1
|
Onda DA, Zhu Y, Yuan X, Loh K. Central and Peripheral Roles of Salt-inducible Kinases in Metabolic Regulation. Endocrinology 2025; 166:bqaf024. [PMID: 39919030 DOI: 10.1210/endocr/bqaf024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/09/2025]
Abstract
Salt-inducible kinases (SIKs), a member of the serine/threonine protein kinase family, have recently garnered considerable research interest as one of the emerging key regulators of metabolism. The 3 SIK isoforms-SIK1, SIK2, and SIK3-exhibit diverse roles both in central and peripheral physiological processes. While early studies focused on their role in inflammation, spurring the development of SIK inhibitors for chronic inflammatory diseases currently in clinical trials, emerging evidence highlights their broader functions in metabolism. In this review, we will summarize the current state of research on the central roles of SIKs in the brain, particularly in regulating energy balance and glucose homeostasis, alongside their peripheral functions in critical metabolic tissues such as the liver, adipose tissue, and pancreas. By integrating insights into their central and peripheral roles, this review underscores the importance of SIKs in maintaining metabolic homeostasis and highlights their therapeutic potential as novel targets for metabolic disease.
Collapse
Affiliation(s)
- Danise-Ann Onda
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
| | - Yifei Zhu
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
| | - XiaoZhuo Yuan
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
| | - Kim Loh
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
2
|
Liu Z, Guo Z, Xu J, Zhou R, Shi B, Chen L, Wu C, Wang H, Wang X, Wang F, Li Q, Liu Q. Regulation of Sleep Amount by CRTC1 via Transcription of Crh in Mice. J Neurosci 2025; 45:e0786242024. [PMID: 39622645 PMCID: PMC11780352 DOI: 10.1523/jneurosci.0786-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 01/31/2025] Open
Abstract
The cAMP response element binding protein (CREB) is required for regulation of daily sleep amount, whereas gain of function of CREB-regulated transcription coactivator 1 (CRTC1) causes severe insomnia in mice. However, the physiological functions of CRTCs and their downstream target genes in the regulation of sleep amount remain unclear. Here, we use an adult brain chimeric (ABC)-expression/knock-out platform for somatic genetic analysis of sleep in adult male mice. ABC expression of constitutively active mutant CRTC1/2CA in the mouse brain neurons significantly reduces the amount of non-rapid eye movement sleep (NREMS) and/or rapid eye movement sleep (REMS). Consistent with the fact that SIK3 phosphorylates and inhibits CRTCs, ABC expression of CRTC1/2/3CA rescues the hypersomnia phenotype of Sleepy (Sik3Slp ) mice. While ABC-Crtc2KO or Crtc3KO causes no sleep phenotype, ABC-Crtc1KO or ABC expression of dominant-negative CRTC (dnCRTC) results in a modest reduction of NREMS amount accompanied with elevated NREMS delta power. Moreover, ABC expression of CRTC1CA or dnCRTC in the excitatory neurons causes bidirectional changes of NREMS/REMS amount and/or NREMS delta power. The ability of CRTC1CA to regulate sleep requires its transactivation domain and CREB-binding domain and is dependent on CREB. Furthermore, we showed that inducible ABC expression of corticotropin-releasing hormone (Crh) and brain-derived neurotrophic factor (Bdnf)-two target genes of CRTCs-significantly reduces daily sleep amount. Notably, ABC-CrhKO , but not BdnfKO , rescues the insomnia phenotype of ABC-CRTC1CA mice. Taken together, these results indicate that the CREB-CRTC1 complex regulates daily sleep amount by modulating the transcription of Crh in the mouse brain neurons.
Collapse
Affiliation(s)
- Zhihao Liu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Zhiyong Guo
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junjie Xu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Rui Zhou
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Bihan Shi
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Lin Chen
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Chongyang Wu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Haiyan Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Xia Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| |
Collapse
|
3
|
Yu K, Ramkumar N, Wong KKL, Tettweiler G, Verheyen EM. The AMPK-like protein kinases Sik2 and Sik3 interact with Hipk and induce synergistic tumorigenesis in a Drosophila cancer model. Front Cell Dev Biol 2023; 11:1214539. [PMID: 37854071 PMCID: PMC10579798 DOI: 10.3389/fcell.2023.1214539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023] Open
Abstract
Homeodomain-interacting protein kinases (Hipks) regulate cell proliferation, apoptosis, and tissue development. Overexpression of Hipk in Drosophila causes tumorigenic phenotypes in larval imaginal discs. We find that depletion of Salt-inducible kinases Sik2 or Sik3 can suppress Hipk-induced overgrowth. Furthermore, co-expression of constitutively active forms of Sik2 or Sik3 with Hipk caused significant tissue hyperplasia and tissue distortion, indicating that both Sik2 and Sik3 can synergize with Hipk to promote tumorous phenotypes, accompanied by elevated dMyc, Armadillo/β-catenin, and the Yorkie target gene expanded. Larvae expressing these hyperplastic growths also display an extended larval phase, characteristic of other Drosophila tumour models. Examination of total protein levels from fly tissues showed that Hipk proteins were reduced when Siks were depleted through RNAi, suggesting that Siks may regulate Hipk protein stability and/or activity. Conversely, expression of constitutively active Siks with Hipk leads to increased Hipk protein levels. Furthermore, Hipk can interact with Sik2 and Sik3 by co-immunoprecipitation. Co-expression of both proteins leads to a mobility shift of Hipk protein, suggesting it is post-translationally modified. In summary, our research demonstrates a novel function of Siks in synergizing with Hipk to promote tumour growth.
Collapse
Affiliation(s)
- Kewei Yu
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Niveditha Ramkumar
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Kenneth Kin Lam Wong
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Gritta Tettweiler
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Esther M. Verheyen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
4
|
Protein Targeting to Glycogen (PTG): A Promising Player in Glucose and Lipid Metabolism. Biomolecules 2022; 12:biom12121755. [PMID: 36551183 PMCID: PMC9775135 DOI: 10.3390/biom12121755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Protein phosphorylation and dephosphorylation are widely considered to be the key regulatory factors of cell function, and are often referred to as "molecular switches" in the regulation of cell metabolic processes. A large number of studies have shown that the phosphorylation/dephosphorylation of related signal molecules plays a key role in the regulation of liver glucose and lipid metabolism. As a new therapeutic strategy for metabolic diseases, the potential of using inhibitor-based therapies to fight diabetes has gained scientific momentum. PTG, a protein phosphatase, also known as glycogen targeting protein, is a member of the protein phosphatase 1 (PP1) family. It can play a role by catalyzing the dephosphorylation of phosphorylated protein molecules, especially regulating many aspects of glucose and lipid metabolism. In this review, we briefly summarize the role of PTG in glucose and lipid metabolism, and update its role in metabolic regulation, with special attention to glucose homeostasis and lipid metabolism.
Collapse
|
5
|
Carper MB, Goel S, Zhang AM, Damrauer JS, Cohen S, Zimmerman MP, Gentile GM, Parag-Sharma K, Murphy RM, Sato K, Nickel KP, Kimple RJ, Yarbrough WG, Amelio AL. Activation of the CREB Coactivator CRTC2 by Aberrant Mitogen Signaling promotes oncogenic functions in HPV16 positive head and neck cancer. Neoplasia 2022; 29:100799. [PMID: 35504112 PMCID: PMC9065880 DOI: 10.1016/j.neo.2022.100799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the 6th most common cancer worldwide and incidence rates are continuing to rise globally. Patients often present with locally advanced disease and a staggering 50% chance of relapse following treatment. Aberrant activation of adaptive response signaling pathways, such as the cAMP/PKA pathway, induce an array of genes associated with known cancer pathways that promote tumorigenesis and drug resistance. We identified the cAMP Regulated Transcription Coactivator 2 (CRTC2) to be overexpressed and constitutively activated in HNSCCs and this confers poor prognosis. CRTCs are regulated through their subcellular localization and we show that CRTC2 is exclusively nuclear in HPV(+) HNSCC, thus constitutively active, due to non-canonical Mitogen-Activated Kinase Kinase 1 (MEKK1)-mediated activation via a MEKK1-p38 signaling axis. Loss-of-function and pharmacologic inhibition experiments decreased CRTC2/CREB transcriptional activity by reducing nuclear CRTC2 via nuclear import inhibition and/or by eviction of CRTC2 from the nucleus. This shift in localization was associated with decreased proliferation, migration, and invasion. Our results suggest that small molecules that inhibit nuclear CRTC2 and p38 activity may provide therapeutic benefit to patients with HPV(+) HNSCC.
Collapse
Affiliation(s)
- Miranda B Carper
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA
| | - Saumya Goel
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, The University of North Carolina at Chapel Hill, NC, USA; Carolina Research Scholar, Undergraduate Curriculum in Biochemistry, The University of North Carolina at Chapel Hill, NC, USA
| | - Anna M Zhang
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, The University of North Carolina at Chapel Hill, NC, USA
| | - Jeffrey S Damrauer
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA
| | - Stephanie Cohen
- Pathology Services Core, Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, USA
| | - Matthew P Zimmerman
- Graduate Curriculum in Genetics & Molecular Biology, Biological & Biomedical Sciences Program, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA
| | - Gabrielle M Gentile
- Graduate Curriculum in Genetics & Molecular Biology, Biological & Biomedical Sciences Program, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA
| | - Kshitij Parag-Sharma
- Graduate Curriculum in Cell Biology & Physiology, Biological & Biomedical Sciences Program, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA
| | - Ryan M Murphy
- Graduate Curriculum in Pharmacology, Biological & Biomedical Sciences Program, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA
| | - Kotaro Sato
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA; Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kwangok P Nickel
- Department of Human Oncology and UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Randall J Kimple
- Department of Human Oncology and UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Wendell G Yarbrough
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA; Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina, Chapel Hill, NC, USA; Department of Pathology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, Chapel Hill, NC, USA
| | - Antonio L Amelio
- Department of Cell Biology and Physiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Biomedical Research Imaging Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, Cancer Cell Biology Program, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
6
|
Ji YX, Wang Y, Li PL, Cai L, Wang XM, Bai L, Liu Z, Tian H, Tian S, Zhang P, Zhang XJ, Cheng X, Yuan Y, She ZG, Hu Y, Li H. A kinome screen reveals that Nemo-like kinase is a key suppressor of hepatic gluconeogenesis. Cell Metab 2021; 33:1171-1186.e9. [PMID: 33951476 DOI: 10.1016/j.cmet.2021.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 04/13/2020] [Accepted: 04/10/2021] [Indexed: 12/12/2022]
Abstract
Antihyperglycemic therapy is an important priority for the treatment of type 2 diabetes (T2D). Excessive hepatic glucose production (HGP) is a major cause of fasting hyperglycemia. Therefore, a better understanding of its regulation would be important to develop effective antihyperglycemic therapies. Using a gluconeogenesis-targeted kinome screening approach combined with transcriptome analyses, we uncovered Nemo-like kinase (NLK) as a potent suppressor of HGP. Mechanistically, NLK phosphorylates and promotes nuclear export of CRTC2 and FOXO1, two key regulators of hepatic gluconeogenesis, resulting in the proteasome-dependent degradation of the former and the inhibition of the self-transcriptional activity and expression of the latter. Importantly, the expression of NLK is downregulated in the liver of individuals with diabetes and in diabetic rodent models and restoring NLK expression in the mouse model ameliorates hyperglycemia. Therefore, our findings uncover NLK as a critical player in the gluconeogenic regulatory network and as a potential therapeutic target for T2D.
Collapse
Affiliation(s)
- Yan-Xiao Ji
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal of Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Yutao Wang
- Institute of Model Animal of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peng-Long Li
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal of Wuhan University, Wuhan, China
| | - Lin Cai
- Institute of Model Animal of Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiao-Ming Wang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal of Wuhan University, Wuhan, China
| | - Lan Bai
- Institute of Model Animal of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen Liu
- Institute of Model Animal of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Han Tian
- Institute of Model Animal of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Song Tian
- Institute of Model Animal of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peng Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xu Cheng
- Institute of Model Animal of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yufeng Yuan
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Zhi-Gang She
- Institute of Model Animal of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Yufeng Hu
- Institute of Model Animal of Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Hongliang Li
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal of Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
7
|
Zeigerer A, Sekar R, Kleinert M, Nason S, Habegger KM, Müller TD. Glucagon's Metabolic Action in Health and Disease. Compr Physiol 2021; 11:1759-1783. [PMID: 33792899 PMCID: PMC8513137 DOI: 10.1002/cphy.c200013] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Discovered almost simultaneously with insulin, glucagon is a pleiotropic hormone with metabolic action that goes far beyond its classical role to increase blood glucose. Albeit best known for its ability to directly act on the liver to increase de novo glucose production and to inhibit glycogen breakdown, glucagon lowers body weight by decreasing food intake and by increasing metabolic rate. Glucagon further promotes lipolysis and lipid oxidation and has positive chronotropic and inotropic effects in the heart. Interestingly, recent decades have witnessed a remarkable renaissance of glucagon's biology with the acknowledgment that glucagon has pharmacological value beyond its classical use as rescue medication to treat severe hypoglycemia. In this article, we summarize the multifaceted nature of glucagon with a special focus on its hepatic action and discuss the pharmacological potential of either agonizing or antagonizing the glucagon receptor for health and disease. © 2021 American Physiological Society. Compr Physiol 11:1759-1783, 2021.
Collapse
Affiliation(s)
- Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Revathi Sekar
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Maximilian Kleinert
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Shelly Nason
- Comprehensive Diabetes Center, Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirk M. Habegger
- Comprehensive Diabetes Center, Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Timo D. Müller
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| |
Collapse
|
8
|
Ma L, Chen S, Wang Z, Guo S, Zhao J, Yi D, Li Q, Liu Z, Guo F, Li X, Jia P, Ding J, Liang C, Cen S. The CREB Regulated Transcription Coactivator 2 Suppresses HIV-1 Transcription by Preventing RNA Pol II from Binding to HIV-1 LTR. Virol Sin 2021; 36:796-809. [PMID: 33723808 DOI: 10.1007/s12250-021-00363-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/09/2020] [Indexed: 10/21/2022] Open
Abstract
The CREB-regulated transcriptional co-activators (CRTCs), including CRTC1, CRTC2 and CRTC3, enhance transcription of CREB-targeted genes. In addition to regulating host gene expression in response to cAMP, CRTCs also increase the infection of several viruses. While human immunodeficiency virus type 1 (HIV-1) long terminal repeat (LTR) promoter harbors a cAMP response element and activation of the cAMP pathway promotes HIV-1 transcription, it remains unknown whether CRTCs have any effect on HIV-1 transcription and HIV-1 infection. Here, we reported that CRTC2 expression was induced by HIV-1 infection, but CRTC2 suppressed HIV-1 infection and diminished viral RNA expression. Mechanistic studies revealed that CRTC2 inhibited transcription from HIV-1 LTR and diminished RNA Pol II occupancy at the LTR independent of its association with CREB. Importantly, CRTC2 inhibits the activation of latent HIV-1. Together, these data suggest that in response to HIV-1 infection, cells increase the expression of CRTC2 which inhibits HIV-1 gene expression and may play a role in driving HIV-1 into latency.
Collapse
Affiliation(s)
- Ling Ma
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China
| | - Shumin Chen
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Zhen Wang
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, H3T 1E2, Canada
| | - Saisai Guo
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China
| | - Jianyuan Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China
| | - Dongrong Yi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China
| | - Quanjie Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China
| | - Zhenlong Liu
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, H3T 1E2, Canada
| | - Fei Guo
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100176, China
| | - Xiaoyu Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China
| | - Pingping Jia
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Jiwei Ding
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China. .,CAMS Key Laboratory of Antiviral Drug Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100050, China.
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, H3T 1E2, Canada
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing, 100050, China. .,CAMS Key Laboratory of Antiviral Drug Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100050, China. .,Beijing Friendship Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
9
|
CREB Coactivator CRTC2 Plays a Crucial Role in Endothelial Function. J Neurosci 2020; 40:9533-9546. [PMID: 33127851 DOI: 10.1523/jneurosci.0407-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 11/21/2022] Open
Abstract
The cAMP pathway is known to stabilize endothelial barrier function and maintain vascular physiology. The family of cAMP-response element binding (CREB)-regulated transcription coactivators (CRTC)1-3 activate transcription by targeting the basic leucine zipper domain of CREB. CRTC2 is a master regulator of glucose metabolism in liver and adipose tissue. However, the role of CRTC2 in endothelium remains unknown. The aim of this study was to evaluate the effect of CRTC2 on endothelial function. We focused the effect of CRTC2 in endothelial cells and its relationship with p190RhoGAP-A. We examined the effect of CRTC2 on endothelial function using a mouse aorta ring assay ex vivo and with photothrombotic stroke in endothelial cell-specific CRTC2-knock-out male mice in vivo CRTC2 was highly expressed in endothelial cells and related to angiogenesis. Among CRTC1-3, only CRTC2 was activated under ischemic conditions at endothelial cells, and CRTC2 maintained endothelial barrier function through p190RhoGAP-A expression. Ser171 was a pivotal regulatory site for CRTC2 intracellular localization, and Ser307 functioned as a crucial phosphorylation site. Endothelial cell-specific CRTC2-knock-out mice showed reduced angiogenesis ex vivo, exacerbated stroke via endothelial dysfunction, and impaired neurologic recovery via reduced vascular beds in vivo These findings suggest that CRTC2 plays a crucial protective role in vascular integrity of the endothelium via p190RhoGAP-A under ischemic conditions.SIGNIFICANCE STATEMENT Previously, the role of CRTC2 in endothelial cells was unknown. In this study, we firstly clarified that CRTC2 was expressed in endothelial cells and among CRTC1-3, only CRTC2 was related to endothelial function. Most importantly, only CRTC2 was activated under ischemic conditions at endothelial cells and maintained endothelial barrier function through p190RhoGAP-A expression. Ser307 in CRTC2 functioned as a crucial phosphorylation site. Endothelial cell-specific CRTC2-knock-out mice showed reduced angiogenesis ex vivo, exacerbated stroke via endothelial dysfunction, and impaired neurologic recovery via reduced vascular beds in vivo These results suggested that CRTC2 maybe a potential therapeutic target for reducing blood-brain barrier (BBB) damage and improving recovery.
Collapse
|
10
|
Wang Y, Liu Q, Zhang H. Phosphorylation of CREB-Specific Coactivator CRTC2 at Ser238 Promotes Proliferation, Migration, and Invasion of Colorectal Cancer Cells. Technol Cancer Res Treat 2020; 19:1533033820962111. [PMID: 33000695 PMCID: PMC7533939 DOI: 10.1177/1533033820962111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
cAMP response element binding protein (CREB)-regulated transcription coactivator 2 (CRTC2), a member of the novel CRTC family of transcriptional coactivators that activates basic leucine zipper transcription factors, including CREB, is overexpressed in many carcinomas, including colon cancer. Phosphorylation of CRTC2 protein at different residues is important for its subcellular localization and activity. However, the functions of some of the serine phosphorylation sites have not been elucidated. This study aimed to investigate the effects of phosphorylation of Ser127, Ser238, and Ser245 sites of CRTC2 in colorectal cancer (CRC) cells. Recombinant lentiviral particles with a CRTC2-targeting small hairpin RNA (shRNA) sequence were transfected into CRC cells to obtained shCRTC2 cell lines. Site-directed mutagenesis of Ser127, Ser238, and Ser245 cells were constructed by transfecting CRTC2 cDNA containing S127A, S238A, and S245A mutations into shCRTC2. Cell proliferation was measured by cell counting kit-8. Cell migration and invasion were examined by transwell assay. mRNA expression was assayed by qRT-PCR, and protein expression was determined by Western blot. Our results indicate that CRTC2 is overexpressed in CRC cells. Knockdown of CRTC2 inhibits the proliferation, migration, and invasion of CRC cells. When the phosphorylation of CRTC2 Ser238 decreases due to the lack of ERK2, the phosphorylation of Ser171 site increases. The proliferation, migration and invasion of CRC cells were inhibited, the nuclear aggregation of CRTC2 in the nucleus was reduced, and the interaction between CRTC2 and CREB was weaken. It is shown that the phosphorylation of CRTC2 Ser238 is important for CREB transcriptional activity. These findings may help in the identification of potentially new targets for CRC therapy.
Collapse
Affiliation(s)
- Yi Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qian Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Hanshuo Zhang
- GeneX health Life Co., Ltd, Beijing, People's Republic of China
| |
Collapse
|
11
|
Ling Q, Huang H, Han Y, Zhang C, Zhang X, Chen K, Wu L, Tang R, Zheng Z, Zheng S, Li L, Wang B. The tacrolimus-induced glucose homeostasis imbalance in terms of the liver: From bench to bedside. Am J Transplant 2020; 20:701-713. [PMID: 31654553 DOI: 10.1111/ajt.15665] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 01/25/2023]
Abstract
Tacrolimus (TAC), the mainstay of maintenance immunosuppressive agents, plays a crucial role in new-onset diabetes after transplant (NODAT). Previous studies investigating the diabetogenic effects of TAC have focused on the β cells of islets. In this study, we found that TAC contributed to NODAT through directly affecting hepatic metabolic homeostasis. In mice, TAC-induced hypoglycemia rather than hyperglycemia during starvation via suppressing gluconeogenetic genes, suggesting the limitation of fasting blood glucose in the diagnosis of NODAT. In addition, TAC caused hepatic insulin resistance and triglyceride accumulation through insulin receptor substrate (IRS)2/AKT and sterol regulatory element binding protein (SREBP1) signaling, respectively. Furthermore, we found a pivotal role of CREB-regulated transcription coactivator 2 (CRTC2) in TAC-induced metabolic disorders. The restoration of hepatic CRTC2 alleviated the metabolic disorders through its downstream molecules (eg, PCK1, IRS2, and SREBP1). Consistent with the findings from bench, low CRTC2 expression in graft hepatocytes was an independent risk factor for NODAT (odds ratio = 2.692, P = .023, n = 135). Integrating grafts' CRTC2 score into the clinical model could significantly increase the predictive capacity (areas under the receiver operating characteristic curve: 0.71 vs 0.79, P = .048). Taken together, in addition to its impact on pancreatic cells, TAC induces "hematogenous diabetes" via CRTC2 signaling. Liver-targeted management may be of help to prevent or heal TAC-associated diabetes.
Collapse
Affiliation(s)
- Qi Ling
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, China
| | - Haitao Huang
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, China
| | - Yuqiu Han
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- State Key Lab for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenzhi Zhang
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, China
| | - Xueyou Zhang
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, China
| | - Kangchen Chen
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, China
| | - Li Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- State Key Lab for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiqi Tang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- State Key Lab for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhipeng Zheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- State Key Lab for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shusen Zheng
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- State Key Lab for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baohong Wang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- State Key Lab for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Smith LIF, Huang V, Olah M, Trinh L, Liu Y, Hazell G, Conway-Campbell B, Zhao Z, Martinez A, Lefrançois-Martinez AM, Lightman S, Spiga F, Aguilera G. Involvement of CREB-regulated transcription coactivators (CRTC) in transcriptional activation of steroidogenic acute regulatory protein (Star) by ACTH. Mol Cell Endocrinol 2020; 499:110612. [PMID: 31604124 PMCID: PMC6899503 DOI: 10.1016/j.mce.2019.110612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/06/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022]
Abstract
Studies in vivo have suggested the involvement of CREB-regulated transcription coactivator (CRTC)2 on ACTH-induced transcription of the key steroidogenic protein, Steroidogenic Acute Regulatory (StAR). The present study uses two ACTH-responsive adrenocortical cell lines, to examine the role of CRTC on Star transcription. Here we show that ACTH-induced Star primary transcript, or heteronuclear RNA (hnRNA), parallels rapid increases in nuclear levels of the 3 isoforms of CRTC; CRTC1, CRTC2 and CRTC3. Furthermore, ACTH promotes recruitment of CRTC2 and CRTC3 by the Star promoter and siRNA knockdown of either CRTC3 or CRTC2 attenuates the increases in ACTH-induced Star hnRNA. Using pharmacological inhibitors of PKA, MAP kinase and calcineurin, we show that the effects of ACTH on Star transcription and CRTC nuclear translocation depend predominantly on the PKA pathway. The data provides evidence that CRTC2 and CRTC3, contribute to activation of Star transcription by ACTH, and that PKA/CRTC-dependent pathways are part of the multifactorial mechanisms regulating Star transcription.
Collapse
Affiliation(s)
- Lorna I F Smith
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA; Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK.
| | - Victoria Huang
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Mark Olah
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Loc Trinh
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Ying Liu
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Georgina Hazell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | - Becky Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | - Zidong Zhao
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | - Antoine Martinez
- Génétique Reproduction & Développement, CNRS UMR 6293, Inserm U1103, Université Clermont Auvergne, 63001, Clermont-Ferrand, France
| | - Anne-Marie Lefrançois-Martinez
- Génétique Reproduction & Développement, CNRS UMR 6293, Inserm U1103, Université Clermont Auvergne, 63001, Clermont-Ferrand, France
| | - Stafford Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | - Francesca Spiga
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | - Greti Aguilera
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| |
Collapse
|
13
|
Ji X, Wang S, Tang H, Zhang Y, Zhou F, Zhang L, Zhu Q, Zhu K, Liu Q, Liu Y, Wang X, Zhou L. PPP1R3C mediates metformin-inhibited hepatic gluconeogenesis. Metabolism 2019; 98:62-75. [PMID: 31181215 DOI: 10.1016/j.metabol.2019.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/15/2019] [Accepted: 06/05/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Metformin has been widely used to alleviate hyperglycemia in patients with type 2 diabetes mainly via suppressing hepatic gluconeogenesis. However, the underlying mechanism remains incompletely clear. Here, we aimed to explore the role of PPP1R3C in metformin-mediated inhibition of hepatic gluconeogenesis. METHODS The differentially expressed genes in primary mouse hepatocytes incubated with 8-Br-cAMP and metformin were analyzed by microarrays. Hepatic glucose production and gluconeogenic gene expressions were detected after adenovirus-mediated overexpression or silence of PPP1R3C in vitro and in vivo. The phosphorylation level and location of transducer of regulated CREB activity 2 (TORC2) were determined by Western blot and immunofluorescence. RESULTS Metformin and adenovirus-mediated activation of AMPK suppressed 8-Br-cAMP-stimulated Ppp1r3c mRNA expression in primary mouse hepatocytes. Overexpression of PPP1R3C in primary mouse hepatocytes or the livers of wild-type mice promoted hepatic glucose production and gluconeogenic gene expressions. On the contrary, adenovirus-mediated knockdown of PPP1R3C in primary mouse hepatocytes decreased hepatic gluconeogenesis, with the suppression of cAMP-stimulated gluconeogenic gene expressions and TORC2 dephosphorylation. Notably, Ppp1r3c expression was increased in the liver of db/db mice. After PPP1R3C silence in the livers of wild-type and db/db mice, blood glucose levels and hepatic glucose production were markedly lowered, with decreased expressions of key gluconeogenic enzymes and transcript factors as well as liver glycogen content. CONCLUSION Metformin-activated AMPK decreases hepatic PPP1R3C expression, leading to the suppression of hepatic gluconeogenesis through blocking cAMP-stimulated TORC2 dephosphorylation. Hepatic specific silence of PPP1R3C provides a promising therapeutic strategy for type 2 diabetes.
Collapse
Affiliation(s)
- Xueying Ji
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Shushu Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Hongju Tang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China; The Department of Geriatrics, The People's of Wenshan Prefecture, Wenshan 663099, PR China
| | - Yuqing Zhang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Feiye Zhou
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Linlin Zhang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Qin Zhu
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Kecheng Zhu
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Qianqian Liu
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Yun Liu
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | - Xiao Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China.
| | - Libin Zhou
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China.
| |
Collapse
|
14
|
Yun CY, Hong SD, Lee YH, Lee J, Jung DE, Kim GH, Kim SH, Jung JK, Kim KH, Lee H, Hong JT, Han SB, Kim Y. Nuclear Entry of CRTC1 as Druggable Target of Acquired Pigmentary Disorder. Am J Cancer Res 2019; 9:646-660. [PMID: 30809299 PMCID: PMC6376463 DOI: 10.7150/thno.30276] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/21/2018] [Indexed: 12/29/2022] Open
Abstract
Rationale: SOX10 (SRY-related HMG-box 10) and MITF-M (microphthalmia-associated transcription factor M) restrict the expression of melanogenic genes, such as TYR (tyrosinase), in melanocytes. DACE (diacetylcaffeic acid cyclohexyl ester) inhibits melanin production in α-MSH (α-melanocyte stimulating hormone)-activated B16-F0 melanoma cells. In this study, we evaluated the antimelanogenic activity of DACE in vivo and elucidated the molecular basis of its action. Methods: We employed melanocyte cultures and hyperpigmented skin samples for pigmentation assays, and applied chromatin immunoprecipitation, immunoblotting, RT-PCR or siRNA-based knockdown for mechanistic analyses. Results: Topical treatment with DACE mitigated UV-B-induced hyperpigmentation in the skin with attenuated expression of MITF-M and TYR. DACE also inhibited melanin production in α-MSH- or ET-1 (endothelin 1)-activated melanocyte cultures. As a mechanism, DACE blocked the nuclear import of CRTC1 (CREB-regulated co-activator 1) in melanocytes. DACE resultantly inhibited SOX10 induction, and suppressed the transcriptional abilities of CREB/CRTC1 heterodimer and SOX10 at MITF-M promoter, thereby ameliorating facultative melanogenesis. Furthermore, this study unveiled new issues in melanocyte biology that i) KPNA1 (Impα5) escorted CRTC1 as a cargo across the nuclear envelope, ii) SOX10 was inducible in the melanogenic process, and iii) CRTC1 could direct SOX10 induction at the transcription level. Conclusion: We propose the targeting of CRTC1 as a unique strategy in the treatment of acquired pigmentary disorders.
Collapse
|
15
|
Sonntag T, Ostojić J, Vaughan JM, Moresco JJ, Yoon YS, Yates JR, Montminy M. Mitogenic Signals Stimulate the CREB Coactivator CRTC3 through PP2A Recruitment. iScience 2018; 11:134-145. [PMID: 30611118 PMCID: PMC6317279 DOI: 10.1016/j.isci.2018.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/12/2018] [Accepted: 12/13/2018] [Indexed: 11/18/2022] Open
Abstract
The second messenger 3',5'-cyclic adenosine monophosphate (cAMP) stimulates gene expression via the cAMP-regulated transcriptional coactivator (CRTC) family of cAMP response element-binding protein coactivators. In the basal state, CRTCs are phosphorylated by salt-inducible kinases (SIKs) and sequestered in the cytoplasm by 14-3-3 proteins. cAMP signaling inhibits the SIKs, leading to CRTC dephosphorylation and nuclear translocation. Here we show that although all CRTCs are regulated by SIKs, their interactions with Ser/Thr-specific protein phosphatases are distinct. CRTC1 and CRTC2 associate selectively with the calcium-dependent phosphatase calcineurin, whereas CRTC3 interacts with B55 PP2A holoenzymes via a conserved PP2A-binding region (amino acids 380-401). CRTC3-PP2A complex formation was induced by phosphorylation of CRTC3 at S391, facilitating the subsequent activation of CRTC3 by dephosphorylation at 14-3-3 binding sites. As stimulation of mitogenic pathways promoted S391 phosphorylation via the activation of ERKs and CDKs, our results demonstrate how a ubiquitous phosphatase enables cross talk between growth factor and cAMP signaling pathways at the level of a transcriptional coactivator.
Collapse
Affiliation(s)
- Tim Sonntag
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jelena Ostojić
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Joan M Vaughan
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - James J Moresco
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Young-Sil Yoon
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Marc Montminy
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
16
|
Wein MN, Foretz M, Fisher DE, Xavier RJ, Kronenberg HM. Salt-Inducible Kinases: Physiology, Regulation by cAMP, and Therapeutic Potential. Trends Endocrinol Metab 2018; 29:723-735. [PMID: 30150136 PMCID: PMC6151151 DOI: 10.1016/j.tem.2018.08.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/02/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022]
Abstract
Salt-inducible kinases (SIKs) represent a subfamily of AMP-activated protein kinase (AMPK) family kinases. Initially named because SIK1 (the founding member of this kinase family) expression is regulated by dietary salt intake in the adrenal gland, it is now apparent that a major biological role of these kinases is to control gene expression in response to extracellular cues that increase intracellular levels of cAMP. Here, we review four physiologically relevant examples of how cAMP signaling impinges upon SIK cellular function. By focusing on examples of cAMP-mediated SIK regulation in gut myeloid cells, bone, liver, and skin, we highlight recent advances in G protein-coupled receptor (GPCR) signal transduction. New knowledge regarding the role of SIKs in GPCR signaling has led to therapeutic applications of novel small molecule SIK inhibitors.
Collapse
Affiliation(s)
- Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Marc Foretz
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ramnik J Xavier
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Henry M Kronenberg
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Yun CY, Mi Ko S, Pyo Choi Y, Kim BJ, Lee J, Mun Kim J, Kim JY, Song JY, Kim SH, Hwang BY, Tae Hong J, Han SB, Kim Y. α-Viniferin Improves Facial Hyperpigmentation via Accelerating Feedback Termination of cAMP/PKA-Signaled Phosphorylation Circuit in Facultative Melanogenesis. Theranostics 2018; 8:2031-2043. [PMID: 29556371 PMCID: PMC5858515 DOI: 10.7150/thno.24385] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 01/24/2018] [Indexed: 12/14/2022] Open
Abstract
Rationale: cAMP up-regulates microphthalmia-associated transcription factor subtype M (MITF-M) and tyrosinase (Tyro) in the generation of heavily pigmented melanosomes. Here, we communicate a therapeutic mechanism of hyperpigmented disorder by α-viniferin, an active constituent of Caragana sinica. Methods: We used cAMP-elevated melanocyte cultures or facial hyperpigmented patches for pigmentation assays, and applied immunoprecipitation, immunobloting, RT-PCR or reporter gene for elucidation of the antimelanogenic mechanism. Results:C. sinica or α-viniferin inhibited melanin production in α-melanocyte-stimulating hormone (α-MSH)-, histamine- or cell-permeable cAMP-activated melanocyte cultures. Moreover, topical application with C. sinica containing α-viniferin, a standard in quality control, decreased melanin index on facial melasma and freckles in patients. As a molecular basis, α-viniferin accelerated protein kinase A (PKA) inactivation via the reassociation between catalytic and regulatory subunits in cAMP-elevated melanocytes, a feedback loop in the melanogenic process. α-Viniferin resultantly inhibited cAMP/PKA-signaled phosphorylation of cAMP-responsive element-binding protein (CREB) coupled with dephosphorylation of cAMP-regulated transcriptional co-activator 1 (CRTC1), thus down-regulating expression of MITF-M or Tyro gene with decreased melanin pigmentation. Conclusion: This study assigned PKA inactivation, a feedback termination in cAMP-induced facultative melanogenesis, as a putative target of α-viniferin in the treatment of melanocyte-specific hyperpigmented disorder. Finally, C. sinica containing α-viniferin was approved as an antimelanogenic agent with topical application in skin hyperpigmentation.
Collapse
|
18
|
Gao WW, Tang HMV, Cheng Y, Chan CP, Chan CP, Jin DY. Suppression of gluconeogenic gene transcription by SIK1-induced ubiquitination and degradation of CRTC1. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:211-223. [PMID: 29408765 DOI: 10.1016/j.bbagrm.2018.01.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 12/21/2022]
Abstract
CRTCs are a group of three transcriptional coactivators required for CREB-dependent transcription. CREB and CRTCs are critically involved in the regulation of various biological processes such as cell proliferation, metabolism, learning and memory. However, whether CRTC1 efficiently induces gluconeogenic gene expression and how CRTC1 is regulated by upstream kinase SIK1 remain to be understood. In this work, we demonstrated SIK1-induced phosphorylation, ubiquitination and degradation of CRTC1 in the context of the regulation of gluconeogenesis. CRTC1 protein was destabilized by SIK1 but not SIK2 or SIK3. This effect was likely mediated by phosphorylation at S155, S167, S188 and S346 residues of CRTC1 followed by K48-linked polyubiquitination and proteasomal degradation. Expression of gluconeogenic genes such as that coding for phosphoenolpyruvate carboxykinase was stimulated by CRTC1, but suppressed by SIK1. Depletion of CRTC1 protein also blocked forskolin-induced gluconeogenic gene expression, knockdown or pharmaceutical inhibition of SIK1 had the opposite effect. Finally, SIK1-induced ubiquitination of CRTC1 was mediated by RFWD2 ubiquitin ligase at a site not equivalent to K628 in CRTC2. Taken together, our work reveals a regulatory circuit in which SIK1 suppresses gluconeogenic gene transcription by inducing ubiquitination and degradation of CRTC1. Our findings have implications in the development of new antihyperglycemic agents.
Collapse
Affiliation(s)
- Wei-Wei Gao
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hei-Man Vincent Tang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yun Cheng
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ching-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
19
|
Li Y, Wang L, Zhou L, Song Y, Ma S, Yu C, Zhao J, Xu C, Gao L. Thyroid stimulating hormone increases hepatic gluconeogenesis via CRTC2. Mol Cell Endocrinol 2017; 446:70-80. [PMID: 28212844 DOI: 10.1016/j.mce.2017.02.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 10/20/2022]
Abstract
Epidemiological evidence indicates that thyroid stimulating hormone (TSH) is positively correlated with abnormal glucose levels. We previously reported that TSH has direct effects on gluconeogenesis. However, the underlying molecular mechanism remains unclear. In this study, we observed increased fasting blood glucose and glucose production in a mouse model of subclinical hypothyroidism (only elevated TSH levels). TSH acts via the classical cAMP/PKA pathway and CRTC2 regulates glucose homeostasis. Thus, we explore whether CRTC2 is involved in the process of TSH-induced gluconeogenesis. We show that TSH increases CRTC2 expression via the TSHR/cAMP/PKA pathway, which in turn upregulates hepatic gluconeogenic genes. Furthermore, TSH stimulates CRTC2 dephosphorylation and upregulates p-CREB (Ser133) in HepG2 cells. Silencing CRTC2 and CREB decreases the effect of TSH on PEPCK-luciferase, the rate-limiting enzyme of gluconeogenesis. Finally, the deletion of TSHR reduces the levels of the CRTC2:CREB complex in mouse livers. This study demonstrates that TSH activates CRTC2 via the TSHR/cAMP/PKA pathway, leading to the formation of a CRTC2:CREB complex and increases hepatic gluconeogenesis.
Collapse
Affiliation(s)
- Yujie Li
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, 324 Jing 5 Rd Jinan, Shandong 250021, PR China
| | - Laicheng Wang
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, 544 Jing 4 Rd Jinan, Shangdong 250021, PR China
| | - Lingyan Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, 324 Jing 5 Rd Jinan, Shandong 250021, PR China
| | - Yongfeng Song
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, 324 Jing 5 Rd Jinan, Shandong 250021, PR China
| | - Shizhan Ma
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, 324 Jing 5 Rd Jinan, Shandong 250021, PR China
| | - Chunxiao Yu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, 324 Jing 5 Rd Jinan, Shandong 250021, PR China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, 324 Jing 5 Rd Jinan, Shandong 250021, PR China
| | - Chao Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, 324 Jing 5 Rd Jinan, Shandong 250021, PR China.
| | - Ling Gao
- Institute of Endocrinology, Shandong Academy of Clinical Medicine, Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, 544 Jing 4 Rd Jinan, Shangdong 250021, PR China.
| |
Collapse
|
20
|
Müller TD, Finan B, Clemmensen C, DiMarchi RD, Tschöp MH. The New Biology and Pharmacology of Glucagon. Physiol Rev 2017; 97:721-766. [PMID: 28275047 DOI: 10.1152/physrev.00025.2016] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last two decades we have witnessed sizable progress in defining the role of gastrointestinal signals in the control of glucose and energy homeostasis. Specifically, the molecular basis of the huge metabolic benefits in bariatric surgery is emerging while novel incretin-based medicines based on endogenous hormones such as glucagon-like peptide 1 and pancreas-derived amylin are improving diabetes management. These and related developments have fostered the discovery of novel insights into endocrine control of systemic metabolism, and in particular a deeper understanding of the importance of communication across vital organs, and specifically the gut-brain-pancreas-liver network. Paradoxically, the pancreatic peptide glucagon has reemerged in this period among a plethora of newly identified metabolic macromolecules, and new data complement and challenge its historical position as a gut hormone involved in metabolic control. The synthesis of glucagon analogs that are biophysically stable and soluble in aqueous solutions has promoted biological study that has enriched our understanding of glucagon biology and ironically recruited glucagon agonism as a central element to lower body weight in the treatment of metabolic disease. This review summarizes the extensive historical record and the more recent provocative direction that integrates the prominent role of glucagon in glucose elevation with its under-acknowledged effects on lipids, body weight, and vascular health that have implications for the pathophysiology of metabolic diseases, and the emergence of precision medicines to treat metabolic diseases.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - B Finan
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - C Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - R D DiMarchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| |
Collapse
|
21
|
Escoubas CC, Silva-García CG, Mair WB. Deregulation of CRTCs in Aging and Age-Related Disease Risk. Trends Genet 2017; 33:303-321. [PMID: 28365140 DOI: 10.1016/j.tig.2017.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/15/2022]
Abstract
Advances in public health in the past century have seen a sharp increase in human life expectancy. With these changes have come an increased prevalence of age-related pathologies and health burdens in the elderly. Patient age is the biggest risk factor for multiple chronic conditions that often occur simultaneously within a single individual. An alternative to disease-centric therapeutic approaches is that of 'geroscience', which aims to define molecular mechanisms that link age to overall disease risk. One such mechanism is deregulation of CREB-regulated transcriptional coactivators (CRTCs). Initially identified for their role in modulating CREB transcription, the past 5 years has seen an expansion in knowledge of new cellular regulators and roles of CRTCs beyond CREB. CRTCs have been shown to modulate organismal aging in Caenorhabditis elegans and to impact on age-related diseases in humans. We discuss CRTC deregulation as a new driver of aging that integrates the link between age and disease risk.
Collapse
Affiliation(s)
- Caroline C Escoubas
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Carlos G Silva-García
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - William B Mair
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Lee J, Foong YH, Musaitif I, Tong T, Jefcoate C. Analysis of specific RNA in cultured cells through quantitative integration of q-PCR and N-SIM single cell FISH images: Application to hormonal stimulation of StAR transcription. Mol Cell Endocrinol 2016; 429:93-105. [PMID: 27091298 PMCID: PMC6181224 DOI: 10.1016/j.mce.2016.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/02/2016] [Accepted: 04/02/2016] [Indexed: 12/21/2022]
Abstract
The steroidogenic acute regulatory protein (StAR) has been proposed to serve as the switch that can turn on/off steroidogenesis. We investigated the events that facilitate dynamic StAR transcription in response to cAMP stimulation in MA-10 Leydig cells, focusing on splicing anomalies at StAR gene loci. We used 3' reverse primers in a single reaction to respectively quantify StAR primary (p-RNA), spliced (sp-RNA/mRNA), and extended 3' untranslated region (UTR) transcripts, which were quantitatively imaged by high-resolution fluorescence in situ hybridization (FISH). This approach delivers spatio-temporal resolution of initiation and splicing at single StAR loci, and transfers individual mRNA molecules to cytoplasmic sites. Gene expression was biphasic, initially showing slow splicing, transitioning to concerted splicing. The alternative 3.5-kb mRNAs were distinguished through the use of extended 3'UTR probes, which exhibited distinctive mitochondrial distribution. Combining quantitative PCR and FISH enables imaging of localization of RNA expression and analysis of RNA processing rates.
Collapse
Affiliation(s)
- Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States.
| | - Yee Hoon Foong
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Ibrahim Musaitif
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Tiegang Tong
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Colin Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States; Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, United States
| |
Collapse
|
23
|
Henriksson E, Säll J, Gormand A, Wasserstrom S, Morrice NA, Fritzen AM, Foretz M, Campbell DG, Sakamoto K, Ekelund M, Degerman E, Stenkula KG, Göransson O. SIK2 regulates CRTCs, HDAC4 and glucose uptake in adipocytes. J Cell Sci 2016; 128:472-86. [PMID: 25472719 DOI: 10.1242/jcs.153932] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Salt-inducible kinase 2 (SIK2) is an AMP-activated protein kinase (AMPK) related kinase abundantly expressed in adipose tissue. Our aim was to identify molecular targets and functions of SIK2 in adipocytes, and to address the role of PKA-mediated phosphorylation of SIK2 on Ser358. Modulation of SIK2 in adipocytes resulted in altered phosphorylation of CREB-regulated transcription co-activator 2 (CRTC2), CRTC3 and class IIa histone deacetylase 4 (HDAC4). Furthermore, CRTC2, CRTC3, HDAC4 and protein phosphatase 2A (PP2A) interacted with SIK2, and the binding of CRTCs and PP2A to wild-type but not Ser358Ala SIK2, was reduced by cAMP elevation. Silencing of SIK2 resulted in reduced GLUT4 (also known as SLC2A4) protein levels, whereas cells treated with CRTC2 or HDAC4 siRNA displayed increased levels of GLUT4. Overexpression or pharmacological inhibition of SIK2 resulted in increased and decreased glucose uptake, respectively. We also describe a SIK2–CRTC2–HDAC4 pathway and its regulation in human adipocytes, strengthening the physiological relevance of our findings. Collectively, we demonstrate that SIK2 acts directly on CRTC2, CRTC3 and HDAC4, and that the cAMP–PKA pathway reduces the interaction of SIK2 with CRTCs and PP2A. Downstream, SIK2 increases GLUT4 levels and glucose uptake in adipocytes.
Collapse
|
24
|
Yahara Y, Takemori H, Okada M, Kosai A, Yamashita A, Kobayashi T, Fujita K, Itoh Y, Nakamura M, Fuchino H, Kawahara N, Fukui N, Watanabe A, Kimura T, Tsumaki N. Pterosin B prevents chondrocyte hypertrophy and osteoarthritis in mice by inhibiting Sik3. Nat Commun 2016; 7:10959. [PMID: 27009967 PMCID: PMC4820810 DOI: 10.1038/ncomms10959] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/05/2016] [Indexed: 01/09/2023] Open
Abstract
Osteoarthritis is a common debilitating joint disorder. Risk factors for osteoarthritis include age, which is associated with thinning of articular cartilage. Here we generate chondrocyte-specific salt-inducible kinase 3 (Sik3) conditional knockout mice that are resistant to osteoarthritis with thickened articular cartilage owing to a larger chondrocyte population. We also identify an edible Pteridium aquilinum compound, pterosin B, as a Sik3 pathway inhibitor. We show that either Sik3 deletion or intraarticular injection of mice with pterosin B inhibits chondrocyte hypertrophy and protects cartilage from osteoarthritis. Collectively, our results suggest Sik3 regulates the homeostasis of articular cartilage and is a target for the treatment of osteoarthritis, with pterosin B as a candidate therapeutic.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, 2630, Sugitani, Toyama 930-0194, Japan
| | - Hiroshi Takemori
- Laboratory of Cell Signaling and Metabolic Disease, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Asagi, Saito, Ibaraki, Osaka 567-0085, Japan
| | - Minoru Okada
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Azuma Kosai
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akihiro Yamashita
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomohito Kobayashi
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kaori Fujita
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yumi Itoh
- Laboratory of Cell Signaling and Metabolic Disease, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Asagi, Saito, Ibaraki, Osaka 567-0085, Japan
| | - Masahiro Nakamura
- Genome/Epigenome Analysis Core Facility, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroyuki Fuchino
- Research Center for Medicinal Plant Resources, Tsukuba Division, National Institutes of Biomedical Innovation, Health and Nutrition, 1-2, Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Nobuo Kawahara
- Research Center for Medicinal Plant Resources, Tsukuba Division, National Institutes of Biomedical Innovation, Health and Nutrition, 1-2, Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Naoshi Fukui
- Graduate School of Arts and Sciences, Department of Life Sciences, The University of Tokyo, Komaba 3-8-1, Meguro-ku, Tokyo 153-8902, Japan
| | - Akira Watanabe
- Genome/Epigenome Analysis Core Facility, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoatsu Kimura
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, 2630, Sugitani, Toyama 930-0194, Japan
| | - Noriyuki Tsumaki
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
25
|
Malm HA, Mollet IG, Berggreen C, Orho-Melander M, Esguerra JLS, Göransson O, Eliasson L. Transcriptional regulation of the miR-212/miR-132 cluster in insulin-secreting β-cells by cAMP-regulated transcriptional co-activator 1 and salt-inducible kinases. Mol Cell Endocrinol 2016; 424:23-33. [PMID: 26797246 DOI: 10.1016/j.mce.2016.01.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/15/2015] [Accepted: 01/11/2016] [Indexed: 12/25/2022]
Abstract
MicroRNAs are central players in the control of insulin secretion, but their transcriptional regulation is poorly understood. Our aim was to investigate cAMP-mediated transcriptional regulation of the miR-212/miR-132 cluster and involvement of further upstream proteins in insulin secreting β-cells. cAMP induced by forskolin+IBMX or GLP-1 caused increased expression of miR-212/miR-132, and elevated phosphorylation of cAMP-response-element-binding-protein (CREB)/Activating-transcription-factor-1 (ATF1) and Salt-Inducible-Kinases (SIKs). CyclicAMP-Regulated Transcriptional Co-activator-1 (CRTC1) was concomitantly dephosphorylated and translocated to the nucleus. Silencing of miR-212/miR-132 reduced, and overexpression of miR-212 increased, glucose-stimulated insulin secretion. Silencing of CRTC1 expression resulted in decreased insulin secretion and miR-212/miR-132 expression, while silencing or inhibition of SIKs was associated with increased expression of the microRNAs and dephosphorylation of CRTC1. CRTC1 protein levels were reduced after silencing of miR-132, suggesting feed-back regulation. Our data propose cAMP-dependent co-regulation of miR-212/miR-132, in part mediated through SIK-regulated CRTC1, as an important factor for fine-tuned regulation of insulin secretion.
Collapse
Affiliation(s)
- Helena Anna Malm
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden; Lund University Diabetes Centre, Lund University, Unit of Diabetes and Cardiovascular Disease, Genetic Epidemiology, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Inês G Mollet
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden; Lund University Diabetes Centre, Lund University, Unit of Diabetes and Cardiovascular Disease, Genetic Epidemiology, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Christine Berggreen
- Lund University Diabetes Centre, Lund University, Protein Phosphorylation Research Unit, Dept. Experimental Medical Science, 221 84 Lund, Sweden
| | - Marju Orho-Melander
- Lund University Diabetes Centre, Lund University, Unit of Diabetes and Cardiovascular Disease, Genetic Epidemiology, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden
| | - Olga Göransson
- Lund University Diabetes Centre, Lund University, Protein Phosphorylation Research Unit, Dept. Experimental Medical Science, 221 84 Lund, Sweden
| | - Lena Eliasson
- Lund University Diabetes Centre, Lund University, Unit of Islet Cell Exocytosis, Dept. Clinical Sciences in Malmö, 205 02 Malmö, Sweden.
| |
Collapse
|
26
|
Hill MJ, Suzuki S, Segars JH, Kino T. CRTC2 Is a Coactivator of GR and Couples GR and CREB in the Regulation of Hepatic Gluconeogenesis. Mol Endocrinol 2016; 30:104-17. [PMID: 26652733 PMCID: PMC4695631 DOI: 10.1210/me.2015-1237] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/25/2015] [Indexed: 12/22/2022] Open
Abstract
Glucocorticoid hormones play essential roles in the regulation of gluconeogenesis in the liver, an adaptive response that is required for the maintenance of circulating glucose levels during fasting. Glucocorticoids do this by cooperating with glucagon, which is secreted from pancreatic islets to activate the cAMP-signaling pathway in hepatocytes. The cAMP-response element-binding protein (CREB)-regulated transcription coactivator 2 (CRTC2) is a coactivator known to be specific to CREB and plays a central role in the glucagon-mediated activation of gluconeogenesis in the early phase of fasting. We show here that CRTC2 also functions as a coactivator for the glucocorticoid receptor (GR). CRTC2 strongly enhances GR-induced transcriptional activity of glucocorticoid-responsive genes. CRTC2 physically interacts with the ligand-binding domain of the GR through a region spanning amino acids 561-693. Further, CRTC2 is required for the glucocorticoid-associated cooperative mRNA expression of the glucose-6-phosphatase, a rate-limiting enzyme for hepatic gluconeogenesis, by facilitating the attraction of GR and itself to its promoter region already occupied by CREB. CRTC2 is required for the maintenance of blood glucose levels during fasting in mice by enhancing the GR transcriptional activity on both the G6p and phosphoenolpyruvate carboxykinase (Pepck) genes. Finally, CRTC2 modulates the transcriptional activity of the progesterone receptor, indicating that it may influence the transcriptional activity of other steroid/nuclear receptors. Taken together, these results reveal that CRTC2 plays an essential role in the regulation of hepatic gluconeogenesis through coordinated regulation of the glucocorticoid/GR- and glucagon/CREB-signaling pathways on the key genes G6P and PEPCK.
Collapse
Affiliation(s)
- Micah J Hill
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| | - Shigeru Suzuki
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| | - James H Segars
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| | - Tomoshige Kino
- Program in Reproductive and Adult Endocrinology (M.J.H., S.S., J.H.S., T.K.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Division of Reproductive Endocrinology and Infertility (M.J.H.), Walter Reed National Military Medical Center, Bethesda, Maryland 20889; Department of Pediatrics (S.S.), Asahikawa Medical University, Asahikawa 078-8510, Japan; Division of Reproductive Sciences and Women's Health Research (J.H.S.), Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205; and Department of Experimental Therapeutics (T.K.), Division of Experimental Biology, Sidra Medical and Research Center, Doha 26999, Qatar
| |
Collapse
|
27
|
McTague J, Ferguson M, Chik CL, Ho AK. The adrenergic-regulated CRTC1 and CRTC2 phosphorylation and cellular distribution is independent of endogenous SIK1 in the male rat pinealocyte. Mol Cell Endocrinol 2015. [PMID: 26210066 DOI: 10.1016/j.mce.2015.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Salt inducible kinase 1 (SIK1) has been reported to repress cAMP-response element binding protein (CREB)-mediated gene transcription by causing the nuclear export of CREB-regulated transcription coactivators (CRTCs) through phosphorylation. Although the repressor role of SIK1 in suppressing the expression of arylalkylamine N-acetyltransferase, the enzyme that controls the daily rhythm in melatonin production in the rat pineal gland, has been established, whether SIK1 regulates the phosphorylation and localization of CRTC1 and CRTC2 in this tissue remains unclear. The present study found that overexpressing SIK1 in NE-stimulated rat pinealocytes could increase the phosphorylation of CRTC1 and CRTC2, reduced selectively the nuclear level of CRTC2 (but not that of CRTC1), and elevated the cytosolic levels of both CRTC1 and CRTC2. In contrast, transient knockdown of endogenous SIK1 had no effect on the phosphorylation or distribution of CRTC1 and CRTC2 in norepinephrine (NE)-stimulated pinealocytes. Our results also showed that adrenergic blockade during NE stimulation led to a rapid rephosphorylation and decline in the nucleus levels of CRTC1 and CRTC2; however SIK1 knockdown had no effect on this rapid rephosphorylation. Moreover, studies with kinase inhibitors revealed that kinase(s) sensitive to KT5823 appeared to be involved in this rapid rephosphorylation. Together, these results indicate that although overexpressing SIK1 can phosphorylate CRTC1 and CRTC2 in the NE-stimulated pinealocyte, the endogenous SIK1, in spite of its induction by NE, does not appear to be the main regulator of the phosphorylation and intracellular localization of these two coactivators.
Collapse
Affiliation(s)
- J McTague
- Department of Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - M Ferguson
- Department of Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - C L Chik
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - A K Ho
- Department of Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
28
|
Hogan MF, Ravnskjaer K, Matsumura S, Huising MO, Hull RL, Kahn SE, Montminy M. Hepatic Insulin Resistance Following Chronic Activation of the CREB Coactivator CRTC2. J Biol Chem 2015; 290:25997-6006. [PMID: 26342077 DOI: 10.1074/jbc.m115.679266] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Indexed: 11/06/2022] Open
Abstract
Under fasting conditions, increases in circulating concentrations of glucagon maintain glucose homeostasis via the induction of hepatic gluconeogenesis. Triggering of the cAMP pathway in hepatocytes stimulates the gluconeogenic program via the PKA-mediated phosphorylation of CREB and dephosphorylation of the cAMP-regulated CREB coactivators CRTC2 and CRTC3. In parallel, decreases in circulating insulin also increase gluconeogenic gene expression via the de-phosphorylation and activation of the forkhead transcription factor FOXO1. Hepatic gluconeogenesis is increased in insulin resistance where it contributes to the attendant hyperglycemia. Whether selective activation of the hepatic CREB/CRTC pathway is sufficient to trigger metabolic changes in other tissues is unclear, however. Modest hepatic expression of a phosphorylation-defective and therefore constitutively active CRTC2S171,275A protein increased gluconeogenic gene expression under fasting as well as feeding conditions. Circulating glucose concentrations were constitutively elevated in CRTC2S171,275A-expressing mice, leading to compensatory increases in circulating insulin concentrations that enhance FOXO1 phosphorylation. Despite accompanying decreases in FOXO1 activity, hepatic gluconeogenic gene expression remained elevated in CRTC2S171,275A mice, demonstrating that chronic increases in CRTC2 activity in the liver are indeed sufficient to promote hepatic insulin resistance and to disrupt glucose homeostasis.
Collapse
Affiliation(s)
- Meghan F Hogan
- From the Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, California 92037, Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, Washington 98108
| | - Kim Ravnskjaer
- From the Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, California 92037, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Shigenobu Matsumura
- From the Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, California 92037, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Oiwake-cho, Kitashirakawa, Sakyo-ku, Kyoto, 606-8502, Japan, and
| | - Mark O Huising
- From the Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, California 92037, Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California 95616
| | - Rebecca L Hull
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, Washington 98108
| | - Steven E Kahn
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, Washington 98108
| | - Marc Montminy
- From the Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, California 92037,
| |
Collapse
|
29
|
Lee J, Tong T, Takemori H, Jefcoate C. Stimulation of StAR expression by cAMP is controlled by inhibition of highly inducible SIK1 via CRTC2, a co-activator of CREB. Mol Cell Endocrinol 2015; 408:80-9. [PMID: 25662274 PMCID: PMC4417451 DOI: 10.1016/j.mce.2015.01.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/15/2015] [Accepted: 01/15/2015] [Indexed: 12/21/2022]
Abstract
In mouse steroidogenic cells the activation of cholesterol metabolism is mediated by steroidogenic acute regulatory protein (StAR). Here, we visualized a coordinated regulation of StAR transcription, splicing and post-transcriptional processing, which are synchronized by salt inducible kinase (SIK1) and CREB-regulated transcription coactivator (CRTC2). To detect primary RNA (pRNA), spliced primary RNA (Sp-RNA) and mRNA in single cells, we generated probe sets by using fluorescence in situ hybridization (FISH). These methods allowed us to address the nature of StAR gene expression and to visualize protein-nucleic acid interactions through direct detection. We show that SIK1 represses StAR expression in Y1 adrenal and MA10 testis cells through inhibition of processing mediated by CRTC2. Digital image analysis matches qPCR analyses of the total cell culture. Evidence is presented for spatially separate accumulation of StAR pRNA and Sp-RNA at the gene loci in the nucleus. These findings establish that cAMP, SIK and CRTC mediate StAR expression through activation of individual StAR gene loci.
Collapse
Affiliation(s)
- Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA
| | - Tiegang Tong
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA
| | | | - Colin Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, USA; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, USA; University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
30
|
Itoh Y, Sanosaka M, Fuchino H, Yahara Y, Kumagai A, Takemoto D, Kagawa M, Doi J, Ohta M, Tsumaki N, Kawahara N, Takemori H. Salt-inducible Kinase 3 Signaling Is Important for the Gluconeogenic Programs in Mouse Hepatocytes. J Biol Chem 2015; 290:17879-17893. [PMID: 26048985 DOI: 10.1074/jbc.m115.640821] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Indexed: 01/24/2023] Open
Abstract
Salt-inducible kinases (SIKs), members of the 5'-AMP-activated protein kinase (AMPK) family, are proposed to be important suppressors of gluconeogenic programs in the liver via the phosphorylation-dependent inactivation of the CREB-specific coactivator CRTC2. Although a dramatic phenotype for glucose metabolism has been found in SIK3-KO mice, additional complex phenotypes, dysregulation of bile acids, cholesterol, and fat homeostasis can render it difficult to discuss the hepatic functions of SIK3. The aim of this study was to examine the cell autonomous actions of SIK3 in hepatocytes. To eliminate systemic effects, we prepared primary hepatocytes and screened the small compounds suppressing SIK3 signaling cascades. SIK3-KO primary hepatocytes produced glucose more quickly after treatment with the cAMP agonist forskolin than the WT hepatocytes, which was accompanied by enhanced gluconeogenic gene expression and CRTC2 dephosphorylation. Reporter-based screening identified pterosin B as a SIK3 signaling-specific inhibitor. Pterosin B suppressed SIK3 downstream cascades by up-regulating the phosphorylation levels in the SIK3 C-terminal regulatory domain. When pterosin B promoted glucose production by up-regulating gluconeogenic gene expression in mouse hepatoma AML-12 cells, it decreased the glycogen content and stimulated an association between the glycogen phosphorylase kinase gamma subunit (PHKG2) and SIK3. PHKG2 phosphorylated the peptides with sequences of the C-terminal domain of SIK3. Here we found that the levels of active AMPK were higher both in the SIK3-KO hepatocytes and in pterosin B-treated AML-12 cells than in their controls. These results suggest that SIK3, rather than SIK1, SIK2, or AMPKs, acts as the predominant suppressor in gluconeogenic gene expression in the hepatocytes.
Collapse
Affiliation(s)
- Yumi Itoh
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Osaka, 567-0085, Japan
| | - Masato Sanosaka
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Osaka, 567-0085, Japan
| | - Hiroyuki Fuchino
- Research Center for Medicinal Plant Resources, Tsukuba Division, Ibaraki, 305-0843, Japan
| | - Yasuhito Yahara
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Ayako Kumagai
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Osaka, 567-0085, Japan
| | - Daisaku Takemoto
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Osaka, 567-0085, Japan; Department of Life Science and Biotechnology, Kansai University, Osaka 564-8680, Japan
| | - Mai Kagawa
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Osaka, 567-0085, Japan
| | - Junko Doi
- Department of Food and Nutrition, Senri Kinran University, Osaka, 565-0873 Japan
| | - Miho Ohta
- Department of Nutrition and Health, Faculty of Human Development, Soai University, Osaka, 559-0033, Japan
| | - Noriyuki Tsumaki
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Nobuo Kawahara
- Research Center for Medicinal Plant Resources, Tsukuba Division, Ibaraki, 305-0843, Japan
| | - Hiroshi Takemori
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Osaka, 567-0085, Japan.
| |
Collapse
|
31
|
Transcriptional coregulators: fine-tuning metabolism. Cell Metab 2014; 20:26-40. [PMID: 24794975 PMCID: PMC4079747 DOI: 10.1016/j.cmet.2014.03.027] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 02/28/2014] [Accepted: 03/18/2014] [Indexed: 12/21/2022]
Abstract
Metabolic homeostasis requires that cellular energy levels are adapted to environmental cues. This adaptation is largely regulated at the transcriptional level, through the interaction between transcription factors, coregulators, and the basal transcriptional machinery. Coregulators, which function as both metabolic sensors and transcriptional effectors, are ideally positioned to synchronize metabolic pathways to environmental stimuli. The balance between inhibitory actions of corepressors and stimulatory effects of coactivators enables the fine-tuning of metabolic processes. This tight regulation opens therapeutic opportunities to manage metabolic dysfunction by directing the activity of cofactors toward specific transcription factors, pathways, or cells/tissues, thereby restoring whole-body metabolic homeostasis.
Collapse
|
32
|
Abstract
The liver is a vital organ responsible for maintaining nutrient homeostasis. After a meal, insulin stimulates glycogen and lipid synthesis in the liver; in the fasted state, glucagon induces gluconeogenesis and ketogenesis, which produce glucose and ketone bodies for other tissues to use as energy sources. These metabolic changes involve spatiotemporally co-ordinated signaling cascades. O-linked β-N-acetylglucosamine (O-GlcNAc) modification has been recognized as a nutrient sensor and regulatory molecular switch. This review highlights mechanistic insights into spatiotemporal regulation of liver metabolism by O-GlcNAc modification and discusses its pathophysiological implications in insulin resistance, non-alcoholic fatty liver disease, and fibrosis.
Collapse
Affiliation(s)
- Kaisi Zhang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruonan Yin
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoyong Yang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- *Correspondence: Xiaoyong Yang, Yale University School of Medicine, 310 Cedar Street, BML 329C, New Haven, CT 06519, USA e-mail:
| |
Collapse
|
33
|
McTague J, Ferguson M, Chik CL, Ho AK. Sustained adrenergic stimulation is required for the nuclear retention of TORC1 in male rat pinealocytes. Endocrinology 2013; 154:3240-50. [PMID: 23751872 DOI: 10.1210/en.2013-1293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The process involved in relocation of the coactivator, transducer of regulated cAMP-regulated element-binding protein (TORC) to the cytoplasm, unlike its activation, is not well understood. Using cultured pineal cells prepared from male rats, we found that although both α- and β-adrenergic stimulation could cause TORC1 dephosphorylation, only α-adrenergic stimulation was effective in the norepinephrine (NE)-mediated translocation of TORC1 into the nucleus. In contrast, blockade of either the α- or the β-adrenergic receptor after NE stimulation was effective in causing the rephosphorylation and rapid relocation of TORC1 into the cytoplasm. Studies with phosphoprotein phosphatase (PP) inhibitors indicated that although both PP2A and PP2B could dephosphorylate TORC1, only PP2B could cause translocation into the nucleus. However, after NE stimulation, treatment with either PP2A or PP2B inhibitors could cause the rephosphorylation and cytoplasmic relocation of TORC1. These results indicate a requirement of continuous activation of both α- and β-adrenergic receptors as well as PP2A and PP2B activities for the nuclear retention of TORC1 during NE stimulation. Knockdown of salt-inducible kinase 1 (SIK1) had no effect on the phosphorylation or localization of TORC1. Although overexpressing SIK1 could induce TORC1 phosphorylation in the nucleus, it did not reduce TORC1 level in the nucleus, indicating that SIK1-mediated TORC1 phosphorylation may not be sufficient for its relocation into the cytoplasm. Together, these results demonstrate that, in the rat pineal gland, different mechanisms are involved in regulating the nuclear entry and exit of TORC1 and that the SIK1-mediated phosphorylation of TORC1 may not lead to its nuclear exit.
Collapse
MESH Headings
- Adrenergic Agonists/pharmacology
- Adrenergic Antagonists/pharmacology
- Animals
- Biological Transport/drug effects
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Cytoplasm/drug effects
- Cytoplasm/metabolism
- Enzyme Inhibitors/pharmacology
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/metabolism
- Male
- Mechanistic Target of Rapamycin Complex 1
- Multiprotein Complexes/metabolism
- Nerve Tissue Proteins/agonists
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/metabolism
- Neuroendocrine Cells/cytology
- Neuroendocrine Cells/drug effects
- Neuroendocrine Cells/metabolism
- Norepinephrine/metabolism
- Phosphorylation/drug effects
- Pineal Gland/cytology
- Pineal Gland/drug effects
- Pineal Gland/metabolism
- Protein Phosphatase 2/antagonists & inhibitors
- Protein Phosphatase 2/metabolism
- Protein Processing, Post-Translational/drug effects
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, alpha/chemistry
- Receptors, Adrenergic, alpha/metabolism
- Receptors, Adrenergic, beta/chemistry
- Receptors, Adrenergic, beta/metabolism
- TOR Serine-Threonine Kinases/metabolism
Collapse
Affiliation(s)
- J McTague
- Department of Physiology, 7-26 Medical Sciences Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | | | | | | |
Collapse
|
34
|
Comandini A, Naro C, Adamo R, Akbar AN, Lanna A, Bonmassar E, Franzese O. Molecular mechanisms involved in HIV-1-Tat mediated inhibition of telomerase activity in human CD4+ T lymphocytes. Mol Immunol 2013; 54:181-92. [DOI: 10.1016/j.molimm.2012.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/29/2012] [Accepted: 12/04/2012] [Indexed: 12/16/2022]
|
35
|
Neurokinin-1 receptor agonists bias therapeutic dendritic cells to induce type 1 immunity by licensing host dendritic cells to produce IL-12. Blood 2013; 121:2923-33. [PMID: 23365459 DOI: 10.1182/blood-2012-07-446054] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Substance-P and hemokinin-1 are proinflammatory neuropeptides with potential to promote type 1 immunity through agonistic binding to neurokinin-1 receptor (NK1R). Dendritic cells (DCs) are professional antigen-presenting cells that initiate and regulate the outcome of innate and adaptive immune responses. Immunostimulatory DCs are highly desired for the development of positive immunization techniques. DCs express functional NK1R; however, regardless of their potential DC-stimulatory function, the ability of NK1R agonists to promote immunostimulatory DCs remains unexplored. Here, we demonstrate that NK1R signaling activates therapeutic DCs capable of biasing type 1 immunity by inhibition of interleukin-10 (IL-10) synthesis and secretion, without affecting their low levels of IL-12 production. The potent type 1 effector immune response observed following cutaneous administration of NK1R-signaled DCs required their homing in skin-draining lymph nodes (sDLNs) where they induced inflammation and licensed endogenous-conventional sDLN-resident and -recruited inflammatory DCs to secrete IL-12. Our data demonstrate that NK1R signaling promotes immunostimulatory DCs, and provide relevant insight into the mechanisms used by neuromediators to regulate innate and adaptive immune responses.
Collapse
|
36
|
Hallenborg P, Feddersen S, Francoz S, Murano I, Sundekilde U, Petersen RK, Akimov V, Olson MV, Lozano G, Cinti S, Gjertsen BT, Madsen L, Marine JC, Blagoev B, Kristiansen K. Mdm2 controls CREB-dependent transactivation and initiation of adipocyte differentiation. Cell Death Differ 2012; 19:1381-1389. [PMID: 22388350 PMCID: PMC3392627 DOI: 10.1038/cdd.2012.15] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 01/23/2012] [Accepted: 01/24/2012] [Indexed: 12/30/2022] Open
Abstract
The role of the E3 ubiquitin ligase murine double minute 2 (Mdm2) in regulating the stability of the p53 tumor suppressor is well documented. By contrast, relatively little is known about p53-independent activities of Mdm2 and the role of Mdm2 in cellular differentiation. Here we report a novel role for Mdm2 in the initiation of adipocyte differentiation that is independent of its ability to regulate p53. We show that Mdm2 is required for cAMP-mediated induction of CCAAT/enhancer-binding protein δ (C/EBPδ) expression by facilitating recruitment of the cAMP regulatory element-binding protein (CREB) coactivator, CREB-regulated transcription coactivator (Crtc2)/TORC2, to the c/ebpδ promoter. Our findings reveal an unexpected role for Mdm2 in the regulation of CREB-dependent transactivation during the initiation of adipogenesis. As Mdm2 is able to promote adipogenesis in the myoblast cell line C2C12, it is conceivable that Mdm2 acts as a switch in cell fate determination.
Collapse
Affiliation(s)
- P Hallenborg
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
- Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - S Feddersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - S Francoz
- Laboratory for Molecular Cancer Biology, VIB-KU Leuven, 3000 Leuven, Belgium
| | - I Murano
- Department of Experimental and Clinical Medicine, Universitá Politecnica della Marche, 60121 Ancona, Italy
| | - U Sundekilde
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - R K Petersen
- Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - V Akimov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - M V Olson
- Department of Genetics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - G Lozano
- Department of Genetics, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - S Cinti
- Department of Experimental and Clinical Medicine, Universitá Politecnica della Marche, 60121 Ancona, Italy
| | - B T Gjertsen
- Institute of Medicine, University of Bergen, Bergen 5021, Norway
| | - L Madsen
- Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen 5021, Norway
| | - J-C Marine
- Laboratory for Molecular Cancer Biology, VIB-KU Leuven, 3000 Leuven, Belgium
| | - B Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - K Kristiansen
- Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
37
|
Uebi T, Itoh Y, Hatano O, Kumagai A, Sanosaka M, Sasaki T, Sasagawa S, Doi J, Tatsumi K, Mitamura K, Morii E, Aozasa K, Kawamura T, Okumura M, Nakae J, Takikawa H, Fukusato T, Koura M, Nish M, Hamsten A, Silveira A, Bertorello AM, Kitagawa K, Nagaoka Y, Kawahara H, Tomonaga T, Naka T, Ikegawa S, Tsumaki N, Matsuda J, Takemori H. Involvement of SIK3 in glucose and lipid homeostasis in mice. PLoS One 2012; 7:e37803. [PMID: 22662228 PMCID: PMC3360605 DOI: 10.1371/journal.pone.0037803] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/24/2012] [Indexed: 01/20/2023] Open
Abstract
Salt-inducible kinase 3 (SIK3), an AMP-activated protein kinase-related kinase, is induced in the murine liver after the consumption of a diet rich in fat, sucrose, and cholesterol. To examine whether SIK3 can modulate glucose and lipid metabolism in the liver, we analyzed phenotypes of SIK3-deficent mice. Sik3(-/-) mice have a malnourished the phenotype (i.e., lipodystrophy, hypolipidemia, hypoglycemia, and hyper-insulin sensitivity) accompanied by cholestasis and cholelithiasis. The hypoglycemic and hyper-insulin-sensitive phenotypes may be due to reduced energy storage, which is represented by the low expression levels of mRNA for components of the fatty acid synthesis pathways in the liver. The biliary disorders in Sik3(-/-) mice are associated with the dysregulation of gene expression programs that respond to nutritional stresses and are probably regulated by nuclear receptors. Retinoic acid plays a role in cholesterol and bile acid homeostasis, wheras ALDH1a which produces retinoic acid, is expressed at low levels in Sik3(-/-) mice. Lipid metabolism disorders in Sik3(-/-) mice are ameliorated by the treatment with 9-cis-retinoic acid. In conclusion, SIK3 is a novel energy regulator that modulates cholesterol and bile acid metabolism by coupling with retinoid metabolism, and may alter the size of energy storage in mice.
Collapse
Affiliation(s)
- Tatsuya Uebi
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Yumi Itoh
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Osamu Hatano
- Department of Anatomy, Nara Medical University, Nara, Japan
| | - Ayako Kumagai
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
- Department of Life Science and Biotechnology, Kansai University, Suita, Osaka, Japan
| | - Masato Sanosaka
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoru Sasagawa
- Department of Bone and Cartilage Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Junko Doi
- Food and Nutrition, Senri Kinran University, Osaka, Japan
| | - Keita Tatsumi
- Department of Laboratory Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kuniko Mitamura
- Faculty of Pharmaceutical Sciences, Kinki University, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Katsuyuki Aozasa
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohiro Kawamura
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Meinoshin Okumura
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Nakae
- Frontier Medicine on Metabolic Syndrome, Keio University School of Medicine, Tokyo, Japan
| | - Hajime Takikawa
- Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Toshio Fukusato
- Department of Pathology, Teikyo University School of Medicine, Tokyo, Japan
| | - Minako Koura
- Animal Models for Human Diseases, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Mayumi Nish
- Department of Anatomy, Nara Medical University, Nara, Japan
| | - Anders Hamsten
- Cardiovascular Genetics and Genomics, Atherosclerosis Research Unit, Karolinska Institutet, CMM, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Angela Silveira
- Cardiovascular Genetics and Genomics, Atherosclerosis Research Unit, Karolinska Institutet, CMM, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Alejandro M. Bertorello
- Membrane Signaling Networks, Atherosclerosis Research Unit, Karolinska Institutet, CMM, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Kazuo Kitagawa
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuo Nagaoka
- Department of Life Science and Biotechnology, Kansai University, Suita, Osaka, Japan
| | - Hidehisa Kawahara
- Department of Life Science and Biotechnology, Kansai University, Suita, Osaka, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Tetsuji Naka
- Laboratory for Immune Signal, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Shigeo Ikegawa
- Faculty of Pharmaceutical Sciences, Kinki University, Osaka, Japan
| | - Noriyuki Tsumaki
- Department of Bone and Cartilage Biology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Junichiro Matsuda
- Animal Models for Human Diseases, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Hiroshi Takemori
- Laboratory of Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
- * E-mail:
| |
Collapse
|
38
|
Peeters A, Fraisl P, van den Berg S, Ver Loren van Themaat E, Van Kampen A, Rider MH, Takemori H, van Dijk KW, Van Veldhoven PP, Carmeliet P, Baes M. Carbohydrate metabolism is perturbed in peroxisome-deficient hepatocytes due to mitochondrial dysfunction, AMP-activated protein kinase (AMPK) activation, and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) suppression. J Biol Chem 2011; 286:42162-42179. [PMID: 22002056 DOI: 10.1074/jbc.m111.299727] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hepatic peroxisomes are essential for lipid conversions that include the formation of mature conjugated bile acids, the degradation of branched chain fatty acids, and the synthesis of docosahexaenoic acid. Through unresolved mechanisms, deletion of functional peroxisomes from mouse hepatocytes (L-Pex5(-/-) mice) causes severe structural and functional abnormalities at the inner mitochondrial membrane. We now demonstrate that the peroxisomal and mitochondrial anomalies trigger energy deficits, as shown by increased AMP/ATP and decreased NAD(+)/NADH ratios. This causes suppression of gluconeogenesis and glycogen synthesis and up-regulation of glycolysis. As a consequence, L-Pex5(-/-) mice combust more carbohydrates resulting in lower body weights despite increased food intake. The perturbation of carbohydrate metabolism does not require a long term adaptation to the absence of functional peroxisomes as similar metabolic changes were also rapidly induced by acute elimination of Pex5 via adenoviral administration of Cre. Despite its marked activation, peroxisome proliferator-activated receptor α (PPARα) was not causally involved in these metabolic perturbations, because all abnormalities still manifested when peroxisomes were eliminated in a peroxisome proliferator-activated receptor α null background. Instead, AMP-activated kinase activation was responsible for the down-regulation of glycogen synthesis and induction of glycolysis. Remarkably, PGC-1α was suppressed despite AMP-activated kinase activation, a paradigm not previously reported, and they jointly contributed to impaired gluconeogenesis. In conclusion, lack of functional peroxisomes from hepatocytes results in marked disturbances of carbohydrate homeostasis, which are consistent with adaptations to an energy deficit. Because this is primarily due to impaired mitochondrial ATP production, these L-Pex5-deficient livers can also be considered as a model for secondary mitochondrial hepatopathies.
Collapse
Affiliation(s)
- Annelies Peeters
- Laboratory of Cell Metabolism, Department of Pharmaceutical Sciences, University of Leuven, B-3000 Leuven, Belgium
| | - Peter Fraisl
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Flanders Institute of Biotechnology, B-3000 Leuven, Belgium; Department of Human Genetics, Leiden University Medical Center, NL-2333 ZA Leiden, The Netherlands
| | - Sjoerd van den Berg
- Department of Human Genetics, Leiden University Medical Center, NL-2333 ZA Leiden, The Netherlands
| | | | - Antoine Van Kampen
- Laboratory of Bioinformatics, Academic Medical Center, NL-1105 AZ Amsterdam, The Netherlands
| | - Mark H Rider
- Université Catholique de Louvain and de Duve Institute, B-1200 Brussels, Belgium
| | - Hiroshi Takemori
- Laboratory of Cell Signaling and Metabolism, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, NL-2333 ZA Leiden, The Netherlands
| | - Paul P Van Veldhoven
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Molecular Cell Biology, University of Leuven, B-3000 Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, University of Leuven, B-3000 Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Flanders Institute of Biotechnology, B-3000 Leuven, Belgium
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical Sciences, University of Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
39
|
A potent inhibitor of SIK2, 3, 3', 7-trihydroxy-4'-methoxyflavon (4'-O-methylfisetin), promotes melanogenesis in B16F10 melanoma cells. PLoS One 2011; 6:e26148. [PMID: 22022544 PMCID: PMC3192784 DOI: 10.1371/journal.pone.0026148] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/20/2011] [Indexed: 11/24/2022] Open
Abstract
Flavonoids, which are plant polyphenols, are now widely used in supplements and cosmetics. Here, we report that 4′-methylflavonoids are potent inducers of melanogenesis in B16F10 melanoma cells and in mice. We recently identified salt inducible kinase 2 (SIK2) as an inhibitor of melanogenesis via the suppression of the cAMP-response element binding protein (CREB)-specific coactivator 1 (TORC1). Using an in vitro kinase assay targeting SIK2, we identified fisetin as a candidate inhibitor, possibly being capable of promoting melanogenesis. However, fisetin neither inhibited the CREB-inhibitory activity of SIK2 nor promoted melanogenesis in B16F10 melanoma cells. Conversely, mono-methyl-flavonoids, such as diosmetin (4′-O-metlylluteolin), efficiently inhibited SIK2 and promoted melanogenesis in this cell line. The cAMP-CREB system is impaired in Ay/a mice and these mice have yellow hair as a result of pheomelanogenesis, while Sik2+/−; Ay/a mice also have yellow hair, but activate eumelanogenesis when they are exposed to CREB stimulators. Feeding Sik2+/−; Ay/a mice with diets supplemented with fisetin resulted in their hair color changing to brown, and metabolite analysis suggested the presence of mono-methylfisetin in their feces. Thus, we decided to synthesize 4′-O-methylfisetin (4′MF) and found that 4′MF strongly induced melanogenesis in B16F10 melanoma cells, which was accompanied by the nuclear translocation of TORC1, and the 4′-O-methylfisetin-induced melanogenic programs were inhibited by the overexpression of dominant negative TORC1. In conclusion, compounds that modulate SIK2 cascades are helpful to regulate melanogenesis via TORC1 without affecting cAMP levels, and the combined analysis of Sik2+/− mice and metabolites from these mice is an effective strategy to identify beneficial compounds to regulate CREB activity in vivo.
Collapse
|
40
|
Altarejos JY, Montminy M. CREB and the CRTC co-activators: sensors for hormonal and metabolic signals. Nat Rev Mol Cell Biol 2011; 12:141-51. [PMID: 21346730 DOI: 10.1038/nrm3072] [Citation(s) in RCA: 792] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cyclic AMP-responsive element-binding protein (CREB) is phosphorylated in response to a wide variety of signals, yet target gene transcription is only increased in a subset of cases. Recent studies indicate that CREB functions in concert with a family of latent cytoplasmic co-activators called cAMP-regulated transcriptional co-activators (CRTCs), which are activated through dephosphorylation. A dual requirement for CREB phosphorylation and CRTC dephosphorylation is likely to explain how these activator-co-activator cognates discriminate between different stimuli. Following their activation, CREB and CRTCs mediate the effects of fasting and feeding signals on the expression of metabolic programmes in insulin-sensitive tissues.
Collapse
Affiliation(s)
- Judith Y Altarejos
- Sanford-Burnham Medical Research Institute at Lake Nona, 6400 Sanger Road, Orlando, Florida 32827, USA
| | | |
Collapse
|
41
|
Sasaki T, Takemori H, Yagita Y, Terasaki Y, Uebi T, Horike N, Takagi H, Susumu T, Teraoka H, Kusano KI, Hatano O, Oyama N, Sugiyama Y, Sakoda S, Kitagawa K. SIK2 is a key regulator for neuronal survival after ischemia via TORC1-CREB. Neuron 2011; 69:106-19. [PMID: 21220102 DOI: 10.1016/j.neuron.2010.12.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2010] [Indexed: 11/16/2022]
Abstract
The cAMP responsive element-binding protein (CREB) functions in a broad array of biological and pathophysiological processes. We found that salt-inducible kinase 2 (SIK2) was abundantly expressed in neurons and suppressed CREB-mediated gene expression after oxygen-glucose deprivation (OGD). OGD induced the degradation of SIK2 protein concomitantly with the dephosphorylation of the CREB-specific coactivator transducer of regulated CREB activity 1 (TORC1), resulting in the activation of CREB and its downstream gene targets. Ca(2+)/calmodulin-dependent protein kinase I/IV are capable of phosphorylating SIK2 at Thr484, resulting in SIK2 degradation in cortical neurons. Neuronal survival after OGD was significantly increased in neurons isolated from sik2(-/-) mice, and ischemic neuronal injury was significantly reduced in the brains of sik2(-)(/-) mice subjected to transient focal ischemia. These findings suggest that SIK2 plays critical roles in neuronal survival, is modulated by CaMK I/IV, and regulates CREB via TORC1.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Spiga F, Liu Y, Aguilera G, Lightman SL. Temporal effect of adrenocorticotrophic hormone on adrenal glucocorticoid steroidogenesis: involvement of the transducer of regulated cyclic AMP-response element-binding protein activity. J Neuroendocrinol 2011; 23:136-42. [PMID: 21083631 PMCID: PMC3189260 DOI: 10.1111/j.1365-2826.2010.02096.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/11/2010] [Accepted: 11/14/2010] [Indexed: 12/31/2022]
Abstract
The availability of active steroidogenic acute regulatory protein (StAR) and side-chain cleavage cytochrome P450 (P450scc) are rate-limiting steps for steroidogenesis. Transcription of StAR and P450scc genes depends on cyclic AMP-response element-binding protein (CREB) phosphorylation and CREB co-activator, transducer of regulated CREB activity (TORC), which is regulated by salt-inducible kinase 1 (SIK1). In the present study, we investigated the relationship between TORC activation and adrenocorticotrophic hormone (ACTH)-induced steroidogenesis in vivo, by examining the time-course of the effect of ACTH injection (4 ng, i.v.) on the transcriptional activity of StAR and P450scc genes and the nuclear accumulation of transducer of regulated CREB activity 2 (TORC2) in rat adrenal cortex. ACTH produced rapid and transient increases in plasma corticosterone, with maximal responses between 5 and 15 min, and a decrease to almost basal values at 30 min. StAR and P450scc hnRNA levels increased 15 min following ACTH and decreased toward basal values at 30 min. Concomitant with an increase in nuclear phospho-CREB, ACTH injection induced nuclear accumulation of TORC2, with maximal levels at 5 min and a return to basal values by 30 min. The decline of nuclear TORC2 was paralleled by increases in SIK1 hnRNA and mRNA 15 and 30 min after injection, respectively. The early rises in plasma corticosterone preceding StAR and P450scc gene transcription suggest that post-transcriptional and post-translational changes in StAR protein mediate the early steroidogenic responses. Furthermore, the direct temporal relationship between nuclear accumulation of TORC2 and the increase in transcription of steroidogenic proteins, implicates TORC2 in the physiological regulation of steroidogenesis in the adrenal cortex. The delayed induction of SIK1 suggests a role for SIK1 in the declining phase of steroidogenesis.
Collapse
Affiliation(s)
- F Spiga
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK.
| | | | | | | |
Collapse
|