Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FAMIN ASSAY METHODS
Document Type and Number:
WIPO Patent Application WO/2020/120406
Kind Code:
A1
Abstract:
This invention relates to the finding that FAMIN (fatty acid metabolism-immunity nexus; LACC1, C13orf31)) is a trifunctional purine salvage enzyme that displays a unique combination of adenosine deaminase, purine nucleoside phosphorylase and methylthioadenosine phosphorylaseactivities. Methodsof measuring FAMIN activity and screening for FAMIN modulators are provided that comprise determining the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activityof an isolated FAMIN protein.

Inventors:
KASER ARTHUR (GB)
CADER ZAEEM (GB)
Application Number:
PCT/EP2019/084256
Publication Date:
June 18, 2020
Filing Date:
December 09, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CAMBRIDGE ENTPR LTD (GB)
International Classes:
C12Q1/34; C12Q1/48
Other References:
M ZAEEM CADER ET AL: "C13orf31 (FAMIN) is a central regulator of immunometabolic function", NATURE IMMUNOLOGY, vol. 17, no. 9, 1 August 2016 (2016-08-01), New York, pages 1046 - 1056, XP055669548, ISSN: 1529-2908, DOI: 10.1038/ni.3532
CADER M ZAEEM ET AL: "FAMIN Is a Multifunctional Purine Enzyme Enabling the Purine Nucleotide Cycle", CELL, ELSEVIER, AMSTERDAM, NL, vol. 180, no. 2, 23 January 2020 (2020-01-23), pages 278, XP085997331, ISSN: 0092-8674, [retrieved on 20200123], DOI: 10.1016/J.CELL.2019.12.017
MORIWAKI Y ET AL: "Enzymes involved in purine metabolism - A review of histochemical localization and functional implications", HISTOLOGY AND HISTOPATHOLOGY: CELLULAR AND MOLECULAR BIOLOGY, GUTENBERG, ES, vol. 14, no. 4, 1 October 1999 (1999-10-01), pages 1321 - 1340, XP009116234, ISSN: 0213-3911
SALMA M. WAKIL ET AL: "Association of a Mutation in LACC1 With a Monogenic Form of Systemic Juvenile Idiopathic Arthritis : Monogenic Systemic Juvenile Idiopathic Arthritis", ARTHRITIS & RHEUMATOLOGY (HOBOKEN), vol. 67, no. 1, 27 December 2014 (2014-12-27), US, pages 288 - 295, XP055669631, ISSN: 2326-5191, DOI: 10.1002/art.38877
"Uniprot", Database accession no. P33644
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 405 - 410
PEARSONLIPMAN, PNAS USA, vol. 85, 1988, pages 2444 - 2448
SMITHWATERMAN, J. MOL BIOI., vol. 147, 1981, pages 195 - 197
S. R. EDDY, CURRENT OPINION IN STRUCTURAL BIOLOGY, vol. 6, 1996, pages 361 - 365
NUCL. ACIDS RES., vol. 25, 1997, pages 3389 - 3402
TERPE, APPL. MICROBIOL. BIOTECHNOL., vol. 60, 2003, pages 523 - 533
FOLCH ET AL., J BIOL CHEM, vol. 226, 1957, pages 497
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 180288-69-1
CHAUHANKUMAR, MED CHEM RES, vol. 24, 2015, pages 2259
BZOWSKA ET AL., PHARMACOL THERAP, vol. 88, 2000, pages 349
FIRESTONE ET AL., ACS CHEM BIOL, vol. 12, 2017, pages 464
M. Z. CADER ET AL., NAT. IMMUNOL., vol. 17, 2016, pages 1046
L. A. O'NEILLE. J. PEARCE, J. EXP. MED., vol. 213, 2016, pages 15
G. A. PROSSERG. LARROUY-MAUMUSL. P. DE CARVALHO, EMBO REPORTS, vol. 15, 2014, pages 657
S. ZHAO ET AL., NATURE, vol. 502, 2013, pages 698
J. FOLCHM. LEESG. H. SLOANE STANLEY, J. BIOL. CHEM., vol. 226, 1957, pages 497
V. GORSHKOV ET AL., J. BASIC MICROBIOL., vol. 57, 2017, pages 998
B. J. LANDGRAFE. L. MCCARTHYS. J. BOOKER, ANNU. REV. BIOCHEM., vol. 85, 2016, pages 485
M. CAMICIS. ALLEGRINIM. G. TOZZI, FEBS J., 2018
A. S. GIZZI ET AL., NATURE, vol. 558, 2018, pages 610
G. ASSADI ET AL., PLOS ONE, vol. 11, 2016, pages e0168276
M. FRIEDKINH. M. KALCKAR: "The Enzymes", vol. 5, 1961, ACADEMIC PRESS, pages: 245
T. P. ZIMMERMANN. B. GERSTENA. F. ROSSR. P. MIECH, CAN. J. BIOCHEM., vol. 49, 1971, pages 1050
J. T. MAYNES ET AL., BIOCHEM. J., vol. 344, no. 2, 1999, pages 585
A. W. MURRAY, ANNU. REV. BIOCHEM., vol. 40, 1971, pages 811
H. ASHIHARAC. STASOLLAT. FUJIMURAA. CROZIER, PHYTOCHEMISTRY, vol. 147, 2018, pages 89
E. R. LINDLEYR. L. PISONI, BIOCHEM. J., vol. 290, no. 2, 1993, pages 457
A. BZOWSKAE. KULIKOWSKAD. SHUGAR, PHARMACOL. THER., vol. 88, 2000, pages 349
F. DELIA RAGIONE ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 223, 1996, pages 514
D. BOISON, PHARMACOL. REV., vol. 65, 2013, pages 906
M. CAMICIM. GARCIA-GILM. G. TOZZI, INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, 2018, pages 19
A. BELOQUI ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 22933
A. I. CANASS. CAMARERO, BIOTECHNOL. ADV., vol. 28, 2010, pages 694
V. PERNAJ. W. AGGERJ. HOLCKA. S. MEYER, SCIENTIFIC REPORTS, vol. 8, 2018, pages 8114
Y. KIM ET AL., PROTEINS, vol. 63, 2006, pages 1097
L. HOLMC. SANDER, PROTEINS, vol. 28, 1997, pages 72
D. K. WILSONF. B. RUDOLPHF. A. QUIOCHO, SCIENCE, vol. 252, 1991, pages 1278
E. JABRIM. B. CARRR. P. HAUSINGERP. A. KARPLUS, SCIENCE, vol. 268, 1995, pages 998
E. ALBERS, IUBMB LIFE, vol. 61, 2009, pages 1132
K. L. BRADFORDF. A. MORETTID. A. CARBONARO-SARRACINOH. B. GASPARD. B. KOHN, J. CLIN. IMMUNOL., vol. 37, 2017, pages 626
E. R. GIBLETTA. J. AMMANND. W. WARAR. SANDMANL. K. DIAMOND, LANCET, vol. 1, 1975, pages 1010
E. R. GIBLETTJ. E. ANDERSONF. COHENB. POLLARAH. J. MEUWISSEN, LANCET, vol. 2, 1972, pages 1067
K. J. MAVRAKIS ET AL., SCIENCE, vol. 351, 2016, pages 1208
T. NOBORI ET AL., PROC. NATL. ACAD. SCI. U. S. A., vol. 93, 1996, pages 6203
Z. H. CHENH. ZHANGT. M. SAVARESE, CANCER RES., vol. 56, 1996, pages 1083
Attorney, Agent or Firm:
MEWBURN ELLIS LLP (GB)
Download PDF:
Claims:
Claims

1 . A method of measuring the activity of a FAMIN protein comprising;

providing an isolated FAMIN protein; and

determining the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein.

2. A method of screening for a compound that modulates the activity of a FAMIN protein comprising; determining the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of an isolated FAMIN protein in the presence and absence of a test compound,

wherein a difference in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound is indicative that the test compound modulates the activity of the FAMIN protein inhibitor

3. A method according to claim 2 wherein a decrease in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound is indicative that the test compound is a FAMIN inhibitor

4. A method according to claim 2 wherein an increase in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound is indicative that the test compound is a FAMIN potentiator.

5. A method according to any one of the preceding claims wherein the purine nucleoside

phosphorylase activity is an adenosine phosphorylase activity.

6. A method according to any one of the preceding claims wherein the adenosine deaminase activity of the FAMIN protein is determined by measuring the conversion of an adenosine molecule into an inosine molecule in the presence of the FAMIN protein.

7. A method according to claim 6 wherein the method comprises contacting the isolated FAMIN protein with adenosine nucleoside in a reaction solution; and determining the amount of adenosine nucleoside or inosine nucleoside in the reaction solution following said contacting.

8. A method according to claim 6 or claim 7 wherein a decrease in the concentration of adenosine nucleoside or an increase in the concentration of inosine nucleoside in the reaction solution is indicative of adenosine deaminase activity.

9. A method according to any one of claims 6 to 8 wherein the adenosine molecule is adenosine and the inosine molecule is inosine.

10. A method according to any one of claims 6 to 8 wherein the adenosine molecule is deoxyadenosine and the inosine molecule is deoxyinosine.

1 1 . A method according to claim 10 wherein the adenosine molecule is 2’-deoxyadenosine and the inosine molecule is 2’deoxyinosine.

12. A method according to claim 10 wherein the adenosine molecule is 5’-deoxyadenosine and the inosine molecule is 5’deoxyinosine.

13. A method according to any one of the preceding claims wherein the purine nucleoside

phosphorylase activity of the FAMIN protein is determined by measuring the conversion of a purine nucleoside into a nucleobase and a ribose-1 -phosphate molecule in the presence of the FAMIN protein.

14. A method according to claim 13 wherein the method comprises contacting the isolated FAMIN protein with a purine nucleoside in a reaction solution; and determining the amount of purine nucleoside; or purine nucleobase and/or ribose-1 -phosphate molecule in the reaction solution following said contacting.

15. A method according to claim 14 wherein the reaction solution comprises inorganic phosphate.

16. A method according to any one of claims 13 to 15 wherein a decrease in the concentration of purine nucleoside or an increase in the concentration of purine nucleobase and/or ribose-1 -phosphate molecule in the reaction solution is indicative of purine nucleoside phosphorylase activity.

17. A method according to any one of claims 1 to 12 wherein the purine nucleoside phosphorylase activity of the FAMIN protein is determined by measuring the conversion of a nucleobase and a ribose-1 - phosphate molecule into a purine nucleoside in the presence of the FAMIN protein.

18. A method according to claim 17 wherein the method comprises contacting the isolated FAMIN protein with a nucleobase and a ribose-1 -phosphate molecule in a reaction solution; and determining the amount of purine nucleoside; or purine nucleobase and/or ribose-1 -phosphate molecule in the reaction solution following said contacting.

19. A method according to claim 17 or 18 wherein a decrease in the concentration of nucleobase and a ribose-1 -phospate molecule or an increase in the concentration of purine nucleoside in the reaction solution is indicative of purine nucleoside phosphorylase activity

20. A method according to any one of claims 13-19 wherein the purine nucleoside is guanosine, the purine nucleobase is guanine and the ribose-1 -phosphate molecule is a-D-ribose-1 -phosphate.

21 . A method according to any one of claims 13-19 wherein the purine nucleoside is adenosine, the purine nucleobase is adenine and the ribose-1 -phosphate molecule is a-D-ribose-1 -phosphate.

22. A method according to any one of claims 13-19 wherein the purine nucleoside is 2’-deoxyadenosine, the purine nucleobase is adenine and the ribose-1 -phosphate molecule is 2’ deoxy a-D-ribose-1 -phosphate.

23. A method according to any one of claims 13-19 wherein the purine nucleoside is 5’-deoxyadenosine, the purine nucleobase is adenine and the ribose-1 -phosphate molecule is 5’ deoxy a-D-ribose-1 -phosphate.

24. A method according to any one of claims 13-19 wherein the purine nucleoside is an inosine molecule.

25. A method according to claim 24 wherein the inosine molecule is inosine, the purine nucleobase is hypoxanthine and the ribose-1 -phosphate molecule is a-D-ribose-1 -phosphate

26. A method according to claim 24 wherein the inosine molecule is deoxyinosine; the purine nucleobase is hypoxanthine and the ribose-1 -phosphate molecule is 2’deoxy-a-D-ribose-1 -phosphate.

27. A method according to any one of claims 1 -26 wherein the methylthioadenosine phosphorylase activity of the FAMIN protein is determined by measuring the conversion of methylthioadenosine into adenine and a methylthioribose-1 -phosphate molecule in the presence of the FAMIN protein.

28. A method according to claim 27 wherein the method comprises contacting the isolated FAMIN protein with a methylthioadenosine nucleoside in a reaction solution; and determining the amount of methylthioadenosine nucleoside; or adenine nucleobase and/or methylthioribose-1 -phosphate molecule in the reaction solution following said contacting.

29. A method according to claim 28 wherein the reaction solution comprises inorganic phosphate.

30. A method according to claim 28 or 29 wherein a decrease in the concentration of

methylthioadenosine nucleoside or an increase in the concentration of adenine nucleobase and/or methylthioribose-1 -phosphate molecule in the reaction solution is indicative of methylthioadenosine phosphorylase activity.

31 . A method according to any one of claims 1 -26 wherein the methylthioadenosine phosphorylase activity of the FAMIN protein is determined by measuring the conversion of adenine nucleobase and a methylthioribose-1 -phosphate molecule into methylthioadenosine nucleoside in the presence of the FAMIN protein.

32. A method according to claim 31 wherein the method comprises contacting the isolated FAMIN protein with adenine nucleobase and a methylthioribose-1 -phosphate molecule in a reaction solution; and determining the amount of methylthioadenosine nucleoside; or adenine nucleobase and/or methylthioribose- 1 -phosphate molecule in the reaction solution following said contacting.

33. A method according to claim 31 or 32 wherein a decrease in the concentration of adenine nucleobase and/or methylthioribose-1 -phosphate molecule or an increase in the concentration of methylthioadenosine nucleoside in the reaction solution is indicative of methylthioadenosine phosphorylase activity.

34. A method according to any one of claims 27-33 wherein the methylthioadenosine nucleoside is 5- methylthioadenosine and the methylthioribose-1 -phosphate molecule is S-methyl-5-thio-a-D-ribose-1 - phosphate.

35 A method according to any one of claims 1 -34 comprising determining the adenosine deaminase activity and purine nucleoside phosphorylase activity of the FAMIN molecule.

36. A method according to claim 35 wherein the method comprising contacting the FAMIN protein with adenosine nucleoside in a reaction solution; and determining the amount of adenosine nucleoside or hypoxanthine and/or ribose-1 -phosphate molecule in the reaction solution following said contacting.

37. A method according to claim 36 wherein the reaction solution comprises inorganic phosphate.

38. A method according to claim 36 or 37 wherein a decrease in the concentration of adenosine nucleoside or an increase in the concentration of hypoxanthine and/or ribose-1 -phosphate molecule in the reaction solution is indicative of adenosine deaminase and purine nucleoside phosphorylase activity.

39. A method according to any one of claims 36-38 wherein the adenosine nucleoside is adenosine and the ribose-1 -phosphate molecule is a-D-ribose-1 -phosphate.

40. A method according to any one of claims 36-38 wherein the adenosine nucleoside is 2’- deoxyadenosine and the ribose-1 -phosphate is 2’deoxy- a-D-ribose-1 -phosphate.

41 . A method according to any one of claims 36-38 wherein the adenosine nucleoside is 5’- deoxyadenosine and the ribose-1 -phosphate is 5’ deoxy- a-D-ribose-1 -phosphate.

42. A method according to any one of the preceding claims comprising determining the adenosine deaminase activity, purine nucleoside phosphorylase activity and methylthioadenosine phosphorylase activity of the FAMIN protein.

43. A method according to any one of claims 1 to 41 comprising determining the adenosine deaminase activity and the methylthioadenosine phosphorylase activity of the FAMIN protein.

44. A method according to any one of claims 1 to 41 comprising determining the purine nucleoside phosphorylase activity and the methylthioadenosine phosphorylase activity of the FAMIN protein.

45. A method according to any one of claims 1 to 44 wherein the FAMIN protein is human FAMIN.

46. A method according to claim 45 wherein the FAMIN protein comprises an amino acid sequence having at least 80% identity to the sequence of amino acids 176-430 of NP_001 121775.1 .

47. A method according to claim 45 wherein the FAMIN protein comprises an lie residue at a position corresponding to position 254 of the amino acid sequence of NP_001 121775.1 (SEQ ID NO: 1).

48. A method according to claim 45 wherein the FAMIN protein comprises a Val residue at a position corresponding to position 254 of the amino acid sequence of NP_001 121775.1 (SEQ ID NO: 1). 49. A method according to any one of the preceding claims comprising identifying a test compound that inhibits FAMIN activity and optionally isolating and/or purifying the identified test compound.

Description:
FAMIN Assay Methods

Field

The present invention relates to assays for determining FAMIN protein activity and methods of screening for compounds that modulate FAMIN protein activity.

Background

The gene encoding fatty acid metabolism-immunity nexus (FAMIN; LACC1, C13orf31 ) is strongly linked to human disease. Highly penetrant mutations cause juvenile idiopathic arthritis (JIA), a spiking fever with rash and arthritis known as Still’s disease, or early-onset inflammatory bowel disease. A common coding polymorphism, I254V, carried by ~1/3 of the world’s population increases risk for Crohn’s disease (CD), an inflammatory bowel disease, and susceptibility to leprosy, a chronic infection with Mycobacterium leprae.

FAMIN, a 430 amino acid protein, has no domains with any reported biochemical function. FAMIN-deficient macrophages, however, are immunologically and metabolically compromised, irrespective of whether they rely on aerobic glycolysis or oxidative phosphorylation ( 1 , 2).

Identifying the biochemical function of orphan proteins, such as FAMIN, is a formidable challenge (3, 4).

Summary

The present inventors have unexpectedly found that FAMIN (fatty acid metabolism-immunity nexus) is a trifunctional purine salvage enzyme that displays a unique combination of adenosine deaminase, purine nucleoside phosphorylase and methylthioadenosine phosphorylase activities. In addition, the purine nucleoside phosphorylase activity of FAMIN was surprisingly found to include a unique adenosine phosphorylase activity not present in other mammalian purine nucleoside phosphorylases, and such activity has been considered absent from eukaryotic cells. Assay methods and methods of screening for FAMIN modulators have been developed based on these findings.

A first aspect of the invention provides a method of measuring the activity of a FAMIN protein comprising; providing an isolated FAMIN protein; and

determining the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the isolated FAMIN protein.

A second aspect of the invention provides a method of screening for a compound that modulates the activity of a FAMIN protein comprising;

determining the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of an isolated FAMIN protein in the presence and absence of a test compound.

A difference in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound is indicative that the test compound modulates the activity of the FAMIN protein. A decrease in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound may be indicative that the test compound is a FAMIN inhibitor. An increase in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound may be indicative that the test compound is a FAMIN potentiator.

A method of the first or second aspects may comprise determining the adenosine deaminase activity of the FAMIN protein. The adenosine deaminase activity of the FAMIN protein may be determined by measuring the conversion of an adenosine molecule into an inosine molecule in the presence of the FAMIN protein.

A method of the first or second aspects may comprise determining the purine nucleoside phosphorylase activity of the FAMIN protein. The purine nucleoside phosphorylase activity of the FAMIN protein may be determined by measuring the conversion of a purine nucleoside into a nucleobase and a ribose-1 -phosphate molecule in the presence of the FAMIN protein; or the conversion of a nucleobase and a ribose-1 -phosphate molecule into a purine nucleoside in the presence of the FAMIN protein.

The purine nucleoside phosphorylase activity of the FAMIN protein may include adenosine phosphorylase activity. In some preferred embodiments, a method of the first or second aspects may comprise determining the adenosine phosphorylase activity of the FAMIN protein. The adenosine phosphorylase activity of the FAMIN protein may be determined by measuring the conversion of adenosine into adenine and ribose-1 - phosphate molecule in the presence of the FAMIN protein; or the conversion of adenine and a ribose-1 - phosphate molecule into adenosine in the presence of the FAMIN protein.

A method of the first or second aspects may comprise determining the methylthioadenosine phosphorylase activity of the FAMIN protein. The methylthioadenosine phosphorylase activity of the FAMIN protein may be determined by measuring the conversion of methylthioadenosine into adenine and a S-methyl-5’-thioribose- 1 -phosphate molecule in the presence of the FAMIN protein; or the conversion of adenine and a S-methyl-5’- thioribose-1 -phosphate molecule into methylthioadenosine in the presence of the FAMIN protein.

Other aspects and embodiments of the invention are described in more detail below.

Brief Description of the Figures

Figure 1 shows that FAMIN is a purine nucleoside metabolising enzyme. Fig. 1 A shows a metabolomic library consisting of the aqueous phase of a methanol:choloroform (Folch) extraction of HepG2 cells 48h after transfection with FAMIN siRNA. High resolution mass spectrometry in positive and negative electrospray ionisation (ESI) modes coupled to either BEH-amide (HILIC) or C18-pfp chromatography resolved ~25,000 individual features. The left panels show representative total mass spectra, and the right panel shows them separated by molecular weight (m/z), chromatography retention time and relative levels. Fig. 1 B shows the change in relative metabolite levels in the library after incubation with either 10 pg recombinant FAMIN or protein buffer control, in 100 pi phosphate-buffered saline (PBS) for 1 h at 37°C. Volcano plot with unadjusted P value and log2 fold change, grey dots labelled with letters indicate compounds whose abundance significantly decreased (candidate substrates;‘A’-‘C’) or increased (candidate products;‘D’-‘F’) in the presence of FAMIN (n = 3 independent reactions). Figure 1 C shows representative extracted chromatograms for candidate substrates (upper) and products (lower) from Figure 1 B. Peak intensity of each extracted chromatogram of given m/z value was normalised to maximum level of 100.

Figure 1 D shows representative mass spectra and extracted chromatograms for compound‘A’ and corresponding standard for adenosine. Figure 1 E shows representative mass spectra and extracted chromatograms for putative FAMIN-catalysed metabolites B, D and F, and corresponding standards for inosine, hypoxanthine and guanine. Figure 1 F shows abundance (area under chromatographic peak, normalised for total ion content) of adenosine, inosine, hypoxanthine and ribose-1 -phosphate within the metabolomic library incubated with 10 pg recombinant FAMIN or protein buffer control (1 h , 37°C, PBS; n = 3, mean + S.E.M.). Figure 1 G shows guanosine and guanine levels following incubation of HepG2 cell aqueous extract with 10 pg recombinant FAMIN in 100 pi PBS. ( n = 3, mean + S.E.M.). *P < 0.05 and **P < 0.01 (unpaired, two-tailed Student’s f-test). Figure 1 H shows on the left side representative extracted

chromatograms for FAMIN-catalysed compound Έ’ and corresponding standards for ribose-1 -phosphate, ribose-5-phosphate, ribulose-5-phosphate and xylulose-5-phosphate. All measurements performed using a BEH amide HILIC column and TSQ Quantiva triple quadrupole. The right panel shows the ratio of selected reaction monitoring (SRM) daughter ions with nominal m/z values of 79 and 97. Figure 11 shows inosine, guanosine, cytidine, uridine and ATP levels following incubation of 0.1 , 1.0, 10.0 or 100.0 pg of recombinant FAMIN with the complete metabolomic library (aqueous phase of methanokchloroform extract of HepG2 cells) in 100 pi PBS ( n = 3, mean + S.E.M.). Figure 1J shows abundance of adenosine within the library after incubation with 0.1 , 1 .0, 10.0 or 100.0 pg of recombinant FAMIN or protein buffer control (1 h , 37°C, PBS; n = 3, mean + S.E.M.). *P < 0.05 and **P < 0.01 (unpaired, two-tailed Student’s f-test). Data are representative of at least 3 independent experiments.

Figure 2 shows that FAMIN has adenosine deaminase, purine nucleoside phosphorylase and S-methyl-5'- thioadenosine phosphorylase activities. Figure 2A shows representative extracted chromatograms for adenosine (upper chromatogram) and inosine, hypoxanthine, ribose-1 -phosphate (lower chromatogram) following incubation of 10 pg recombinant FAMIN or appropriate control with 100 pM adenosine for 1 h at 37°C in 100 pi phosphate-buffered saline (PBS). Peak intensity of each extracted chromatogram per given m/z value normalised to maximum level of 100. Controls as indicated include reaction buffer (PBS), protein elution buffer as described in methods, recombinant FAMIN without substrate or protein buffer with adenosine substrate. Figure 2B shows adenosine, inosine, hypoxanthine and ribose-1 -phosphate levels following incubation of 10 pg recombinant FAMIN or appropriate control with 100 pM adenosine as per Figure 2A (n = 3 each, mean + S.E.M.). Figure 2C, 2D, 2E, and 2F show representative chromatograms and mass spectra for (2C) adenine, (2D) hypoxanthine, (2E) inosine and (2F) ribose-1 -phosphate. Upper panels: (2C-E) extracted positive ion chromatogram showing injection of (2C) 1 pM standard of adenine; (2D) 5 pM standard of hypoxanthine; (2E) 5 pM standard of inosine on a BEH C8 column; (2F) extracted negative ion chromatogram showing injection of a 5 pg/mL standard of ribose-1 -phosphate on a BEH amide HILIC column. All measurements performed using the Q Exactive Orbitrap and extracted at m/z values of (2C) adenine [M+H] + : 136.0617, (2D) hypoxanthine [M+H] + : 137.0457, (2E) inosine [M+H] + : 269.0877 and (2F) ribose-1 -phosphate [M-H]-: 229.01 19 at a tolerance of 2 ppm. Middle panels: Corresponding extracted chromatograms of a representative sample (10 pg recombinant FAMIN 2541 incubated with 10 mM adenosine for 1 h in 100 mI phosphate-buffered saline). Peaks at indicated retention times perfectly match those of adenine, hypoxanthine, inosine and ribose-1 -phosphate standards. Lower panels: high resolution Fourier transform mass spectrometry (FTMS) mass spectrum of the representative FAMIN 254 ' incubated sample during the peaks putatively identified as (2C) adenine, (2D) hypoxanthine, (2E) inosine, or (2F) ribose-1 - phosphate. Nominal m/z values of (2C) 136, (2D) 137, (2E) 269 and (2F) 229, respectively, were selectively targeted and fragmented using a higher-energy collision dissociation (HCD) collision voltage of 25 eV to give the fragments shown. Figure 2G shows inosine, hypoxanthine and ribose-1 -phosphate levels following incubation of 10 mg recombinant FAMIN or equimolar cholesterol oxidase with 10 mM adenosine for 1 h in 100 pi PBS (n = 3 each, mean + S.E.M.). Figure 2H shows representative extracted chromatograms using a modified CSH-C18 method to allow separation and measurement of adenine following incubation of 10 pg recombinant FAMIN or appropriate control with 10 mM adenosine. Figure 2I shows fractional conversion of adenosine into adenine versus inosine versus hypoxanthine following incubation of 10 pg recombinant FAMIN with 10 mM adenosine for 1 h in 100 pi PBS. Calculated from absolute levels of hypoxanthine and inosine quantified using 13 C2, 15 Ni-hypoxanthine and 15 N 4 -inosine internal standards, and adenine levels estimated using adenine external standard. Figure 2J shows detection of [ 13 C5 15 Ns] adenine, [ 13 C IO 15 N 4 ] inosine, [ 13 Cs 15 N 4 ] hypoxanthine and [ 13 Cs] ribose-1 -phosphate levels following incubation of 10 pg recombinant FAMIN 2541 with 10 mM [ 13 Cio 15 Ns] adenosine for 1 h in 100 pi PBS (n = 3 each, mean + S.E.M.). Figure 2K shows adenosine levels in reactions of 50 mM adenine and 50 mM ribose-1 -phosphate in 100 pi PBS in the presence of 10 pg FAMIN or control (1 h, 37°C). Figure 2L shows adenine, inosine and ribose-1 - phosphate levels following incubation of 10 pg recombinant FAMIN or appropriate controls, including heat- denatured recombinant FAMIN, with 10 mM adenosine for 1 h in 100 pi PBS or HEPES buffer ( n = 3, mean + S.E.M.). Figure 2M shows FAMIN catalysed enzymatic reactions. Figure 2N shows specificity of FAMIN activity towards purine and pyrimidine nucleosides, measured as substrate (each added at 100 pM) consumption following incubation with 10 pg recombinant FAMIN or protein control in 100 pi PBS for 1 h at 37°C (SAM; S-adenosylmethionine; MTA, S-methyl-5'-thioadenosine; 2’-dA, 2’-deoxyadenosine; n = 3, mean + S.E.M.). Figure 20 shows representative extracted chromatograms for inosine, hypoxanthine and ribose-1 - phosphate (upper chromatogram); and MTA, adenine and methylthioribose-1 -phosphate (lower

chromatogram) for reactions in Figure 2N. Figure 2P and 2Q show on the left FAMIN catalysed enzymatic reactions. On the right they show levels of guanine or hypoxanthine and ribose-1 -phosphate in reactions containing 100 mM guanosine or inosine and recombinant FAMIN or buffer control in 100 pi after 1 h at 37°C ( n = 3, mean + S.E.M.). Figure 2R shows on the upper panel FAMIN catalysed enzymatic reaction. On the lower panels adenine, 2’-deoxyinosine, hypoxanthine and deoxyribose-1 -phosphate levels following incubation of 10 pg recombinant FAMIN or buffer control with 10 mM 2’deoxyadenosine for 1 h in 100 pi PBS (n = 3 each, mean + S.E.M.). Figure 2S shows adenine and deoxyribose-1 -phosphate levels following incubation of 10 pg recombinant FAMIN or buffer control with 10 pM 5’deoxyadenosine (5’dA) for 1 h in 100 pi PBS (n = 3 each, mean + S.E.M.). Figure 2T shows adenine and methylthioribose-1 -phosphate levels upon incubation of 100 mM MTA in PBS with recombinant FAMIN or buffer control for 1 h (n = 3, mean + S.E.M.). Figure 2U shows MTA levels following incubation of 0.1 , 1.0, 10.0 or 100.0 pg of recombinant FAMIN with the complete metabolomic library (aqueous phase of methanokchloroform extract of HepG2 cells) in 100 pi PBS ( n = 3, mean + S.E.M.). Figure 2V shows further FAMIN catalysed enzymatic reaction. All metabolite level data in this figure measured as area under the chromatographic peak, normalised for total ion content. *P < 0.05 (unpaired, two-tailed Student’s f-test).

Figure 3 shows the enzyme characteristics of the two FAMIN variants (FAMIN 2541 and FAMIN 254V ) that are common in humans. Figure 3A shows Michaelis-Menten enzyme kinetics for FAMIN-dependent activities using a range of 500 nM to 100 mM adenosine, inosine and MTA as substrate. Activity measured as inosine, hypoxanthine and adenine production following incubation with protein for 1 h at 37°C at pH 7.4. Figure 3B shows consumption of adenosine and Figure 3C production of adenine, inosine and ribose-1 -phosphate following incubation of 10 pg recombinant FAMIN 2541 , FAMIN 254V or buffer control with 10 mM adenosine for 1 h in 100 pi PBS. Figure 3D shows hypoxanthine levels following incubation of 10 mg recombinant FAMIN 2541 or FAMIN 254V with 10 mM adenosine for 1 h in 100 mI phosphate buffered saline (PBS), (n = 3 each, mean + S.E.M.). Figure 3E shows fractional conversion of adenosine into adenine versus inosine versus

hypoxanthine following incubation of 10 mM adenosine with 10 pg recombinant FAMIN 2541 or FAMIN 254V for 1 h in 100 pi PBS. Figure 3F shows adenosine levels following incubation of 10 pg FAMIN 2541 , FAMIN 254V or control with 50 mM adenine and 50 mM ribose-1 -phosphate for 1 h in 100 mI PBS. Figure 3G shows EC 2.4.2.1 (purine nucleoside phosphorylase) and EC 2.4.2.28 (MTA phosphorylase) activities of FAMIN 2541 and FAMIN 254V as measured by hypoxanthine and adenine following incubation of recombinant protein with 10 mM inosine and methylthioadenosine (MTA), respectively, in PBS ( n = 3, mean + S.E.M.). Figure 3H shows inosine monophosphate (IMP), hypoxanthine and guanine levels in HEK293T cells 24 hours after transient transfection with FAMIN 2541 , FAMIN 254V , FAMIN 284R plasmids or empty vector ( n = 3, mean + S.E.M.). Figure 31 shows adenosine, inosine and guanosine levels in HEK293T cells 24 hours after transient transfection with FAMIN 2541 , FAMIN 254V , FAMIN 284R plasmids or empty vector ( n = 3, mean + S.E.M.). Figure 3J shows IMP levels in control and FAMIN silenced HepG2 cells 24 h after transfection ( n = 3, mean + S.E.M.). Figure 3K shows inosine and hypoxanthine levels in control and FAMIN silenced HepG2 cells 48 h after transfection ( n = 6, mean + S.E.M.). *P < 0.05, **P < 0.01 and ***p < 0.001 (unpaired, two-tailed Student’s f-test). Figure 3L shows guanosine and S-methyl-5'-thioadenosine levels in control and FAMIN silenced HepG2 cells 48 h after transfection ( n = 6, mean + S.E.M.). ***p < 0.001 (unpaired, two-tailed Student’s f-test).

Figure 4 shows that FAMIN affects levels of purine nucleobases, nucleosides, nucleotides and cofactors in cells. Figure 4A shows adenine, adenosine and MTA levels in Famin p 2b Famin p 2bA , Famin p 28AR M1 macrophages (n = 12, mean + S.E.M.). Figure 4B shows inosine, hypoxanthine, guanosine and guanine levels in Famin p 25 Famin p 25A , Famin p 28AR M1 macrophages (n = 12, mean + S.E.M.). Figure 4C shows 2’ and 5’deoxyadenosine levels in Famin p 25 Famin p 25A , Famin p 28AR macrophages with or without treatment with interferon-a ( n = 6, mean + S.E.M.). *P < 0.05 (ANOVA; post-hoc Tukey’s multiple comparisons test). Figure 4D shows ATP, FAD levels and NAD7NADH ratio in Famin p 2bA Famin p 2bAV , Famin p 28AR M1 macrophages (n = 12, mean + S.E.M.). NAD + and NADH each quantified as relative level, hence relative ratio. Figure 4E shows the phylogenetic tree of FAMIN paralogues as generated using the HMMER database using human FAMIN protein sequence as the search input. Figure 4F shows EC 3.5.4.4 (adenosine deaminase), EC 2.4.2.1 (purine nucleoside phosphorylase) and EC 2.4.2.28 (MTA phosphorylase) activities of E. coli expressed recombinant full-length FAMIN and FAMIN A176 as measured by inosine, hypoxanthine and adenine production following incubation of protein with 10 mM adenosine, inosine and MTA, respectively, in PBS. Figure 4G shows EC 3.5.4.4 (Adenosine deaminase) and EC 2.4.2.28 (MTA phosphorylase) activities of E. coli expressed YlmD (1 T8H), YfiH (1 Z9T) as measured by inosine, and adenine and hypoxanthine production, respectively, following incubation of 10 mM adenosine or MTA in with 10 pg of recombinant proteins as indicated in PBS at 37°C for 1 h (n = 3, mean + S.E.M.).

Figure 5 shows modulation of FAMIN enzymatic function with small molecules. Figure 5A shows levels of inosine after incubating 10 pg recombinant FAMIN or buffer control with 10 pM adenosine in PBS for 1 h at 37°C in the presence or absence of pentostatin, erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA), and 8- aminoguanosine at the indicated concentrations. Figure 5B and 5C show levels of adenine and ribose-1 - phosphate (R-1 -P) in the reaction described in Figure 5A. Figure 5D shows levels of hypoxanthine after incubating 10 pg recombinant FAMIN or buffer control with 10 pM inosine in PBS for 1 h at 37°C in the presence or absence of 8-aminoguanosine. Figure 5E shows R-1 -P levels in the reaction described in Figure 5D.

Detailed Description

This invention relates to the finding that FAMIN (fatty acid metabolism-immunity nexus) has a trifunctional adenosine deaminase, purine nucleoside phosphorylase and methylthioadenosine phosphorylase activity. In addition, the purine nucleoside phosphorylase activity of FAMIN is found to include an adenosine phosphorylase activity that is unique in mammals. Assay methods to determine the activity of the FAMIN protein may comprise measuring one or more of these activities. Methods of screening for modulators of FAMIN protein may comprise measuring one or more of these activities in the presence and absence of test compound.

A FAMIN (fatty acid metabolism-immunity nexus) protein as described herein may be a eukaryotic FAMIN protein, preferably a mammalian FAMIN, such as human FAMIN.

Human FAMIN (also known as LACC1 or C13orf3; Gene ID: 14481 1) may have the amino acid sequence of NP_001 121775.1 (SEQ ID NO: 1) or a variant thereof and may be encoded by the nucleotide sequence of NM_001 128303.2 (SEQ ID NO: 2), NM_001350638.1 , NM_001350639.1 , NM_001350640.1 ,

NM_001350641 .1 or NM_001350642.1 , or a variant thereof. In some embodiments, a FAMIN protein may comprise amino acids 176-430 (FAMIN A176 ) of human FAMIN.

In some embodiments, a FAMIN protein may comprise an lie residue at a position corresponding to position 254 of the amino acid sequence of NP_001 121775.1 (SEQ ID NO: 1 ). In other embodiments, a FAMIN protein may comprise a Val residue at a position corresponding to position 254 of the amino acid sequence of NP_001 121775.1 (SEQ ID NO: 1).

A FAMIN protein as described herein may be a prokaryotic FAMIN protein, preferably a bacterial FAMIN. Bacterial FAMIN proteins may include YlmD (Uniprot P84138) from Geobacillus stearothermophilus and YfiH (Uniprot P33644) from Escherichia coli strain K12. A FAMIN protein as described herein may comprise or consist of a DUF152 domain (Pfam02578, Cluster of Orthologous Group [COG] 1496). The DUF152 domain of a FAMIN protein may be identified using standard sequence analysis techniques. For example, the DUF152 domain is located in the C terminal portion (amino acids 176-430) of human FAMIN. The DUF152 domain may be responsible for the enzymatic activity of the FAMIN protein and the sequence of the DUF152 domain may be conserved between different FAMIN proteins.

A variant of a FAMIN protein or nucleotide sequence may share at least 50% sequence identity with the wild- type FAMIN amino acid or nucleotide sequence, for example the sequence of human FAMIN or FAMIN A176 , at least 55%, at least 60%, at least 65%, at least 70%, at least about 80%, at least 90%, at least 95%, at least 98% or at least 99% sequence identity. Particular amino acid sequence variants may differ from a wild- type FAMIN protein sequence by insertion, addition, substitution or deletion of 1 amino acid, 2, 3, 4, 5, 6, 7,

8, 9, or 10 or more than 10 amino acids.

Sequence identity is commonly defined with reference to the algorithm GAP (Wisconsin GCG package, Accelerys Inc, San Diego USA). GAP uses the Needleman and Wunsch algorithm to align two complete sequences that maximizes the number of matches and minimizes the number of gaps. Generally, default parameters are used, with a gap creation penalty = 12 and gap extension penalty = 4. Use of GAP may be preferred but other algorithms may be used, e.g. BLAST (which uses the method of Altschul et al. (1990) J. Mol. Biol. 215: 405-410), FASTA (which uses the method of Pearson and Lipman (1988) PNAS USA 85: 2444-2448), the Smith-Waterman algorithm (Smith and Waterman (1981) J. Mol Biol. 147: 195-197), the TBLASTN program, of Altschul et al. (1990) supra, or HMMER (S. R. Eddy. Current Opinion in Structural Biology , 6:361 -365, 1996) generally employing default parameters. In particular, the psi-Blast algorithm may be used (Nucl. Acids Res. (1997) 25 3389-3402).

Sequence comparisons are preferably made over the full-length of the relevant sequence described herein.

In some embodiments, the FAMIN protein may be an isolated FAMIN protein, for example a recombinant FAMIN protein.

An isolated FAMIN protein may be free or substantially free of contaminants or material with which it is naturally associated, such as other components of the cells or organelles with which it is found in its natural environment, or the environment in which it is prepared (e.g. cell culture), when such preparation is by recombinant DNA technology practised in vitro or in vivo.

FAMIN protein as described herein may be provided using synthetic or recombinant techniques which are standard in the art. Alternatively, endogenous FAMIN protein may be purified from cells.

A recombinant FAMIN protein may be expressed in insoluble inclusion bodies in a prokaryotic expression system such as E coli and Lactococcus lactis. Following expression, the inclusion bodies may be isolated and solubilised with a denaturant to generate the FAMIN protein re-folded into soluble form. Alternatively, a recombinant FAMIN protein may be expressed in a eukaryotic expression system. Suitable eukaryotic host cells include mammalian cells such as CHO and CHO-derived cell lines (Lec cells), HeLa, COS, HEK293, HEK293T, and HEK-EBNA cells, amphibian cells such as Xenopus oocytes, insect cells, such as

Thchoplusia ni, Sf9 and Sf21 and yeast cells, such as Pichia pastohs. In some preferred embodiments, recombinant FAMIN protein may be expressed in a mammalian cell line, such as HEK293T.

Recombinant techniques for the expression of proteins are standard in the art. The FAMIN protein may be coupled to a signal leader peptide to direct secretion of the fusion polypeptide from a eukaryotic cell into the culture medium. A range of suitable signal leader peptides are known in the art. The signal leader peptide may be a FAMIN signal sequence or may be heterologous to the FAMIN protein i.e. it may be a non-FAMIN signal sequence. For example, an a-factor secretion signal or BiP signal sequence may be employed.

Preferably, the signal peptide is removed by post-translational processing after expression of the FAMIN protein.

The expressed FAMIN protein may be isolated and/or purified, after production. This may be achieved using any convenient method known in the art. Techniques for the purification of recombinant polypeptides are well known in the art and include, for example HPLC, FPLC, size exclusion chromatography or affinity chromatography. In some embodiments, the expressed FAMIN protein may be partially purified before use in an assay method described herein.

In some embodiments, the FAMIN protein may be produced as a fusion protein further comprising an affinity tag, which may, for example, be useful for purification. An affinity tag is a heterologous peptide sequence which forms one member of a specific binding pair. Polypeptides containing the tag may be purified by the binding of the other member of the specific binding pair to the polypeptide, for example in an affinity column. For example, the tag sequence may form an epitope which is bound by an antibody molecule. Suitable affinity tags are well known in the art and are reviewed in Terpe (2003) Appl. Microbiol. Biotechnol. 60 523- 533. The affinity tag sequence may be separated from the FAMIN protein after purification, for example, using a site-specific protease.

In some preferred embodiments, recombinant human FAMIN protein with an affinity tag may be expressed in mammalian cells, such as HEK293T cells, and then purified by affinity chromatography followed by tag cleavage and size exclusion chromatography.

In other embodiments, the FAMIN protein may be comprised within a cell extract. Adenosine phosphorylase activity, which is unique to FAMIN in mammalian cells, may be determined in the cell extract. Other purine nucleoside phosphorylase activities, as well as adenosine deaminase and methylthioadenosine

phosphorylase activity may be determined in the cell extract relative to a control cell extract and/or in the presence of selective inhibition of other cellular enzymes with the determined activity, such as adenosine deaminase (ADA), methylthioadenosine phosphorylase (MTAP), and purine nucleoside phosphorylase (PNP).

A control cell extract may be obtained from a cell in which FAMIN is inactivated. FAMIN may be inactivated in a control cell by any convenient means, such as RNA silencing using RNAi transfection or gene knock-out. In other embodiments, a control cell extract may be obtained from a cell which expresses a FAMIN-I254V mutant. The adenosine phosphorylase activity of the FAMIN-I254V mutant is selectively inactivated and a control cell extract comprising the FAMIN-I254V mutant may be useful in screening for compounds that have relative selectivity for adenosine phosphorylase activity.

One, two or all three of the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the isolated FAMIN protein may be determined. In some embodiments, these activities may be determined individually, for example to screen for selective inhibitors of an individual activity.

The adenosine deaminase activity of FAMIN converts adenosine nucleosides into inosine nucleosides (EC3.5.4.4). Adenosine deamination activity may be determined by;

contacting the FAMIN protein with an adenosine nucleoside in a reaction solution; and

determining the amount of adenosine nucleoside, inosine nucleoside or NFh in the reaction solution, following said contacting.

Depletion of adenosine nucleoside or generation of NFh or inosine nucleoside in the reaction solution is indicative of the adenosine deamination activity of the FAMIN protein. The amount of decrease in the concentration of adenosine nucleoside or increase in the concentration of inosine nucleoside in the reaction solution or the amount of NFh generated may be indicative of the amount of adenosine deamination activity in the reaction solution. Depletion of adenosine nucleoside or the generation of NFh or the inosine nucleoside in the reaction solution may be determined in the presence relative to the absence of the isolated FAMIN protein.

An adenosine nucleoside may include adenosine or an analogue or derivative thereof. An inosine nucleoside may include inosine or an analogue or derivative thereof.

In some embodiments, the adenosine nucleoside is adenosine (PubChem CID 60961) and the inosine nucleoside is inosine (CID 6021 ). In other embodiments, the adenosine nucleoside is deoxyadenosine and the inosine nucleoside is deoxyinosine. For example, the adenosine nucleoside may be 2’-deoxyadenosine (CID 13730) and the inosine nucleoside may be 2’-deoxyinosine (CID 65058) or the adenosine nucleoside may be 5’-deoxyadenosine (CID 439182) and the inosine nucleoside may be 5’-deoxyinosine (CID

14693755).

The purine nucleoside phosphorylase activity of FAMIN converts purine nucleosides into purine nucleobases and ribose-1 -phosphate molecules in the presence of phosphate (e.g. orthophosphate PO4 3 ). For example, inosine nucleosides may be converted into hypoxanthine and a-D-ribose-1 -phosphate (EC2.4.2.1) and guanosine nucleosides may be converted into guanine and a-D-ribose-1 -phosphate (EC2.4.2.15) by the FAMIN protein. In addition, adenosine nucleosides may be converted by a FAMIN protein into adenine and a-D-ribose-1 -phosphate (EC2.4.2.1 KEGG R01561 ; adenosine: phosphate a-D-ribosyltransferase; also known as adenosine phosphorylase) by the FAMIN protein. The reaction is reversible and the purine nucleoside phosphorylase activity of FAMIN may also convert a purine nucleobase and ribose-1 -phosphate molecule into a purine nucleoside and phosphate.

The purine nucleoside phosphorylase activity of the FAMIN protein may be determined by measuring the conversion of purine nucleoside and phosphate into a nucleobase and a ribose-1 -phosphate molecule; or a nucleobase and a ribose-1 -phospate molecule into a purine nucleoside and phosphate in the presence of the FAMIN protein.

In some embodiments, purine nucleoside phosphorylase activity may be determined by;

contacting the FAMIN protein with a purine nucleoside in a reaction solution; and

determining the amount of purine nucleoside; or purine nucleobase and/or ribose-1 -phosphate molecule in the reaction solution following said contacting.

The FAMIN protein may be contacted with the purine nucleoside in the presence of inorganic phosphate.

For example, the reaction solution may comprise phosphate buffered saline. In some embodiments, the amount of phosphate in the reaction solution following said contacting may be determined.

Depletion of purine nucleoside and/or phosphate or generation of the purine nucleobase and/or ribose-1 - phosphate molecule in the reaction solution is indicative of the purine nucleoside phosphorylase activity of the FAMIN protein. The amount or rate of decrease in the concentration of purine nucleoside or increase in the concentration of purine nucleobase and/or ribose-1 -phosphate molecule in the reaction solution is indicative of the amount of purine nucleoside phosphorylase activity in the reaction solution. Depletion of purine nucleoside or generation of the purine nucleobase and/or ribose-1 -phosphate molecule in the reaction solution may be determined in the presence relative to the absence of the isolated FAMIN protein.

In some embodiments, purine nucleoside phosphorylase activity may be determined by;

contacting the FAMIN protein with purine nucleobase and a ribose-1 -phosphate molecule in a reaction solution; and

determining the amount of purine nucleoside and/or phosphate; or purine nucleobase and/or ribose- 1 -phosphate molecule in the reaction solution following said contacting.

Depletion of purine nucleobase and/or ribose-1 -phosphate molecule or generation of the purine nucleoside in the reaction solution by the FAMIN protein is indicative of purine nucleoside phosphorylase activity. The amount or rate of decrease in the concentration of purine nucleobase and/or ribose-1 -phosphate molecule or increase in the concentration of purine nucleoside in the reaction solution may be indicative of the amount of purine nucleoside phosphorylase activity in the reaction solution. Depletion of purine nucleobase and/or ribose-1 -phosphate molecule or generation of the purine nucleoside in the reaction solution may be determined in the presence relative to the absence of the isolated FAMIN protein.

A purine nucleoside may include guanosine, adenosine and inosine molecule, or analogues or derivatives thereof. An inosine nucleoside may include inosine, deoxy-inosine, or analogues or derivatives thereof. In some embodiments, the FAMIN protein may be contacted with multiple different purine nucleosides or purine nucleobases, either simultaneously or in series, and the depletion of the multiple purine nucleosides or purine nucleobases and/or the generation of multiple reaction products may be determined.

In some embodiments, the purine nucleoside is an inosine molecule. For example, the purine nucleoside may be inosine (CID 6021), the purine nucleobase may be hypoxanthine (CID 790) and the ribose-1 - phosphate molecule may be a-D-ribose-1 -phosphate (CID 439236). Alternatively, the purine nucleoside may be deoxy-inosine; the purine nucleobase may be hypoxanthine and the ribose-1 -phosphate molecule may be 2’deoxy-a-D-ribose-1 -phosphate (CID 439287).

In other embodiments, the purine nucleoside is guanosine (CID 6802), the purine nucleobase is guanine and the ribose-1 -phosphate molecule is a-D-ribose-1 -phosphate. In other embodiments, the purine nucleoside is adenosine (CID 60961), the purine nucleobase is adenine and the ribose-1 -phosphate molecule is a-D- ribose-1 -phosphate.

The purine nucleoside phosphorylase activity of FAMIN may convert adenosine into adenine and a ribose-1 - phosphate molecule (i.e. adenosine phosphorylase activity). A FAMIN protein may therefore convert adenosine into inosine through its adenosine deaminase activity or adenine through its adenosine phosphorylase activity. Inosine may be further converted into hypoxanthine by the FAMIN protein, as described above.

The adenosine phosphorylase activity may be determined by;

contacting the FAMIN protein with adenosine in a reaction solution; and

determining the amount of adenosine or the amount of adenine and/or ribose-1 -phosphate molecule in the reaction solution following said contacting.

The FAMIN protein may be contacted with the adenosine in the presence of inorganic phosphate. For example, the reaction solution may comprise phosphate buffered saline. In some embodiments, the amount or rate of phosphate in the reaction solution following said contacting may be determined. Depletion of adenosine and/or phosphate or generation of adenine and/or ribose-1 -phosphate in the reaction solution by the FAMIN protein is indicative of adenosine phosphorylase activity. For example, a decrease in the concentration of adenosine or an increase in the concentration of adenine and/or ribose-1 -phosphate in the reaction solution may be indicative of activity.

In some embodiments, depletion of adenosine and/or phosphate or generation of adenine and/or ribose-1 - phosphate in the reaction solution may be determined in the presence relative to the absence of the isolated FAMIN protein. In other embodiments, depletion of adenine and/or ribose-1 -phosphate molecule or generation of adenosine and/or phosphate in the reaction solution may be determined in the presence of wild type FAMIN protein relative to the FAMIN-I254V mutant, which has selectively impaired adenosine phosphorylase activity.

In some embodiments, adenosine phosphorylase activity may be determined by; contacting the FAMIN protein with adenine and a ribose-1 -phosphate molecule in a reaction solution; and

determining the amount of adenosine and/or phosphate; or adenine and/or ribose-1 -phosphate molecule in the reaction solution following said contacting.

Depletion of adenine and/or ribose-1 -phosphate molecule or generation of adenosine and/or phosphate in the reaction solution by the FAMIN protein is indicative of adenosine phosphorylase activity. The amount or rate of decrease in the concentration of adenine and/or ribose-1 -phosphate molecule or increase in the concentration of adenosine and/or phosphate in the reaction solution may be indicative of the amount of adenosine phosphorylase activity in the reaction solution.

In some embodiments, depletion of adenine and/or ribose-1 -phosphate molecule or generation of the adenosine and/or phosphate in the reaction solution may be determined in the presence relative to the absence of the isolated FAMIN protein. In other embodiments, depletion of adenine and/or ribose-1 - phosphate molecule or generation of adenosine and/or phosphate in the reaction solution may be determined in the presence of wild type FAMIN protein relative to the FAMIN-I254V mutant, which has selectively impaired adenosine phosphorylase activity.

Methylthioadenosine phosphorylase activity converts methylthioadenosine nucleosides into adenine nucleobases and S-methyl-5’-thioribose-1 -phosphate (i.e. methylthioribose-1 -phosphate) molecules. A methylthioadenosine nucleoside may include S-methyl-5’-thioadenosine (i.e. 5-methylthioadenosine) and analogues or derivatives thereof. An adenine nucleobase may include adenine and analogues or derivatives thereof. A methylthioribose-1 -phosphate molecule may include S-methyl-5-thio-a-D-ribose-1 -phosphate and analogues or derivatives thereof. For example, 5-methylthioadenosine (CID 149) may be converted into adenine (CID 190) and S-methyl-5-thio-a-D-ribose-1 -phosphate (CID 45266677) (EC2.4.2.28). The reaction is reversible and methylthioadenosine phosphorylase activity may also convert adenine nucleobases and methylthioribose-1 -phosphate molecules into methylthioadenosine nucleosides.

In some embodiments, methylthioadenosine phosphorylase activity may be determined by;

contacting the FAMIN protein with a methylthioadenosine nucleoside in a reaction solution; and determining the amount of methylthioadenosine nucleoside; or adenine nucleobase and/or methylthioribose-1 -phosphate molecule in the reaction solution following said contacting.

The FAMIN protein may be contacted with the methylthioadenosine nucleoside in the presence of inorganic phosphate. For example, the reaction solution may comprise phosphate buffered saline. In some embodiments, the amount of phosphate in the reaction solution following said contacting may be determined.

Depletion of methylthioadenosine nucleoside or generation of the adenine nucleobase and/or

methylthioribose-1 -phosphate molecule in the reaction solution by the FAMIN protein is indicative of methylthioadenosine phosphorylase activity. For example, the amount of decrease in the concentration of methylthioadenosine nucleoside or increase in the concentration of adenine nucleobase and/or

methylthioribose-1 -phosphate molecule in the reaction solution is indicative of the amount of methylthioadenosine phosphorylase activity in the reaction solution. Depletion of methylthioadenosine nucleoside or generation of the adenine nucleobase and/or methylthioribose-1 -phosphate molecule in the reaction solution may be determined in the presence relative to the absence of the isolated FAMIN protein.

The adenosine deaminase and purine nucleoside phosphorylase activities of FAMIN in combination may convert adenosine nucleosides into hypoxanthine and a ribose-1 -phosphate molecule.

Adenosine deaminase and purine nucleoside phosphorylase activity may be determined by;

contacting the FAMIN protein with adenosine nucleoside in a reaction solution; and

determining the amount of adenosine nucleoside or hypoxanthine and/or ribose-1 -phosphate molecule in the reaction solution following said contacting.

The FAMIN protein may be contacted with the adenosine nucleoside in the presence of inorganic phosphate (e.g. orthophosphate). For example, the reaction solution may comprise phosphate buffered saline. In some embodiments, the amount of phosphate in the reaction solution following said contacting may be determined

Depletion of adenosine nucleoside or generation of hypoxanthine and/or ribose-1 -phosphate in the reaction solution by the FAMIN protein is indicative of combined adenosine deamination and purine nucleoside phosphorylase activity. For example, a decrease in the concentration of adenosine nucleoside or an increase in the concentration of hypoxanthine and/or ribose-1 -phosphate in the reaction solution is indicative of the combined activity. Depletion of adenosine nucleoside or generation of hypoxanthine and/or ribose-1 - phosphate in the reaction solution may be determined in the presence relative to the absence of the isolated FAMIN protein.

In some embodiments, the adenosine nucleoside is adenosine and the ribose-1 -phosphate is a-D-ribose-1 - phosphate. In other embodiments, the adenosine nucleoside is deoxyadenosine and the ribose-1 -phosphate is deoxy- a-D-ribose-1 -phosphate. For example, the adenosine nucleoside may be 2’-deoxyadenosine and the ribose-1 -phosphate may be 2’-deoxy- a-D-ribose-1 -phosphate or the 5’-deoxyadenosine and the ribose- 1 -phosphate may be 5’-deoxy- a-D-ribose-1 -phosphate.

In some embodiments, the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of an isolated FAMIN protein may be determined in the presence and absence of test compound. A decrease in activity in the presence relative to the absence of test compound is indicative that the test compound is a FAMIN inhibitor. This may be useful in screening for compounds that inhibit FAMIN as described below.

In some embodiments, the reactants in the reactions described herein may be isolated molecules. Suitable reactant molecules may be synthesised using conventional techniques or obtained from commercial suppliers (e.g. Sigma-Aldrich Corp, St. Louis, MO, USA). In other embodiments, the reactants in the reactions described herein may be within a cell extract, for example an aqueous phase extract of cell metabolites (as shown for example in Figures 1A-1 F). Suitable cell extracts may be devoid of protein and may include for example aqueous extracts obtained by Folch extraction (Folch et al., J Biol Chem 1957, 226, 497). An aqueous extract of reactants may be freeze-dried and re-suspended in phosphate-buffered saline.

The reactions may be performed in a reaction solution comprising inorganic phosphate (Pi). For example a phosphate containing buffer, such as PBS, may be used. The reaction solution may be devoid of co-factors or reactants other than inorganic phosphate.

The reactions may be performed at any convenient temperature, for example 32°C to 42°C, such as 37°C.

The reactions may be performed at any convenient pH, for example pH 7-8, such as pH 7.4.

The depletion of a substrate or the generation of a product in the adenosine deaminase, purine nucleoside phosphorylase and/or methylthioadenosine phosphorylase reactions described herein may be determined by any convenient technique. Suitable techniques include, for example, absorbance, chromatography-coupled mass spectrometry (e.g. LC-MS/MS) or NMR. In some preferred embodiments, the reactants or the generation of products in the reaction solution may be measured by LC-MS/MS.

In some embodiments, depletion of a substrate or the generation of a product in a FAMIN-mediated adenosine deaminase, purine nucleoside phosphorylase and/or methylthioadenosine phosphorylase reaction described herein may be determined by measuring absorbance. The substrates and products described above may display different absorbance spectra and changes in absorbance at one or more selected wavelengths in the reaction solution may be indicative of the depletion of a substrate or the generation of a product.

In some embodiments, the depletion of a substrate or generation of product in a FAMIN-mediated reaction described herein may be determined using a substituted analogue of the substrate or product which displays characteristic spectrophotometric properties, such that the concentration of the analogue in the reaction solution can be directly measured.

In some embodiments, a substrate may be labelled with a detectable label. Conversion of the substrate into products by the FAMIN protein as described herein may release the detectable label, which may then be measured by any convenient means.

In some embodiments, the depletion of substrate or the generation of product may be determined using a detection reaction which generates a detectable compound in the presence of the reactant or product.

For example, the depletion or generation of xanthine may be determined using xanthine oxidase that generates H2O2, which can be detected by detection reagents, such as luminescence or fluorescence reagents. The detection reaction may be coupled with the FAMIN mediated reaction. For example, both FAMIN and xanthine oxidase may be present in the reaction solution. Alternatively, the detection reaction may be performed after the FAMIN mediated reaction. Alternatively, the depletion or generation of inorganic phosphate may be determined using purine nucleoside phosphorylase (PNP). Suitable assay methods and kits are well known in the art and commercially available (for example EnzChek® Phosphate Assay Kit, ThermoFisher Scientific). For example, inorganic phosphate in the reaction solution may be determined by the conversion of the 2-amino-6-mercapto-7-methylpurine riboside (MESG) into ribose 1 -phosphate and 2-amino-6-mercapto-7-methylpurine product, which can be detected through a spectrophotometric shift in maximum absorbance from 330 nm (substrate) to 360 nm (product).

Methods of measuring FAMIN activity as described herein may be useful in screening for compounds that modulate, e.g. promote or inhibit, FAMIN activity.

In some embodiments, screening may be performed using an isolated FAMIN protein. For example, a method of screening for a compound that modulates the activity of a FAMIN protein may comprise;

determining the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of an isolated FAMIN protein in the presence and absence of a test compound.

A difference in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein that is measured in the presence of test compound relative to the absence of test compound may be indicative that the test compound modulates the activity of FAMIN protein.

A decrease in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound is indicative that the test compound is a FAMIN inhibitor or antagonist.

An increase in the adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of the FAMIN protein in the presence relative to the absence of test compound is indicative that the test compound is a FAMIN potentiator or agonist.

The adenosine deaminase activity, purine nucleoside phosphorylase activity and/or methylthioadenosine phosphorylase activity of an isolated FAMIN protein may be determined as described above.

An increase or decrease in one or more of (i) the conversion of an adenosine molecule into an inosine molecule, (ii) the conversion of a purine nucleoside into a nucleobase and a ribose-1 -phosphate molecule in the presence of the FAMIN protein, (iii) the conversion of a nucleobase and a ribose-1 -phosphate molecule into a purine nucleoside (iv) the conversion of adenosine into adenine and a ribose-1 -phosphate molecule in the presence of the FAMIN protein, (v) the conversion of adenine and a ribose-1 -phosphate molecule into adenosine (vi) the conversion of methylthioadenosine into adenine and a S-methyl-5’-thioribose-1-phosphate molecule, or (vii) the conversion of adenine and a S-methyl-5’-thioribose-1 -phosphate molecule into methylthioadenosine; by the FAMIN protein in the presence relative to the absence of the test compound is indicative that the test compound is a FAMIN potentiator or inhibitor, respectively. The precise format of any of the screening or assay methods of the present invention may be varied by those of skill in the art using routine skill and knowledge. The skilled person is well aware of the need to employ appropriate control experiments. For example, in some embodiments, the amount of the above compounds may also be determined in a control in which FAMIN is inactivated.

A test compound may be an isolated molecule or may be comprised in a sample, mixture or extract, for example, a biological sample. Compounds which may be screened using the methods described herein may be natural or synthetic chemical compounds used in drug screening programmes. Extracts of plants, microbes or other organisms, which contain several characterised or uncharacterised components may also be used.

Combinatorial library technology provides an efficient way of testing a potentially vast number of different compounds for ability to modulate FAMIN activity. Such libraries and their use are known in the art, for all manner of natural products, small molecules and peptides, among others. The use of peptide libraries may be preferred in certain circumstances

The amount of test compound which may be added to an assay of the invention will normally be determined by trial and error depending upon the type of compound used. Typically, from about 0.001 nM to 1 mM or more concentrations of putative inhibitor compound may be used, for example from 0.01 nM to 100pM, e.g. 0.1 to 50 pM, such as about 10 pM. Even a compound which has a weak effect may be a useful lead compound for further investigation and development.

Suitable test compounds for screening include compounds that inhibit similar activities to the adenosine deaminase, purine nucleoside phosphorylase and methylthioadenosine phosphorylase activities of FAMIN. Suitable test compounds include ADA inhibitors, such as 3'-deoxy-N-(1 -oxododecyl) adenosine, pentostatin (PubchemID 439693, CAS ID 53910-25-1) and EHNA (erythro-9-(2-hydroxy-3-nonyl)adenine (PubchemID 3206, CAS ID 59262-86-1) (see for example Chauhan and Kumar (2015) Med Chem Res 24:2259); PNP inhibitors, such as 8-aminoguanosine (PubchemID 96849; CAS ID 180288-69-1) (see for example Bzowska et al (2000) Pharmacol Therap 88:349); MTAP inhibitors, such as MT-lmmA and MT-DADMe-lmmA (MTDIA) (see for example Firestone et al (2017) ACS Chem Biol 12:464); and purine nucleosides.

Suitable test compounds also include analogues, derivatives, variants and mimetics of any of the compounds listed above, for example compounds produced using rational drug design to provide test candidate compounds with particular molecular shape, size and charge characteristics suitable for modulating FAMIN activity.

A test compound identified as modulating FAMIN activity may be investigated further. For example, the selectivity of a compound for FAMIN may be determined by screening against other isolated ADA, PNP or MTAP enzymes. Suitable methods for determining the effect of a compound on the activity of recombinant enzymes are well known in the art. A test compound identified as modulating FAMIN activity may be isolated and/or purified or alternatively, it may be synthesised using conventional techniques of recombinant expression or chemical synthesis.

Furthermore, it may be manufactured and/or used in preparation, i.e. manufacture or formulation, of a composition such as a medicament, pharmaceutical composition or drug. Methods described herein may thus comprise formulating the test compound in a pharmaceutical composition with a pharmaceutically acceptable excipient, vehicle or carrier for therapeutic application.

Following identification of a compound which modulates FAMIN activity as described herein, a method may further comprise modifying the compound to optimise its pharmaceutical properties. Suitable methods of optimisation, for example by structural modelling, are well known in the art.

Further optimisation or modification can then be carried out to arrive at one or more final compounds for in vivo or clinical testing.

Other aspects and embodiments of the invention provide the aspects and embodiments described above with the term“comprising” replaced by the term“consisting of and the aspects and embodiments described above with the term“comprising” replaced by the term’’consisting essentially of.

It is to be understood that the application discloses all combinations of any of the above aspects and embodiments described above with each other, unless the context demands otherwise. Similarly, the application discloses all combinations of the preferred and/or optional features either singly or together with any of the other aspects, unless the context demands otherwise.

Modifications of the above embodiments, further embodiments and modifications thereof will be apparent to the skilled person on reading this disclosure, and as such, these are within the scope of the present invention.

All documents and sequence database entries mentioned in this specification are incorporated herein by reference in their entirety for all purposes.

“and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. For example“A and/or B” is to be taken as specific disclosure of each of (i) A, (ii) B and (iii) A and B, just as if each is set out individually herein.

Experimental

Materials and Methods

Cell lines

HepG2 (ATCC HB-8065) and HEK293T (ATCC CRL-3216) cell lines were maintained in complete DMEM with 10% FBS. Sanger sequencing of the FAMIN gene identified the HepG2 cell line as heterozygous at SNP rs3764147, encoding FAMIN (p.254l/p.254V). Silencing experiments were carried out using reverse transfection with Lipofectamine RNAiMAX reagent and 25nM of small interfering RNA (siRNA) according to the manufacturer’s guidelines. HepG2 cells were silenced using siRNA (SMART pool technology; Dharmacon) against human FAMIN (LACC1 ; M-015653-00). Silencing efficiency was verified using qPCR and gene expression confirmed to be reduced by at least 80% at 48 hours following transfection. Nontargeting siRNA (Dharmacon) was used as negative control.

Extraction of aqueous and lipid metabolites

Cells were harvested as indicated using either direct extraction with 4:1 methanofwater or Trypsin-EDTA (0.25%). For direct methanol extraction, cells were washed with PBS followed by addition of pre-chilled 500 pi 4:1 methanofwater. Cells were scraped and the resulting methanol mixture transferred to 2 mL-flat- bottomed screw cap tubes. Samples were then vortexed, sonicated and lastly centrifuged at 21 ,000 g for 10 minutes to pellet any debris. The supernatant was transferred to new 2 ml_ tubes for drying as described below. All solvents used were HPLC grade or higher and obtained from Honeywell (Fisher Scientific). Cell pellets harvested using trypsinization were washed with PBS and then subjected to extraction using the methanoLchloroform method described by Folch (5). Briefly, a stainless steel ball (Qiagen) was added to each washed cell pellet on dry ice along with 1 ml_ of ice cold 2:1 chlorofornrmethanol inside a 2 mL-flat- bottomed screw cap tube (Starlab). The samples were homogenised using a Tissue Lyser (Qiagen) for 10 min at 25 Hz to ensure optimum extraction and ascertaining to freeze the tissue lyser plates prior to homogenisation in order to keep samples cold during extraction. 400 pi of ice cold water was added and the samples thoroughly vortexed and sonicated for 5 minutes before centrifugation at 21 ,000 g for 5 min. After centrifugation the aqueous (top layer) and organic (bottom layer) fractions were separated and aliquoted into separate screw-cap tubes both kept on dry ice. A further 1 ml_ of 2:1 chlorofornrmethanol was added to the original tube containing the protein pellet and the extraction repeated as described above. The resulting layers were combined, dried (as described below) and stored at -20°C prior to further preparation and analysis.

LC-MS sample preparation

Aqueous extracts of cells or protein reaction mixtures were lyophilised using a centrifugal evaporator (Labconco) and reconstituted in 100 pi (for cell extracts) and 200 mI (for protein reaction mixtures) of 70:30 acetonitrile: 0.1 M aqueous ammonium carbonate water containing 2 mM [ 13 Cio, 15 Ns] adenosine monophosphate, adenosine triphosphate [ 13 Cio, 15 Ns], succinic acid 13 Cio and glutamic acid [ 13 Cs, 15 Ns] (all from Sigma Aldrich except the glutamic acid from Cambridge Isotope Laboratories) as internal standards, although it should be noted that for experiments using labelled substrates internal standards were omitted to avoid contamination of metabolites. The resulting solution was vortexed then sonicated for 15 min followed by centrifugation at 21 ,000 g to pellet any remaining undissolved material. After centrifugation the supernatant was transferred with an automatic pipette into a 300 mI vial with insert (Fisher Scientific) and capped ready for analysis.

LC-MS analysis of aqueous metabolites

For untargeted analysis, a Velos Pro Elite orbitrap mass spectrometer coupled to a U3000 chromatography system or a Q Exactive Plus orbitrap coupled to a Vanquish Horizon ultra-high performance liquid chromatography system was used. For targeted analysis, samples were analysed using a Quantiva triple stage quadrupole mass spectrometer coupled to a Vanquish Horizon (all analytical instrument combinations supplied by Thermo Fisher Scientific). Samples were then analysed using a bridged ethylene hybrid (BEH) amide hydrophilic interaction liquid chromatography (HILIC) approach for the highly polar aqueous metabolites. For this analysis the strong mobile phase (A) was 100 mM ammonium carbonate, the weak mobile phase was acetonitrile (B) with 9:1 water: acetonitrile being used for the needle wash. The LC column used was the BEH amide column (150 x 2.1 mm, 1 .7 pm, Waters). The following linear gradient was used: 20% A in acetonitrile for 1 .5 min followed by an increase to 60% A over 2.5 min with a further 1 min at 60% A after which the column was reequilibrated for 1 .9 min. After each chromatographic run the column was washed with 30 column volumes of 6:4 watenacetonitrile followed by a further 10 column volumes of 95:5 acetonitrile:water for storage. The total run time was 7 min, the flow rate was 0.6 mL/min and the injection volume was 5 pL. In order to resolve pentose phosphates for identification of ribose-1 -phosphate a shallower gradient was employed: 30% A in acetonitrile for 2.0 minutes followed by an increase to 50% A over 3.0 minutes with re-equilibration for 1 .9 minutes. After HILIC analysis samples were dried and reconstituted in the same volume of 10 mM ammonium acetate prior to orthogonal mixed mode analysis using an ACE Excel C18-PFP column

(150 x 2.1 mm, 2.0 pm, Hichrom). Mobile phase A consisted of water with 0.1 % formic acid and mobile phase B was acetonitrile with 0.1 % formic acid. For gradient elution mobile phase B was held at 0% for 1 .6 min. followed by a linear gradient to 30% B over 4.0 minutes, a further increase to 90% over 1 min and a hold at 90% B for 1 min with re-equilibration for 1 .5 minutes giving a total run time of 6.5 minutes. The flow rate was 0.5 mL/min and the injection volume was 2 pL. The needle wash used was 4:1 watenacetonitrile with 0.1 % formic acid. Both chromatography modes were used for both targeted and untargeted analysis. For enzyme kinetic assays a further chromatographic approach was used to allow for analysis of all substrates and products using a single assay. A Waters BEH C8 column was used (100 c 2.1 mm, 1 .7 pm) with a weak mobile phase (A) of aqueous 10 mM ammonium acetate with 0.1 % ammonia and a strong mobile phase (B) of acetonitrile. For gradient elution mobile phase B was held at 0% for 1 .6 min, followed by a linear gradient to 30% B over 4.0 minutes, a further increase to 90% over 1 min, and a hold at 90% B for 1 min, with re-equilibration for 1 .5 minutes giving a total run time of 6.5 minutes. The flow rate was 0.5 mL/min and the injection volume was 2 pL. The needle wash used was 4:1 watenacetonitrile.

Untargeted analysis on the Elite used a high resolution FTMS full scan of 60-1500 m/z with a resolution of 60,000 ppm where due to positive mode negative mode equilibration times each mode was run

independently. Source parameters used for the orbitrap were a vaporizer temperature of 400°C and ion transfer tube temperature of 300°C, an ion spray voltage of 3.5 kV (2.5 kV for negative ion mode) and a sheath gas, auxiliary gas and sweep gas of 55, 15 and 3 arbitrary units respectively with an S-lens RF (radio frequency) of 60%. For untargeted analysis using the Q Exactive Plus a full scan of 60-900 m/z was used at a resolution of 70,000 ppm where positive and negative ion mode assays were run separately in order to maximise data points across a peak at the chosen resolution. The source parameters were the same as those used for the Elite. For analysis of CoA species using the Q Exactive orbitrap unique mass spectrometry methodology was employed where the full scan mass range was reduced to 800-1000 m/z, the capillary temperature was increased to 350°C and the S-lens RF to 100%.

Targeted analysis on the Quantiva utilised selected reaction monitoring (SRM) employing fast polarity switching with mass transitions and compound dependent parameters (collision energy voltage and RF lens voltage) determined on infusion of 1 mM standards at a flow rate of 10 mI/min in 4:1 acetonitrile:water with 0.1 % acetic acid. Source parameters used were a vaporizer temperature of 440°C and ion transfer tube temperature of 362 °C, an ion spray voltage of 3.5 kV (2.5 kV for negative ion mode) and a sheath gas, auxiliary gas and sweep gas of 54, 17 and 2 arbitrary units respectively.

LC-MS data processing

Data were acquired, processed and integrated using Xcalibur (Version 3.0, Thermo Fisher Scientific) and Compound Discoverer (Version 2.1 , Thermo Fisher Scientific). For untargeted analysis, metabolites of interest were identified using high resolution m/z values as specified in the METLIN database (Scripps Research Institute) corresponding to their [M+H] + or [M-H]- ion adducts in positive or negative ionisation modes, respectively. Compound retention time and fragmentation pattern were validated against known external standards. Peak areas corresponding to metabolite levels were manually quantified and normalised to internal standard or total ion content (as appropriate) and presented as relative areas. All sample data were processed using Compound Discoverer (Version 2.1 , Thermo Fisher Scientific) to accurately calculate total ion content for normalisation.

For multivariate analysis, data were processed using Compound Discoverer (Version 2.1 , Thermo Fisher Scientific) to determine putative compounds with differential abundance between sample groups. For each differential MS feature, chromatogram peaks were manually verified using Xcalibur (Version 3.0, Thermo Fisher Scientific). Accurate m/z values were compared against the METLIN database (Scripps Research Institute) including [M+H] + , [M+Na] + , [M+NFU]* for positive mode and [M-H]-, [M+CI]- for negative mode ion adducts with a mass tolerance of 2ppm. A combination of MS/MS fragmentation profile, molecular formulae calculation based on isotope pattern and expected chromatographic chemical behaviour was then used to attribute metabolite identity. In case of ambiguity, and for all proposed FAMIN products and substrates, external standards were used to confirm metabolite identification. Data from positive and negative ionisation modes were combined and duplicate metabolite identities removed. Data was normalised to total ion content and fold change graphically depicted as volcano plots. Metabolite levels between groups were compared using a two-tailed, unpaired Student’s f-test.

Reagents

The following reagents were used: M-CSF (Peprotech, 300-25), LPS (from E. coli K12, InvivoGen, tlrl- peklps), recombinant mouse IFN-a (BIO-RAD, PMP29Z), murine IFN-g (Peprotech, 315-05), adenosine (Sigma Aldrich, A9251), inosine (Sigma Aldrich, 14125), hypoxanthine (Sigma Aldrich, H9377),

methylthioadenosine (Sigma Aldrich, D501 1 ), S-adenosylmethionine (Sigma Aldrich, A7007), S- adenosylhomocysteine (Sigma Aldrich, A9384), cytidine (Sigma Aldrich, C122106), uridine (Sigma Aldrich, U3750), xanthosine (Sigma Aldrich, X0750), 2’deoxyadenosine (Sigma Aldrich, D7400), 5’deoxyadenosine (Sigma Aldrich, D1771), cholesterol oxidase (Sigma Aldrich, C8649), 13 Cio, 15 Ns-adenosine (Cambridge Isotope Laboratories), 15 Ns-adenine (Cambridge Isotope Laboratories) for isotopic tracing experiments; all standards for validation of mass spectrometry compound identification were purchased through Sigma Aldrich. Plasmids

A pESG-IBA105 vector (IBA Life Sciences 5-4505-001) was used for the mammalian expression of

FAMIN 2541 , allowing expression of recombinant protein containing a twin Strep-tag at the N-terminus under CMV enhancer and promoter elements. The DNA sequence of human FAMIN was amplified with Phusion DNA polymerase (NEB M0530) from an IMAGE clone (MHS1010-7508636) using a forward primer containing a leader sequence with Esp3l targeting site followed by TEV cleavage sequence, and reverse primer containing an Esp3l targeting site. The purified PCR product was digested with Esp3l (NEB R0734) and verified on 1 % agarose gel. The gel-purified amplicon was then cloned into Esp3l- digested pESG- IBA105 with T4 DNA ligase (NEB M0202). The ligated plasmid was verified by Sanger sequencing, with a final construct consisting of, from the 5’ to 3’ ends, a Kozak sequence just upstream of the start codon of twin Strep-tag, followed by a short linker and a TEV cleavage motif sequence and human FAMIN sequence. A similar protocol was adapted for the YfiH and YlmD constructs, using pPSG-IBA105 plasmid vector (IBA Life Sciences 5-4305-001) that allows expression of recombinant protein containing twin Strep-tag at the N- terminus under constitutive T7 promoter. The DNA sequences of yfiH and ylmD were custom-synthesised by Origene and codon-optimised for prokaryotic expression, with Esp3l target sites flanking the sequence and TEV cleavage motif immediately upstream of the gene. The Esp3l- digested yfiH and ylmD sequences were then cloned into Esp3l- digested pPSG-IBA105 with T4 DNA ligase (NEB). Site-directed mutagenesis was subsequently performed using Q5 Site-Directed Mutagenesis Kit (NEB E0554), following the manufacturer’s instruction, to generate FAMIN 254V for both prokaryotic and mammalian expression. All constructs generated were verified by Sanger sequencing.

Protein expression and purification

HEK293T cells were transfected with PEI-DNA complex containing plasmids for mammalian expression of Strep-tagged FAMIN. After 48-72 hours, transfected cells were harvested and resuspended in detergent-free pre-chilled suspension buffer (100 mM NaCI, 20 mM HEPES, 5 mM TCEP and 10% glycerol pH 8.0) supplemented with Complete Mini EDTA-free protease inhibitor cocktail (Sigma 1 1836170001). Following sonication and clearance by centrifugation at 30,000 rpm, the lysates were then loaded onto a Streptactin XT Superflow column (IBA Life Sciences 2-4012-001 ) pre-equilibrated with 2 column volumes of lysis buffer.

The column was then washed with 10 column volumes of wash buffers and the Strep-tagged protein was eluted from the column with 6 column volumes of 50 mM biotin. The purified recombinant protein was then incubated with N-terminally 6xHis-tagged TEV protease (Sigma TEV protease) at a ratio of 10:1 as determined by A280 quantification using NanoDrop spectrophotometer. The mixture was incubated overnight at 4°C. The TEV protease was then removed by passing the mixture through a 1 ml Ni-NTA Superflow column (IBA Life Sciences 2-3206-025), after pre-equilibrating the column with the protein buffer. TEV protease binds to the column and the recombinant protein, in the flow-through, was collected. The flowthrough constituting tag-free recombinant protein was collected. This was then concentrated with a 10 kDa column filter (Amicon) and further purified by size exclusion using an AKTA Superdex 200 increase (10/300) column (GE Life Sciences), followed by copious washing. Eluted fractions corresponding to positive peaks on the chromatogram were confirmed on Coomassie SDS-PAGE.

For prokaryotic expression, T7 Express lysY/lq High Competent E. coli (NEB C3013) were transformed with plasmids containing the DNA sequence of recombinant protein. Fresh colonies (less than 2 weeks after plating) were inoculated into an LB broth supplemented with 0.5% glucose and grown at 30°C in a shaking incubator. Upon reaching OD600 of 0.4-0.7, protein expression was induced with 0.3 mM of IPTG. After 4-6 hours, bacterial culture was centrifuged at 14000 rpm for 15 minutes. The cell pellet was re-suspended in 5 ml of pre-chilled hypotonic lysis buffer (20 mM NaCI, 20 mM HEPES, 10% glycerol, pH 8.0) supplemented with protease inhibitors. Following sonication, the lysates were cleared by centrifugation at 30,000 rpm for 15 minutes and treated with 5 pg/ml of DNase I (Sigma) and 10 ug/ml of RNAse A (Sigma) for 15 minutes on ice. Subsequent protein purification steps were similar to those employed above in the mammalian expression system.

Enzyme assays

Putative enzymatic function of recombinant human strep-tagged FAMIN was investigated against aqueous HepG2 metabolite extracts (dried, Folch extracted from 5 c 10 6 cells) or nucleoside substrates at indicated concentrations and detected using HPLC-MS as described above. Unless otherwise indicated in the figure legends, HEK293T cell-expressed protein was used for all assays. The reaction mixture (final volume 100 pL) consisted of 10 pg of recombinant protein and 10 pM nucleoside substrate in Dulbecco’s PBS (Thermo Fisher), pH 7.4 unless otherwise indicated. The samples were incubated at 37 °C for 1 h and then quenched with x5 volume of ice-cold methanol. Samples were centrifuged at 21 ,000 g for 5 min transferred to fresh tubes and then dried down prior to analysis as described above.

Mice

6- to 10-week-old mice were used for all experiments and were age and gender matched for individual experiments. Famin +I+ , Famin ~ ' ~ , Famin p 25 Famin p 254V and Famin p 284R mice have previously been described ( 1 ). Mice were bred and maintained under specific pathogen-free conditions at the Central Biomedical Services facility, University of Cambridge. All procedures performed had local ethics and UK Home Office approval.

Murine bone marrow-derived macrophage isolation

Bone marrow derived macrophages (BMDMs) were prepared by flushing mouse femurs and tibias with PBS. Cells were filtered through a 70 pm cell strainer and re-suspended in complete RPMI-1640 medium

(containing 100 U/mL of penicillin-streptomycin, 1 mM HEPES buffer and 10% FBS). To generate BMDMs, cells were cultured for 6 days in complete medium containing 100 ng/ml_ of M-CSF with media exchanged after 3 days. Macrophages were harvested, seeded and polarized for 24 h toward M1 F with IFN-g (50 ng/ml_) plus LPS (20 ng/mL). As indicated, recombinant IFNa was added to undifferentiated BMDMs at 500 lU/mL for 16 h prior to direct methanol extraction.

Statistical analysis

Statistical analyses were performed using Graphpad Prism 6.0 or, as described in LC-MS analysis methods, Compound Discoverer (Thermo Scientific). Unless otherwise stated, statistical significance was calculated as appropriate using unpaired, two-tailed Student’s f-test as described in the figure legends. Data are represented as mean and standard error of the mean (S.E.M.). A P value of <0.05 was considered significant. Results

We hypothesised that FAMIN is an enzyme and devised an unbiased screen for activity against an extensive library of metabolites without a priori assumptions on putative function. We optimised a strategy for large- scale production of highly purified, recombinant human FAMIN from transiently transfected HEK293T cells, using Strep-tag affinity purification, TEV protease-mediated tag cleavage and size-exclusion

chromatography. The resultant FAMIN protein exhibited stable properties in solution consistent with correct folding and lack of aggregation. To generate a suitable metabolomic library of substrates and potential cofactors, we tested a variety of cell lines for FAMIN functionality. The human hepatocellular carcinoma cell line HepG2 entered growth arrest after transfection with FAMIN siRNA, which became evident after 72 h. HepG2 cells also exhibited reduced glycolysis and oxidative phosphorylation 48h after FAMIN siRNA transfection, resembling observations in macrophages ( 1 ). This indicated that FAMIN is active and performs a non-redundant role in HepG2 cells, which could thus be reasonably expected to contain all cofactors and substrates necessary for FAMIN function.

We adopted bespoke, quantitative, high-sensitivity and high-resolution orthogonal liquid chromatography mass spectrometry (LC-MS) methodology to capture and accurately resolve a wide range of chemically highly diverse metabolites in aqueous extracts of FAM/A/-silenced HepG2 cells. We identified over 25,000 unique LC-MS features in freeze-dried extracts across the different chromatography modalities and ionisation modes (Fig. 1A) and hypothesized that one or more of these might correspond to FAMIN substrates. We incubated 10 pg recombinant human FAMIN, or an equivalent volume of protein buffer control, with the metabolite library resuspended in a final reaction volume of 100 pi phosphate-buffered saline (PBS) pH 7.4 for 1 h at 37°C. After quenching the reaction, samples were re-extracted and only 3 and 4 compounds within the library were significantly decreased and increased, respectively, in the FAMIN- compared to the mock reaction (Fig. 1 B, 1 C).

We next sought to elucidate the identities of these candidate substrates and products. We searched against the METLIN database, the m/z values {A: 268.104, B: 269.089, C: 284.099 in positive ESI} of the 3 LC-MS features within the screened library whose levels were reduced in the presence of recombinant FAMIN (Fig. 1 B, 1 C). Intriguingly, all three of these putative substrates, A-C, exactly matched purine nucleosides, and could not be matched to any other compound. Molecular formula determination, using accurate mass and supported by isotopic mass distribution, also indicated compounds A-C were purine nucleosides. However, m/z and molecular formulae alone could not discriminate their identity. Compound A, with formula

C10H13N5O4, might correspond to either adenosine or deoxyguanosine. Comparing chromatography characteristics of the candidate metabolites A-C against nucleoside standards we demonstrated that the retention times exactly matched adenosine, guanosine and inosine (Fig. 1 D, 1 E), suggesting that FAMIN was an enzyme catabolising the major cellular purine nucleosides (Fig. 1 F, G). Consistent with this, the m/z values of the LC-MS features, D-F (Fig. 1 B, 1 C), whose levels increased upon reaction with recombinant FAMIN {137.046, 152.057 in positive ESI and 229.012 in negative ESI} matched the purine nucleobases hypoxanthine, guanine and a pentose-phosphate, respectively (Fig. 1 F, 1 G). The other LC-MS feature, G, with m/z value of {153.041 in positive ESI} that was also increased in the screen corresponded to the purine nucleobase xanthine, though its absolute levels were extremely low. Modified chromatography allowed faithful separation of isomeric pentose-phosphate compounds and identified compound E as ribose-1 - phosphate (Fig. 1 H). FAMIN did not affect any other nucleosides or nucleotides, e.g. ATP nor pyrimidine metabolites such as cytidine or uridine that were also present in our metabolomics library (Fig. 11).

Furthermore, adenosine, inosine, and guanosine consumption increased with the amount of recombinant FAMIN present in the reaction (Fig. 11, 1 J). Altogether, this suggested that FAMIN may be an enzyme that acts upon purine nucleosides to generate purine nucleobases and ribose-1 -phosphate.

To unambiguously validate results from the library screen, we examined enzyme activity in a fully reductionist system using pure substrate, assayed with recombinant FAMIN in PBS at pH 7.4 and 37°C. FAMIN consumed adenosine and generated inosine, hypoxanthine and ribose-1 -phosphate, confirmed by authentic standards (Fig. 2A, B, C, D, E, F). No spontaneous degradation of adenosine or formation of products in the absence of FAMIN or adenosine was observed (Fig. 2A, B), nor did equimolar amounts of an unrelated enzyme, cholesterol oxidase, generate inosine, hypoxanthine or ribose-1 -phosphate (Fig. 2G). Since our orthogonal LC methods (HILIC and C18-PFP) did not resolve adenosine and adenine well, and adenosine undergoes source fragmentation to adenine, we included further chromatography methods to faithfully separate these two metabolites. This demonstrated that FAMIN also converted adenosine to adenine (Fig. 2H). Quantification indicated that ~85% of consumed adenosine was converted by FAMIN to adenine and ~15% to inosine (Fig. 2I). FAMIN-catalysed activities were further confirmed by tracing

[ 15 N 5 13 C IO ] adenosine-derived stable isotopes into reaction products (Fig. 2J). Incubating recombinant FAMIN with adenine and ribose-1 -phosphate yielded adenosine, demonstrating the reaction can also operate in reverse, thereby also corroborating the identities of the products of the enzymatic forward reaction (Fig. 2 K). FAMIN did not produce any other detectable products from adenosine, and heat-denaturing rendered the protein completely inactive (Fig. 2L). Consistent with FAMIN using inorganic orthophosphate (Pi) present in PBS to generate ribose-1 -phosphate, the enzymatic reaction progressed only to inosine without producing adenine or ribose-1 -phosphate when performed in HEPES buffer instead of PBS (Fig. 2L). We concluded that FAMIN exhibited two distinct enzymatic activities as adenosine deaminase (EC 3.5.4.4) and purine nucleoside phosphorylase (EC 2.4.2.1), and that reactions can proceed independently from each other (Fig. 2M).

We next investigated FAMIN’s activity spectrum with pure purine substrates. FAMIN metabolised pure inosine and guanosine, also identified in the library screen, into ribose-1 -phosphate and their respective nucleobases, hypoxanthine and guanine (Fig. 2N, 20, 2P, 2Q), consistent with purine nucleoside phosphorylase activity. No activity towards the pyrimidine cytidine was detected (Fig. 2N). FAMIN also metabolised 2’-deoxy-adenosine, producing adenine, 2’-deoxy-inosine, hypoxanthine, and 2’-deoxy-ribose-1- phosphate (Fig. 2N, 2R). 5’-deoxy-adenosine, a by-product of radical S-adenosylmethionine (SAM) enzymes (7), was also a substrate of FAMIN (Fig. 2S). FAMIN further metabolised S-methyl-5’-thioadenosine (MTA), though not SAM (Fig. 2N). MTA was not converted into S-methyl-5’-thioinosine and hypoxanthine as would be predicted from sequential adenosine deaminase and purine nucleoside phosphorylase activity, but into adenine and S-methyl-5’-thioribose-1 -phosphate (Fig. 20, 2T). Revisiting data from the library screen, we indeed observed a 50% reduction of MTA compared to baseline when 100 pg FAMIN was present in the 100 pi reaction volume (Fig. 2U). Hence this added a third activity of FAMIN as a S-methyl-5'-thioadenosine phosphorylase, EC 2.4.2.28 (Fig. 2V). The apparent adenosine K m for FAMIN’s deaminase activity was 1 1 pM, whereas its phosphorylase activities demonstrated K m values for adenosine, inosine, and MTA of 36 pM, 41 pM, and 1 pM, respectively (Fig. 3A). We next addressed how the I254V substitution, associated with risk for CD and leprosy, affected FAMIN activity. We compared recombinant FAMIN 2541 and the‘risk-variant’ FAMIN 254V for their adenosine metabolising activity. Compared with FAMIN 2541 , adenosine consumption was approximately halved with FAMIN 254V (Fig. 3B). Conversion to adenine was 10-fold lower with FAMIN 254V compared to FAMIN 2541 (Fig. 3C). In contrast, the production of inosine and hypoxanthine by FAMIN 254V was slightly higher and similar, respectively, compared to FAMIN 2541 (Fig. 3C, 3D). Ribose-1 -phosphate generation was ~10-fold reduced with FAMIN 254V compared to FAMIN 2541 (Fig. 3C), hence corresponding to the reduction in conversion to adenine. Thus, FAMIN 254V markedly differed from FAMIN 2541 in how it metabolised adenosine, with only ~20% rather than 85% of consumed adenosine converted to adenine, instead diverting it to inosine and thence hypoxanthine (Fig. 3E). Consistent with the forward reaction, FAMIN 254V generated 10-fold less adenosine from adenine and ribose-1 -phosphate compared with FAMIN 2541 (Fig. 3F). FAMIN 254V also demonstrated reduced phosphorylase activities towards inosine and MTA, since generation of hypoxanthine from inosine was ~50% lower, and adenine from MTA ~25% less compared to FAMIN 2541 (Fig. 3G). In summary, amongst FAMIN’s enzymatic functions, its adenosine phosphorylase activity was the most attenuated in the FAMIN 254V variant, whereas the adenosine deaminase activity appeared intact.

We next explored whether FAMIN affects cellular levels of purine metabolites. Transient transfection of FAMIN cDNA in HEK293T cells results in equally high protein expression of FAMIN 2541 and FAMIN 254V and only low expression of FAMIN 284R ( 1 ). 24 h after transfection with FAMIN 2541 plasmid, cellular hypoxanthine levels increased by 50% (Fig. 3H). Hypoxanthine modestly increased with FAMIN 254V , and remained unaltered in FAMIN 284R compared to control transfectants (Fig. 3H). Cellular guanine levels were elevated in FAMIN 2541 transfected HEK293T cells (Fig. 3H), while total purine nucleoside levels were unaffected by expressing any of the FAMIN plasmids (Fig. 31). Moreover, transfection with either FAMIN 2541 or FAMIN 254V doubled the cellular levels of inosine monophosphate (IMP) (Fig. 3H). IMP is at the juncture of purine salvage and de novo synthesis and the obligatory intermediate of both (8). Conversely to the increase upon overexpression, cellular IMP levels were halved in HepG2 cells 24 h after transfection with FAMIN siRNA (Fig. 3J), which was amongst very few changes in metabolite levels observed that early. Acute silencing of FAMIN in HepG2 cells also reduced hypoxanthine levels, while it increased levels of inosine, guanosine and MTA (Fig. 3K, 3L). This demonstrated that FAMIN bolsters the generation of nucleobases for purine nucleotide salvage, and confirmed the activities established with pure recombinant protein as physiologically relevant.

We next turned to terminally differentiated murine bone marrow (BM)-derived macrophages, which in contrast to proliferating HepG2 cells only employ purine nucleotide salvage, with hardly any de novo synthesis. Gene-editing human polymorphic FAMIN variants into the murine genome had established that compared to FAMIN 2541 (Fam/n p 2541 ), reduced (Fam/n p 254V ) or absent ( Famin p 284R or Faminr^) FAMIN activity in macrophages causes a progressive impairment in reactive oxygen species (ROS) production, bactericidal activity, inflammasome activation and cytokine secretion. This was linked to diminished de novo lipogenesis, oxidative phosphorylation, aerobic glycolysis and levels of adenosine triphosphate (ATP) ( 1 ). It was therefore interesting to observe that cellular levels of adenosine, adenine, and MTA were lowest in Fam/n p 284R , intermediate in Fam/n p 254V , and highest in Famin p 2541 classically-activated bone marrow-derived macrophages (Fig. 4A). In contrast, cellular levels of inosine, hypoxanthine, guanosine and guanine were comparable across genotypes (Fig. 4B). Levels of 5’-deoxyadenosine, induced by interferon-a (9), were not significantly different between Famin genotypes, while 2’-deoxyadenosine levels were higher in Fam/n p 2541 and Fam/n p 254V compared to Famin p 284R cells (Fig. 4C). Although total cellular metabolite levels may underestimate highly compartmentalised processes, such as with FAMIN that localises to the cytoplasmic surface of peroxisomes ( 1 , 10), these data demonstrated that FAMIN promoted adenyl metabolism in macrophages.

Eukaryotic cells have been considered devoid of an activity that phosphorolytically converts adenosine or 2’- deoxyadenosine to adenine and ribose-phosphate (11-13). FAMIN adds such activity and furthermore combines in one single enzyme activities that are essential and non-redundant for purine nucleotide salvage. These had been thought to be encoded by single genes that are ubiquitously expressed: adenosine deaminase (ADA; secreted ADA2 is expressed from a separate gene), purine nucleoside phosphorylase (PA/P) and S-methyl-5'-thioadenosine phosphorylase ( MTAP ) (14, 15). The reported apparent K m of these enzymes for their primary substrates is within the same range as FAMIN, indeed those of FAMIN appear slightly lower (16-18). The principal purine nucleosides, adenosine, inosine and guanosine, and their nucleobases, adenine, hypoxanthine and guanine, are neither precursors nor intermediates of cfe novo purine synthesis, but are generated by the reactions that supply purine nucleotide salvage (8). Purine nucleotide salvage proceeds from nucleobases via hypoxanthine guanine phosphoribosyl transferase (HPRT) and adenine phosphoribosyl transferase (APRT), and adenosine is the only nucleoside that can additionally be directly salvaged via adenosine kinase (ADK) (8). We show that FAMIN’s trifunctionality expands core purine metabolism and adds an entirely novel layer of interconnectivity. This is associated with a gradual increase in total cellular ATP, FAD, and relative NAD/NADH ratio from loss-of-function Famin p 284R via hypomorphic Fam/n p 254V to Famin p 2541 genotypes (Fig. 4D).

Finally, we investigated whether FAMIN’s trifunctionality in purine metabolism is evolutionarily conserved. The C terminal portion of FAMIN exhibits structural homology to Cluster of Orthologous Group (COG) 1496 proteins that contain the Pfam motif Domain of Unknown Function (DUF) 152, with paralogues widely distributed across species (Fig. 4E). An E. coli expressed maltose binding protein (MBP) fusion protein of truncated FAMIN, which contains the DUF152 homology domain only (FAMIN A176 , amino acids 176-430), exhibited all three enzymatic activities, similar to MBP-tagged full-length FAMIN (Fig. 4F). This demonstrated that the DUF152 homology region contains all enzymatic activity. Consistent with this, recombinantly expressed Strep-tagged COG1496 bacterial proteins YlmD (Uniprot P84138; from Geobacillus

stearothermophilus) and YfiH (Uniprot P33644; from Escherichia coli strain K12), metabolise adenosine to both inosine and adenine, MTA to adenine, and inosine to hypoxanthine (Fig. 4G). Structures of YlmD (Protein Data Bank [PDB] accession code 1T8H) and YfiH (PDB 1Z9T), and other bacterial COG1496 proteins contain at least one Zn 2+ ion coordination, as do cytosine and cytidine deaminases (24). FAMIN, YlmD and YfiH contain a conserved cysteine-histidine-histidine triad, which in the structure of YfiH coordinates a zinc ion. Notably, the active sites of purine and pyrimidine amidohydrolases, including cytosine deaminase, ADA, and urease contain a zinc ion (25-27), which is often coordinated by a combination of histidine and cysteine side chains. However, the overall folds of these enzymes are different from YlmD and YfiH. Regarding genome context, yfiH is flanked in E. coli and S. flexneri by clpB and rluD, which encode an adenosine triphosphatase subunit of a chaperone and a ribosomal pseudouridine synthase, respectively (24). We conclude that FAMIN is indeed a prototype of a new class of trifunctional‘all-in-one’ purine nucleoside metabolising enzymes that is conserved from bacteria to man.

Previously, MTAP had been thought to be the sole route of adenine generation and MTA metabolism (8, 28), and ADA and ADK the only routes of adenosine conversion (8, 19). Associated with peroxisomes, FAMIN may act on a pool of purine nucleosides which is partially or completely distinct from those ADA, PNP and MTAP act upon. Mutations in ADA and PNP cause severe combined immunodeficiency (SCID) (29-31). MTAP is frequently deleted in cancers, accounting for their altered methionine metabolism (32-35).

Decreased or complete loss of FAMIN function, in contrast, presents with auto-inflammatory diseases. While expression of FAMIN is highest in macrophages, it is present in several other cell types, but not in T and B lymphocytes that are affected in SCID. FAMIN may hence re-shape concepts of organismal purine metabolism in addition to those on immune function.

In summary, FAMIN adds a surprising new layer to central adenyl metabolism, a highly interconnected and exquisitely tuned ancient metabolic circuitry that has adenosine and adenine at its centre and supplies adenyl groups for cofactors that are critically involved throughout metabolic processes.

Applying methods disclosed herein, we identified pentostatin (PubchemID 439693, CAS ID 53910-25-1) and erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA; PubchemID 3206, CAS ID 59262-86-1) as FAMIN inhibitors (Fig. 5A). Specifically, pentostatin and EHNA inhibited the adenosine deaminase activity of FAMIN (Fig. 5A), while leaving FAMIN’s adenosine phosphorylase activity intact (Fig. 5B, C). 8-aminoguanosine (PubchemID 96849, CAS ID 3868-32-4) inhibited FAMIN’s purine nucleoside phosphorylase activity as demonstrated by reduced generation of hypoxanthine and ribose-1 -phosphate (R-1 -P) from inosine (Fig. 5D, E). Importantly, 8-aminoguanosine did not affect FAMIN’s adenosine phosphorylase activity as demonstrated by unimpaired generation of adenine and R-1 -P from adenosine (Fig. 5C, D). This demonstrates that individual activities of FAMIN can be selectively targeted with small molecule modulators.

References

1. M. Z. Cader et al., Nat. Immunol. 17, 1046 (2016).

2. L. A. O'Neill, E. J. Pearce, J. Exp. Med. 213, 15 (2016).

3. G. A. Prosser, G. Larrouy-Maumus, L. P. de Carvalho, EMBO reports 15, 657 (2014).

4. S. Zhao et al., Nature 502, 698 (2013).

5. J. Folch, M. Lees, G. H. Sloane Stanley, J. Biol. Chem. 226, 497 (1957).

6. V. Gorshkov et al. , J. Basic Microbiol. 57, 998 (2017).

7. B. J. Landgraf, E. L. McCarthy, S. J. Booker, Annu. Rev. Biochem. 85, 485 (2016).

8. M. Camici, S. Allegrini, M. G. Tozzi, FEBS J., (2018).

9. A. S. Gizzi et al., Nature 558, 610 (2018).

10. G. Assadi et al., PloS one 11 , e0168276 (2016).

11. M. Friedkin, H. M. Kalckar, in The Enzymes, P. D. Boyer, H. Lardy, K. Myrback, Eds. (Academic Press, New York, 1961), vol. 5, pp. 245.

12. T. P. Zimmerman, N. B. Gersten, A. F. Ross, R. P. Miech, Can. J. Biochem. 49, 1050 (1971).

13. J. T. Maynes et al., Biochem. J. 344 Pt 2, 585 (1999).

14. A. W. Murray, Annu. Rev. Biochem. 40, 811 (1971).

15. H. Ashihara, C. Stasolla, T. Fujimura, A. Crazier, Phytochemistry 147, 89 (2018).

16. E. R. Lindley, R. L. Pisoni, Biochem. J. 290 ( Pt 2), 457 (1993).

17. A. Bzowska, E. Kulikowska, D. Shugar, Pharmacol. Ther. 88, 349 (2000).

18. F. Della Ragione et al., Biochem. Biophys. Res. Commun. 223, 514 (1996).

19. D. Boison, Pharmacol. Rev. 65, 906 (2013).

20. M. Camici, M. Garcia-Gil, M. G. Tozzi, International journal of molecular sciences 19, (2018).

21. A. Beloqui et al., J. Biol. Chem. 281 , 22933 (2006).

22. A. I. Canas, S. Camarero, Biotechnol. Adv. 28, 694 (2010).

23. V. Perna, J. W. Agger, J. Hoick, A. S. Meyer, Scientific reports 8, 81 14 (2018).

24. Y. Kim et al., Proteins 63, 1097 (2006).

25. L. Holm, C. Sander, Proteins 28, 72 (1997).

26. D. K. Wilson, F. B. Rudolph, F. A. Quiocho, Science 252, 1278 (1991).

27. E. Jabri, M. B. Carr, R. P. Hausinger, P. A. Karplus, Science 268, 998 (1995).

28. E. Albers, IUBMB life 61 , 1132 (2009).

29. K. L. Bradford, F. A. Moretti, D. A. Carbonaro-Sarracino, H. B. Gaspar, D. B. Kohn, J. Clin. Immunol.

37, 626 (2017).

30. E. R. Giblett, A. J. Ammann, D. W. Wara, R. Sandman, L. K. Diamond, Lancet 1 , 1010 (1975).

31. E. R. Giblett, J. E. Anderson, F. Cohen, B. Pollara, H. J. Meuwissen, Lancet 2, 1067 (1972).

32. G. V. Kryukov et al., Science 351 , 1214 (2016).

33. K. J. Mavrakis et al., Science 351 , 1208 (2016).

34. T. Nobori et al., Proc. Natl. Acad. Sci. U. S. A. 93, 6203 (1996).

35. Z. H. Chen, H. Zhang, T. M. Savarese, Cancer Res. 56, 1083 (1996).

Sequences

1 maeavlidlf glklnsqknc hqtllktlna vqyhhaakak flcimccsni syerdgeqdn 61 ceietsngls alleefeivs cpsmaatlyt ikqkideknl ssikvivprh rktlmkafid 121 qlftdvynfe fedlqvtfrg glfkqsiein vitaqelrgi qneietflrs lpalrgklti 181 itsslipdif ihgfttrtgg isyiptlssf nlfssskrrd pkvvvqenlr rlanaagfnv 241 ekfyrikthh sndiwimgrk epdsydgitt nqrgvtiaal gadcipivfa dpvkkacgva 301 hagwkgtllg vamatvnami aeygcsledi vvvlgpsvgp ccftlpresa eafhnlhpac 361 vqlfdspnpc idirkatril leqggilpqn iqdqnqdlnl ctschpdkff shvrdglnfg 421 tqigfisike

SEQ ID NO: 1 Homo sapiens FAMIN amino acid sequence (NP_001 121775.1)

1 agacctgcag ctcctgccgc cctgcgcccg ctcccagggc ccgtcgttcc gccgccctat

61 ccctcctcaa ggggccccta gctgcctcct cgcgaccctt tccggactcg gcctgcccac

121 tcctgcccgc taacccgcct ggctcccggg cgagagccct cgcgcggctc tggttcctgt

181 tcctctaacg ccgccggggc tgcgggatgc cgactccgcg gaccgcccag acccggaact

241 gctgaggcag cagcgggctc gcggcgcttg gctcatcccg ggattcccca gctctcgcgc

301 tgggcccgcc gcgttcgcac caagcacgcc aggcggccct ggcctacctc cctcccgcct

361 cccggcagct ggcacgaggg aacctggccg tcaggtttcc cctgggatcc tgggacggta

421 tcaggcgggg aatctgtgcg gccgcggcga ggtgatttat ttggcataaa agtattcttt

481 caaggatggc agaagctgtt ttgattgatc tttttggttt gaaattgaac tctcaaaaaa

541 actgccatca gacattactg aagactttga atgctgtcca ataccaccat gctgccaagg

601 ccaagtttct ctgtataatg tgttgcagta acatcagcta tgaaagggat ggagaacaag

661 ataattgtga aatagaaaca agcaatggat tatcagctct cttggaagaa tttgagattg

721 ttagctgtcc cagcatggct gccactttgt ataccattaa acagaaaatt gatgaaaaaa

781 atctgagcag cattaaggta attgtaccca ggcacaggaa gacattaatg aaagctttta

841 ttgatcaact cttcactgat gtttacaatt ttgaatttga agatttgcaa gtgactttta

901 ggggagggct ttttaaacag tccattgaaa taaacgtaat cacagctcaa gaactaagag

961 gaattcagaa tgaaatagaa acatttttga gaagtctgcc agcactgaga ggaaaattaa

1021 ctattatcac ttcttctttg atcccagata ttttcataca tggatttact acaagaacag

1081 gtgggatatc ttatatacca actcttagct cattcaatct cttcagtagt tccaaacgga

1141 gagatcccaa ggtagtggtt caagaaaatc tgcgtaggtt ggcgaatgct gcaggattta

1201 atgtggagaa attttaccga ataaagactc atcattccaa tgacatctgg attatgggaa

1261 gaaaggagcc tgactcttat gatggaataa ccacaaatca gagaggagtc acaatagcag

1321 ctcttggtgc agactgtata ccgatagttt ttgcagatcc agtcaaaaaa gcatgtgggg

1381 ttgctcacgc tggttggaaa ggtactttgt tgggtgttgc tatggctaca gtgaatgcta

1441 tgatagcaga atatggctgc agtttggaag acattgttgt tgtacttgga ccttcagtag

1501 gaccttgctg ttttactctt ccaagggaat cagcagaggc atttcataat cttcatcctg

1561 catgtgtaca actatttgat tcaccaaatc cctgtatcga catccgtaaa gccacaagga

1621 ttcttctaga acagggagga attcttccac agaatattca ggaccagaac caagatctca

1681 acctctgtac atcttgccat cctgacaagt ttttctccca tgtccgagat ggccttaatt

1741 ttggtacaca gattggcttc atatcaatta aagaatgaga tacttgactg gatttttgta

1801 taactgcttc ctgcctcctt ccaaactgac tgcaagagag aaatttagct gtttgattta

1861 cttaaaacca aatggattac aatggataat tcatcttttg ggtatatttt tactattatt

1921 caaagccaaa tgattttcat ttaattgtaa taataactga caaaaatcag tatgttgtag 1981 ctaatatgtt ttatgcatga gaattattct taaagtttgt tctccctgtt tattacacag

2041 atcaggaata gatttgttca gttcagtatt tattggatac cctctattgg tcaggcattg

2101 tgttaagcat atgtgaatca aaatgaacac aactttttcc tttgagtctg atacagtgaa

2161 ggagataaac acttctacaa cttaaattta attttaatag cagtagaaga gaacataagg

2221 aatagaggtt aattttaccc agaagcagga tagagaaaat attacagaga aaatcacata

2281 tcacatgggc tcgaaagatg tagaggtttt tgacaaatga agaacaacca taacaggtag

2341 agggaacacc atgaaccagg gcatgaaact gaaagtgcat aacatattct agagagagaa

2401 gggtgtgggc atgagttagg gctggaaaaa caggttggaa acagataagt aagggtctca

2461 aatgcaatgt caaagagctt gcagtttatt ttccaggcaa tgagtaggca gccaaaaaaa

2521 aaaaagtaag gatgtttttt ttttttttcc catggcatca tatttaagag gatggattta

2581 aattgtgtga gaccaaagca tagagactag ataagaggcg atcaaaatat ttcaaaaaga

2641 aataatgaag atccaatgaa ggaagtggaa attaaaatag ggaagagagt agatggatta

2701 gagagacatt taagagatgg aatcaataga tcctgttact agataatgga agtaagaggt

2761 gaggaagagt ggaaaagtca ttaatgactc taagatttct gcttggctgc ttaccaagat

2821 tggcaacaaa gggagggaga aggtttggaa aaagagagaa ggataatgag tttgacttta

2881 catagaatga agggcatcca gatagaaatc tttggttaat aattagaaat atagacctag

2941 aaattaggag gaaacctgag acagagacaa atatttcaaa gcttacaata cagagatgat

3001 acctgattct attggagcag gtttgatcat ctaggcagaa attaggatga gaaaaaagga

3061 gatccaataa tacaacctta tagtcacaga agtaagaaaa aaagggtagt tgttttgaag

3121 aagccaggat aggtgtggaa agtactcaaa aagaaatctt cagggataaa ataaagtgat

3181 aatttaaaag aaatcaatgg attaaacata ttgaaactgt tctataggca gtggtcattg

3241 agtcagcttt cagtgcatta ggaagaagat gcataggtgt caactctttt ctgacagcat

3301 ttactagaga agagaaaaag ctggggacta catcttcaag gaagggactt tttttggatg

3361 agcagttttg agtgtgtttg tcagttaaag agaggaatta ggttagtttt catttgggaa

3421 aaattgtata tatatttaat gtaagttatc acattgcatc ttaaaaatat tcttatttaa

3481 tacatatatt tcctacatgt atatgtggta gcatgatagc aaataacatt tgtttggtat

3541 ttccaaagga ctttcatgta cattgcctca ttttaccttt acagctactc tgaaatacac

3601 aggcattatc ccttttattc agctgagaaa actgagcttc attgaggtgg aggtcaaaaa

3661 tcacaaaatt tgtgatgaat taagatttga acatatgttt tgtgactcca gttttccttt

3721 cagattttaa aattaattaa agggatcttc attatacttt tattgttaac tttttgttaa

3781 cataatttat tcatacattc agtgaaaatt ttgttgaggt actgggacag gttaaaaaat

3841 acagttgtag ccctcaggat atttaatatc cagtgaaaag tgacagtcag taaaccaaca

3901 atctcaatac tttgatatat gttgtgaggt tgtgataacc gattcttgtt tagtttaatt

3961 ctatatctcc cttagaccag tgttaaattt aaataaaaca cctcattttt tccaattcag

4021 ggaaggcact aaacataaag cataggatag aaatgttgaa ctcatccaaa atattatttt

4081 gtttaatgaa aatgatgaag attaaggaat acttccatgt attgagtaag gttgataatt

4141 ttctaattct tcactgtgca ttattttgtt tgaagttggt aaatttggag tatcctgcag

4201 acacattttg ctttatgtac tacaacattc tacaaccaaa taaaaattat tttgattatc

4261 tgaaaaaaaa aaaaaaaaaa aaaaaaaa

SEQ ID NO: 2 Homo sapiens FAMIN coding sequence (NM_001 128303.1)