GET THE APP

CDKs: Much More Than Just the Control of Cell Cycle Progression
Journal of Cell Science & Therapy

Journal of Cell Science & Therapy
Open Access

ISSN: 2157-7013

+44 1300 500008

Commentary - (2014) Volume 5, Issue 4

CDKs: Much More Than Just the Control of Cell Cycle Progression

Felicia Hanzu and Xavier Escoté*
Endocrine Disorders Research Group, Hospital Clínic & IDIBAPS, Barcelona, Spain, E-mail: fhanzu@clinic.ub.es
*Corresponding Author: Xavier Escoté, Endocrine Disorders Research Group, Hospital Clínic & IDIBAPS, Barcelona, 149-153, 5th Floor, 08036 Barcelona, Spain, Tel: 3493227570 Email:

Abstract

Lee and colleagues have shown that CDK4 plays a central role in glucose homeostasis. Insulin activates cyclin D1–CDK4, leading to a decrease in circulating glucose as a consequence of the down-regulation of main genes involved in liver gluconeogenesis.

Commentary

In recent years, our knowledge of CDK regulation has improved significantly and is no longer restricted to just its role in the control of cell cycle progression. Cyclin-dependent kinases (CDKs) are serine/threonine protein kinases that are associated with their specific partners, cyclins which phosphorylate their protein targets. Thus, the cyclin-CDK complex works as a holoenzyme. The control of cell cycle progression by cyclin-CDK complexes was first studied several years ago, in the context of high proliferation rates and cancer. Understanding how CDKs function was one of the main aims of these studies, as the loss of this regulation can alter cell proliferation rates which, in the worst case scenario, may translate into malignant tumours.

The cell cycle is mainly divided into G1, S, G2, and M phases, and transition between these phases is strongly controlled. Transition from G1 to S is tightly regulated and depends on the activity of the G1 phase cyclin-CDK complexes, where CDK can be CDK4 or CDK6 and is associated with G1 D-type cyclins (D1, D2 and D3). In addition, there is an upper-level control of the activity of cyclin-CDK complexes determined by the presence or absence of two families of CDK inhibitors (CKIs), INK4 proteins and the Cip/Kip family, respectively. When the G1 phase cyclin-CDK complexes are active, they regulate the transition to the S phase by a direct phosphorylation of the retinoblastoma protein (Rb). Rb hyperphosphorylation mediates the release of the E2F transcription factor, allowing its transit to the nucleus, where it can promote the transcription of several genes involved in cell cycle progression, apoptosis, and DNA synthesis [1].

Besides their well-described role in the control of cell cycle progression, in recent years a new role for CDKs has emerged beyond the context of cell cycle progression, as a master regulator of metabolism. In this sense, the CDK4-RB-E2F axis and other cell cycle components have acquired a main role in the control of insulin secretion by pancreatic β-cells [2,3], of oxidative metabolism [4] and of adipogenesis [5-8].

Much more recently, a unique study designed by the Puigserver group [9] demonstrated that CDK4 plays a central role in glucose homeostasis, specifically in liver gluconeogenesis. Briefly, insulin activates AKT (also known as PKB) through a cascade of correlative phosphorylation with the insulin receptor, insulin receptor substrates, PI3K and finally PDK1 (Figure 1). Once AKT is activated, AKT phosphorylates several substrates, among them, Glycogen synthase kinase 3 (GSK3), at Ser 21 in α subunit and at Ser 9 in β subunit, resulting in the inhibition of GSK3 kinase activity [10-13]. GSK3 is a well-conserved kinase, originally identified as an enzyme that regulates glycogen synthesis in response to insulin [14].Further studies have demonstrated that GSK3 phosphorylates a broad range of substrates, including several transcription factors [15,16]. Moreover, GSK3 was eventually identified as being capable of phosphorylating cyclin D1 on T286 and inducing its rapid turnover in a proteasomal-dependent manner [17,18]. Consequently, GSK3 inhibition by AKT phosphorylation leads to an accumulation of cyclin D1.Furthermore, Lee et al. have demonstrated the increased mRNA expression of cyclin D1 after re-feeding, with amino acids specifically responsible for enhancing cyclin D1 mRNA levels [9] and (Figure 1). However, the molecular mechanisms underlying this increase remain elusive. Thus, we consider that it is necessary to investigate which amino acid oracids are responsible for cyclin D1 overexpression in hepatocytes after feeding. Subsequently, this knowledge may lead to the development of non-invasive therapies to control liver gluconeogenesis.

cell-science-therapy-gluconeogenesis-CDK4

Figure 1: Schematic diagram showing the regulation of gluconeogenesis by CDK4 in liver. Adapted from (10)

The scenario described above promotes an activation of the holoenzyme cyclin D1–CDK4, which translates into decreased circulating glucose as a consequence of the inhibited expression of main genes involved in liver gluconeogenesis, namely Pck1 and G6pc. Expression of Pck1 and G6pc is mainly regulated by PGC-1α (peroxisome-proliferator-activated receptor-γ-coactivator-1α), whose activity is regulated by acetylation (inhibition) and deacetylation (activation).The histone deacetyl transferasesirtuin 1 (SIRT-1) deacetylates and activates PGC-1α [19],whereas GCN5 (general control non-repressed protein 5) acetylates PGC-1α and represses its co-transcriptional activity [20].

Activation of cyclin D1-CDK4 by insulin does not affect SIRT-1 activity. On the contrary, once the CDK4-cyclin D1 complex is active, this directly phosphorylates GCN5, increasing its acetyl transferase activity and suppressing hepatic glucose production by acetylation and inhibition of PGC-1α, which, in turn, inhibits the transcription of gluconeogenic genes (Figure 1). These results were corroborated with several molecular strategies: knock-down experiments, the use of non-phosphorytable GCN5 alleles, CDK4 chemical inhibition, and the use of different cyclin D1mutants. Consequently, the loss of holoenzyme activity resulted inaugmented gluconeogenesis and in higher levels of circulating glucose. In addition, in many in vivo studies, an involvement of PPARs was observed in the regulation of energy metabolism by the liver [21,22].

These discoveries indicate a potential crosstalk between the proteins of cell cycle control and metabolic machinery. Very recently, Lee and colleagues [9] contributed striking evidence toward these discoveries. They ingeniously showed using cells and animal models that insulin is capable of activating cell cycle machinery in hepatocytes to control glucose homeostasis. Thus, a new rolefor CDKs emerges, independently of previous ones related to cell division. This fact, combined with the role of CDK4 in pancreatic β-cell proliferation and insulin secretion, points to cell cycle machinery as a central regulator of systemic glucose homeostasis.

Many interesting questions remain open, and answering them can help provide a more complete view of CDK control in liver gluconeogenesis. Future investigations should determine whether CDK4 phosphorylations over FoxO3A trigger any physiological effects, perhaps modifying in some way cyclin D1 expression and consequently affecting CDK4 activity. Another point to consider is if the implication of CDKs is restricted to liver gluconeogenesis or if they are also involved in the metabolic regulation of other tissues such as skeletal muscle or adipose tissue. Finally, another unanswered question is if CDK6, also activated by D type cyclins, may play a similar role in the liver.

These results open a new field of research aimed at deciphering the metabolic functions of cell cycle components, as well as, in an opposing manner, the regulation of cell cycle progression by metabolic genes, not only in the liver but also in other organs and tissues. A greater understanding of CDKs may pave the way for developing the use of these proteins as therapeutic targets for metabolic diseases.

References

  1. Attwooll C, LazzeriniDenchi E, Helin K (2004) The E2F family: specific functions and overlapping interests. See comment in PubMed Commons below EMBO J 23: 4709-4716.
  2. Fajas L, Annicotte JS, Miard S, Sarruf D, Watanabe M, et al. (2004) Impaired pancreatic growth, beta cell mass, and beta cell function in E2F1 (-/- )mice. See comment in PubMed Commons below J Clin Invest 113: 1288-1295.
  3. Annicotte JS, Blanchet E, Chavey C, Iankova I, Costes S, et al. (2009) The CDK4-pRB-E2F1 pathway controls insulin secretion. See comment in PubMed Commons below Nat Cell Biol 11: 1017-1023.
  4. Blanchet E, Annicotte JS, Lagarrigue S, Aguilar V, Clapé C, et al. (2011) E2F transcription factor-1 regulates oxidative metabolism. See comment in PubMed Commons below Nat Cell Biol 13: 1146-1152.
  5. Sarruf DA, Iankova I, Abella A, Assou S, Miard S, et al. (2005) Cyclin D3 promotes adipogenesis through activation of peroxisome proliferator-activated receptor gamma. See comment in PubMed Commons below Mol Cell Biol 25: 9985-9995.
  6. Fajas L, Landsberg RL, Huss-Garcia Y, Sardet C, Lees JA, et al. (2002) E2Fs regulate adipocyte differentiation. See comment in PubMed Commons below Dev Cell 3: 39-49.
  7. Abella A, Dubus P, Malumbres M, Rane SG, Kiyokawa H, et al. (2005) Cdk4 promotes adipogenesis through PPARgamma activation. See comment in PubMed Commons below Cell Metab 2: 239-249.
  8. Aguilar V, Annicotte JS, Escote X, Vendrell J, Langin D, et al. (2010) Cyclin G2 regulates adipogenesis through PPAR gamma coactivation. See comment in PubMed Commons below Endocrinology 151: 5247-5254.
  9. Lee Y, Dominy JE, Choi YJ, Jurczak M, Tolliday N, et al. (2014) Cyclin D1-Cdk4 controls glucose metabolism independently of cell cycle progression. See comment in PubMed Commons below Nature 510: 547-551.
  10. Sutherland C, Leighton IA, Cohen P (1993) Inactivation of glycogen synthase kinase-3 beta by phosphorylation: new kinase connections in insulin and growth-factor signalling. See comment in PubMed Commons below Biochem J 296 : 15-19.
  11. Cross DA, Alessi DR, Vandenheede JR, McDowell HE, Hundal HS, et al. (1994) The inhibition of glycogen synthase kinase-3 by insulin or insulin-like growth factor 1 in the rat skeletal muscle cell line L6 is blocked by wortmannin, but not by rapamycin: evidence that wortmannin blocks activation of the mitogen-activated protein kinase pathway in L6 cells between Ras and Raf. See comment in PubMed Commons below Biochem J 303 : 21-26.
  12. Stambolic V, Woodgett JR (1994) Mitogen inactivation of glycogen synthase kinase-3 beta in intact cells via serine 9 phosphorylation. See comment in PubMed Commons below Biochem J 303 : 701-704.
  13. Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA (1995) Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. See comment in PubMed Commons below Nature 378: 785-789.
  14. Hughes K, Nikolakaki E, Plyte SE, Totty NF, Woodgett JR (1993) Modulation of the glycogen synthase kinase-3 family by tyrosine phosphorylation. See comment in PubMed Commons below EMBO J 12: 803-808.
  15. Plyte SE, Hughes K, Nikolakaki E, Pulverer BJ, Woodgett JR (1992) Glycogen synthase kinase-3: functions in oncogenesis and development. See comment in PubMed Commons below BiochimBiophysActa 1114: 147-162.
  16. Welsh GI, Wilson C, Proud CG (1996) GSK3: a SHAGGY frog story. See comment in PubMed Commons below Trends Cell Biol 6: 274-279.
  17. Diehl JA, Cheng M, Roussel MF, Sherr CJ (1998) Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. See comment in PubMed Commons below Genes Dev 12: 3499-3511.
  18. Diehl JA, Zindy F, Sherr CJ (1997) Inhibition of cyclin D1 phosphorylation on threonine-286 prevents its rapid degradation via the ubiquitin-proteasome pathway. See comment in PubMed Commons below Genes Dev 11: 957-972.
  19. Lerin C, Rodgers JT, Kalume DE, Kim SH, Pandey A, et al. (2006) GCN5 acetyltransferase complex controls glucose metabolism through transcriptional repression of PGC-1alpha. See comment in PubMed Commons below Cell Metab 3: 429-438.
  20. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, et al. (2005) Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. See comment in PubMed Commons below Nature 434: 113-118.
  21. Mastinu A, Pira M, Pani L, Pinna GA, Lazzari P (2012) NESS038C6, a novel selective CB1 antagonist agent with anti-obesity activity and improved molecular profile. See comment in PubMed Commons below Behav Brain Res 234: 192-204.
  22. Mastinu A, Pira M, Pinna GA, Pisu C, Casu MA, et al. (2013) NESS06SM reduces body weight with an improved profile relative to SR141716A. See comment in PubMed Commons below Pharmacol Res 74: 94-108.
Citation: Hanzu F, Escoté X (2014) CDKs: Much More Than Just the Control of Cell Cycle Progression. J Cell Sci Ther 5:170.

Copyright: © 2014 Hanzu F, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top