Suppression of gluconeogenic gene transcription by SIK1-induced ubiquitination and degradation of CRTC1

https://doi.org/10.1016/j.bbagrm.2018.01.021Get rights and content

Highlights

  • SIK1 specifically destabilizes CRTC1 protein.

  • S155, S167, S188 and S346 of CRTC1 are critical for destabilization.

  • SIK1 suppresses but CRTC1 stimulates gluconeogenic gene expression.

  • SIK1 induces RFWD2-mediated and K628-independent ubiquitination of CRTC1.

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.

Introduction

cAMP response element (CRE)-binding protein (CREB) is a multifaceted transcription factor regulating the expression of about 4000 target genes [1], which collectively exert a substantial impact on metabolism [2,3], cell proliferation [4], immune response [5], learning and memory [6], as well as other physiological and pathological processes [7]. CREB activity is regulated by two distinct but interconnected mechanisms. First, protein kinase A (PKA)-mediated phosphorylation of CREB at S133 promotes the recruitment of transcriptional coactivators in the family of histone acetyltransferases [8]. Second, CREB activation is achieved through another group of obligate transcriptional coactivators termed CREB-regulated transcriptional coactivator (CRTCs), also known as transducer of regulated CREB activities (TORCs), consisted of three isoforms CRTC1, CRTC2 and CRTC3 [9].

Although the three CRTC isoforms are structurally and functionally related, their tissue distribution patterns are distinct and some of their biological functions are non-redundant. Whereas CRTC2 and CRTC3 are more abundant in the liver, CRTC1 is more concentrated in certain areas of the brain. However, all isoforms are widely expressed but not restricted to particular types of tissue [10]. In addition, their expression can be substantially increased in response to cellular stress and other conditions. For example, CRTC1 expression is highly induced in many cancer cells [11,12] and by hepatitis B virus [13]. CRTC2 is most studied among the three isoforms. Both CRTC2 and CRTC3 are thought to be key regulators of gluconeogenesis [14], glucose uptake [15], energy homeostasis [16,17] and macrophage polarization [5,18]. A role for CRTC2 in lipid metabolism [19,20] and the requirement of CRTC3 for hormonal control of stress response [21] have also been described. In contrast to CRTC2 and CRTC3, CRTC1 has been shown to have a neuronal function. Particularly, CRTC1 is essential for long term memory [22,23], circadian rhythm [24], dendritic growth [25] and neuronal survival after ischemia [26]. CRTC1-null mice are hyperphagic, obese and infertile [28]. They might also develop hepatic steatosis [27]. They can serve as a model for depression [29]. Disruption of CRTC2 in mice results in increased insulin sensitivity [30]. Knockout of mouse CRTC3 leads to resistance to obesity plausibly due to increased energy expenditure and increased β-adrenergic receptor signaling [31]. These phenotypes highlight the unique biological functions of the three CRTC isoforms.

The activity of CRTCs is tightly regulated by phosphorylation. Hyperphosphorylated CRTCs are bound with 14-3-3 proteins in the cytoplasm and their rapid dephosphorylation are required for activation of CREB-dependent transcription [32]. CRTC phosphorylation is catalyzed by salt-inducible kinases (SIKs), which are AMP-activated protein kinase (AMPK)-related kinases regulated by tumor suppressor kinase LKB1 [33]. SIKs comprising three isoforms were initially identified from adrenal glands of rats fed with high-salt diet [34]. They function as master regulators of sodium sensing [35], bone formation [36] and cAMP signaling [37,38]. Particularly, SIK2 directly phosphorylates CRTC2 at S171 [39] and it also phosphorylates p300 to disrupt its acetylation of CRTC2 at K628 [40]. Deacetylated and phosphorylated CRTC2 undergoes ubiquitination at K628 catalyzed by E3 ubiquitin ligase RFWD2, also known as COP1, leading to proteasomal degradation [39]. Although regulation of other CRTC isoforms by SIKs is assumed [14], it remains to be elucidated whether the modification might be isoform-specific. Particularly, whether CRTC1 is phosphorylated and regulated by SIK1 has not been determined experimentally. In this regard, genetic evidence suggests that the regulation of CRTCs by SIKs is not promiscuous [41,42]. Our previous analysis of the role of LKB1 and SIKs in human T-cell leukemia virus type 1 (HTLV-1) transcription suggests that the three SIK isoforms cooperate with each other in the regulation of CRTC activity [43]. SIK1-knockout mice are viable and normoglycemic on regular chow diet, but they exhibit increased sensitivity to insulin and their plasma insulin levels are elevated on high fat diet. They also have high arterial blood pressure [44,45]. Tissue-specific knockout reveals this specific function of SIK1 in skeletal muscle [45]. SIK2-null mice are hyperglycemic and hypertriglyceridemic [46]. Knockout of SIK3 in mice causes dwarfism. The mice are hypoglycemic and hypolipidemic, with increased insulin sensitivity and aberrant circadian rhythms [41,[47], [48], [49]]. Thus, the three SIK isoforms have distinct and related biological functions.

Regulatory phosphorylation of CRTC2 has also been shown to occur at several other sites including S70 [50], S275 [50,51] and S307 [52]. Additional phosphorylation sites on CRTC1 have also been suggested [53]. These findings prompted us to identify additional regulatory sites on CRTC1. Our gain-of-function and loss-of-function experiments in this study revealed SIK1-induced ubiquitination and degradation of CRTC1. The influence of this regulatory mechanism on gluconeogenic gene expression was also assessed.

Section snippets

Plasmids, antibodies and reagents

Expression plasmids for CRTC1 and its mutants based on pCAGEN-V5 as well as reporter plasmids pCRE-Luc and pPEPCK-Luc have been described previously [13,43,54]. Expression plasmids pCAGEN-FLAG-SIK1/2/3 were constructed by subcloning of SIK1/2/3 cDNA from pCMV-Tag2B-SIK1/2/3 [43] to pCAGEN vector. Expression plasmid pCAGEN-RFWD2-HA was constructed by cloning of RFWD2 cDNA into pCAGEN vector. Plasmids pCMV-myc-ubiquitin and derivatives were made from constructs provided by Dr. Dirk Bohmann

Kinase-dependent induction of CRTC1 degradation by SIK1

Phosphorylation of CRTC2 by SIK2 triggers ubiquitination and degradation [39]. To investigate whether CRTC1 might be subjected to similar mode of regulation, we expressed wild-type (WT) CRTC1 and constitutively active (CA) SIKs (SIK1 T182D, SIK2 T175D and SIK3 T163D) in HeLa cells. A significant reduction of CRTC1 protein was only observed when SIK1 T182D was expressed (Fig. 1A, lane 2 compared to 1). Neither SIK2 T175D nor SIK3 T163D had a substantial influence on the steady-state level of

Discussion

In this study, we demonstrated SIK1-induced ubiquitination and degradation of the transcriptional coactivator CRTC1. In addition to S167, three other residues S155, S188 and S346 were identified to be critical in SIK1-induced degradation of CRTC1. A regulatory role of SIK1-CRTC1 signaling in gluconeogenesis was characterized. Whereas SIK1 functioned as a gluconeogenic suppressor, CRTC1 and two pharmaceutical inhibitors of SIKs exerted the opposite effect. Furthermore, RFWD2 served as an E3

Author contributions

WWG, HMVT, Chi-PC and DYJ conceptualized and designed the study. WWG performed experiments with the help from YC and Ching-PC. All authors contributed to data analysis. WWG and DYJ wrote the paper with input from all authors.

Transparency document

Transparency document

Acknowledgments

We thank Kristopher Clark, Ted Dawson, Zhijian James Chen and Dirk Dohmann for reagents; and members of Jin Laboratory for critical reading of the manuscript.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

References (87)

  • J. McTague et al.

    The adrenergic-regulated CRTC1 and CRTC2 phosphorylation and cellular distribution is independent of endogenous SIK1 in the male rat pinealocyte

    Mol. Cell. Endocrinol.

    (2015)
  • M. Treier et al.

    Ubiquitin-dependent c-Jun degradation in vivo is mediated by the δ domain

    Cell

    (1994)
  • W. Zeng et al.

    Key role of Ubc5 and lysine-63 polyubiquitination in viral activation of IRF3

    Mol. Cell

    (2009)
  • T.H. Ch'ng et al.

    Activity-dependent transport of the transcriptional coactivator CRTC1 from synapse to nucleus

    Cell

    (2012)
  • M.A. Bittinger et al.

    Activation of cAMP response element-mediated gene expression by regulated nuclear transport of TORC proteins

    Curr. Biol.

    (2004)
  • T. Okamoto et al.

    Establishment and characterization of a novel method for evaluating gluconeogenesis using hepatic cell lines, H4IIE and HepG2

    Arch. Biochem. Biophys.

    (2009)
  • A. Jagannath et al.

    The CRTC1-SIK1 pathway regulates entrainment of the circadian clock

    Cell

    (2013)
  • C.C. Escoubas et al.

    Deregulation of CRTCs in aging and age-related disease risk

    Trends Genet.

    (2017)
  • I.D. O'Neill

    t(11;19) translocation and CRTC1-MAML2 fusion oncogene in mucoepidermoid carcinoma

    Oral Oncol.

    (2009)
  • D. Bell et al.

    CRTC1/MAML2 fusion transcript in central mucoepidermoid carcinoma of mandible–diagnostic and histogenetic implications

    Ann. Diagn. Pathol.

    (2010)
  • F. Miranda et al.

    Salt-inducible kinase 2 couples ovarian cancer cell metabolism with survival at the adipocyte-rich metastatic niche

    Cancer Cell

    (2016)
  • X. Zhang et al.

    Genome-wide analysis of cAMP-response element binding protein occupancy, phosphorylation, and target gene activation in human tissues

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • K.J. Oh et al.

    CREB and FoxO1: two transcription factors for the regulation of hepatic gluconeogenesis

    BMB Rep.

    (2013)
  • K. Ravnskjaer et al.

    Role of the cAMP pathway in glucose and lipid metabolism

    Handb. Exp. Pharmacol.

    (2016)
  • Y.-T. Siu et al.

    CREB–a real culprit in oncogenesis

    FEBS J.

    (2007)
  • B. Luan et al.

    CREB pathway links PGE2 signaling with macrophage polarization

    Proc. Natl. Acad. Sci. U. S. A.

    (2015)
  • K. Sakamoto et al.

    CREB: a multifaceted regulator of neuronal plasticity and protection

    J. Neurochem.

    (2011)
  • J.Y. Altarejos et al.

    CREB and the CRTC co-activators: sensors for hormonal and metabolic signals

    Nat. Rev. Mol. Cell Biol.

    (2011)
  • R.P. Kwok et al.

    Nuclear protein CBP is a coactivator for the transcription factor CREB

    Nature

    (1994)
  • V. Iourgenko et al.

    Identification of a family of cAMP response element-binding protein coactivators by genome-scale functional analysis in mammalian cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • T. Komiya et al.

    Enhanced activity of the CREB co-activator Crtc1 in LKB1 null lung cancer

    Oncogene

    (2010)
  • Y. Schumacher et al.

    Dysregulated CRTC1 activity is a novel component of PGE2 signaling that contributes to colon cancer growth

    Oncogene

    (2016)
  • H.-M.V. Tang et al.

    Requirement of CRTC1 coactivator for hepatitis B virus transcription

    Nucleic Acids Res.

    (2014)
  • K. Patel et al.

    The LKB1-salt-inducible kinase pathway functions as a key gluconeogenic suppressor in the liver

    Nat. Commun.

    (2014)
  • E. Henriksson et al.

    SIK2 regulates CRTCs, HDAC4 and glucose uptake in adipocytes

    J. Cell Sci.

    (2015)
  • N.E. Bruno et al.

    CREB coactivators direct anabolic responses and enhance performance of skeletal muscle

    EMBO J.

    (2014)
  • T. Shan et al.

    Lkb1 controls brown adipose tissue growth and thermogenesis by regulating the intracellular localization of CRTC3

    Nat. Commun.

    (2016)
  • K. Clark et al.

    Phosphorylation of CRTC3 by the salt-inducible kinases controls the interconversion of classically activated and regulatory macrophages

    Proc. Natl. Acad. Sci. U. S. A.

    (2012)
  • J. Han et al.

    The CREB coactivator CRTC2 controls hepatic lipid metabolism by regulating SREBP1

    Nature

    (2015)
  • Y. Li et al.

    A novel role for CRTC2 in hepatic cholesterol synthesis through SREBP-2

    Hepatology

    (2017)
  • B. Jurek et al.

    Oxytocin regulates stress-induced Crf gene transcription through CREB-regulated transcription coactivator 3

    J. Neurosci.

    (2015)
  • K. Sakamoto et al.

    Clock and light regulation of the CREB coactivator CRTC1 in the suprachiasmatic circadian clock

    J. Neurosci.

    (2013)
  • S. Li et al.

    TORC1 regulates activity-dependent CREB-target gene transcription and dendritic growth of developing cortical neurons

    J. Neurosci.

    (2009)
  • Cited by (0)

    View full text