Diabetes Metab J.  2018 Apr;42(2):117-127. 10.4093/dmj.2018.42.2.117.

Primary Cilia as a Signaling Platform for Control of Energy Metabolism

Affiliations
  • 1Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. mskim@amc.seoul.kr
  • 2Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul, Korea.
  • 3Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.

Abstract

Obesity has become a common healthcare problem worldwide. Cilia are tiny hair-like organelles on the cell surface that are generated and anchored by the basal body. Non-motile primary cilia have been considered to be evolutionary rudiments until a few decades, but they are now considered as important signaling organelles because many receptors, channels, and signaling molecules are highly expressed in primary cilia. A potential role of primary cilia in metabolic regulation and body weight maintenance has been suspected based on rare genetic disorders termed as ciliopathy, such as Bardet-Biedl syndrome and Alström syndrome, which manifest as obesity. Recent studies have demonstrated involvement of cilia-related cellular signaling pathways in transducing metabolic information in hypothalamic neurons and in determining cellular fate during adipose tissue development. In this review, we summarize the current knowledge about cilia and cilia-associated signaling pathways in the regulation of body metabolism.

Keyword

Cilia; Hedgehogs; Metabolism; Obesity; Wnt signaling pathway

MeSH Terms

Adipose Tissue
Alstrom Syndrome
Bardet-Biedl Syndrome
Basal Bodies
Body Weight Maintenance
Cilia*
Delivery of Health Care
Energy Metabolism*
Hedgehogs
Metabolism
Neurons
Obesity
Organelles
Wnt Signaling Pathway

Figure

  • Fig. 1 Structure of the primary cilia and intraflagellar transport (IFT) system. A primary cilium is composed of cytoskeleton, called the ciliary axoneme (green), and the ciliary membrane (dark blue line). The basal body acts as a microtubule-organizing center of the ciliary axoneme. The ciliary membrane is enriched for many receptors, ion channels, effector proteins, and transcription factors, which are produced in the cytoplasmic endoplasmic reticulum and transported to the transition zone of cilia via the specialized vesicular trafficking system. The axonemal precursor and ciliary membrane proteins are transported from the base to the tip of the cilia by antegrade IFT (red arrow), which is mediated by the IFT complex B (IFT-B) and motor protein kinesin-2. Retrograde transport (blue arrow) returns turnover products from the tip of the cilia back to the base and is performed by the IFT complex A (IFT-A) with dynein-2 as a motor protein. KAP3, kinesin-associated protein 3; KIF3A, a subunit A of heterotrimeric motor protein kinesin-2; KIF3B/C, a subunit B/C of kinesin 2; ER, endoplasmic reticulum.

  • Fig. 2 Signaling pathways related to the primary cilia. (A) Canonical Hedgehog (HH) signaling pathway: under basal condition without HH ligands, the HH receptor Patched (Ptch) inhibits the activity of smoothened (SMO). In this condition, the suppressor of fused (SUFU) binds to the GLI transcription factor and prevents GLI activation. If HH binds to Ptch, SMO migrates to the cilia and activates GLI. GLI leaves cilia and activates the transcription of GLI target genes in the nucleus. (B) Canonical Wnt/β-catenin signaling pathway: without Wnt ligands, β-catenin is ubiquitinated by a destruction complex and degraded by the proteasome. The β-catenin destruction complex is composed of axin, casein kinase-1α (CK-1α), adenomatosis polyposis coli (APC), protein phosphatase 2A (PP2A), and glycogen synthase kinase-3 (GSK-3β). When Wnt ligands bind the Frizzled (Fzd) family receptor and its coreceptor low density lipoprotein receptor-related protein-5/6 (LRP-5/6), the Fzd receptor transmits the signal to inactivate the β-catenin destruction complex. Therefore, β-catenin accumulates in the cytoplasm and translocates to the nucleus where it regulates target gene expression along with the transcriptional coactivator T-cell-specific transcription factor (TCF) family. Dsh, Dishevelled.

  • Fig. 3 Potential role of primary cilia in metabolic regulation and body weight control. In hypothalamic neurons, primary cilia may be involved in sensing metabolic signals. In the adipose tissue, primary cilia control the process of adipocyte differentiation by transducing canonical Hedgehog (HH) signaling and Wnt signaling. Moreover, non-canonical HH signaling in the skeletal muscle and brown adipocytes controls glucose and energy metabolism. Therefore, dysfunction or dysgenesis of cilia can lead to obesity and abnormal glucose metabolism.


Reference

1. Bhurosy T, Jeewon R. Overweight and obesity epidemic in developing countries: a problem with diet, physical activity, or socioeconomic status? ScientificWorldJournal. 2014; 2014:964236.
Article
2. Hruby A, Hu FB. The epidemiology of obesity: a big picture. Pharmacoeconomics. 2015; 33:673–689.
Article
3. Maes HH, Neale MC, Eaves LJ. Genetic and environmental factors in relative body weight and human adiposity. Behav Genet. 1997; 27:325–351.
4. Oh EC, Vasanth S, Katsanis N. Metabolic regulation and energy homeostasis through the primary Cilium. Cell Metab. 2015; 21:21–31.
Article
5. Kebede MA, Attie AD. Insights into obesity and diabetes at the intersection of mouse and human genetics. Trends Endocrinol Metab. 2014; 25:493–501.
Article
6. Ingalls AM, Dickie MM, Snell GD. Obese, a new mutation in the house mouse. J Hered. 1950; 41:317–318.
Article
7. Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Wool EA, Monroe CA, Tepper RI. Identification and expression cloning of a leptin receptor, OB-R. Cell. 1995; 83:1263–1271.
Article
8. Bloodgood RA. From central to rudimentary to primary: the history of an underappreciated organelle whose time has come. The primary cilium. Methods Cell Biol. 2009; 94:3–52.
9. Berbari NF, O'Connor AK, Haycraft CJ, Yoder BK. The primary cilium as a complex signaling center. Curr Biol. 2009; 19:R526–R535.
Article
10. Hildebrandt F, Benzing T, Katsanis N. Ciliopathies. N Engl J Med. 2011; 364:1533–1543.
Article
11. Louvi A, Grove EA. Cilia in the CNS: the quiet organelle claims center stage. Neuron. 2011; 69:1046–1060.
Article
12. Christensen ST, Pedersen LB, Schneider L, Satir P. Sensory cilia and integration of signal transduction in human health and disease. Traffic. 2007; 8:97–109.
Article
13. Hsiao YC, Tuz K, Ferland RJ. Trafficking in and to the primary cilium. Cilia. 2012; 1:4.
Article
14. Cole DG, Diener DR, Himelblau AL, Beech PL, Fuster JC, Rosenbaum JL. Chlamydomonas kinesin-II-dependent intraflagellar transport (IFT): IFT particles contain proteins required for ciliary assembly in Caenorhabditis elegans sensory neurons. J Cell Biol. 1998; 141:993–1008.
Article
15. Pazour GJ, Wilkerson CG, Witman GB. A dynein light chain is essential for the retrograde particle movement of intraflagellar transport (IFT). J Cell Biol. 1998; 141:979–992.
Article
16. Pazour GJ, Dickert BL, Vucica Y, Seeley ES, Rosenbaum JL, Witman GB, Cole DG. Chlamydomonas IFT88 and its mouse homologue, polycystic kidney disease gene tg737, are required for assembly of cilia and flagella. J Cell Biol. 2000; 151:709–718.
Article
17. Cortellino S, Wang C, Wang B, Bassi MR, Caretti E, Champeval D, Calmont A, Jarnik M, Burch J, Zaret KS, Larue L, Bellacosa A. Defective ciliogenesis, embryonic lethality and severe impairment of the sonic Hedgehog pathway caused by inactivation of the mouse complex A intraflagellar transport gene Ift122/Wdr10, partially overlapping with the DNA repair gene Med1/Mbd4. Dev Biol. 2009; 325:225–237.
Article
18. Takeda S, Yonekawa Y, Tanaka Y, Okada Y, Nonaka S, Hirokawa N. Left-right asymmetry and kinesin superfamily protein KIF3A: new insights in determination of laterality and mesoderm induction by kif3A-/- mice analysis. J Cell Biol. 1999; 145:825–836.
Article
19. Jiang J, Hui CC. Hedgehog signaling in development and cancer. Dev Cell. 2008; 15:801–812.
Article
20. Varjosalo M, Taipale J. Hedgehog: functions and mechanisms. Genes Dev. 2008; 22:2454–2472.
Article
21. Echelard Y, Epstein DJ, St-Jacques B, Shen L, Mohler J, McMahon JA, McMahon AP. Sonic hedgehog, a member of a family of putative signaling molecules, is implicated in the regulation of CNS polarity. Cell. 1993; 75:1417–1430.
Article
22. Krauss S, Concordet JP, Ingham PW. A functionally conserved homolog of the Drosophila segment polarity gene hh is expressed in tissues with polarizing activity in zebrafish embryos. Cell. 1993; 75:1431–1444.
Article
23. Roelink H, Augsburger A, Heemskerk J, Korzh V, Norlin S, Ruiz i, Tanabe Y, Placzek M, Edlund T, Jessell TM, Dodd J. Floor plate and motor neuron induction by vhh-1, a vertebrate homolog of hedgehog expressed by the notochord. Cell. 1994; 76:761–775.
Article
24. Kim J, Kato M, Beachy PA. Gli2 trafficking links Hedgehog-dependent activation of Smoothened in the primary cilium to transcriptional activation in the nucleus. Proc Natl Acad Sci U S A. 2009; 106:21666–21671.
Article
25. Brennan D, Chen X, Cheng L, Mahoney M, Riobo NA. Noncanonical Hedgehog signaling. Vitam Horm. 2012; 88:55–72.
Article
26. Teperino R, Aberger F, Esterbauer H, Riobo N, Pospisilik JA. Canonical and non-canonical Hedgehog signalling and the control of metabolism. Semin Cell Dev Biol. 2014; 33:81–92.
Article
27. Benzler J, Andrews ZB, Pracht C, Stohr S, Shepherd PR, Grattan DR, Tups A. Hypothalamic WNT signalling is impaired during obesity and reinstated by leptin treatment in male mice. Endocrinology. 2013; 154:4737–4745.
Article
28. Nusse R, Varmus H. Three decades of Wnts: a personal perspective on how a scientific field developed. EMBO J. 2012; 31:2670–2684.
Article
29. Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: a prelude for more. Circ Res. 2010; 106:1798–1806.
30. Molenaar M, van de Wetering M, Oosterwegel M, Peterson-Maduro J, Godsave S, Korinek V, Roose J, Destree O, Clevers H. XTcf-3 transcription factor mediates beta-catenin-induced axis formation in Xenopus embryos. Cell. 1996; 86:391–399.
31. Helfer G, Tups A. Hypothalamic Wnt signalling and its role in energy balance regulation. J Neuroendocrinol. 2016; 28:12368.
Article
32. Aznar N, Billaud M. Primary cilia bend LKB1 and mTOR to their will. Dev Cell. 2010; 19:792–794.
Article
33. Ravindran S, Kuruvilla V, Wilbur K, Munusamy S. Nephroprotective effects of metformin in diabetic nephropathy. J Cell Physiol. 2017; 232:731–742.
Article
34. Pampliega O, Cuervo AM. Autophagy and primary cilia: dual interplay. Curr Opin Cell Biol. 2016; 39:1–7.
Article
35. Tang Z, Lin MG, Stowe TR, Chen S, Zhu M, Stearns T, Franco B, Zhong Q. Autophagy promotes primary ciliogenesis by removing OFD1 from centriolar satellites. Nature. 2013; 502:254–257.
Article
36. Pampliega O, Orhon I, Patel B, Sridhar S, Diaz-Carretero A, Beau I, Codogno P, Satir BH, Satir P, Cuervo AM. Functional interaction between autophagy and ciliogenesis. Nature. 2013; 502:194–200.
Article
37. Jin H, White SR, Shida T, Schulz S, Aguiar M, Gygi SP, Bazan JF, Nachury MV. The conserved Bardet-Biedl syndrome proteins assemble a coat that traffics membrane proteins to cilia. Cell. 2010; 141:1208–1219.
Article
38. Badano JL, Mitsuma N, Beales PL, Katsanis N. The ciliopathies: an emerging class of human genetic disorders. Annu Rev Genomics Hum Genet. 2006; 7:125–148.
Article
39. Jacobsson JA, Schioth HB, Fredriksson R. The impact of intronic single nucleotide polymorphisms and ethnic diversity for studies on the obesity gene FTO. Obes Rev. 2012; 13:1096–1109.
40. Stratigopoulos G, Martin Carli JF, O'Day DR, Wang L, Leduc CA, Lanzano P, Chung WK, Rosenbaum M, Egli D, Doherty DA, Leibel RL. Hypomorphism for RPGRIP1L, a ciliary gene vicinal to the FTO locus, causes increased adiposity in mice. Cell Metab. 2014; 19:767–779.
Article
41. Millan MJ, Millan MH, Reid LD, Herz A. The role of the mediobasal arcuate hypothalamus in relation to opioid systems in the control of ingestive behaviour in the rat. Brain Res. 1986; 381:29–42.
Article
42. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW. Central nervous system control of food intake and body weight. Nature. 2006; 443:289–295.
Article
43. Kang GM, Han YM, Ko HW, Kim J, Oh BC, Kwon I, Kim MS. Leptin elongates hypothalamic neuronal cilia via transcriptional regulation and actin destabilization. J Biol Chem. 2015; 290:18146–18155.
Article
44. Davenport JR, Watts AJ, Roper VC, Croyle MJ, van Groen T, Wyss JM, Nagy TR, Kesterson RA, Yoder BK. Disruption of intraflagellar transport in adult mice leads to obesity and slow-onset cystic kidney disease. Curr Biol. 2007; 17:1586–1594.
Article
45. Han YM, Kang GM, Byun K, Ko HW, Kim J, Shin MS, Kim HK, Gil SY, Yu JH, Lee B, Kim MS. Leptin-promoted cilia assembly is critical for normal energy balance. J Clin Invest. 2014; 124:2193–2197.
Article
46. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000; 404:661–671.
Article
47. Rahmouni K, Fath MA, Seo S, Thedens DR, Berry CJ, Weiss R, Nishimura DY, Sheffield VC. Leptin resistance contributes to obesity and hypertension in mouse models of Bardet-Biedl syndrome. J Clin Invest. 2008; 118:1458–1467.
Article
48. Berbari NF, Pasek RC, Malarkey EB, Yazdi SM, McNair AD, Lewis WR, Nagy TR, Kesterson RA, Yoder BK. Leptin resistance is a secondary consequence of the obesity in ciliopathy mutant mice. Proc Natl Acad Sci U S A. 2013; 110:7796–7801.
Article
49. Seo S, Guo DF, Bugge K, Morgan DA, Rahmouni K, Sheffield VC. Requirement of Bardet-Biedl syndrome proteins for leptin receptor signaling. Hum Mol Genet. 2009; 18:1323–1331.
Article
50. Berbari NF, Lewis JS, Bishop GA, Askwith CC, Mykytyn K. Bardet-Biedl syndrome proteins are required for the localization of G protein-coupled receptors to primary cilia. Proc Natl Acad Sci U S A. 2008; 105:4242–4246.
Article
51. Seeley RJ, Woods SC. Monitoring of stored and available fuel by the CNS: implications for obesity. Nat Rev Neurosci. 2003; 4:901–909.
Article
52. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999; 284:143–147.
Article
53. Marion V, Stoetzel C, Schlicht D, Messaddeq N, Koch M, Flori E, Danse JM, Mandel JL, Dollfus H. Transient ciliogenesis involving Bardet-Biedl syndrome proteins is a fundamental characteristic of adipogenic differentiation. Proc Natl Acad Sci U S A. 2009; 106:1820–1825.
Article
54. Ross SE, Hemati N, Longo KA, Bennett CN, Lucas PC, Erickson RL, MacDougald OA. Inhibition of adipogenesis by Wnt signaling. Science. 2000; 289:950–953.
Article
55. Wright WS, Longo KA, Dolinsky VW, Gerin I, Kang S, Bennett CN, Chiang SH, Prestwich TC, Gress C, Burant CF, Susulic VS, MacDougald OA. Wnt10b inhibits obesity in ob/ob and agouti mice. Diabetes. 2007; 56:295–303.
Article
56. Suh JM, Gao X, McKay J, McKay R, Salo Z, Graff JM. Hedgehog signaling plays a conserved role in inhibiting fat formation. Cell Metab. 2006; 3:25–34.
Article
57. Pospisilik JA, Schramek D, Schnidar H, Cronin SJ, Nehme NT, Zhang X, Knauf C, Cani PD, Aumayr K, Todoric J, Bayer M, Haschemi A, Puviindran V, Tar K, Orthofer M, Neely GG, Dietzl G, Manoukian A, Funovics M, Prager G, Wagner O, Ferrandon D, Aberger F, Hui CC, Esterbauer H, Penninger JM. Drosophila genome-wide obesity screen reveals hedgehog as a determinant of brown versus white adipose cell fate. Cell. 2010; 140:148–160.
Article
58. Teperino R, Amann S, Bayer M, McGee SL, Loipetzberger A, Connor T, Jaeger C, Kammerer B, Winter L, Wiche G, Dalgaard K, Selvaraj M, Gaster M, Lee-Young RS, Febbraio MA, Knauf C, Cani PD, Aberger F, Penninger JM, Pospisilik JA, Esterbauer H. Hedgehog partial agonism drives Warburg-like metabolism in muscle and brown fat. Cell. 2012; 151:414–426.
Article
59. Ogden SK, Fei DL, Schilling NS, Ahmed YF, Hwa J, Robbins DJ. G protein Galphai functions immediately downstream of smoothened in Hedgehog signalling. Nature. 2008; 456:967–970.
60. Polizio AH, Chinchilla P, Chen X, Manning DR, Riobo NA. Sonic Hedgehog activates the GTPases Rac1 and RhoA in a Gli-independent manner through coupling of smoothened to Gi proteins. Sci Signal. 2011; 4:pt7.
Article
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr