Loading

Commentary Open Access
Volume 3 | Issue 2 | DOI: https://doi.org/10.33696/Signaling.3.073

Insights from Natural Product PHGDH Inhibitor Studies

  • 1Department of Pathology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
+ Affiliations - Affiliations

*Corresponding Author

Qingxiang Sun, qingxiang.sun@scu.edu.cn

Received Date: April 29, 2022

Accepted Date: May 26, 2022

Abstract

Cancer cells often exhibit a reprogrammed metabolism to accommodate their higher proliferation rates. Serine is a building block for de novo nucleic acids synthesis, and cancer cells typically require more of it. PHGDH is the rate limiting enzyme in human serine synthesis steps and is often highly expressed in cancer. About a dozen PHGDH inhibitors have been reported, including three natural products, azacoccone E, ixocarpalactone A, and oridonin. The inhibition mechanism of most inhibitors is still lacking, and none of these inhibitors have entered clinical trials. In this commentary, we present new data on withaferin A, another natural product inhibitor of PHGDH, and discuss the inhibition mechanism by these natural products. The knowledge gained from these inhibitor studies may help guide the development of future PHGDHtargeted anticancer drugs.

Commentary

The synthetic activity of serine is significantly upregulated in several cancers [1]. Homo sapiens 3-phosphoglycerate dehydrogenase (PHGDH) catalyzes the rate-limiting step of serine synthesis, which converts 3-phosphoglycerate (3PG) and NAD+ to 3-phosphohydroxypyruvate and NADH [2,3]. PHGDH inhibitors were extensively pursued recently for their use in the treatment of affected cancers [4,5]. Among the inhibitors identified, three natural products including azacoccone E [6], ixocarpalactone A [7], withaferin A and oridonin [8] were shown to effectively inhibit PHGDH enzyme activity and cancer cell growth.

Azacoccone E, an aza-epicoccone derivative from the culture of Aspergillus flavipes, is the first reported natural product inhibitor that was non-competitive against 3PG, but reduced the Vmax of the reaction [6]. In serine-depleted media, Azacoccone E selectively inhibited the growth of cells expressing high PHGDH, but had little inhibitory effect on PHGDH low-expressing cells. The fact that it inhibited PHGDH enzyme activity time-dependently [9] suggests that it was likely a covalent inhibitor, especially the structure of azacoccone E contains a functional group capable of forming covalent bonds [10].

Ixocarpalactone A, a natural withanolide obtained from dietary tomatillo (Physalis ixocarpa), not only selectively inhibited PHGDH high-expressing cells, but also exhibited anti-cancer activity in a xenograft mouse model [7]. It reduced Vmax but not Km of 3PG and NAD+. Ixocarpalactone A contains a Michael acceptor, which can form covalent bonds with cysteine residues in a protein. At the enzyme activity level, the IC50 value of azacoccone E (9.8 ± 4.3 μM) was slightly higher than that of ixocarpalactone (1.66 ± 0.28 μM).

Through a screen of about 300 natural products, we recently reported two new PHGDH inhibitors, oridonin and withaferin A, but the study only focused on oridonin [8]. Here, we are presenting new data on withaferin A, which is a steroidal lactone derived from Acnistus arborescens. Withaferin A is structurally similar to ixocarpalactone A, especially the portion containing the Michael acceptor (Figure 1A). At the enzyme level, the IC50 of withaferin A (0.59 ± 0.01 μM) is similar to that of oridonin (0.50 ± 0.02 μM) (Figure 1B). These values should not be directly compared with the values for the two aforementioned inhibitors because the experimental conditions (e.g. pre-incubation time) are different. Withaferin A was shown to be a covalent inhibitor of PHGDH (Figure 2), arguing that ixocarpalactone A is also a covalent inhibitor.

 

Figure 1: Chemical structures and inhibition curves of PHGDH natural product inhibitors. A) Chemical structures for known natural product inhibitors of PHGDH. B) Concentration-dependent inhibition of PHGDH by oridonin and withaferin A. The compounds were pre-incubated with PHGDH (3-314) for 2 hours before reaction.

 

 

Figure 2: Locations of cysteine residues in the structure of PHGDH (3-314). The SBD and NBD of one PHGDH monomer is shown in green and cyan, respectively. Another PHGDH monomer is colored grey. Cysteine residues are shown as ball and stick structures.

Oridonin is a kaurene-type diterpenoid isolated from Rabdosia rubescens. Oridonin was a covalent PHGDH inhibitor with antiproliferative activity on cancer cells [8]. Unlike azacoccone E and ixocarpalactone A, oridonin notably reduced the Km of 3PG, but barely affected the Vmax of PHGDH.

Crystal structures often provide powerful insights into the inhibition mechanism of compounds [11]. The cocrystallization of several PHGDH-inhibitor complexes was attempted, but only the complex crystal structure of PHGDH and oridonin was successfully obtained [8]. Oridonin bound in the substrate binding domain (SBD) of PGHDH, covalently linked to C18. Structural analysis revealed that a 3PGinteracting residue R54 was dislocated upon oridonin binding. R54 and C18 mutations (R54A or C18W or C18Y) that mimic the binding effect of oridonin, were shown to reduce the Km of 3PG. Therefore, oridonin binding to C18 allosterically reduced the substrate affinity of PHGDH. This newly identified allostery may help the design of allosteric drugs, which are sometimes more advantageous over orthosteric drugs [12].

Cysteine to serine mutation is often used to validate whether a site is responsible for the observed effects of a covalent inhibitor [13]. Surprisingly, oridonin inhibited C18S and WT PHGDH with similar IC50 values [8]. Isothermal titration calorimetry (ITC) also indicated that more oridonin molecules were bound per PHGDH molecule. Therefore, there are more oridonin binding sites in PHGDH, and binding to these sites played a redundant role in PHGDH inhibition.

Oridonin contains a Michael acceptor that can covalently bind to cysteine residues [14]. There are altogether 11 cysteine residues in SBD and the NAD+ binding domain (NBD) of PHGDH (Figure 2). Only the crystallographically oridonin-conjugated C18 is solvent exposed. Other residues are mostly buried inside the protein or in the dimerization interface. Covalent modifications to C295 and C234 are expected to disrupt the binding of 3PG and NAD+, respectively. Mass spectrometry showed that oridonin bound to multiple cysteine residues in PHGDH, and the number of sites depended on the concentration of oridonin [8]. A low concentration oridonin bound to C18 and C281. Binding to C281 is expected to disrupt dimerization and inhibit PHGDH, since the enzyme activity is dimerization dependent [15].

Except for NAD+ competitive inhibitors, most known PHGDH inhibitors including CBR-5884, PKUMDL-WQ-2101, disulfiram, DNS-pE, and the four natural products contain covalentbinding functional groups [6-8,16-19]. A high concentration withaferin A bound to C18, C234, C254, and C295 (Figure 2), suggesting that it may disrupt binding of 3PG and NAD+ to PHGDH. Disulfiram, an anti-alcohol drug, bound to C111, C116, and C281 [18], thus would disrupt protein dimerization. DNSpE is likely bound to C234, C281, and C295 [19]. In a proteomic survey, multiple PHGDH cysteine residues including C18, C48, C116, C234, C254, C281, and C295 were reported to be reactive to high concentrations of small covalent modifiers [20]. Although different compounds bound to different sites, they all bound to multiple cysteine residues in PHGDH. This suggests that PHGDH is susceptible to cysteine covalent modification, and that other covalent inhibitors probably also inhibit PHGDH by binding to multiple cysteine residues.

It has been reported that oridonin degraded several cellular proteins including BCL-ABL, Glut1, MCT1, FAS [21-23]. Oridonin also induced proteasome-dependent PHGDH degradation in human cells [8]. Analysis using differential scanning fluorescence, GTA cross-linking, and size-exclusion chromatography showed that oridonin reduced the stability of PHGDH, and caused protein aggregation/precipitation [8]. These observations could explain why PHGDH was degraded in oridonin-treated cells, as cellular aggregates are often cleared by degradation machineries [24]. The other cellular targets of oridonin were probably cleared by the same aggregation-degradation mechanism.

Oridonin-induced protein aggregation may be due to two reasons. As a highly hydrophobic molecule [25], oridonin can increase the hydrophobicity of conjugated proteins, thereby promoting their aggregation/precipitation. Alternatively, binding to buried cysteine residues disrupts the protein’s local folding, causing it to aggregate. At 20 μM concentration, NCT-503 (non-covalent), but not BI-4916 (NAD competitive) and CBR-5884 were shown to induced PHGDH clearance [8]. This suggests that NCT503 (at 20 μM) is ‘on target’ in cells, and that covalent-binding is not essential for PHGDH clearance. The aggregating-degrading ability of oridonin may be shared by other natural product PHGDH inhibitors, but more investigations are needed.

While testing whether cancer mutants were sensitive to oridonin, many cancer mutants (6 out of 8) were found to have impaired enzyme activity [8]. This seems to contradict the notion that cancer cells require more serine synthesis [26-28]. Several studies have demonstrated non-metabolic roles of PHGDH in different cancers [29-31]. For example, PHGDH interacted with the translation initiation factors eIF4A1 and eIF4E to facilitate the assembly of the translation initiation complex, thereby promoting the development of pancreatic cancer [30]. PHGDH was also reported to interact with and stabilize FOXM1 at the protein level, promoting the proliferation, invasion, and tumorigenicity of glioma cells [32]. Therefore, rather than focusing on inhibiting enzyme activity, it may be more desirable to develop PHGDH-degrading inhibitors, which may have a broader application in PHGDHdependent cancers.

In summary, four natural products were shown to directly engage and inhibit PHGDH. However, the inhibition mechanisms are quite different. Unlike Vmax type inhibitors azacoccone E and ixocarpalactone A, oridonin reduce the affinity of the substrate 3PG. The crystal structure indicates that a critical substrate-binding residue was relocated upon oridonin binding. Furthermore, multiple cysteines in PHGDH were prone to covalent binding by different covalent inhibitors. Selective clearance of cellular PHGDH may represent a new strategy for the treatment of PHGDH-dependent cancers. The insights generated may facilitate the mechanistic study for other known inhibitors and the development of new PHGDH inhibitors.

Acknowledgement

We thank Dr. Xiaofei Shen from the Hospital of Chengdu University of Traditional Chinese Medicine for helpful discussions.

Conflict of Interest

The authors declare no conflict of interest.

References

1. Mattaini KR, Sullivan MR, Vander Heiden MG. The importance of serine metabolism in cancer. Journal of Cell Biology. 2016 Aug 1;214(3):249-57.

2. Walsh DA, Sallach HJ. Purification and properties of chicken liver D-3-phosphoglycerate dehydrogenase. Biochemistry. 1965 Jun 1;4(6):1076-85.

3. Pizer LI. The pathway and control of serine biosynthesis in Escherichia coli. Journal of Biological Chemistry. 1963 Dec 1;238(12):3934-44.

4. Zhu J, Ma J, Wang X, Ma T, Zhang S, Wang W, et al. High Expression of PHGDH Predicts Poor Prognosis in Non–Small Cell Lung Cancer. Translational Oncology. 2016 Dec 1;9(6):592-9.

5. Song Z, Feng C, Lu Y, Lin Y, Dong C. PHGDH is an independent prognosis marker and contributes cell proliferation, migration and invasion in human pancreatic cancer. Gene. 2018 Feb 5;642:43-50.

6. Guo J, Gu X, Zheng M, Zhang Y, Chen L, Li H. Azacoccone E inhibits cancer cell growth by targeting 3-phosphoglycerate dehydrogenase. Bioorganic Chemistry. 2019 Jun 1;87:16-22.

7. Zheng M, Guo J, Xu J, Yang K, Tang R, Gu X, Li H, Chen L. Ixocarpalactone A from dietary tomatillo inhibits pancreatic cancer growth by targeting PHGDH. Food & Function. 2019;10(6):3386-95.

8. Tan Y, Zhou X, Gong Y, Gou K, Luo Y, Jia D, et al. Biophysical and biochemical properties of PHGDH revealed by studies on PHGDH inhibitors. Cellular and Molecular Life Sciences. 2022 Jan;79(1):1-3.

9. Harris CM, Foley SE, Goedken ER, Michalak M, Murdock S, Wilson NS. Merits and pitfalls in the characterization of covalent inhibitors of Bruton’s tyrosine kinase. SLAS DISCOVERY: Advancing Life Sciences R&D. 2018 Dec;23(10):1040-50.

10. Velander P, Wu L, Ray WK, Helm RF, Xu B. Amylin amyloid inhibition by flavonoid baicalein: key roles of its vicinal dihydroxyl groups of the catechol moiety. Biochemistry. 2016 Aug 9;55(31):4255-8.

11. Lonhienne T, Garcia MD, Pierens G, Mobli M, Nouwens A, Guddat LW. Structural insights into the mechanism of inhibition of AHAS by herbicides. Proceedings of the National Academy of Sciences. 2018 Feb 27;115(9):E1945-54.

12. Nussinov R, Tsai CJ. Allostery in disease and in drug discovery. Cell. 2013 Apr 11;153(2):293-305.

13. Lei Y, An Q, Shen XF, Sui M, Li C, Jia D, et al. Structure-guided design of the first noncovalent small-molecule inhibitor of CRM1. Journal of Medicinal Chemistry. 2021 May 11;64(10):6596-607.

14. Ding Y, Li D, Ding C, Wang P, Liu Z, Wold EA, et al. Regio-and stereospecific synthesis of oridonin D-ring aziridinated analogues for the treatment of triple-negative breast cancer via mediated irreversible covalent warheads. Journal of Medicinal Chemistry. 2018 Mar 12;61(7):2737-52.

15. Xu H, Qing X, Wang Q, Li C, Lai L. Dimerization of PHGDH via the catalytic unit is essential for its enzymatic function. Journal of Biological Chemistry. 2021 Jan 1;296.

16. Mullarky E, Lucki NC, Zavareh RB, Anglin JL, Gomes AP, Nicolay BN, et al. Identification of a small molecule inhibitor of 3-phosphoglycerate dehydrogenase to target serine biosynthesis in cancers. Proceedings of the National Academy of Sciences. 2016 Feb 16;113(7):1778-83.

17. Wang Q, Liberti MV, Liu P, Deng X, Liu Y, Locasale JW, et al. Rational design of selective allosteric inhibitors of PHGDH and serine synthesis with anti-tumor activity. Cell Chemical Biology. 2017 Jan 19;24(1):55-65.

18. Spillier Q, Vertommen D, Ravez S, Marteau R, Thémans Q, Corbet C, et al. Anti-alcohol abuse drug disulfiram inhibits human PHGDH via disruption of its active tetrameric form through a specific cysteine oxidation. Scientific Reports. 2019 Mar 18;9(1):4737.

19. Pan S, Jang SY, Liew SS, Fu J, Wang D, Lee JS, et al. A Vinyl Sulfone-Based Fluorogenic Probe Capable of Selective Labeling of PHGDH in Live Mammalian Cells. Angewandte Chemie International Edition. 2018 Jan 8;57(2):579-83.

20. Backus KM, Correia BE, Lum KM, Forli S, Horning BD, González- Páez GE, et al. Proteome-wide covalent ligand discovery in native biological systems. Nature. 2016 Jun;534(7608):570-4.

21. Yao Z, Xie F, Li M, Liang Z, Xu W, Yang J, et al. Oridonin induces autophagy via inhibition of glucose metabolism in p53-mutated colorectal cancer cells. Cell Death & Disease. 2017 Feb;8(2):e2633.

22. Huang H, Weng H, Dong B, Zhao P, Zhou H, Qu L. Oridonin triggers chaperon-mediated proteasomal degradation of BCR-ABL in leukemia. Scientific Reports. 2017 Jan 27;7:41525.

23. Kwan HY, Yang Z, Fong WF, Hu YM, Yu ZL, Hsiao WL. The anticancer effect of oridonin is mediated by fatty acid synthase suppression in human colorectal cancer cells. Journal of Gastroenterology. 2013 Feb;48(2):182-92.

24. Sullivan MR, Mattaini KR, Dennstedt EA, Nguyen AA, Sivanand S, Reilly MF, et al. Increased serine synthesis provides an advantage for tumors arising in tissues where serine levels are limiting. Cell Metabolism. 2019 Jun 4;29(6):1410-21.

25. Liu J, Zhang N, Li N, Fan X, Li Y. Influence of verapamil on the pharmacokinetics of oridonin in rats. Pharmaceutical Biology. 2019 Jan 1;57(1):787-91.

26. Possemato R, Marks KM, Shaul YD, Pacold ME, Kim D, Birsoy K, et al. Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature. 2011 Aug;476(7360):346-50.

27. Maddocks OD, Athineos D, Cheung EC, Lee P, Zhang T, van den Broek NJ, et al. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature. 2017 Apr;544(7650):372-6.

28. Reid MA, Allen AE, Liu S, Liberti MV, Liu P, Liu X, et al. Serine synthesis through PHGDH coordinates nucleotide levels by maintaining central carbon metabolism. Nature Communications. 2018 Dec 21;9(1): 5442.

29. Zhao X, Fu J, Du J, Xu W. The role of D-3-phosphoglycerate dehydrogenase in cancer. International Journal of Biological Sciences. 2020;16(9):1495-1506.

30. Ma X, Li B, Liu J, Fu Y, Luo Y. Phosphoglycerate dehydrogenase promotes pancreatic cancer development by interacting with eIF4A1 and eIF4E. Journal of Experimental & Clinical Cancer Research. 2019 Dec;38(1):1-5.

31. Wang Z, Pan Q, Yang L, Zhou H, Xu C, Yu F, et al. Automatic crystal centring procedure at the SSRF macromolecular crystallography beamline. Journal of Synchrotron Radiation. 2016 Nov 1;23(6):1323-32.

32. Liu J, Guo S, Li Q, Yang L, Xia Z, Zhang L, et al. Phosphoglycerate dehydrogenase induces glioma cells proliferation and invasion by stabilizing forkhead box M1. Journal of Neuro-Oncology. 2013 Feb;111(3):245-55.

Author Information X