AMPK activation mediates KCa2.3 phosphorylation in endothelial cells with enhanced channel activity. This effect ameliorates endothelial dysfunction of mesenteric resistance arteries and alleviates angiotensin II-induced early phase hypertension in mice.
Collapse
Affiliation(s)
- Zheng-Da Pang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Yan Wang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Meng-Zhuan Han
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Gang She
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Xia Sun
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Ru-Yue Bai
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Bao-Chang Lai
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Yi Zhang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Xiao-Jun Du
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613, USA.
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China.
| |
Collapse
|
2
|
Marin TL, Wilson CG, Ramirez ML, Sun W, Malhotra A, Gongol B. AMPK Phosphorylates LMX1b to Regulate a Brainstem Neurogenic Network Important for Control of Breathing in Neonatal Mice. Int J Mol Sci 2024; 26:213. [PMID: 39796072 PMCID: PMC11720625 DOI: 10.3390/ijms26010213] [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: 12/07/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Ventilatory drive is modulated by a variety of neurochemical inputs that converge on spatially oriented clusters of cells within the brainstem. This regulation is required to maintain energy homeostasis and is essential to sustain life across all mammalian organisms. Therefore, the anatomical orientation of these cellular clusters during development must have a defined mechanistic basis with redundant genomic variants. Failure to completely develop these features causes several conditions including apnea of prematurity (AOP) and sudden infant death syndrome (SIDS). AOP is associated with many adverse outcomes including increased risk of interventricular hemorrhage. However, there are no pharmacological interventions that reduce SIDS and AOP prevalence by promoting brainstem development. AMP-activated protein kinase (AMPK) is a kinase that regulates ventilatory control to maintain homeostasis. This study identifies a signaling axis in which the pharmacological activation of AMPK in vivo via metformin in brainstem ventilatory control centers results in the phosphorylation of LIM homeobox transcription factor 1-beta (Lmx1b), a key player in dorsal-ventral patterning during fetal development. The phosphorylation of Lmx1b transactivates a neurogenic interactome important for the development and regulation of ventilatory control centers. These findings highlight the potential for metformin in the treatment and prevention of AOP.
Collapse
Affiliation(s)
- Traci L. Marin
- Department of Respiratory Therapy, Victor Valley College, Victorville, CA 92395, USA
- Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Christopher G. Wilson
- Department of Basic Sciences, Division of Physiology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Miguel Lopez Ramirez
- Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
- Department of Pharmacology, University of California San Diego, San Diego, CA 92093, USA
| | - Wei Sun
- Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
- VA San Deigo Medical Center, San Diego, CA 92161, USA
| | - Atul Malhotra
- Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Brendan Gongol
- Department of Respiratory Therapy, Victor Valley College, Victorville, CA 92395, USA
- Institute for Integrative Genome Biology, University of California, Riverside, CA 92697, USA
| |
Collapse
|
3
|
Zhao M, Cao N, Gu H, Xu J, Xu W, Zhang D, Wei TYW, Wang K, Guo R, Cui H, Wang X, Guo X, Li Z, He K, Li Z, Zhang Y, Shyy JYJ, Dong E, Xiao H. AMPK Attenuation of β-Adrenergic Receptor-Induced Cardiac Injury via Phosphorylation of β-Arrestin-1-ser330. Circ Res 2024; 135:651-667. [PMID: 39082138 DOI: 10.1161/circresaha.124.324762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND β-adrenergic receptor (β-AR) overactivation is a major pathological cue associated with cardiac injury and diseases. AMPK (AMP-activated protein kinase), a conserved energy sensor, regulates energy metabolism and is cardioprotective. However, whether AMPK exerts cardioprotective effects via regulating the signaling pathway downstream of β-AR remains unclear. METHODS Using immunoprecipitation, mass spectrometry, site-specific mutation, in vitro kinase assay, and in vivo animal studies, we determined whether AMPK phosphorylates β-arrestin-1 at serine (Ser) 330. Wild-type mice and mice with site-specific mutagenesis (S330A knock-in [KI]/S330D KI) were subcutaneously injected with the β-AR agonist isoproterenol (5 mg/kg) to evaluate the causality between β-adrenergic insult and β-arrestin-1 Ser330 phosphorylation. Cardiac transcriptomics was used to identify changes in gene expression from β-arrestin-1-S330A/S330D mutation and β-adrenergic insult. RESULTS Metformin could decrease cAMP/PKA (protein kinase A) signaling induced by isoproterenol. AMPK bound to β-arrestin-1 and phosphorylated Ser330 with the highest phosphorylated mass spectrometry score. AMPK activation promoted β-arrestin-1 Ser330 phosphorylation in vitro and in vivo. Neonatal mouse cardiomyocytes overexpressing β-arrestin-1-S330D (active form) inhibited the β-AR/cAMP/PKA axis by increasing PDE (phosphodiesterase) 4 expression and activity. Cardiac transcriptomics revealed that the differentially expressed genes between isoproterenol-treated S330A KI and S330D KI mice were mainly involved in immune processes and inflammatory response. β-arrestin-1 Ser330 phosphorylation inhibited isoproterenol-induced reactive oxygen species production and NLRP3 (NOD-like receptor protein 3) inflammasome activation in neonatal mouse cardiomyocytes. In S330D KI mice, the β-AR-activated cAMP/PKA pathways were attenuated, leading to repressed inflammasome activation, reduced expression of proinflammatory cytokines, and mitigated macrophage infiltration. Compared with S330A KI mice, S330D KI mice showed diminished cardiac fibrosis and improved cardiac function upon isoproterenol exposure. However, the cardiac protection exerted by AMPK was abolished in S330A KI mice. CONCLUSIONS AMPK phosphorylation of β-arrestin-1 Ser330 potentiated PDE4 expression and activity, thereby inhibiting β-AR/cAMP/PKA activation. Subsequently, β-arrestin-1 Ser330 phosphorylation blocks β-AR-induced cardiac inflammasome activation and remodeling.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Ning Cao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital (N.C.), Capital Medical University, Beijing, China
| | - Huijun Gu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Jiachao Xu
- Laboratory for Clinical Medicine (N.C.), Capital Medical University, Beijing, China
| | - Wenli Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| | - Di Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (D.Z., Zhiyuan Li), Peking University, Beijing, China
| | - Tong-You Wade Wei
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-Y.W.W., J.Y.-J.S.)
| | - Kang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Ruiping Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Hongtu Cui
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Xiaofeng Wang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (X.W.)
| | - Xin Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Zhiyuan Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (D.Z., Zhiyuan Li), Peking University, Beijing, China
| | - Kangmin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China (J.X., K.H.)
- University of Chinese Academy of Sciences, Beijing, China (K.H.)
| | - Zijian Li
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-Y.W.W., J.Y.-J.S.)
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Institute of Cardiovascular Sciences (E.D.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| |
Collapse
|
4
|
Østvold AC, Grundt K, Wiese C. NUCKS1 is a highly modified, chromatin-associated protein involved in a diverse set of biological and pathophysiological processes. Biochem J 2022; 479:1205-1220. [PMID: 35695515 PMCID: PMC10016235 DOI: 10.1042/bcj20220075] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/17/2022] [Accepted: 05/26/2022] [Indexed: 11/17/2022]
Abstract
The Nuclear Casein and Cyclin-dependent Kinase Substrate 1 (NUCKS1) protein is highly conserved in vertebrates, predominantly localized to the nucleus and one of the most heavily modified proteins in the human proteome. NUCKS1 expression is high in stem cells and the brain, developmentally regulated in mice and associated with several diverse malignancies in humans, including cancer, metabolic syndrome and Parkinson's disease. NUCKS1 function has been linked to modulating chromatin architecture and transcription, DNA repair and cell cycle regulation. In this review, we summarize and discuss the published information on NUCKS1 and highlight the questions that remain to be addressed to better understand the complex biology of this multifaceted protein.
Collapse
Affiliation(s)
- Anne Carine Østvold
- Institute of Basic Medical Science, Dept. of Biochemistry, University of Oslo, P.O box 1110 Blindern, 0317 Oslo, Norway
| | - Kirsten Grundt
- Institute of Basic Medical Science, Dept. of Biochemistry, University of Oslo, P.O box 1110 Blindern, 0317 Oslo, Norway
| | - Claudia Wiese
- Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| |
Collapse
|
5
|
Yan Y, Mukherjee S, Harikumar KG, Strutzenberg TS, Zhou XE, Suino-Powell K, Xu TH, Sheldon RD, Lamp J, Brunzelle JS, Radziwon K, Ellis A, Novick SJ, Vega IE, Jones RG, Miller LJ, Xu HE, Griffin PR, Kossiakoff AA, Melcher K. Structure of an AMPK complex in an inactive, ATP-bound state. Science 2021; 373:413-419. [PMID: 34437114 PMCID: PMC8428800 DOI: 10.1126/science.abe7565] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 03/31/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022]
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK) regulates metabolism in response to the cellular energy states. Under energy stress, AMP stabilizes the active AMPK conformation, in which the kinase activation loop (AL) is protected from protein phosphatases, thus keeping the AL in its active, phosphorylated state. At low AMP:ATP (adenosine triphosphate) ratios, ATP inhibits AMPK by increasing AL dynamics and accessibility. We developed conformation-specific antibodies to trap ATP-bound AMPK in a fully inactive, dynamic state and determined its structure at 3.5-angstrom resolution using cryo-electron microscopy. A 180° rotation and 100-angstrom displacement of the kinase domain fully exposes the AL. On the basis of the structure and supporting biophysical data, we propose a multistep mechanism explaining how adenine nucleotides and pharmacological agonists modulate AMPK activity by altering AL phosphorylation and accessibility.
Collapse
Affiliation(s)
- Yan Yan
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Somnath Mukherjee
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Kaleeckal G Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Timothy S Strutzenberg
- Department of Molecular Medicine, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - X Edward Zhou
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Kelly Suino-Powell
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ting-Hai Xu
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ryan D Sheldon
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Jared Lamp
- Integrated Mass Spectrometry Unit, Department of Translational Neuroscience, Michigan State University College of Human Medicine, Grand Rapids Research Center, Grand Rapids, MI 49503, USA
| | - Joseph S Brunzelle
- Life Sciences Collaborative Access Team, Northwestern University Synchrotron Research Center, Northwestern University, Argonne, IL 60439, USA
| | - Katarzyna Radziwon
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Abigail Ellis
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Scott J Novick
- Department of Molecular Medicine, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Irving E Vega
- Integrated Mass Spectrometry Unit, Department of Translational Neuroscience, Michigan State University College of Human Medicine, Grand Rapids Research Center, Grand Rapids, MI 49503, USA
| | - Russell G Jones
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - H Eric Xu
- Center for Structure and Function of Drug Targets, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
| | - Patrick R Griffin
- Department of Molecular Medicine, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
- Institute of Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Karsten Melcher
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
6
|
Shi Y, Shen HM, Gopalakrishnan V, Gordon N. Epigenetic Regulation of Autophagy Beyond the Cytoplasm: A Review. Front Cell Dev Biol 2021; 9:675599. [PMID: 34195194 PMCID: PMC8237754 DOI: 10.3389/fcell.2021.675599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process induced under various stress conditions to protect the cell from harm and allow survival in the face of nutrient- or energy-deficient states. Regulation of autophagy is complex, as cells need to adapt to a continuously changing microenvironment. It is well recognized that the AMPK and mTOR signaling pathways are the main regulators of autophagy. However, various other signaling pathways have also been described to regulate the autophagic process. A better understanding of these complex autophagy regulatory mechanisms will allow the discovery of new potential therapeutic targets. Here, we present a brief overview of autophagy and its regulatory pathways with emphasis on the epigenetic control mechanisms.
Collapse
Affiliation(s)
- Yin Shi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Vidya Gopalakrishnan
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nancy Gordon
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
7
|
Paquette M, El-Houjeiri L, C Zirden L, Puustinen P, Blanchette P, Jeong H, Dejgaard K, Siegel PM, Pause A. AMPK-dependent phosphorylation is required for transcriptional activation of TFEB and TFE3. Autophagy 2021; 17:3957-3975. [PMID: 33734022 DOI: 10.1080/15548627.2021.1898748] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Increased macroautophagy/autophagy and lysosomal activity promote tumor growth, survival and chemo-resistance. During acute starvation, autophagy is rapidly engaged by AMPK (AMP-activated protein kinase) activation and MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) inhibition to maintain energy homeostasis and cell survival. TFEB (transcription factor E3) and TFE3 (transcription factor binding to IGHM enhancer 3) are master transcriptional regulators of autophagy and lysosomal activity and their cytoplasm/nuclear shuttling is controlled by MTORC1-dependent multisite phosphorylation. However, it is not known whether and how the transcriptional activity of TFEB or TFE3 is regulated. We show that AMPK mediates phosphorylation of TFEB and TFE3 on three serine residues, leading to TFEB and TFE3 transcriptional activity upon nutrient starvation, FLCN (folliculin) depletion and pharmacological manipulation of MTORC1 or AMPK. Collectively, we show that MTORC1 specifically controls TFEB and TFE3 cytosolic retention, whereas AMPK is essential for TFEB and TFE3 transcriptional activity. This dual and opposing regulation of TFEB and TFE3 by MTORC1 and AMPK is reminiscent of the regulation of another critical regulator of autophagy, ULK1 (unc-51 like autophagy activating kinase 1). Surprisingly, we show that chemoresistance is mediated by AMPK-dependent activation of TFEB, which is abolished by pharmacological inhibition of AMPK or mutation of serine 466, 467 and 469 to alanine residues within TFEB. Altogether, we show that AMPK is a key regulator of TFEB and TFE3 transcriptional activity, and we validate AMPK as a promising target in cancer therapy to evade chemotherapeutic resistance.AbbreviationsACACA: acetyl-CoA carboxylase alpha; ACTB: actin beta; AICAR: 5-aminoimidazole-4-carboxamide ribonucleotide; AMPK: AMP-activated protein kinase; AMPKi: AMPK inhibitor, SBI-0206965; CA: constitutively active; CARM1: coactivator-associated arginine methyltransferase 1; CFP: cyan fluorescent protein; CLEAR: coordinated lysosomal expression and regulation; DKO: double knock-out; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethyl sulfoxide; DQ-BSA: self-quenched BODIPY® dye conjugates of bovine serum albumin; EBSS: Earle's balanced salt solution; FLCN: folliculin; GFP: green fluorescent protein; GST: glutathione S-transferases; HD: Huntington disease; HTT: huntingtin; KO: knock-out; LAMP1: lysosomal associated membrane protein 1; MEF: mouse embryonic fibroblasts; MITF: melanocyte inducing transcription factor; MTORC1: MTOR complex 1; PolyQ: polyglutamine; RPS6: ribosomal protein S6; RT-qPCR: reverse transcription quantitative polymerase chain reaction; TCL: total cell lysates; TFE3: transcription factor binding to IGHM enhancer 3; TFEB: transcription factor EB; TKO: triple knock-out; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Mathieu Paquette
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada.,Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Leeanna El-Houjeiri
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada.,Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Linda C Zirden
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada.,Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Pietri Puustinen
- Cell Death and Metabolism, Danish Cancer Society Research Center (DCRC), Copenhagen, Denmark
| | - Paola Blanchette
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada.,Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Hyeonju Jeong
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada.,Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Kurt Dejgaard
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Peter M Siegel
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada.,Department of Biochemistry, McGill University, Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Arnim Pause
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada.,Department of Biochemistry, McGill University, Montréal, Québec, Canada
| |
Collapse
|
8
|
Chatham JC, Zhang J, Wende AR. Role of O-Linked N-Acetylglucosamine Protein Modification in Cellular (Patho)Physiology. Physiol Rev 2020; 101:427-493. [PMID: 32730113 DOI: 10.1152/physrev.00043.2019] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the mid-1980s, the identification of serine and threonine residues on nuclear and cytoplasmic proteins modified by a N-acetylglucosamine moiety (O-GlcNAc) via an O-linkage overturned the widely held assumption that glycosylation only occurred in the endoplasmic reticulum, Golgi apparatus, and secretory pathways. In contrast to traditional glycosylation, the O-GlcNAc modification does not lead to complex, branched glycan structures and is rapidly cycled on and off proteins by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery, O-GlcNAcylation has been shown to contribute to numerous cellular functions, including signaling, protein localization and stability, transcription, chromatin remodeling, mitochondrial function, and cell survival. Dysregulation in O-GlcNAc cycling has been implicated in the progression of a wide range of diseases, such as diabetes, diabetic complications, cancer, cardiovascular, and neurodegenerative diseases. This review will outline our current understanding of the processes involved in regulating O-GlcNAc turnover, the role of O-GlcNAcylation in regulating cellular physiology, and how dysregulation in O-GlcNAc cycling contributes to pathophysiological processes.
Collapse
Affiliation(s)
- John C Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Jianhua Zhang
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
9
|
Ly CH, Lynch GS, Ryall JG. A Metabolic Roadmap for Somatic Stem Cell Fate. Cell Metab 2020; 31:1052-1067. [PMID: 32433923 DOI: 10.1016/j.cmet.2020.04.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/13/2020] [Accepted: 04/29/2020] [Indexed: 01/14/2023]
Abstract
While metabolism was initially thought to play a passive role in cell biology by generating ATP to meet bioenergetic demands, recent studies have identified critical roles for metabolism in the generation of new biomass and provision of obligate substrates for the epigenetic modification of histones and DNA. This review details how metabolites generated through glycolysis and the tricarboxylic acid cycle are utilized by somatic stem cells to support cell proliferation and lineage commitment. Importantly, we also discuss the evolving hypothesis that histones can act as an energy reservoir during times of energy stress. Finally, we discuss how cells integrate both extrinsic metabolic cues and intrinsic metabolic machinery to regulate cell fate.
Collapse
Affiliation(s)
- C Hai Ly
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - James G Ryall
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
10
|
He M, Martin M, Marin T, Chen Z, Gongol B. Endothelial mechanobiology. APL Bioeng 2020; 4:010904. [PMID: 32095737 PMCID: PMC7032971 DOI: 10.1063/1.5129563] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/19/2020] [Indexed: 12/11/2022] Open
Abstract
Lining the luminal surface of the vasculature, endothelial cells (ECs) are in direct
contact with and differentially respond to hemodynamic forces depending on their anatomic
location. Pulsatile shear stress (PS) is defined by laminar flow and is predominantly
located in straight vascular regions, while disturbed or oscillatory shear stress (OS) is
localized to branch points and bifurcations. Such flow patterns have become a central
focus of vascular diseases, such as atherosclerosis, because the focal distribution of
endothelial dysfunction corresponds to regions exposed to OS, whereas endothelial
homeostasis is maintained in regions defined by PS. Deciphering the mechanotransduction
events that occur in ECs in response to differential flow patterns has required the
innovation of multidisciplinary approaches in both in vitro and
in vivo systems. The results from these studies have identified a
multitude of shear stress-regulated molecular networks in the endothelium that are
implicated in health and disease. This review outlines the significance of scientific
findings generated in collaboration with Dr. Shu Chien.
Collapse
Affiliation(s)
- Ming He
- Department of Medicine, University of California, San Diego, California 92093, USA
| | - Marcy Martin
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Traci Marin
- Department of Health Sciences, Victor Valley College, Victorville, California 92395, USA
| | - Zhen Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, California 91010, USA
| | - Brendan Gongol
- Department of Medicine, University of California, San Diego, California 92093, USA
| |
Collapse
|
11
|
Saline M, Badertscher L, Wolter M, Lau R, Gunnarsson A, Jacso T, Norris T, Ottmann C, Snijder A. AMPK and AKT protein kinases hierarchically phosphorylate the N-terminus of the FOXO1 transcription factor, modulating interactions with 14-3-3 proteins. J Biol Chem 2019; 294:13106-13116. [PMID: 31308176 DOI: 10.1074/jbc.ra119.008649] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/10/2019] [Indexed: 11/06/2022] Open
Abstract
Forkhead box protein O1 (FOXO1) is a transcription factor involved in various cellular processes such as glucose metabolism, development, stress resistance, and tumor suppression. FOXO1's transcriptional activity is controlled by different environmental cues through a myriad of posttranslational modifications. In response to growth factors, the serine/threonine kinase AKT phosphorylates Thr24 and Ser256 in FOXO1 to stimulate binding of 14-3-3 proteins, causing FOXO1 inactivation. In contrast, low nutrient and energy levels induce FOXO1 activity. AMP-activated protein kinase (AMPK), a master regulator of cellular energy homeostasis, partly mediates this effect through phosphorylation of Ser383 and Thr649 in FOXO1. In this study, we identified Ser22 as an additional AMPK phosphorylation site in FOXO1's N terminus, with Ser22 phosphorylation preventing binding of 14-3-3 proteins. The crystal structure of a FOXO1 peptide in complex with 14-3-3 σ at 2.3 Å resolution revealed that this is a consequence of both steric hindrance and electrostatic repulsion. Furthermore, we found that AMPK-mediated Ser22 phosphorylation impairs Thr24 phosphorylation by AKT in a hierarchical manner. Thus, numerous mechanisms maintain FOXO1 activity via AMPK signaling. AMPK-mediated Ser22 phosphorylation directly and indirectly averts binding of 14-3-3 proteins, whereas phosphorylation of Ser383 and Thr649 complementarily stimulates FOXO1 activity. Our results shed light on a mechanism that integrates inputs from both AMPK and AKT signaling pathways in a small motif to fine-tune FOXO1 transcriptional activity.
Collapse
Affiliation(s)
- Maria Saline
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Lukas Badertscher
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Madita Wolter
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P. O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Roxanne Lau
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P. O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Anders Gunnarsson
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Tomas Jacso
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Tyrrell Norris
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P. O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Arjan Snijder
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
12
|
Gongol B, Marin T, Zhang J, Wang SC, Sun W, He M, Chen S, Chen L, Li J, Liu JH, Martin M, Han Y, Kang J, Johnson DA, Lytle C, Li YS, Huang PH, Chien S, Shyy JYJ. Shear stress regulation of miR-93 and miR-484 maturation through nucleolin. Proc Natl Acad Sci U S A 2019; 116:12974-12979. [PMID: 31182601 PMCID: PMC6600934 DOI: 10.1073/pnas.1902844116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pulsatile shear (PS) and oscillatory shear (OS) elicit distinct mechanotransduction signals that maintain endothelial homeostasis or induce endothelial dysfunction, respectively. A subset of microRNAs (miRs) in vascular endothelial cells (ECs) are differentially regulated by PS and OS, but the regulation of the miR processing and its implications in EC biology by shear stress are poorly understood. From a systematic in silico analysis for RNA binding proteins that regulate miR processing, we found that nucleolin (NCL) is a major regulator of miR processing in response to OS and essential for the maturation of miR-93 and miR-484 that target mRNAs encoding Krüppel-like factor 2 (KLF2) and endothelial nitric oxide synthase (eNOS). Additionally, anti-miR-93 and anti-miR-484 restore KLF2 and eNOS expression and NO bioavailability in ECs under OS. Analysis of posttranslational modifications of NCL identified that serine 328 (S328) phosphorylation by AMP-activated protein kinase (AMPK) was a major PS-activated event. AMPK phosphorylation of NCL sequesters it in the nucleus, thereby inhibiting miR-93 and miR-484 processing and their subsequent targeting of KLF2 and eNOS mRNA. Elevated levels of miR-93 and miR-484 were found in sera collected from individuals afflicted with coronary artery disease in two cohorts. These findings provide translational relevance of the AMPK-NCL-miR-93/miR-484 axis in miRNA processing in EC health and coronary artery disease.
Collapse
Affiliation(s)
- Brendan Gongol
- Department of Medicine, University of California, San Diego, CA 92093
| | - Traci Marin
- Department of Health Sciences, Victor Valley College, Victorville, CA 92395
| | - Jiao Zhang
- Department of Medicine, University of California, San Diego, CA 92093
| | - Shen-Chih Wang
- Department of Anesthesiology, Taipei Veterans General Hospital, 115 Taipei, Taiwan
| | - Wei Sun
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
| | - Ming He
- Department of Medicine, University of California, San Diego, CA 92093
| | - Shanshan Chen
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China Xi'an, 710029 Xi'an, China
| | - Lili Chen
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China Xi'an, 710029 Xi'an, China
| | - Jie Li
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China Xi'an, 710029 Xi'an, China
| | - Jun-Hui Liu
- Department of Clinical Laboratory, First Affiliated Hospital of the Medical School, Xi'an Jiaotong University, 710029 Xi'an, China
| | - Marcy Martin
- Department of Medicine, University of California, San Diego, CA 92093
| | - Yue Han
- Institute of Mechanobiology and Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Jian Kang
- Department of Medicine, University of California, San Diego, CA 92093
| | - David A Johnson
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
| | - Christian Lytle
- Division of Biomedical Sciences, University of California, Riverside, CA 92521
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Po-Hsun Huang
- Cardiovascular Research Center, National Yang-Ming University, 112 Taipei, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, 115 Taipei, Taiwan
| | - Shu Chien
- Department of Medicine, University of California, San Diego, CA 92093;
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
| | - John Y-J Shyy
- Department of Medicine, University of California, San Diego, CA 92093;
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China Xi'an, 710029 Xi'an, China
| |
Collapse
|
13
|
Zhao S, Feng J, Li C, Gao H, Lv P, Li J, Liu Q, He Y, Wang H, Gong L, Li D, Zhang Y. Phosphoproteome profiling revealed abnormally phosphorylated AMPK and ATF2 involved in glucose metabolism and tumorigenesis of GH-PAs. J Endocrinol Invest 2019; 42:137-148. [PMID: 29691806 DOI: 10.1007/s40618-018-0890-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/11/2018] [Indexed: 01/04/2023]
Abstract
PURPOSE Protein phosphorylation plays a key role in tumorigenesis and progression. However, little is known about the phosphoproteome profiles of growth hormone-secreting pituitary adenomas (GH-PAs). The aim of this study was to identify critical biomarkers and signaling pathways that might play important roles in GH-PAs and may, therefore, represent potential therapeutic targets. METHODS The differential phosphoprotein expression patterns involved in GH-PAs were investigated by nano-LC-MS/MS in a group of samples. The phosphoprotein expression data were analyzed by bioinformatics. The expression levels of the candidate phosphorylated AMPK (ser496) and ATF2 (ser112) were validated by Western blot analysis in another group of samples. RESULTS A total of 1213 phosphorylated protein sites corresponding to 667 proteins were significantly different between GH-PAs and healthy pituitary glands. Among these phosphorylated sites, 871 exhibited lower levels of phosphorylation in GH-PAs. Moreover, 140 novel phosphosites corresponding to 93 proteins were differentially phosphorylated between GH-PAs and healthy pituitary glands, 101 of which showed decreased phosphorylation in GH-PAs. The majority of differentially expressed phosphorylated proteins were significantly enriched in glycolysis and the AMPK signaling pathway in GH-PAs. The AMPK signaling pathway was demonstrated to be inhibited in GH-PAs by pathway activity analysis (z score = - 2.324). Notably, the phosphorylated levels of AMPK (ser496) and ATF2 (ser112) were significantly lower in GH-PAs than in healthy pituitary glands. CONCLUSION These findings suggest that decreased phosphorylation of the AMPK/ATF2 pathway may be critical for glucose metabolism and tumorigenesis in GH-PAs.
Collapse
Affiliation(s)
- S Zhao
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China.
| | - J Feng
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - C Li
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - H Gao
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - P Lv
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
- Chinese Medical Association, Beijing, 100710, China
| | - J Li
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - Q Liu
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - Y He
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - H Wang
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - L Gong
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - D Li
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China
| | - Y Zhang
- Beijing Neurosurgical Institute, Capital Medical University, TianTanXiLi6, Beijing, 100050, China.
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
- Beijing Institute for Brain Disorders Brain Tumor Center, Capital Medical University, Beijing, 100050, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, 100050, China.
| |
Collapse
|
14
|
Gongol B, Sari I, Bryant T, Rosete G, Marin T. AMPK: An Epigenetic Landscape Modulator. Int J Mol Sci 2018; 19:ijms19103238. [PMID: 30347687 PMCID: PMC6214086 DOI: 10.3390/ijms19103238] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/14/2018] [Accepted: 10/17/2018] [Indexed: 12/22/2022] Open
Abstract
Activated by AMP-dependent and -independent mechanisms, AMP-activated protein kinase (AMPK) plays a central role in the regulation of cellular bioenergetics and cellular survival. AMPK regulates a diverse set of signaling networks that converge to epigenetically mediate transcriptional events. Reversible histone and DNA modifications, such as acetylation and methylation, result in structural chromatin alterations that influence transcriptional machinery access to genomic regulatory elements. The orchestration of these epigenetic events differentiates physiological from pathophysiological phenotypes. AMPK phosphorylation of histones, DNA methyltransferases and histone post-translational modifiers establish AMPK as a key player in epigenetic regulation. This review focuses on the role of AMPK as a mediator of cellular survival through its regulation of chromatin remodeling and the implications this has for health and disease.
Collapse
Affiliation(s)
- Brendan Gongol
- Department of Medicine, University of California, San Diego, CA 92093, USA.
- Department of Cardiopulmonary Sciences, School of Allied Health Professions, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Indah Sari
- Department of Cardiopulmonary Sciences, School of Allied Health Professions, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Tiffany Bryant
- Department of Cardiopulmonary Sciences, School of Allied Health Professions, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Geraldine Rosete
- Department of Cardiopulmonary Sciences, School of Allied Health Professions, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Traci Marin
- Department of Medicine, University of California, San Diego, CA 92093, USA.
- Department of Health Sciences, Victor Valley College, Victorville, CA 92395, USA.
| |
Collapse
|
15
|
Zhang J, Dong J, Martin M, He M, Gongol B, Marin TL, Chen L, Shi X, Yin Y, Shang F, Wu Y, Huang HY, Zhang J, Zhang Y, Kang J, Moya EA, Huang HD, Powell FL, Chen Z, Thistlethwaite PA, Yuan ZY, Shyy JYJ. AMP-activated Protein Kinase Phosphorylation of Angiotensin-Converting Enzyme 2 in Endothelium Mitigates Pulmonary Hypertension. Am J Respir Crit Care Med 2018; 198:509-520. [PMID: 29570986 PMCID: PMC6118028 DOI: 10.1164/rccm.201712-2570oc] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/23/2018] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Endothelial dysfunction plays an integral role in pulmonary hypertension (PH). AMPK (AMP-activated protein kinase) and ACE2 (angiotensin-converting enzyme 2) are crucial in endothelial homeostasis. The mechanism by which AMPK regulates ACE2 in the pulmonary endothelium and its protective role in PH remain elusive. OBJECTIVES We investigated the role of AMPK phosphorylation of ACE2 Ser680 in ACE2 stability and deciphered the functional consequences of this post-translational modification of ACE2 in endothelial homeostasis and PH. METHODS Bioinformatics prediction, kinase assay, and antibody against phospho-ACE2 Ser680 (p-ACE2 S680) were used to investigate AMPK phosphorylation of ACE2 Ser680 in endothelial cells. Using CRISPR-Cas9 genomic editing, we created gain-of-function ACE2 S680D knock-in and loss-of-function ACE2 knockout (ACE2-/-) mouse lines to address the involvement of p-ACE2 S680 and ACE2 in PH. The AMPK-p-ACE2 S680 axis was also validated in lung tissue from humans with idiopathic pulmonary arterial hypertension. MEASUREMENTS AND MAIN RESULTS Phosphorylation of ACE2 by AMPK enhanced the stability of ACE2, which increased Ang (angiotensin) 1-7 and endothelial nitric oxide synthase-derived NO bioavailability. ACE2 S680D knock-in mice were resistant to PH as compared with wild-type littermates. In contrast, ACE2-knockout mice exacerbated PH, a similar phenotype found in mice with endothelial cell-specific deletion of AMPKα2. Consistently, the concentrations of phosphorylated AMPK, p-ACE2 S680, and ACE2 were decreased in human lungs with idiopathic pulmonary arterial hypertension. CONCLUSIONS Impaired phosphorylation of ACE2 Ser680 by AMPK in pulmonary endothelium leads to a labile ACE2 and hence is associated with the pathogenesis of PH. Thus, AMPK regulation of the vasoprotective ACE2 is a potential target for PH treatment.
Collapse
Affiliation(s)
- Jiao Zhang
- Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Division of Cardiology and
| | - Jianjie Dong
- Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Marcy Martin
- Division of Cardiology and
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, California
| | | | - Brendan Gongol
- Department of Cardiopulmonary Sciences, Loma Linda University, Loma Linda, California; and
| | - Traci L. Marin
- Department of Cardiopulmonary Sciences, Loma Linda University, Loma Linda, California; and
| | - Lili Chen
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xinxing Shi
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yanjun Yin
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Fenqing Shang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yan Wu
- Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hsi-Yuan Huang
- Institute of Bioinformatics and Systems Biology and
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City, Taiwan
| | - Jin Zhang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yu Zhang
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| | | | | | - Hsien-Da Huang
- Institute of Bioinformatics and Systems Biology and
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu City, Taiwan
| | | | | | - Patricia A. Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| | - Zu-Yi Yuan
- Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - John Y.-J. Shyy
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Division of Cardiology and
| |
Collapse
|
16
|
Lee MH, Koh D, Na H, Ka NL, Kim S, Kim HJ, Hong S, Shin YK, Seong JK, Lee MO. MTA1 is a novel regulator of autophagy that induces tamoxifen resistance in breast cancer cells. Autophagy 2018; 14:812-824. [PMID: 29130361 PMCID: PMC6070012 DOI: 10.1080/15548627.2017.1388476] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 09/18/2017] [Accepted: 10/02/2017] [Indexed: 12/22/2022] Open
Abstract
Tamoxifen is commonly used to treat patients with ESR/ER-positive breast cancer, but its therapeutic benefit is limited by the development of resistance. Recently, alterations in macroautophagy/autophagy function were demonstrated to be a potential mechanism for tamoxifen resistance. Although MTA1 (metastasis-associated 1) has been implicated in breast tumorigenesis and metastasis, its role in endocrine resistance has not been studied. Here, we report that the level of MTA1 expression was upregulated in the tamoxifen resistant breast cancer cell lines MCF7/TAMR and T47D/TR, and knockdown of MTA1 sensitized the cells to 4-hydroxytamoxifen (4OHT). Moreover, knockdown of MTA1 significantly decreased the enhanced autophagy flux in the tamoxifen resistant cell lines. To confirm the role of MTA1 in the development of tamoxifen resistance, we established a cell line, MCF7/MTA1, which stably expressed MTA1. Compared with parental MCF7, MCF7/MTA1 cells were more resistant to 4OHT-induced growth inhibition in vitro and in vivo, and showed increased autophagy flux and higher numbers of autophagosomes. Knockdown of ATG7 or cotreatment with hydroxychloroquine, an autophagy inhibitor, restored sensitivity to 4OHT in both the MCF7/MTA1 and tamoxifen resistant cells. In addition, AMP-activated protein kinase (AMPK) was activated, probably because of an increased AMP:ATP ratio and decreased expression of mitochondrial electron transport complex components. Finally, publicly available breast cancer patient datasets indicate that MTA1 levels correlate with poor prognosis and development of recurrence in patients with breast cancer treated with tamoxifen. Overall, our findings demonstrated that MTA1 induces AMPK activation and subsequent autophagy that could contribute to tamoxifen resistance in breast cancer.
Collapse
Affiliation(s)
- Min-Ho Lee
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Dahae Koh
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Hyelin Na
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Na-Lee Ka
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Seungsu Kim
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Hyeon-Ji Kim
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Sungyoul Hong
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Young Kee Shin
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University and Korea Mouse Phenotyping Center, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Mi-Ock Lee
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| |
Collapse
|
17
|
Khan AS, Frigo DE. A spatiotemporal hypothesis for the regulation, role, and targeting of AMPK in prostate cancer. Nat Rev Urol 2017; 14:164-180. [PMID: 28169991 PMCID: PMC5672799 DOI: 10.1038/nrurol.2016.272] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The 5'-AMP-activated protein kinase (AMPK) is a master regulator of cellular homeostasis. Despite AMPK's known function in physiology, its role in pathological processes such as prostate cancer is enigmatic. However, emerging evidence is now beginning to decode the paradoxical role of AMPK in cancer and, therefore, inform clinicians if - and how - AMPK could be therapeutically targeted. Spatiotemporal regulation of AMPK complexes could be one of the mechanisms that governs this kinase's role in cancer. We hypothesize that different upstream stimuli will activate select subcellular AMPK complexes. This hypothesis is supported by the distinct subcellular locations of the various AMPK subunits. Each of these unique AMPK complexes regulates discrete downstream processes that can be tumour suppressive or oncogenic. AMPK's final biological output is then determined by the weighted net function of these downstream signalling events, influenced by additional prostate-specific signalling.
Collapse
Affiliation(s)
- Ayesha S. Khan
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX USA
| | - Daniel E. Frigo
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX USA
- Genomic Medicine Program, The Houston Methodist Research Institute, Houston, TX USA
| |
Collapse
|
18
|
Marin TL, Gongol B, Zhang F, Martin M, Johnson DA, Xiao H, Wang Y, Subramaniam S, Chien S, Shyy JYJ. AMPK promotes mitochondrial biogenesis and function by phosphorylating the epigenetic factors DNMT1, RBBP7, and HAT1. Sci Signal 2017; 10:10/464/eaaf7478. [PMID: 28143904 DOI: 10.1126/scisignal.aaf7478] [Citation(s) in RCA: 406] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK) acts as a master regulator of cellular energy homeostasis by directly phosphorylating metabolic enzymes and nutrient transporters and by indirectly promoting the transactivation of nuclear genes involved in mitochondrial biogenesis and function. We explored the mechanism of AMPK-mediated induction of gene expression. We identified AMPK consensus phosphorylation sequences in three proteins involved in nucleosome remodeling: DNA methyltransferase 1 (DNMT1), retinoblastoma binding protein 7 (RBBP7), and histone acetyltransferase 1 (HAT1). DNMT1 mediates DNA methylation that limits transcription factor access to promoters and is inhibited by RBBP7. Acetylation of histones by HAT1 creates a more relaxed chromatin-DNA structure that favors transcription. AMPK-mediated phosphorylation resulted in the activation of HAT1 and inhibition of DNMT1. For DNMT1, this inhibition was both a direct effect of phosphorylation and the result of increased interaction with RBBP7. In human umbilical vein cells, pharmacological AMPK activation or pulsatile shear stress triggered nucleosome remodeling and decreased cytosine methylation, leading to increased expression of nuclear genes encoding factors involved in mitochondrial biogenesis and function, such as peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), transcription factor A (Tfam), and uncoupling proteins 2 and 3 (UCP2 and UCP3). Similar effects were seen in the aortas of mice given pharmacological AMPK activators, and these effects required AMPK2α. These results enhance our understanding of AMPK-mediated mitochondrial gene expression through nucleosome remodeling.
Collapse
Affiliation(s)
- Traci L Marin
- Department of Biochemistry, University of California, Riverside, Riverside, CA 92521, USA.,Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA.,Department of Cardiopulmonary Sciences, Loma Linda University, Loma Linda, CA 92350, USA
| | - Brendan Gongol
- Department of Biochemistry, University of California, Riverside, Riverside, CA 92521, USA.,Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA.,Department of Cardiopulmonary Sciences, Loma Linda University, Loma Linda, CA 92350, USA
| | - Fan Zhang
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521, USA
| | - Marcy Martin
- Department of Biochemistry, University of California, Riverside, Riverside, CA 92521, USA.,Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - David A Johnson
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Han Xiao
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521, USA
| | - Shankar Subramaniam
- Departments of Bioengineering, Cellular and Molecular Medicine, and Computer Science and Engineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Shu Chien
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA.,Departments of Bioengineering, Cellular and Molecular Medicine, and Computer Science and Engineering, University of California, San Diego, San Diego, CA 92093, USA
| | - John Y-J Shyy
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
19
|
Aznar N, Patel A, Rohena CC, Dunkel Y, Joosen LP, Taupin V, Kufareva I, Farquhar MG, Ghosh P. AMP-activated protein kinase fortifies epithelial tight junctions during energetic stress via its effector GIV/Girdin. eLife 2016; 5. [PMID: 27813479 PMCID: PMC5119889 DOI: 10.7554/elife.20795] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 11/03/2016] [Indexed: 02/06/2023] Open
Abstract
Loss of epithelial polarity impacts organ development and function; it is also oncogenic. AMPK, a key sensor of metabolic stress stabilizes cell-cell junctions and maintains epithelial polarity; its activation by Metformin protects the epithelial barrier against stress and suppresses tumorigenesis. How AMPK protects the epithelium remains unknown. Here, we identify GIV/Girdin as a novel effector of AMPK, whose phosphorylation at a single site is both necessary and sufficient for strengthening mammalian epithelial tight junctions and preserving cell polarity and barrier function in the face of energetic stress. Expression of an oncogenic mutant of GIV (cataloged in TCGA) that cannot be phosphorylated by AMPK increased anchorage-independent growth of tumor cells and helped these cells to evade the tumor-suppressive action of Metformin. This work defines a fundamental homeostatic mechanism by which the AMPK-GIV axis reinforces cell junctions against stress-induced collapse and also provides mechanistic insight into the tumor-suppressive action of Metformin.
Collapse
Affiliation(s)
- Nicolas Aznar
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Arjun Patel
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Cristina C Rohena
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Ying Dunkel
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Linda P Joosen
- Department of Medicine, University of California, San Diego, San Diego, United States
| | - Vanessa Taupin
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, United States
| | - Marilyn G Farquhar
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Pradipta Ghosh
- Department of Medicine, University of California, San Diego, San Diego, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States.,Moores Cancer Center, University of California, San Diego, San Diego, United States
| |
Collapse
|
20
|
Shentu TP, He M, Sun X, Zhang J, Zhang F, Gongol B, Marin TL, Zhang J, Wen L, Wang Y, Geary GG, Zhu Y, Johnson DA, Shyy JYJ. AMP-Activated Protein Kinase and Sirtuin 1 Coregulation of Cortactin Contributes to Endothelial Function. Arterioscler Thromb Vasc Biol 2016; 36:2358-2368. [PMID: 27758765 DOI: 10.1161/atvbaha.116.307871] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/12/2016] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Cortactin translocates to the cell periphery in vascular endothelial cells (ECs) on cortical-actin assembly in response to pulsatile shear stress. Because cortactin has putative sites for AMP-activated protein kinase (AMPK) phosphorylation and sirtuin 1 (SIRT1) deacetylation, we examined the hypothesis that AMPK and SIRT1 coregulate cortactin dynamics in response to shear stress. APPROACH AND RESULTS Analysis of the ability of AMPK to phosphorylate recombinant cortactin and oligopeptides whose sequences matched AMPK consensus sequences in cortactin pointed to Thr-401 as the site of AMPK phosphorylation. Mass spectrometry confirmed Thr-401 as the site of AMPK phosphorylation. Immunoblot analysis with AMPK siRNA and SIRT1 siRNA in human umbilical vein ECs and EC-specific AMPKα2 knockout mice showed that AMPK phosphorylation of cortactin primes SIRT1 deacetylation in response to shear stress. Immunoblot analyses with cortactin siRNA in human umbilical vein ECs, phospho-deficient T401A and phospho-mimetic T401D mutant, or aceto-deficient (9K/R) and aceto-mimetic (9K/Q) showed that cortactin regulates endothelial nitric oxide synthase activity. Confocal imaging and sucrose-density gradient analyses revealed that the phosphorylated/deacetylated cortactin translocates to the EC periphery facilitating endothelial nitric oxide synthase translocation from lipid to nonlipid raft domains. Knockdown of cortactin in vitro or genetic reduction of cortactin expression in vivo in mice substantially decreased the endothelial nitric oxide synthase-derived NO bioavailability. In vivo, atherosclerotic lesions increase in ApoE-/-/cortactin+/- mice, when compared with ApoE-/-/cortactin+/+ littermates. CONCLUSIONS AMPK phosphorylation of cortactin followed by SIRT1 deacetylation modulates the interaction of cortactin and cortical-actin in response to shear stress. Functionally, this AMPK/SIRT1 coregulated cortactin-F-actin dynamics is required for endothelial nitric oxide synthase subcellular translocation/activation and is atheroprotective.
Collapse
Affiliation(s)
- Tzu-Pin Shentu
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Ming He
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Xiaoli Sun
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Jianlin Zhang
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Fan Zhang
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Brendan Gongol
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Traci L Marin
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Jiao Zhang
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Liang Wen
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Yinsheng Wang
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Gregory G Geary
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - Yi Zhu
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - David A Johnson
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.)
| | - John Y-J Shyy
- From the Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-P.S., M.H., J.Z., J.Z.; L.W., J.Y.-J.S.); Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China (X.S., Y.Z.); Department of Chemistry, University of California, Riverside (F.Z., Y.W.); Department of Cardiopulmonary Sciences, Schools of Allied Health, Loma Linda University, CA (B.G., T.L.M.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.G.); and Division of Biomedical Sciences, University of California, Riverside (D.A.J.).
| |
Collapse
|
21
|
Hardie DG, Schaffer BE, Brunet A. AMPK: An Energy-Sensing Pathway with Multiple Inputs and Outputs. Trends Cell Biol 2016; 26:190-201. [PMID: 26616193 PMCID: PMC5881568 DOI: 10.1016/j.tcb.2015.10.013] [Citation(s) in RCA: 675] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 12/20/2022]
Abstract
AMP-activated protein kinase (AMPK) is a key regulator of energy balance expressed ubiquitously in eukaryotic cells. Here we review the canonical adenine nucleotide-dependent mechanism that activates AMPK when cellular energy status is compromised, as well as other, noncanonical activation mechanisms. Once activated, AMPK acts to restore energy homeostasis by promoting catabolic pathways, resulting in ATP generation, and inhibiting anabolic pathways that consume ATP. We also review the various hypothesis-driven and unbiased approaches that have been used to identify AMPK substrates and have revealed substrates involved in both metabolic and non-metabolic processes. We particularly focus on methods for identifying the AMPK target recognition motif and how it can be used to predict new substrates.
Collapse
Affiliation(s)
- D Grahame Hardie
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK.
| | - Bethany E Schaffer
- Department of Genetics and the Cancer Biology Program, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| | - Anne Brunet
- Department of Genetics and the Cancer Biology Program, Stanford University, 300 Pasteur Drive, Stanford, CA, USA
| |
Collapse
|
22
|
Schaffer BE, Levin RS, Hertz NT, Maures TJ, Schoof ML, Hollstein PE, Benayoun BA, Banko MR, Shaw RJ, Shokat KM, Brunet A. Identification of AMPK Phosphorylation Sites Reveals a Network of Proteins Involved in Cell Invasion and Facilitates Large-Scale Substrate Prediction. Cell Metab 2015; 22:907-21. [PMID: 26456332 PMCID: PMC4635044 DOI: 10.1016/j.cmet.2015.09.009] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 07/28/2015] [Accepted: 09/08/2015] [Indexed: 12/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is a central energy gauge that regulates metabolism and has been increasingly involved in non-metabolic processes and diseases. However, AMPK's direct substrates in non-metabolic contexts are largely unknown. To better understand the AMPK network, we use a chemical genetics screen coupled to a peptide capture approach in whole cells, resulting in identification of direct AMPK phosphorylation sites. Interestingly, the high-confidence AMPK substrates contain many proteins involved in cell motility, adhesion, and invasion. AMPK phosphorylation of the RHOA guanine nucleotide exchange factor NET1A inhibits extracellular matrix degradation, an early step in cell invasion. The identification of direct AMPK phosphorylation sites also facilitates large-scale prediction of AMPK substrates. We provide an AMPK motif matrix and a pipeline to predict additional AMPK substrates from quantitative phosphoproteomics datasets. As AMPK is emerging as a critical node in aging and pathological processes, our study identifies potential targets for therapeutic strategies.
Collapse
Affiliation(s)
- Bethany E Schaffer
- Cancer Biology Program, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Rebecca S Levin
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA
| | - Nicholas T Hertz
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA
| | - Travis J Maures
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Michael L Schoof
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Pablo E Hollstein
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Max R Banko
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Reuben J Shaw
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA
| | - Anne Brunet
- Cancer Biology Program, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford, CA 94305, USA.
| |
Collapse
|