Mechanisms of Gaucher disease pathogenesis
Editorial

Mechanisms of Gaucher disease pathogenesis

Simon Wheeler, Dan John Sillence

School of Pharmacy, Hawthorn Building, De Montfort University, Leicester, UK

Correspondence to: Dan John Sillence. School of Pharmacy, Faculty of Health and Life Sciences, Hawthorn Building, De Montfort University, Leicester, LE1 9BH, UK. Email: dsillence@dmu.ac.uk.

Submitted Jan 07, 2015. Accepted for publication Feb 06, 2015.

doi: 10.3978/j.issn.2305-5839.2015.03.42


What is Gaucher disease?

Gaucher disease is caused by mutations in the Gba1 gene encoding an acid β-glucocerebrosidase (GBA1), the lysosomal hydrolase which breaks down glucosylceramide (GlcCer). In Gaucher type 1 disease the accumulation of this simple glycolipid is mainly restricted to tissue phagocyte lysosomes resulting ultimately in hepatomegaly, splenomegaly and osteopenia. Lower residual GBA1 levels leads to neuronal storage, in types 2 and 3 neurological symptoms are characterised by acute (death at age 2) or sub-acute onset, respectively. The links between cellular changes and clinical manifestations are largely unknown but are the key to the development and monitoring of new therapies.

The newcomer to Gaucher disease is likely attracted to the apparent simplicity of an autosomal recessive disorder which promises to unravel the critical GlcCer function in normal cells (GlcCer is widespread, it’s even present in some bacteria—also, mouse and fly GlcCer knockouts die at embryo stage). However, closer acquaintance reveals not a classic Mendelian disorder—sometimes even monozygotic twins have different symptoms—and studies at the cellular level have so far failed to reveal clear GlcCer functions. Now a team led by Ellen Sidransky at the NIH has taken what appears to be a big step forward by producing two in vitro models of Gaucher cells (1).


How has Gaucher disease been investigated?

Research has been hampered by the inaccessibility of Gaucher macrophages and the lack of in vitro models. The simplest approach has been to induce a Gaucher phenotype by treating cells with the GBA1 inhibitor, conditurol-β-epoxide. Whilst this method has the virtues of being cheap and experimentally easy, off-target effects are not controlled for. For instance, conduritol-β-epoxide also inhibits a related enzyme, GBA2 (2). Inhibition of this non-lysosomal enzyme has been reported to rescue mutations in lysosomal GBA1 (3). GlcCer is unusual amongst glycolipids with intracellular trafficking connecting both lysosomal and non-lysosomal pools on both sides of the bilayer membrane (4). GlcCer transporters have been identified (5-7) but the relationships between different pools of GlcCer are still unclear.

A second approach has been to use fibroblasts from Gaucher patients. Although the macrophage-centric view of Gaucher disease has recently been questioned (8), Gaucher skin fibroblasts are not important in Gaucher disease and don’t store GlcCer.


What has now been achieved?

The researchers selected 20 Gaucher patients representing a total of 4 genotypes. Monocytes were extracted from these patients and differentiated into macrophages by the use of M-CSF: the resulting cells being termed hMacs. This method, whilst relatively quick and cheap, does not lead to a sustainable cell line. This was addressed by taking Gaucher fibroblasts from 4 patients and transformed them into induced pluripotent stem cells (iPSCs), then to monocytes and finally macrophages, referred to as iMacs. Whilst expensive and difficult, the use of stem cell technology means that this method does generate a sustainable cell line. Control cells of both types were produced from blood and fibroblasts donated by healthy volunteers.

Researchers examined the two cell types produced, hand-in-hand with an evaluation of previously disclosed (9) prototype drug NCGC00188758. This belongs to the class of molecules known as molecular chaperones: binding to the enzyme (in this case GBA1) correcting the misfolding. This results in repaired transport to the lysosome and enhanced GBA1 function.

Gratifyingly the phenotypes of hMacs and iMacs resembled genuine Gaucher cells. Compared with control macrophages, both cell types showed reduced impaired GBA1 activity and impaired transport of mutant GBA1 to lysosomes, indicated by colocalisation with the lysosomal marker LAMP2. Crucially, and in marked contrast to Gaucher fibroblasts, both cell types accumulated GlcCer and glucosylsphingosine. Cellular defects were rescued by treatment with NCGC00188758. Indeed, this small molecule drug was slightly more effective at restoring GBA1 activity than imiglucerase, an enzyme commonly used in enzyme replacement therapy.

Chemotaxis was found to be reduced versus controls, an observation previously reported for some, though not all, Gaucher patients (10). Whilst the Gaucher model cells were found to phagocytose IgG-opsonised erythrocytes and bacteria normally, dysfunction was found in the production of reactive oxygen species (ROS). Thus iMacs and hMacs had lower concentration of ROS in the resting state, and no further generation of ROS upon phagocytosis. These findings mirror previous reports on impaired superoxide generation in Gaucher cells (10,11). Importantly both chemotaxis and ROS production were restored on treatment with NCGC00188758.


The potential medical significance of Gaucher disease does not end with the condition itself. Most attention has been focussed on the unexpected finding that having even one mutant copy of the Gba1 gene is a significant risk factor for Parkinson’s disease (12). This has prompted research interest into the possible links between Gaucher disease and Parkinson’s disease. The most fundamental observation is that poorly functional GBA1 is associated with the accumulation of α-synuclein (α-syn) (13) leading to neuronal death. This protein can fold and aggregate in many different ways and a possible mechanism for its accumulation is the stabilisation of oligomers by GlcCer (13). In turn, α-syn can inhibit GBA1 (14), an observation that may well account for the reduced levels of GBA1 activity seen in post mortem brains of sporadic Parkinson’s disease patients (15). Furthermore, α-syn can interfere with vesicular traffic of GBA1 from the ER to the Golgi (13). Thus, by means of a bi-directional loop, even a slight loss of GBA1 function can become magnified. A qualification to the above discussion arises from the observation that post-mortem brains of patients suffering from all types of Gaucher disease had monomeric, but not oligomeric α-syn (16). Further work is needed to unravel the exact mechanism by which mutant Gba1 gives rise to α-syn aggregates. Further explorations of the consequences of this accumulation of cytosolic, insoluble α-syn are also required. It has been shown, for example, that in normal neurones α-syn is localised at the synaptic membrane [where it plays a role in regulating synaptic vesicles (17)] and that this localisation is mediated by lipid rafts (18). How these changes relate to increased raft-forming GlcCer has yet to be addressed.


Future research using Gaucher cell models

Recent research has revealed interdependence of phagosome pH and ROS generation (19) hence decreased generation of ROS might be linked to increased pH of Gaucher lysosomes (20). Increased pH may also explain reduced lysosomal proteolysis (13), co-storage of cholesterol and disrupted membrane trafficking in Gaucher cells (21). Alternatively, glucosylsphingosine (GlcSph) may mediate decreased ROS (22). Whilst interest has generally focussed on GlcSph as a biomarker for Gaucher disease, it’s still an open question whether enough GlcSph escapes the lysosome to inhibit Protein kinase C [IC50 =85 µM (23)]. However, PKC has also been implicated in the phagocytosis of opsonised bacteria (24).

Several workers have reported increased levels of inflammatory markers, including M-CSF, in the serum of patients with Gaucher disease. These observations raise the possibility that this could be the cause of the reported proliferation of osteoclasts associated with Gaucher disease (25,26) and the consequent occurrence of bone symptoms in some patients.

In conclusion it appears that the researchers have produced both a realistic model of Gaucher cells and a promising prototype drug. Although much work is required before NCGC00188758 can be considered as a usable drug in patients, there is a particular lack of treatment options for the neurodegenerative forms of Gaucher disease (12,27).


Acknowledgements

Disclosure: The authors declare no conflict of interest.


References

  1. Aflaki E, Stubblefield BK, Maniwang E, et al. Macrophage models of Gaucher disease for evaluating disease pathogenesis and candidate drugs. Sci Transl Med 2014;6:240ra73.
  2. Ridley CM, Thur KE, Shanahan J, et al. β-Glucosidase 2 (GBA2) activity and imino sugar pharmacology. J Biol Chem 2013;288:26052-66. [PubMed]
  3. Mistry PK, Liu J, Sun L, et al. Glucocerebrosidase 2 gene deletion rescues type 1 Gaucher disease. Proc Natl Acad Sci U S A 2014;111:4934-9. [PubMed]
  4. Halter D, Neumann S, van Dijk SM, et al. Pre- and post-Golgi translocation of glucosylceramide in glycosphingolipid synthesis. J Cell Biol 2007;179:101-15. [PubMed]
  5. D'Angelo G, Uemura T, Chuang CC, et al. Vesicular and non-vesicular transport feed distinct glycosylation pathways in the Golgi. Nature 2013;501:116-20. [PubMed]
  6. Tuuf J, Mattjus P. Membranes and mammalian glycolipid transferring proteins. Chem Phys Lipids 2014;178:27-37. [PubMed]
  7. Malinina L, Malakhova ML, Teplov A, et al. Structural basis for glycosphingolipid transfer specificity. Nature 2004;430:1048-53. [PubMed]
  8. Mistry PK, Liu J, Yang M, et al. Glucocerebrosidase gene-deficient mouse recapitulates Gaucher disease displaying cellular and molecular dysregulation beyond the macrophage. Proc Natl Acad Sci U S A 2010;107:19473-8. [PubMed]
  9. Patnaik S, Zheng W, Choi JH, et al. Discovery, structure-activity relationship, and biological evaluation of noninhibitory small molecule chaperones of glucocerebrosidase. J Med Chem 2012;55:5734-48. [PubMed]
  10. Liel Y, Rudich A, Nagauker-Shriker O, et al. Monocyte dysfunction in patients with Gaucher disease: evidence for interference of glucocerebroside with superoxide generation. Blood 1994;83:2646-53. [PubMed]
  11. Maródi L, Káposzta R, Tóth J, et al. Impaired microbicidal capacity of mononuclear phagocytes from patients with type I Gaucher disease: partial correction by enzyme replacement therapy. Blood 1995;86:4645-9. [PubMed]
  12. Sidransky E, Lopez G. The link between the GBA gene and parkinsonism. Lancet Neurol 2012;11:986-98. [PubMed]
  13. Mazzulli JR, Xu YH, Sun Y, et al. Gaucher disease glucocerebrosidase and α-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell 2011;146:37-52. [PubMed]
  14. Yap TL, Velayati A, Sidransky E, et al. Membrane-bound α-synuclein interacts with glucocerebrosidase and inhibits enzyme activity. Mol Genet Metab 2013;108:56-64. [PubMed]
  15. Gegg ME, Burke D, Heales SJ, et al. Glucocerebrosidase deficiency in substantia nigra of parkinson disease brains. Ann Neurol 2012;72:455-63. [PubMed]
  16. Choi JH, Stubblefield B, Cookson MR, et al. Aggregation of α-synuclein in brain samples from subjects with glucocerebrosidase mutations. Mol Genet Metab 2011;104:185-8. [PubMed]
  17. Murphy DD, Rueter SM, Trojanowski JQ, et al. Synucleins are developmentally expressed, and alpha-synuclein regulates the size of the presynaptic vesicular pool in primary hippocampal neurons. J Neurosci 2000;20:3214-20. [PubMed]
  18. Fortin DL, Troyer MD, Nakamura K, et al. Lipid rafts mediate the synaptic localization of alpha-synuclein. J Neurosci 2004;24:6715-23. [PubMed]
  19. Nunes P, Demaurex N, Dinauer MC. Regulation of the NADPH oxidase and associated ion fluxes during phagocytosis. Traffic 2013;14:1118-31. [PubMed]
  20. Sillence DJ. Glucosylceramide modulates endolysosomal pH in Gaucher disease. Mol Genet Metab 2013;109:194-200. [PubMed]
  21. Sillence DJ, Puri V, Marks DL, et al. Glucosylceramide modulates membrane traffic along the endocytic pathway. J Lipid Res 2002;43:1837-45. [PubMed]
  22. Dekker N, van Dussen L, Hollak CE, et al. Elevated plasma glucosylsphingosine in Gaucher disease: relation to phenotype, storage cell markers, and therapeutic response. Blood 2011;118:e118-27. [PubMed]
  23. Hannun YA, Bell RM. Lysosphingolipids inhibit protein kinase C: implications for the sphingolipidoses. Science 1987;235:670-4. [PubMed]
  24. Zheleznyak A, Brown EJ. Immunoglobulin-mediated phagocytosis by human monocytes requires protein kinase C activation. Evidence for protein kinase C translocation to phagosomes. J Biol Chem 1992;267:12042-8. [PubMed]
  25. Mucci JM, Scian R, De Francesco PN, et al. Induction of osteoclastogenesis in an in vitro model of Gaucher disease is mediated by T cells via TNF-α. Gene 2012;509:51-9. [PubMed]
  26. Reed M, Baker RJ, Mehta AB, et al. Enhanced differentiation of osteoclasts from mononuclear precursors in patients with Gaucher disease. Blood Cells Mol Dis 2013;51:185-94. [PubMed]
  27. Schiffmann R, Fitzgibbon EJ, Harris C, et al. Randomized, controlled trial of miglustat in Gaucher's disease type 3. Ann Neurol 2008;64:514-22. [PubMed]
Cite this article as: Wheeler S, Sillence DJ. Mechanisms of Gaucher disease pathogenesis. Ann Transl Med 2015;3(S1):S1. doi: 10.3978/j.issn.2305-5839.2015.03.42

Download Citation