1
|
Paquet Luzy C, Doppler E, Polasek TM, Giorgino R. First-in-human single-dose study of nizubaglustat, a dual inhibitor of ceramide glucosyltransferase and non-lysosomal glucosylceramidase: Safety, tolerability, pharmacokinetics, and pharmacodynamics of single ascending and multiple doses in healthy adults. Mol Genet Metab 2024; 141:108113. [PMID: 38113551 DOI: 10.1016/j.ymgme.2023.108113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
Nizubaglustat is a novel, orally available, brain penetrant, potent, and selective dual inhibitor of ceramide glucosyltranferase and non-lysosomal neutral glucosylceramidase (NLGase), which is currently under development for the treatment of subjects with neurological manifestations in primary and secondary gangliosidoses. The objectives of this first-in-human study were to evaluate the safety and tolerability, pharmacokinetics, and pharmacodynamics (PD) of single oral doses of nizubaglustat after single (1, 3, and 9 mg) and multiple oral doses (9 mg once per day (QD) over 14 days) in healthy adults. Nizubaglustat was rapidly absorbed and systemic exposure was dose-proportional. Steady-state was achieved after three days of QD multiple dosing with minimal accumulation. Renal clearance accounted for around 15% of nizubaglustat elimination. Following multiple dosing, plasma concentrations of glucosylceramide (GlcCer), lactosylceramide (LacCer), and monosialodihexosylganglioside (GM3) decreased to a nadir at Day 10. PD target engagement of GCS inhibition was shown by a median decrease from baseline of plasma concentrations of GlcCer, LacCer, and GM3 ganglioside by 70%, 50%, and 48%, respectively. NLGase inhibition was also manifested by increased concentrations of GlcCer in cerebrospinal fluid from Day 1 to Day 14. Nizubaglustat was safe and well-tolerated at all doses tested. Consistent with the high selectivity, and the absence of intestinal disaccharidases inhibition, no cases of diarrhea were reported. No decreased appetite or weight loss was noted. Only treatment-emergent adverse events with preferred terms belonging to the system organ class skin and subcutaneous disorders of mild intensity were reported as drug-related in the nizubaglustat arm, in line with the pharmacological mechanism targeting glucosylceramide metabolism. Taken together, these data support QD dosing of nizubaglustat and its ongoing development in patients with primary and secondary forms of gangliosidoses.
Collapse
Affiliation(s)
| | | | - Thomas M Polasek
- Principal Investigator, CMAX Research Phase 1 Unit, Ground Floor 21-24 North Terrace, Adelaide, 5000, SA, Australia; Department of Clinical Pharmacology, Royal Adelaide Hospital, Port Rd, Adelaide, SA 5000, Australia
| | | |
Collapse
|
2
|
Harzer K, Yildiz Y, Beck-Wödl S. Assay of β-glucosidase 2 (GBA2) activity using lithocholic acid β-3-O-glucoside substrate for cultured fibroblasts and glucosylceramide for brain tissue. Biol Chem 2019; 400:745-752. [PMID: 30864417 DOI: 10.1515/hsz-2018-0438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/27/2018] [Indexed: 11/15/2022]
Abstract
Beta (β)-glucosidase 2 (GBA2) is deficient in a form of human spastic paraplegia due to defects in GBA2 (SPG46). GBA2 was proposed as a modifier of Gaucher disease, a lysosomal storage disease resulting from deficient β-glucosidase 1; GBA1. Current GBA2 activity assays using artificial substrates incompletely model the activity encountered in vivo. We studied GBA2 activity, using lithocholic acid β-glucoside or glucosylceramide as natural β-glucosidase substrates in murine tissues or cultured patient fibroblasts with the pathologic genotypes: Gba1-/-; Gba2-/-; GBA1-/-; GBA2+/- and found expected and unexpected deviations from normal controls.
Collapse
Affiliation(s)
- Klaus Harzer
- Department of Neuropediadrics, Neurometabolic Laboratory, Children's Hospital, University of Tübingen, Hoppe-Seyler-Str. 1, D-72076 Tübingen, Germany
| | - Yildiz Yildiz
- Internal Medicine, Medicnova Hospital, Selemad 10, FL-9487 Gamprin-Bendern, Liechtenstein
| | - Stefanie Beck-Wödl
- Department of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstr. 7, D-72076 Tübingen, Germany
| |
Collapse
|
3
|
Woeste MA, Wachten D. The Enigmatic Role of GBA2 in Controlling Locomotor Function. Front Mol Neurosci 2017; 10:386. [PMID: 29234271 PMCID: PMC5712312 DOI: 10.3389/fnmol.2017.00386] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 11/06/2017] [Indexed: 01/22/2023] Open
Abstract
The non-lysosomal glucosylceramidase GBA2 catalyzes the hydrolysis of glucosylceramide to glucose and ceramide. Loss of GBA2 function results in accumulation of glucosylceramide. Mutations in the human GBA2 gene have been associated with hereditary spastic paraplegia (HSP) and autosomal-recessive cerebellar ataxia (ARCA). Patients suffering from these disorders exhibit impaired locomotion and neurological abnormalities. GBA2 mutations found in these patients have been proposed to impair GBA2 function. However, the molecular mechanism underlying the occurrence of mutations in the GBA2 gene and the development of locomotor dysfunction is not well-understood. In this review, we aim to summarize recent findings regarding mutations in the GBA2 gene and their impact on GBA2 function in health and disease.
Collapse
Affiliation(s)
- Marina A Woeste
- Institute of Innate Immunity, University Hospital, University of Bonn, Bonn, Germany
| | - Dagmar Wachten
- Institute of Innate Immunity, University Hospital, University of Bonn, Bonn, Germany.,Molecular Physiology, Center of Advanced European Studies and Research, Minerva Max Planck Research Group, Bonn, Germany
| |
Collapse
|
4
|
Current and Novel Aspects on the Non-lysosomal β-Glucosylceramidase GBA2. Neurochem Res 2015; 41:210-20. [DOI: 10.1007/s11064-015-1763-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 10/22/2022]
|
5
|
Sansenya S, Mutoh R, Charoenwattanasatien R, Kurisu G, Ketudat Cairns JR. Expression and crystallization of a bacterial glycoside hydrolase family 116 β-glucosidase from Thermoanaerobacterium xylanolyticum. Acta Crystallogr F Struct Biol Commun 2015; 71:41-4. [PMID: 25615966 PMCID: PMC4304745 DOI: 10.1107/s2053230x14025461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 11/20/2014] [Indexed: 11/10/2022] Open
Abstract
The Thermoanaerobacterium xylanolyticum gene product TxGH116, a glycoside hydrolase family 116 protein of 806 amino-acid residues sharing 37% amino-acid sequence identity over 783 residues with human glucosylceramidase 2 (GBA2), was expressed in Escherichia coli. Purification by heating, immobilized metal-affinity and size-exclusion chromatography produced >90% pure TxGH116 protein with an apparent molecular mass of 90 kDa on SDS-PAGE. The purified TxGH116 enzyme hydrolyzed the p-nitrophenyl (pNP) glycosides pNP-β-D-glucoside, pNP-β-D-galactoside and pNP-N-acetyl-β-D-glucopyranoside, as well as cellobiose and cellotriose. The TxGH116 protein was crystallized using a precipitant consisting of 0.6 M sodium citrate tribasic, 0.1 M Tris-HCl pH 7.0 by vapour diffusion with micro-seeding to form crystals with maximum dimensions of 120×25×5 µm. The TxGH116 crystals diffracted X-rays to 3.15 Å resolution and belonged to the monoclinic space group P2(1). Structure solution will allow a structural explanation of the effects of human GBA2 mutations.
Collapse
Affiliation(s)
- Sompong Sansenya
- Institute of Science, School of Biochemistry and Center for Biomolecular Structure, Function and Application, Suranaree University of Technology, 111 University Avenue, Nakhon Ratchasima Muang District, Nakhon Ratchasima 30000, Thailand
- Department of Chemistry, Faculty of Science, Rajamangala University of Technology, Thanyaburi, 39 Moo 1, Rangsit-Nakhon Nayok Road, Klong 6, Thanyaburi, Pathum Thani 12110, Thailand
| | - Risa Mutoh
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ratana Charoenwattanasatien
- Institute of Science, School of Biochemistry and Center for Biomolecular Structure, Function and Application, Suranaree University of Technology, 111 University Avenue, Nakhon Ratchasima Muang District, Nakhon Ratchasima 30000, Thailand
| | - Genji Kurisu
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - James R. Ketudat Cairns
- Institute of Science, School of Biochemistry and Center for Biomolecular Structure, Function and Application, Suranaree University of Technology, 111 University Avenue, Nakhon Ratchasima Muang District, Nakhon Ratchasima 30000, Thailand
- Laboratory of Biochemistry, Chulabhorn Research Institute, Vipavadee-Rangsit Highway, Bangkok 10210, Thailand
| |
Collapse
|
6
|
Martin E, Schüle R, Smets K, Rastetter A, Boukhris A, Loureiro JL, Gonzalez MA, Mundwiller E, Deconinck T, Wessner M, Jornea L, Oteyza AC, Durr A, Martin JJ, Schöls L, Mhiri C, Lamari F, Züchner S, De Jonghe P, Kabashi E, Brice A, Stevanin G. Loss of function of glucocerebrosidase GBA2 is responsible for motor neuron defects in hereditary spastic paraplegia. Am J Hum Genet 2013; 92:238-44. [PMID: 23332916 DOI: 10.1016/j.ajhg.2012.11.021] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/15/2012] [Accepted: 11/30/2012] [Indexed: 01/30/2023] Open
Abstract
Spastic paraplegia 46 refers to a locus mapped to chromosome 9 that accounts for a complicated autosomal-recessive form of hereditary spastic paraplegia (HSP). With next-generation sequencing in three independent families, we identified four different mutations in GBA2 (three truncating variants and one missense variant), which were found to cosegregate with the disease and were absent in controls. GBA2 encodes a microsomal nonlysosomal glucosylceramidase that catalyzes the conversion of glucosylceramide to free glucose and ceramide and the hydrolysis of bile acid 3-O-glucosides. The missense variant was also found at the homozygous state in a simplex subject in whom no residual glucocerebrosidase activity of GBA2 could be evidenced in blood cells, opening the way to a possible measurement of this enzyme activity in clinical practice. The overall phenotype was a complex HSP with mental impairment, cataract, and hypogonadism in males associated with various degrees of corpus callosum and cerebellar atrophy on brain imaging. Antisense morpholino oligonucleotides targeting the zebrafish GBA2 orthologous gene led to abnormal motor behavior and axonal shortening/branching of motoneurons that were rescued by the human wild-type mRNA but not by applying the same mRNA containing the missense mutation. This study highlights the role of ceramide metabolism in HSP pathology.
Collapse
Affiliation(s)
- Elodie Martin
- Unité Mixte de Recherche S975, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epinière, Pitie-Salpêtrière Hospital, Université Pierre et Marie Curie (Paris 6), Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Harzer K, Blech-Hermoni Y, Goldin E, Felderhoff-Mueser U, Igney C, Sidransky E, Yildiz Y. Beta-glucosidase 1 (GBA1) is a second bile acid β-glucosidase in addition to β-glucosidase 2 (GBA2). Study in β-glucosidase deficient mice and humans. Biochem Biophys Res Commun 2012; 423:308-12. [PMID: 22659419 DOI: 10.1016/j.bbrc.2012.05.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 05/18/2012] [Indexed: 10/28/2022]
Abstract
Beta-glucosidase 1 (GBA1; lysosomal glucocerebrosidase) and β-glucosidase 2 (GBA2, non-lysosomal glucocerebrosidase) both have glucosylceramide as a main natural substrate. The enzyme-deficient conditions with glucosylceramide accumulation are Gaucher disease (GBA-/- in humans), modelled by the Gba-/- mouse, and the syndrome with male infertility in the Gba2-/- mouse, respectively. Before the leading role of glucosylceramide was recognised for both deficient conditions, bile acid-3-O-β-glucoside (BG), another natural substrate, was viewed as the main substrate of GBA2. Given that GBA2 hydrolyses both BG and glucosylceramide, it was asked whether vice versa GBA1 hydrolyses both glucosylceramide and BG. Here we show that GBA1 also hydrolyses BG. We compared the residual BG hydrolysing activities in the GBA1-/-, Gba1-/- conditions (where GBA2 is the almost only active β-glucosidase) and those in the Gba2-/- condition (GBA1 active), with wild-type activities, but we used also the GBA1 inhibitor isofagomine. GBA1 and GBA2 activities had characteristic differences between the studied fibroblast, liver and brain samples. Independently, the hydrolysis of BG by pure recombinant GBA1 was shown. The fact that both GBA1 and GBA2 are glucocerebrosidases as well as bile acid β-glucosidases raises the question, why lysosomal accumulation of glucosylceramide in GBA1 deficiency, and extra-lysosomal accumulation in GBA2 deficiency, are not associated with an accumulation of BG in either condition.
Collapse
Affiliation(s)
- Klaus Harzer
- Neurometabolic Laboratory, Klinik für Kinder- und Jugendmedizin, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
8
|
Wang Y, Li J, Xu Y. Characterization of novel β-glucosidases with transglycosylation properties from Trichosporon asahii. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:11219-27. [PMID: 21916502 DOI: 10.1021/jf203693v] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Two novel β-glucosidases from Trichosporon asahii, named BG1 and BG2, were purified to electrophoretic homogeneity using ammonium sulfate precipitation, hydrophobic interaction, ion exchange, and gelfiltration chromatography. The molecular weight of BG1 and BG2 were estimated as 160 kDa and 30 kDa, respectively. The K(m), V(max), K(cat), and K(cat)/K(m) values of the two β-glucosidases for p-nitrophenyl-β-D-glucopyranoside were determined. Both enzymes showed relatively high affinity to p-nitrophenyl-β-D-glucopyranoside in 4-nitrophenol glycosides and gentiobiose in saccharide substrates. The enzymes exhibited optimum activity at pH 6.0 and pH 5.5, respectively. Their respective optimum temperatures were 70 and 50 °C. Metal ions and inhibitors had different effects on the enzymes activities. Circular dichroism (CD) spectroscopy demonstrated that the purified BG1 exhibited a β-sheet-rich structure and that BG2 displayed a high random coil conformation. HPLC analysis of transglycosylation and reverse hydrolysis assays revealed that only BG1 possessed transglycosylation activity and synthesized cello-oligosaccharides by the addition of glucose. This suggested that BG1 could be used to produce complex bioactive glycosides and could be considered as a potential enzyme for industrial application.
Collapse
Affiliation(s)
- Yuxia Wang
- State Key Laboratory of Food Science and Technology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China
| | | | | |
Collapse
|
9
|
Walden CM, Sandhoff R, Chuang CC, Yildiz Y, Butters TD, Dwek RA, Platt FM, van der Spoel AC. Accumulation of Glucosylceramide in Murine Testis, Caused by Inhibition of β-Glucosidase 2. J Biol Chem 2007; 282:32655-64. [PMID: 17848577 DOI: 10.1074/jbc.m702387200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
One of the hallmarks of male germ cell development is the formation of a specialized secretory organelle, the acrosome. This process can be pharmacologically disturbed in C57BL/6 mice, and thus infertility can be induced, by small molecular sugar-like compounds (alkylated imino sugars). Here the biochemical basis of this effect has been investigated. Our findings suggest that in vivo alkylated imino sugars primarily interact with the non-lysosomal glucosylceramidase. This enzyme cleaves glucosylceramide into glucose and ceramide, is sensitive to imino sugars in vitro, and has been characterized as beta-glucosidase 2 (GBA2). Imino sugars raised the level of glucosylceramide in brain, spleen, and testis, in a dose-dependent fashion. In testis, multiple species of glucosylceramide were similarly elevated, those having long acyl chains (C16-24), as well as those with very long polyunsaturated acyl chains (C28-30:5). Both of these GlcCer species were also increased in the testes from GBA2-deficient mice. When considering that the very long polyunsaturated sphingolipids are restricted to germ cells, these results indicate that in the testis GBA2 is present in both somatic and germ cells. Furthermore, in all mouse strains tested imino sugar treatment caused a rise in testicular glucosylceramide, even in a number of strains, of which the males remain fertile after drug administration. Therefore, it appears that acrosome formation can be derailed by accumulation of glucosylceramide in an extralysosomal localization, and that the sensitivity of male germ cells to glucosylceramide is genetically determined.
Collapse
Affiliation(s)
- Charlotte M Walden
- Departments of Biochemistry and Pharmacology, University of Oxford, Mansfield Road, Oxford, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Boot RG, Verhoek M, Donker-Koopman W, Strijland A, van Marle J, Overkleeft HS, Wennekes T, Aerts JMFG. Identification of the Non-lysosomal Glucosylceramidase as β-Glucosidase 2. J Biol Chem 2007; 282:1305-12. [PMID: 17105727 DOI: 10.1074/jbc.m610544200] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The primary catabolic pathway for glucosylceramide is catalyzed by the lysosomal enzyme glucocerebrosidase that is defective in Gaucher disease patients. A distinct non-lysosomal glucosylceramidase has been described but its identity remained enigmatic for years. We here report that the non-lysosomal glucosylceramidase is identical to the earlier described bile acid beta-glucosidase, being beta-glucosidase 2 (GBA2). Expressed GBA2 is identical to the native non-lysosomal glucosylceramidase in various enzymatic features such as substrate specificity and inhibitor sensitivity. Expression of GBA2 coincides with increased non-lysosomal glucosylceramidase activity, and GBA2-targeted RNA interference reduces endogenous non-lysosomal glucosylceramidase activity in cells. GBA2 is found to be located at or close to the cell surface, and its activity is linked to sphingomyelin generation. Hydrophobic deoxynojirimycins are extremely potent inhibitors for GBA2. In mice pharmacological inhibition of GBA2 activity is associated with impaired spermatogenesis, a phenomenon also very recently reported for GBA2 knock-out mice (Yildiz, Y., Matern, H., Thompson, B., Allegood, J. C., Warren, R. L., Ramirez, D. M., Hammer, R. E., Hamra, F. K., Matern, S., and Russell, D. W. (2006) J. Clin. Invest. 116, 2985-2994). In conclusion, GBA2 plays a role in cellular glucosylceramide metabolism.
Collapse
Affiliation(s)
- Rolf G Boot
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Yildiz Y, Matern H, Thompson B, Allegood JC, Warren RL, Ramirez DM, Hammer RE, Hamra FK, Matern S, Russell DW. Mutation of beta-glucosidase 2 causes glycolipid storage disease and impaired male fertility. J Clin Invest 2006; 116:2985-94. [PMID: 17080196 PMCID: PMC1626112 DOI: 10.1172/jci29224] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Accepted: 07/25/2006] [Indexed: 01/24/2023] Open
Abstract
beta-Glucosidase 2 (GBA2) is a resident enzyme of the endoplasmic reticulum thought to play a role in the metabolism of bile acid-glucose conjugates. To gain insight into the biological function of this enzyme and its substrates, we generated mice deficient in GBA2 and found that these animals had normal bile acid metabolism. Knockout males exhibited impaired fertility. Microscopic examination of sperm revealed large round heads (globozoospermia), abnormal acrosomes, and defective mobility. Glycolipids, identified as glucosylceramides by mass spectrometry, accumulated in the testes, brains, and livers of the knockout mice but did not cause obvious neurological symptoms, organomegaly, or a reduction in lifespan. Recombinant GBA2 hydrolyzed glucosylceramide to glucose and ceramide; the same reaction catalyzed by the beta-glucosidase acid 1 (GBA1) defective in subjects with the Gaucher's form of lysosomal storage disease. We conclude that GBA2 is a glucosylceramidase whose loss causes accumulation of glycolipids and an endoplasmic reticulum storage disease.
Collapse
Affiliation(s)
- Yildiz Yildiz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heidrun Matern
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bonne Thompson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jeremy C. Allegood
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rebekkah L. Warren
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Denise M.O. Ramirez
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert E. Hammer
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - F. Kent Hamra
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Siegfried Matern
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David W. Russell
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Biochemistry and
Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
12
|
Gebhardt R. Inhibition of cholesterol biosynthesis in HepG2 cells by artichoke extracts is reinforced by glucosidase pretreatment. Phytother Res 2002; 16:368-72. [PMID: 12112295 DOI: 10.1002/ptr.960] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
High-dose aqueous extracts from artichoke leaves were found to inhibit cholesterol biosynthesis from (14)C-acetate rather moderately in HepG2 cells in contrast to primary cultured rat hepatocytes in which the inhibition was stronger. Preincubation of the extracts with several glycohydrolases revealed that pretreatment with beta-glucosidase considerably reinforced the inhibition. A significant reduction of acetate incorporation was found above extract concentrations of 0.01 mg/mL and at 0.2 mg/mL almost 60% inhibition was observed. Cytotoxic effects detected by the MTT-assay were restricted to higher concentrations of the extracts with and without beta-glucosidase pretreatment. Since cynaroside represents a major glucoside in artichoke extracts, both cynaroside and its aglycone luteolin were tested. It could be demonstrated that cynaroside is indeed one of the targets of beta-glucosidase and that the liberated luteolin is responsible for the inhibitory effect. Direct measurements of beta-glucosidase activity in rat hepatocytes and HepG2 cells revealed that endogenous enzyme activity in hepatocytes may be sufficient to convert cynaroside to its aglycone, while in HepG2 cells this may not be the case. These findings emphasize the importance of beta-glucosidase-dependent liberation of luteolin for the ability of artichoke extracts to inhibit hepatic cholesterol biosynthesis.
Collapse
Affiliation(s)
- Rolf Gebhardt
- Institut für Biochemie, Universitätsklinikum Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
13
|
Matern H, Boermans H, Lottspeich F, Matern S. Molecular cloning and expression of human bile acid beta-glucosidase. J Biol Chem 2001; 276:37929-33. [PMID: 11489889 DOI: 10.1074/jbc.m104290200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A novel microsomal beta-glucosidase was recently purified and characterized from human liver that catalyzes the hydrolysis of bile acid 3-O-glucosides as endogenous compounds. The primary structure of this bile acid beta-glucosidase was deduced by cDNA cloning on the basis of the amino acid sequences of peptides obtained from the purified enzyme by proteinase digestion. The isolated cDNA comprises 3639 base pairs containing 524 nucleotides of 5'-untranslated and 334 nucleotides of 3'-untranslated sequences including the poly(A) tail. The open reading frame predicts a 927-amino acid protein with a calculated M(r) of 104,648 containing one putative transmembrane domain. Data base searches revealed no homology with any known glycosyl hydrolase or other functionally identified protein. The cDNA sequence was found with significant identity in the human chromosome 9 clone RP11-112J3 of the human genome project. The recombinant enzyme was expressed in a tagged form in COS-7 cells where it displayed bile acid beta-glucosidase activity. Northern blot analysis of various human tissues revealed high levels of expression of the bile acid beta-glucosidase mRNA (3.6-kilobase message) in brain, heart, skeletal muscle, kidney, and placenta and lower levels of expression in the liver and other organs.
Collapse
Affiliation(s)
- H Matern
- Department of Internal Medicine III, Rheinisch-Westfälische Technische Hochschule Aachen, 52057 Aachen, Germany.
| | | | | | | |
Collapse
|
14
|
Hays WS, Wheeler DE, Eghtesad B, Glew RH, Johnston DE. Expression of cytosolic beta-glucosidase in guinea pig liver cells. Hepatology 1998; 28:156-63. [PMID: 9657108 DOI: 10.1002/hep.510280121] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytosolic beta-glucosidase of mammalian liver has been implicated in the metabolic transformation of plant glycosides, such as vicine and amygdalin, which are associated with the development of toxic syndromes. We investigated which cell types express cytosolic beta-glucosidase in guinea pig liver, and characterized the contribution of this enzyme to the hydrolysis of aromatic glucosides in cultured cells and in tissue slices. Cytosolic beta-glucosidase was expressed in hepatocytes and not in Kupffer or endothelial cells as determined by enzyme-specific activity and Western blots of liver cell extracts. Intracellular beta-glucosidase activity was visualized using the fluorescent beta-glucosidase substrate, resorufin beta-D-glucoside, and shown to be caused by the cytosolic beta-glucosidase using the inhibitors, conduritol beta-epoxide and dinitrophenol-2-deoxy-2-fluoro-beta-D-glucopyranoside (DNP2FGlc). Staining of fresh liver slices with resorufin beta-glucoside revealed that cytosolic beta-glucosidase is expressed in all hepatocytes, with no significant portal-central gradient. These data indicate that cytosolic beta-glucosidase is a hepatocyte-specific enzyme, and support the hypothesis that cytosolic beta-glucosidase in the liver functions to hydrolyze small glucosides absorbed by the intestine. Furthermore, toxic injury to cultured hepatocytes by CCl4 resulted in release of cytosolic beta-glucosidase in parallel with the hepatocyte marker enzymes alanine transaminase and lactate dehydrogenase. This suggests that acute increases in serum levels of cytosolic beta-glucosidase in animal models of liver injury may reflect direct injury of hepatocytes.
Collapse
Affiliation(s)
- W S Hays
- Department of Biochemistry, University of New Mexico Health Sciences Center, Albuquerque, 87131-5271 USA
| | | | | | | | | |
Collapse
|
15
|
Matern H, Heinemann H, Legler G, Matern S. Purification and characterization of a microsomal bile acid beta-glucosidase from human liver. J Biol Chem 1997; 272:11261-7. [PMID: 9111029 DOI: 10.1074/jbc.272.17.11261] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A human liver microsomal beta-glucosidase has been purified to apparent homogeneity in sodium dodecyl sulfate-polyacrylamide gel electrophoresis where a single protein band of Mr 100,000 was obtained under reducing conditions. The enzyme was enriched about 73, 000-fold over starting microsomal membranes by polyethylene glycol fractionation, anion exchange chromatographies on DEAE-Trisacryl, and Mono Q followed by affinity chromatography on N-(9-carboxynonyl)-1-deoxynojirimycin-AH-Sepharose 4B. The purified enzyme had a pH optimum between 5.0 and 6.4, was activated by divalent metal ions, and required phospholipids for exhibition of activity. The enzyme catalyzed the hydrolysis of 3beta-D-glucosido-lithocholic and 3beta-D-glucosido-chenodeoxycholic acids with high affinity (Km, 1.7 and 6.2 microM, respectively) and of the beta-D-glucoside (Km, 210 microM) and the beta-D-galactoside of 4-methylumbelliferone. The ratio of relative reaction rates for these substrates was about 6:3:11:1. No activity was detectable toward 6beta-D-glucosido-hyodeoxycholic acid, glucocerebroside, and the following glycosides of 4-methylumbelliferone: alpha-D-glucoside, alpha-L-arabinoside, beta-D-fucoside or beta-D-xyloside. Immunoinhibition and immunoprecipitation studies using antibodies prepared against lysosomal glucocerebrosidase showed no cross-reactivity with microsomal beta-glucosidase suggesting that these two enzymes are antigenically unrelated.
Collapse
Affiliation(s)
- H Matern
- Department of Internal Medicine III, Aachen University of Technology, D-52074 Aachen, Federal Republic of Germany
| | | | | | | |
Collapse
|
16
|
Iida T, Nishida S, Yamaguchi Y, Kodake M, Chang FC, Niwa T, Goto J, Nambara T. Potential bile acid metabolites. 23. Syntheses of 3-glucosides of nonamidated and glycine- and taurine-amidated bile acids. J Lipid Res 1995. [DOI: 10.1016/s0022-2275(20)39897-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|