リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Regulation of the orphan receptor Gpr176 activity via post-translational modifications in the central circadian clock」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Regulation of the orphan receptor Gpr176 activity via post-translational modifications in the central circadian clock

Wang, Tianyu 京都大学 DOI:10.14989/doctor.k24558

2023.03.23

概要

GPCRs are the largest family of cell-surface receptors and are the therapeutic targets of nearly
a third of clinically marketed drugs1,2. Despite their importance, more than one hundred
human GPCRs remain poorly characterised due to the lack of useful information on their
ligands3. Included among these so-called orphan GPCRs is Gpr176, which is predicted to be a
56-kDa seven-transmembrane protein of class A GPCR with potential sites for N-glycosylation. ...

この論文で使われている画像

参考文献

10

11

12

13

14

15

16

17

18

Santos, R. et al. A comprehensive map of molecular drug targets. Nat Rev Drug

Discov 16, 19-34, doi:10.1038/nrd.2016.230 (2017).

Hauser, A. S., Attwood, M. M., Rask-Andersen, M., Schioth, H. B. & Gloriam, D. E.

Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug

Discov 16, 829-842, doi:10.1038/nrd.2017.178 (2017).

Wacker, D., Stevens, R. C. & Roth, B. L. How Ligands Illuminate GPCR Molecular

Pharmacology. Cell 170, 414-427, doi:10.1016/j.cell.2017.07.009 (2017).

Hata, S., Emi, Y., Iyanagi, T. & Osumi, T. cDNA cloning of a putative G proteincoupled receptor from brain. Biochim Biophys Acta 1261, 121-125 (1995).

Doi, M. et al. Gpr176 is a Gz-linked orphan G-protein-coupled receptor that sets the

pace of circadian behaviour. Nat Commun 7, 10583, doi:10.1038/ncomms10583

(2016).

Schultz, D. J. et al. Transcriptomic response of breast cancer cells to anacardic acid.

Sci Rep 8, 8063, doi:10.1038/s41598-018-26429-x (2018).

Martin, A. L., Steurer, M. A. & Aronstam, R. S. Constitutive Activity among Orphan

Class-A G Protein Coupled Receptors. PLoS One 10, e0138463,

doi:10.1371/journal.pone.0138463 (2015).

Sawutz, D. G., Lanier, S. M., Warren, C. D. & Graham, R. M. Glycosylation of the

mammalian alpha 1-adrenergic receptor by complex type N-linked oligosaccharides.

Mol Pharmacol 32, 565-571 (1987).

van Koppen, C. J. & Nathanson, N. M. Site-directed mutagenesis of the m2

muscarinic acetylcholine receptor. Analysis of the role of N-glycosylation in receptor

expression and function. J Biol Chem 265, 20887-20892 (1990).

Fukushima, Y. et al. Structural and functional analysis of the canine histamine H2

receptor by site-directed mutagenesis: N-glycosylation is not vital for its action.

Biochem J 310 ( Pt 2), 553-558, doi:10.1042/bj3100553 (1995).

Innamorati, G., Sadeghi, H. & Birnbaumer, M. A fully active nonglycosylated V2

vasopressin receptor. Mol Pharmacol 50, 467-473 (1996).

Bisello, A. et al. Role of glycosylation in expression and function of the human

parathyroid hormone/parathyroid hormone-related protein receptor. Biochemistry 35,

15890-15895, doi:10.1021/bi962111+ (1996).

Davis, D. P., Rozell, T. G., Liu, X. & Segaloff, D. L. The six N-linked carbohydrates

of the lutropin/choriogonadotropin receptor are not absolutely required for correct

folding, cell surface expression, hormone binding, or signal transduction. Mol

Endocrinol 11, 550-562, doi:10.1210/mend.11.5.9927 (1997).

Kimura, T. et al. The role of N-terminal glycosylation in the human oxytocin

receptor. Mol Hum Reprod 3, 957-963, doi:10.1093/molehr/3.11.957 (1997).

Kaushal, S., Ridge, K. D. & Khorana, H. G. Structure and function in rhodopsin: the

role of asparagine-linked glycosylation. Proc Natl Acad Sci U S A 91, 4024-4028,

doi:10.1073/pnas.91.9.4024 (1994).

Rands, E. et al. Mutational analysis of beta-adrenergic receptor glycosylation. J Biol

Chem 265, 10759-10764 (1990).

Deslauriers, B. et al. N-glycosylation requirements for the AT1a angiotensin II

receptor delivery to the plasma membrane. Biochem J 339 ( Pt 2), 397-405 (1999).

Servant, G., Dudley, D. T., Escher, E. & Guillemette, G. Analysis of the role of Nglycosylation in cell-surface expression and binding properties of angiotensin II type2 receptor of rat pheochromocytoma cells. Biochem J 313 ( Pt 1), 297-304,

doi:10.1042/bj3130297 (1996).

27

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

Fan, G. et al. N-linked glycosylation of the human Ca2+ receptor is essential for its

expression at the cell surface. Endocrinology 138, 1916-1922,

doi:10.1210/endo.138.5.5131 (1997).

Garcia Rodriguez, C. et al. The role of N-glycosylation for functional expression of

the human platelet-activating factor receptor. Glycosylation is required for efficient

membrane trafficking. J Biol Chem 270, 25178-25184, doi:10.1074/jbc.270.42.25178

(1995).

Davidson, J. S. et al. Identification of N-glycosylation sites in the gonadotropinreleasing hormone receptor: role in receptor expression but not ligand binding. Mol

Cell Endocrinol 107, 241-245 (1995).

Michineau, S., Muller, L., Pizard, A., Alhenc-Gelas, F. & Rajerison, R. M. N-linked

glycosylation of the human bradykinin B2 receptor is required for optimal cellsurface expression and coupling. Biol Chem 385, 49-57, doi:10.1515/BC.2004.007

(2004).

Tansky, M. F., Pothoulakis, C. & Leeman, S. E. Functional consequences of

alteration of N-linked glycosylation sites on the neurokinin 1 receptor. Proc Natl

Acad Sci U S A 104, 10691-10696, doi:10.1073/pnas.0703394104 (2007).

Roy, S., Perron, B. & Gallo-Payet, N. Role of asparagine-linked glycosylation in cell

surface expression and function of the human adrenocorticotropin receptor

(melanocortin 2 receptor) in 293/FRT cells. Endocrinology 151, 660-670,

doi:10.1210/en.2009-0826 (2010).

Lee, K. H. et al. Effect of N-glycosylation on ligand binding affinity of rat V1a

vasopressin receptor. Biochem Biophys Res Commun 286, 707-713,

doi:10.1006/bbrc.2001.5456 (2001).

Zhong, X., Kriz, R., Seehra, J. & Kumar, R. N-linked glycosylation of platelet

P2Y12 ADP receptor is essential for signal transduction but not for ligand binding or

cell surface expression. FEBS Lett 562, 111-117, doi:10.1016/S0014-5793(04)001917 (2004).

Kozielewicz, P. et al. N-glycosylation and expression in human tissues of the orphan

GPR61 receptor. FEBS Open Bio 7, 1982-1993, doi:10.1002/2211-5463.12339

(2017).

Cherepanova, N., Shrimal, S. & Gilmore, R. N-linked glycosylation and homeostasis

of the endoplasmic reticulum. Curr Opin Cell Biol 41, 57-65,

doi:10.1016/j.ceb.2016.03.021 (2016).

Hebert, D. N., Lamriben, L., Powers, E. T. & Kelly, J. W. The intrinsic and extrinsic

effects of N-linked glycans on glycoproteostasis. Nat Chem Biol 10, 902-910,

doi:10.1038/nchembio.1651 (2014).

Brodsky, J. L. Cleaning up: ER-associated degradation to the rescue. Cell 151, 11631167, doi:10.1016/j.cell.2012.11.012 (2012).

Smith, M. H., Ploegh, H. L. & Weissman, J. S. Road to ruin: targeting proteins for

degradation in the endoplasmic reticulum. Science 334, 1086-1090,

doi:10.1126/science.1209235 (2011).

Helenius, A. & Aebi, M. Roles of N-linked glycans in the endoplasmic reticulum.

Annu Rev Biochem 73, 1019-1049, doi:10.1146/annurev.biochem.73.011303.073752

(2004).

Cormack, B. Directed mutagenesis using the polymerase chain reaction. Curr Protoc

Mol Biol Chapter 8, Unit8 5, doi:10.1002/0471142727.mb0805s37 (2001).

Doi, M. et al. Salt-sensitive hypertension in circadian clock-deficient Cry-null mice

involves dysregulated adrenal Hsd3b6. Nat Med 16, 67-74, doi:10.1038/nm.2061

(2010).

28

35

36

37

38

39

40

Hambrock, A., Loffler-Walz, C. & Quast, U. Glibenclamide binding to

sulphonylurea receptor subtypes: dependence on adenine nucleotides. Br J

Pharmacol 136, 995-1004, doi:10.1038/sj.bjp.0704801 (2002).

Yamamura, K. et al. Immunolocalization of murine type VI 3beta-hydroxysteroid

dehydrogenase in the adrenal gland, testis, skin, and placenta. Mol. Cell. Endocrinol.

382, 131-138, doi:10.1016/j.mce.2013.09.014 (2014).

Doi, M. et al. Circadian regulation of intracellular G-protein signalling mediates

intercellular synchrony and rhythmicity in the suprachiasmatic nucleus. Nat.

Commun. 2, 327, doi:10.1038/ncomms1316 (2011).

Nakagawa, S., Nguyen Pham, K. T., Shao, X. & Doi, M. Time-Restricted G-Protein

Signaling Pathways via GPR176, G(z), and RGS16 Set the Pace of the Master

Circadian Clock in the Suprachiasmatic Nucleus. Int J Mol Sci 21,

doi:10.3390/ijms21145055 (2020).

Wang, T. et al. Identification and functional characterisation of N-linked

glycosylation of the orphan G protein-coupled receptor Gpr176. Sci Rep 10, 4429,

doi:10.1038/s41598-020-61370-y (2020).

Hu, Y. et al. GWAS of 89,283 individuals identifies genetic variants associated with

self-reporting of being a morning person. Nat Commun 7, 10448,

doi:10.1038/ncomms10448 (2016).

29

Supplementary Fig. 1 | Comparison of SDS-PAGE migration profiles of WT Gpr176

and mutants containing a single Asn (N) to Gln (Q) substitution of the potential Nterminal glycosylation sites. The WT Gpr176 and respective N4Q, N11Q, N17Q, and

N26Q mutants were separately expressed in Flp-In TREx293 cells and subjected to SDSPAGE/immunoblot analysis with anti-Gpr176 antibody. A longer exposure of the same

immunoblot is shown in lower panel. A dashed white horizontal line is overlaid on the

high exposure images to emphasize the differences in size between WT and mutants. The

WT Gpr176 was recognized as a broad band with a molecular weight slightly higher than

75 kDa. The broadness of the band implies N-linked glycosylation heterogeneity. All

tested mutants exhibited a small but noticeable change in mobility, with N4Q and N26Q

evoking larger mobility changes than N11Q and N17Q. These results suggest that all four

N-terminal consensus sites under N-glycosylation.

30

Supplementary Fig. 2 (related to Fig. 3) | N-glycosylation deficiency leads to

decreased expression of GFP-fused Gpr176. Flp-In TREx293 cells expressing either

WT or N-ter mut Gpr176-GFP fusion protein were immunoblotted for Gpr176 (upper)

and β-actin (lower). The protein extracts from N-ter mut cells were loaded five-fold over

WT cells to increase the sensitivity of protein detection.

31

Supplementary Figure 3 (related to Fig. 3) | Partially overlapped localization of Nter mut Gpr176 and calnexin in Flp-In TREx293 cells. Cells expressing N-ter mut

Gpr176-GFP (green) were immunolabeled for calnexin (red). Calnexin is an ER-resident

lectin chaperone that binds with N-linked oligosaccharides containing terminal glucose

residues. Merged image is a combined image of GFP and calnexin with DAPI (blue).

Cells are representative of a population with independent experiments. Scale bars, 10 μm.

Note that in the ER, calnexin (red) and N-ter mut Gpr176 (green) were not completely

overlapped, likely consistent with the idea that N-ter mutation of Gpr176 results in the

loss of N-glycans to which calnexin binds.

32

Supplementary Fig. 4 (related to Fig. 3) | Increased level of glycosylation-deficient

Gpr176 by treatment with MG132 but not with bafilomycin A1. In this assay, I

analyzed non-tagged Gpr176 (i.e., no C-terminal GFP). Flp-In TREx293 cells that

express N-glycosylation deficient Gpr176 (i.e., N-ter mut) were treated with either

MG132 or bafilomycin A1 at the indicated concentrations for 6 hours and immunoblotted

for Gpr176 (upper) and β-Actin (lower). Arrowhead indicates the position of nonglycosylated Gpr176. Asterisk, a non-specific band (see Fig. 2B).

33

Supplementary Fig. 5 (related to Fig. 4) | Comparable basal GloSensor levels

between Dox-treated (+) and untreated (−) cells. The GloSensor system relies on the

biosensor expression. To be sure that the intracellular functional GloSensor level does not

differ between Dox-treated and non-treated cells, I verified whether their cell lysates are

able to show similar GloSensor activities when incubated in vitro with fixed amounts of

cAMP. The cells were lysed with or without (w/o) standard cAMP (final conc.: 1 or 4

µM) in buffer containing D-luciferin, Mg2+, and ATP. Luminometry showed that both

cell lysates exhibit equivalent dose-dependent GloSensor activities, providing evidence

that Dox treatment does not affect GloSensor expression in the cell. Data shown are

biological replicates (n = 2, for each condition).

34

Supplementary Fig. 6 (related to Fig. 4) | Correlation between Gpr176 protein

expression level and its cellular cAMP-repressing activity. (A) Immunoblot of Flp-In

TREx293-Gpr176 (tet-on) cells. Cells were treated with increasing doses of Dox (0. 1, 5,

10, 102, and 103 ng ml−1) and immunoblotted for Gpr176 (upper) and β-actin (lower).

Asterisk, nonspecific bands. (B) GloSensor activity traces of Flp-In TREx293-Gpr176

(tet-on) cells. Cells were treated with increasing doses of Dox (0. 1, 5, 10, 102, and 103

ng/mL). For comparison, GloSensor traces of Dox untreated cells (i.e., 0 ng ml−1) are

displayed in parallel. Data represent three independent biological replicates per condition.

RLU, relative light units. (C) Relative protein levels of Gpr176 dependent on Dox doses.

Values are the means ± s.d. (n = 3 for each) of the relative band intensities in (A). *P <

0.05, ***P < 0.005, versus Dox-untreated cells, one-way ANOVA with Bonferroni post

hoc test. (D) Relative area under the curve (AUC) of luminescence values in (B). Light

emissions were integrated and normalized with those of the untreated control. **P <

0.001, ****P < 0.0001, versus Dox-untreated cells, one-way ANOVA with

Bonferroni post hoc test. (n = 3 for each condition). Error bars indicate s.d. (E)

Relationship between protein expression level (x axis) and inhibition activity (y axis) of

Gpr176 in cells treated with different doses of Dox. Values are the means ± s.d. (n = 3 for

each). Correlation coefficient (r) and P value were calculated by Pearson product moment

correlation coefficient analysis (r = 0.9920, P<0.01). The linear regression equation is

shown at the top of figure.

35

Chapter 2

Identification of regulation of Gpr176 activity via

phosphorylation by T-207219 and other non-orphan GPCRs

Gpr176 is an SCN-enriched orphan receptor that is able to set the pace of circadian

behavioural activity rhythm. Therefore, this orphan receptor may be a promising therapeutic

target for circadian disorders such as insomnia and lifestyle-related diseases. However,

biochemical features that control the activity of Gpr176 are still largely unknown. In

Chapter 1, I showed that Gpr176 undergoes N-glycosylation, which is required for proper

cell-surface expression of Gpr176. However, N-glycosylation itself is dispensable for

Gpr176 to keep its basal (constitutive) activity. In this chapter, using a high-throughput

chemical library screening, I identified T-207219 as a modulator for Gpr176 basal activity.

Phosphorylation is a key regulatory post-translational modification described for GPCRs.

However, much less is known about phosphorylation and its potential function(s) in orphan

GPCRs. I found that T-207219 can induce phosphorylation at two separate conserved serine

residues in the C-terminal region of Gpr176, causing receptor internalization. Moreover,

using a receptor co-expression system, I obtained evidence that ligand-mediated activation of

non-orphan GPCRs leads to phosphorylation of the same residues in Gpr176. Induced

phosphorylation, importantly, caused reduced cell-surface expression and second messenger

signaling of Gpr176. My data therefore identified a previously uncharacterised inter-GPCRs

mechanism in which the orphan receptor is regulated by non-orphan receptors through

phosphorylation.

36

ACKNOWLEDGEMENTS

First and foremost, I would like to express my sincere gratitude to Prof. Masao Doi and Prof.

Hitoshi Okamura for accepting me as a member of their laboratory and giving me the

opportunity to meet many wonderful people in this world of chronobiology.

In particular, I am deeply grateful to my direct supervisor, Prof. Masao Doi, for his patient

guidance, warm encouragement, valuable discussions, and all kinds of advice in research and

life. Under Prof. Doi 's instruction, I have learned how to work independently, think logically

and solve difficulties in my research. I would not have been able to complete my Ph.D. course

without his continuous efforts.

I would like to extend my sincere thanks to Associate prof. Akira Hirasawa from the

Department of Genomic Drug Discovery Science for his insightful comments in cell line

preparation and Associate prof. Naoyuki Sugiyama from Laboratory of Molecular Systems

BioAnalysis for his suggestions and technical assistance in mass spectrometry.

I am grateful to all the members of Doi’s lab for their help in life and experiments, which

are lovely memories that I treasure. I would like to especially thank Dr. Iori Murai, Dr. Yukari

Takahashi, Mr. Genzui Setsu, Mr. Shumpei Nakagawa, Mr. Kaoru Goto and Ms. Kaho Tanaka

for their technical contributions in this study.

I would like to thank the Monbukagakusho Honors Scholarship, the AEON Scholarship,

the Kyoto University Doctoral Program (KSPD), the Fujitajinsei Scholarship generously

established by Prof. Emeritus Tetsuro Fujita and his family, and the Japan Society for the

Promotion of Science for financial support during my 10 years of study in Japan. I also thank

the Kyoto International Community for helping me with general advice on living in Japan.

To my dearest friends, Dianhui Wei, Shuheng Yan, Yalin Zhang, Chunmei Zou, Jiachen

Wang, and Yue Song, your companies made me alive.

Last but not least, I would like to express my gratitude to my parents for their unselfish

love and continuous support during my doctoral study and my life in general. Writing this

paper is the hardest thing I have ever encountered, and I cannot overcome this without them.

Thank for raising me and giving me the strength to go forward fearlessly.

37

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る