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 「Allosteric regulation accompanied by oligomeric state changes of Trypanosoma brucei GMP reductase through cystathionine-β-synthase domain」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

Allosteric regulation accompanied by oligomeric state changes of Trypanosoma brucei GMP reductase through cystathionine-β-synthase domain

Imamura Akira Okada Tetsuya Mase Hikaru Otani Takuya Kobayashi Tomoka Tamura Manatsu Kubata Bruno Kilunga Inoue Katsuaki Rambo Robert P Uchiyama Susumu Ishii Kentaro Nishimura Shigenori Inui Takashi 大阪府立大学

2021.07.29

概要

Allosteric regulation accompanied by
oligomeric state changes of Trypanosoma brucei
GMP reductase through cystathionine-β
-synthase domain
著者

journal or
publication title
volume
number
page range
year
権利

URL

Imamura Akira, Okada Tetsuya, Mase Hikaru,
Otani Takuya, Kobayashi Tomoka, Tamura
Manatsu, Kubata Bruno Kilunga, Inoue Katsuaki,
Rambo Robert P, Uchiyama Susumu, Ishii
Kentaro, Nishimura Shigenori, Inui Takashi
Nature Communications
11
Article number: 1837
1-10
2020-04-15
(C) The Author(s) 2020. This article is
licensed under a Creative Commons Attribution
4.0 International License, which permits use,
sharing, adaptation, distribution and
reproduction in any medium or format, as long
as you give appropriate credit to the original
author(s) and the source, provide a link to
the Creative Commons license, and indicate if
changes were made. The images or other third
party material in this article are included in
the article’s Creative Commons license,
unless indicated otherwise in a credit line to
the material. If material is not included in
the article’s Creative Commons license and
your intended use is not permitted by
statutory regulation or exceeds the permitted
use, you will need to obtain permission
directly from the copyright holder. To view a
copy of this license, visit
http://creativecommons.org/licenses/by/4.0/.
http://hdl.handle.net/10466/00017469
doi: https://doi.org/10.1038/s41467-020-15611-3

ARTICLE
https://doi.org/10.1038/s41467-020-15611-3

OPEN

1234567890():,;

Allosteric regulation accompanied by oligomeric
state changes of Trypanosoma brucei GMP
reductase through cystathionine-β-synthase
domain
Akira Imamura1,7, Tetsuya Okada 1,7, Hikaru Mase1, Takuya Otani1, Tomoka Kobayashi 1, Manatsu Tamura1,
Bruno Kilunga Kubata2, Katsuaki Inoue 3, Robert P. Rambo 3, Susumu Uchiyama 4,5, Kentaro Ishii 5,6,
Shigenori Nishimura 1 & Takashi Inui 1 ✉

Guanosine 5′-monophosphate reductase (GMPR) is involved in the purine salvage pathway
and is conserved throughout evolution. Nonetheless, the GMPR of Trypanosoma brucei
(TbGMPR) includes a unique structure known as the cystathionine-β-synthase (CBS) domain,
though the role of this domain is not fully understood. Here, we show that guanine and
adenine nucleotides exert positive and negative effects, respectively, on TbGMPR activity by
binding allosterically to the CBS domain. The present structural analyses revealed that
TbGMPR forms an octamer that shows a transition between relaxed and twisted conformations in the absence and presence of guanine nucleotides, respectively, whereas the
TbGMPR octamer dissociates into two tetramers when ATP is available instead of guanine
nucleotides. These findings demonstrate that the CBS domain plays a key role in the allosteric
regulation of TbGMPR by facilitating the transition of its oligomeric state depending on ligand
nucleotide availability.

1 Department

of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai,
Osaka 599-8531, Japan. 2 AU/NEPAD Agency Regional Office for Eastern and Central Africa, Nairobi, Kenya. 3 Diamond Light Source, Harwell Science and
Innovation Campus, Didcot, Oxfordshire OX11 0DE, UK. 4 Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka,
Suita, Osaka 565-0871, Japan. 5 Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama,
Myodaiji-cho, Okazaki 444-8787, Japan. 6Present address: Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka,
Suita, Osaka 565-0871, Japan. 7These authors contributed equally: Akira Imamura, Tetsuya Okada. ✉email: inuit@bioinfo.osakafu-u.ac.jp
NATURE COMMUNICATIONS | (2020)11:1837 | https://doi.org/10.1038/s41467-020-15611-3 | www.nature.com/naturecommunications

1

ARTICLE

NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-15611-3

T

he appropriate regulation of intracellular concentrations of
purine nucleotides is critical for the health of cells, and
most cells have the mechanism of either synthesizing de
novo or salvaging purine nucleotides to maintain their intracellular concentrations; nevertheless, this is not the case for most
parasitic protozoa. Trypanosoma brucei is one such parasite that
causes African trypanosomiasis by infecting to both human and
animals, and depends solely on salvaging the purines produced by
the host animals; interconversion between adenine and guanine
nucleotides is indispensable for this parasite1–4.
We recently have characterized the enzymes involved in purine
nucleotide salvage in T. brucei: guanosine 5ʹ-monophosphate
reductase (GMPR) and inosine 5ʹ-monophosphate dehydrogenase
(IMPDH)5,6. The former catalyzes the conversion of guanosine 5ʹmonophosphate (GMP) to inosine 5ʹ-monophosphate (IMP),
whereas the latter utilizes IMP to produce xanthosine 5ʹmonophosphate. Our previous studies have demonstrated that a
purine nucleotide analog, ribavirin 5ʹ-monophosphate, acts as an
inhibitor for both T. brucei GMPR (TbGMPR) and IMPDH
(TbIMPDH), but also shows an anti-trypanosomal effect in culture
when provided in the nucleoside form, ribavirin5,6. In general, both
GMPRs and IMPDHs have strong similarities in amino acid
sequence, and their catalytic domains share the common structure of
a (β/α)8 barrel, also referred to as a TIM barrel7; nevertheless, these
two enzymes are still distinctive each other by the presence or
absence of an additional domain8. This additional domain is known
as a cystathionine-β-synthase (CBS) or Bateman domain that consists of a tandem repeat of α-β-β-α folds, and is found in IMPDHs of
all organisms reported to date. The CBS domain of IMPDHs has
been shown to participate in the regulation of its activity and conformation in response to the concentrations of purine nucleotides8.
Despite of these previous findings, we and others have recently
revealed that GMPRs of trypanosomatids, including Trypanosoma
and Leishmania species, uniquely possesses a CBS domain that is
absent from the GMPRs of other species6,9, although the structure of
a GMPR harboring a CBS domain still remains undetermined.
These observations prompted us to investigate the structure and
reaction mechanism of TbGMPR.
In the present study, we investigated the biochemical and
structural effects of adenine and guanine nucleotides on TbGMPR.
We found that the binding of adenine and guanine nucleotides to
the CBS domain have opposing effect on the allosteric regulation
of TbGMPR activity. A combination of X-ray crystallography and
size-exclusion chromatography small-angle X-ray scattering (SECSAXS) analysis clearly revealed that TbGMPR can exist as a tetramer or an octamer depending on the nucleotide species that is
bound to the CBS domain. Our findings suggest that the change in
the oligomeric state of TbGMPR is responsible for allosteric regulation by nucleotide binding to the CBS domain.
Results
Opposite modulation of TbGMPR activity by purine nucleotides. Enzymes harboring a CBS domain usually change their
activities in the presence of purine nucleotides10–12; therefore, we
sought to examine whether purine nucleotides modify the activity
of TbGMPR. In the presence of GTP, the initial velocity of
TbGMPR was upregulated in a concentration-dependent manner
(Fig. 1a), and the EC50 value was estimated to be 4.8 µM. In
contrast, TbGMPR exhibited a decrease in its initial velocity in
the presence of ATP with an EC50 value of 160 µM (Fig. 1b). The
effect of each triphosphate nucleotide was maintained when
added as a magnesium complex, i.e. Mg-GTP or Mg-ATP
(Fig. 1a, b). These results indicate that TbGMPR is positively and
negatively regulated by GTP and ATP, respectively, with and
without magnesium ions. Kinetic analysis demonstrated that the
2

initial reaction velocity of TbGMPR without ligand nucleotides
showed a sigmoidal curve when plotted against the concentrations of GMP, and the plots were well-fitted to the Hill equation
(Fig. 1c, open circles). The kinetic parameters K0.5 and kcat values
for GMP were determined as 184 ± 3 µM and 16.7 ± 0.15 min−1,
respectively (mean ± s.d.; Table 1). The Hill constant (nHill) was
calculated to be 3.04 ± 0.12, meaning that GMP induced a positive
cooperativity effect on TbGMPR. A similar sigmoidal curve was
observed using recombinant TbGMPR prepared via an affinity
purification with glutathione-S-transferase (GST) tag and subsequent tag-removal (Supplementary Fig. 1). These results indicate that GMP induces a positive cooperative effect on TbGMPR,
both with and without a terminal tag.
The addition of GTP to the reaction mixture enhanced
TbGMPR activity by decreasing the K0.5 accompanied with the
increase of the kcat value in a concentration-dependent manner
(Table 1, Fig. 1c and Supplementary Fig. 2). The nHill value in the
presence of 1 mM GTP was 1.00 ± 0.34. In contrast, ATP showed
an inhibitory effect on TbGMPR activity by lowering the
substrate–enzyme affinity as indicated by the K0.5 value of 1200
± 12 µM, while the kcat and nHill values were relatively unchanged
compared to those obtained in the absence of ligands (Table 1,
Fig. 1c and Supplementary Fig. 2). The increase in the K0.5 value
was observed in an ATP concentration-dependent manner
(Supplementary Fig. 2a). The opposed effects of GTP and ATP
on TbGMPR activity were clearly observed when the values of
catalytic efficiency (kcat/K0.5) were plotted against each ligand
concentration. Although the kcat/K0.5 value of TbGMPR in the
absence of the ligands was determined as 0.913 × 105 M−1 min−1,
it was increased by the addition of 10 µM GTP, and finally
showed 4.36 × 105 M−1 min−1 with 1 mM GTP (Fig. 1d and
Table 1). In contrary, the kcat/K0.5 value was decreased by the
addition of ATP and reached to 0.111 × 105 M−1 min−1 with
1 mM ATP (Fig. 1d and Table 1). ...

この論文で使われている画像

参考文献

1.

2.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

Fish, W. R., Marr, J. J. & Berens, R. L. Purine metabolism in Trypanosoma

brucei gambiense. Biochim Biophys. Acta 714, 422–428 (1982).

Fish, W. R., Looker, D. L., Marr, J. J. & Berens, R. L. Purine metabolism in the

bloodstream forms of Trypanosoma gambiense and Trypanosoma rhodesiense.

Biochim. Biophys. Acta 719, 223–231 (1982).

Hammond, D. J. & Gutteridge, W. E. Purine and pyrimidine metabolism in

the trypanosomatidae. Mol. Biochem. Parasitol. 13, 243–261 (1984).

Berriman, M. et al. The genome of the African trypanosome Trypanosoma

brucei. Science 309, 416–422 (2005).

Bessho, T. et al. Characterization of the novel Trypanosoma brucei inosine 5’monophosphate dehydrogenase. Parasitology 140, 735–745 (2013).

Bessho, T. et al. Novel characteristics of Trypanosoma brucei guanosine 5′monophosphate reductase distinct from host animals. PLoS Negl. Trop. Dis.

10, e0004339, https://doi.org/10.1371/journal.pntd.0004339 (2016).

Hedstrom, L. The dynamic determinants of reaction specificity in the

IMPDH/GMPR family of (β/α)8 barrel enzymes. Crit. Rev. Biochem. Mol. Biol.

47, 250–263 (2012).

Hedstrom, L. IMP dehydrogenase: mechanism of action and inhibition. Chem.

Rev. 109, 2903–2928 (2009).

Smith, S. et al. The cystathionine-β-synthase domains on the guanosine 5′monophosphate reductase and inosine 5′-monophosphate dehydrogenase

enzymes from Leishmania regulate enzymatic activity in response to guanylate

and adenylate nucleotide levels. Mol. Microbiol. 100, 824–840 (2016).

Anashkin, V. A., Baykov, A. A. & Lahti, R. Enzymes regulated via

cystathionine β-synthase domains. Biochem 82, 1079–1087 (2017).

Baykov, A. A., Tuominen, H. K. & Lahti, R. The CBS domain: A protein

module with an emerging prominent role in regulation. ACS Chem. Biol. 6,

1156–1163 (2011).

Ereño-Orbea, J., Oyenarte, I. & Martínez-Cruz, L. A. CBS domains: Ligand

binding sites and conformational variability. Arch. Biochem. Biophys. 540,

70–81 (2013).

Krissinel, E. Stock-based detection of protein oligomeric states in jsPISA.

Nucleic Acids Res. 43, W314–W319 (2015).

Buey, R. M. et al. A nucleotide-controlled conformational switch modulates

the activity of eukaryotic IMP dehydrogenases. Sci. Rep. 7, 2648, https://doi.

org/10.1038/s41598-017-02805-x (2017).

Buey, R. M. et al. Guanine nucleotide binding to the Bateman domain

mediates the allosteric inhibition of eukaryotic IMP dehydrogenases. Nat.

Commun. 6, 8923, https://doi.org/10.1038/ncomms9923 (2015).

Li, J. et al. Crystal structure of human guanosine monophosphate reductase 2

(GMPR2) in complex with GMP. J. Mol. Biol. 355, 980–988 (2006).

ARTICLE

17. Anthony, S. A. et al. Reconstituted IMPDH polymers accommodate both

catalytically active and inactive conformations. Mol. Biol. Cell 28, 2600–2608

(2017).

18. Hochstetter, A. et al. Microfluidics-based single cell analysis reveals drugdependent motility changes in trypanosomes. Lab Chip 15, 1961–1968 (2015).

19. Langousis, G. & Hill, K. L. Motility and more: the flagellum of Trypanosoma

brucei. Nat. Rev. Microbiol. 12, 505–518 (2014).

20. Fairlamb, A. H. & Opperdoes, F. R. in Carbohydrate Metabolism in Cultured

Cells (ed. Morgan, M. J.) 183–224 (Springer, Boston, MA, 1986).

21. Bringaud, F., Rivière, L. & Coustou, V. Energy metabolism of

trypanosomatids: adaptation to available carbon sources. Mol. Biochem.

Parasitol. 149, 1–9 (2006).

22. Graven, P., Tambalo, M., Scapozza, L. & Perozzo, R. Purine metabolite and

energy charge analysis of Trypanosoma brucei cells in different growth phases

using an optimized ion-pair RP-HPLC/UV for the quantification of adenine

and guanine pools. Exp. Parasitol. 141, 28–38 (2014).

23. Baba, S., Hoshino, T., Ito, L. & Kumasaka, T. Humidity control and

hydrophilic glue coating applied to mounted protein crystals improves X-ray

diffraction experiments. Acta Crystallogr. Sect. D Biol. Crystallogr. 69,

1839–1849 (2013).

24. Kabsch, W. Xds. Acta Crystallogr. Sect. D Biol. Crystallogr. 66, 125–132 (2010).

25. Labesse, G., Alexandre, T., Gelin, M., Haouz, A. & Munier-Lehmann, H.

Crystallographic studies of two variants of Pseudomonas aeruginosa IMPDH

with impaired allosteric regulation. Acta Crystallogr. Sect. D Biol. Crystallogr.

71, 1890–1899 (2015).

26. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40,

658–674 (2007).

27. Vagin, A. & Teplyakov, A. MOLREP: an automated program for molecular

replacement. J. Appl. Crystallogr. 30, 1022–1025 (1997).

28. Winn, M. D. et al. Overview of the CCP4 suite and current developments. Acta

Crystallogr. Sect. D Biol. Crystallogr. 67, 235–242 (2011).

29. Adams, P. D. et al. PHENIX: A comprehensive Python-based system for

macromolecular structure solution. Acta Crystallogr. Sect. D Biol. Crystallogr.

66, 213–221 (2010).

30. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics.

Acta Crystallogr. Sect. D Biol. Crystallogr. 60, 2126–2132 (2004).

31. Murshudov, G. N., Vagin, A. A. & Dodson, E. J. Refinement of

macromolecular structures by the maximum-likelihood method. Acta

Crystallogr. Sect. D Biol. Crystallogr. 53, 240–255 (1997).

32. Basham, M. et al. Data analysis WorkbeNch (DAWN). J. Synchrotron Radiat.

22, 853–858 (2015).

33. Förster, S., Apostol, L. & Bras, W. Scatter: software for the analysis of

nano- and mesoscale small-angle scattering. J. Appl. Crystallogr. 43, 639–646

(2010).

34. Rambo, R. P. & Tainer, J. A. Accurate assessment of mass, models and

resolution by small-angle scattering. Nature 496, 477–481 (2013).

35. Konarev, P. V., Volkov, V. V., Sokolova, A. V., Koch, M. H. J. J. & Svergun, D.

I. PRIMUS: a Windows PC-based system for small-angle scattering data

analysis. J. Appl. Crystallogr. 36, 1277–1282 (2003).

Acknowledgements

We thank Drs. O. Ishibashi, T. Bessho, and T. Kodama for helpful discussions. This work

was supported by the program Grants-in-Aid for Scientific Research of the Ministry of

Education, Culture, Sports, Science and Technology of Japan (Grant Nos. 25660231,

25242046, and 17K19329 to T.I.), and Osaka Prefecture. The synchrotron radiation

experiments for the crystal structure analysis were performed under the approval of the

Japan Synchrotron Radiation Research Institute (JASRI) (Proposal Nos. 2015B2099,

2016B2561, 2016A2699, 2016B2699, 2017A2504, 2017B2504, 2017A2696, 2017B2696,

2018A2550, 2018B2550, 2018A2551, 2018B2551, 2019A2571, and 2019B2571). We

thank Diamond Light Source (Didcot, UK) for allowing access to the BioSAXS beamline

B21. All the SEC-SAXS works were done in the in-house beamtime under the approval of

Diamond Light Source. This research was supported by Joint Research by Exploratory

Research Center on Life and Living Systems (ExCELLS).

Author contributions

A.I. designed and performed the kinetic experiments and SEC-SAXS analysis, and wrote

the manuscript. Tet.O. provided support for the kinetic experiments, and wrote the

manuscript. M.T. and T.K. performed the kinetic experiments. Tak.O. and H.M. performed protein crystallization and structure solution. Kat.I. and R.R. performed SECSAXS analysis. Ken.I. and S.U. performed mass spectrometry. B.K.K. prepared materials.

S.N. provided support for protein crystallization and structure solution, and wrote the

manuscript. T.I. managed the project, designed experiments, and wrote the manuscript,

with contributions from all co-authors.

Competing interests

The authors declare no competing interests.

NATURE COMMUNICATIONS | (2020)11:1837 | https://doi.org/10.1038/s41467-020-15611-3 | www.nature.com/naturecommunications

ARTICLE

NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-15611-3

Additional information

Supplementary information is available for this paper at https://doi.org/10.1038/s41467020-15611-3.

Correspondence and requests for materials should be addressed to T.I.

Peer review information Nature Communications thanks the anonymous reviewer(s) for

their contribution to the peer review of this work. Peer reviewer reports are available.

Reprints and permission information is available at http://www.nature.com/reprints

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in

published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons

Attribution 4.0 International License, which permits use, sharing,

adaptation, distribution and reproduction in any medium or format, as long as you give

appropriate credit to the original author(s) and the source, provide a link to the Creative

Commons license, and indicate if changes were made. The images or other third party

material in this article are included in the article’s Creative Commons license, unless

indicated otherwise in a credit line to the material. If material is not included in the

article’s Creative Commons license and your intended use is not permitted by statutory

regulation or exceeds the permitted use, you will need to obtain permission directly from

the copyright holder. To view a copy of this license, visit http://creativecommons.org/

licenses/by/4.0/.

© The Author(s) 2020

10

NATURE COMMUNICATIONS | (2020)11:1837 | https://doi.org/10.1038/s41467-020-15611-3 | www.nature.com/naturecommunications

...

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