1
|
Yang Y, Zhang W, Wang F, Li D, Meng X, Sun X, Xu J. Construction of biocatalysts based on P450BM3 for the degradation of non-steroidal anti-inflammatory drugs. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136097. [PMID: 39405679 DOI: 10.1016/j.jhazmat.2024.136097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/23/2024] [Accepted: 10/05/2024] [Indexed: 12/01/2024]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are widespread pollutants in aquatic environments, posing significant risks to both ecosystems and human health due to their persistence and bioaccumulation. Effective and sustainable degradation methods are urgently required to address this environmental challenge. This study aims to design and optimize a cytochrome P450BM3-based biocatalyst for the rapid and efficient degradation of NSAIDs by direct chemical intervention and protein engineering. The novel biocatalyst achieved efficient biodegradation of four common NSAIDs. Notably, the F87I/T268D mutant achieved 99.22 % degradation of diclofenac (DCF) within 10 min, and degraded meloxicam (MEL) and phenylbutazone (PBZ) at rates of 98.86 % and 90.51 % within 5 min, respectively. Furthermore, the F87G mutant accomplished 99.08 % degradation of acetaminophen (APAP) within just 2 min. The catalytic properties of P450BM3 and its mutants were evaluated through kinetic studies, and potential degradation pathways of the four NSAIDs were proposed in conjunction with UPLC-MS. This study provides a novel biocatalytic approach for the rapid degradation of NSAIDs in aquatic systems, offering considerable environmental benefits for pollution mitigation.
Collapse
Affiliation(s)
- Yadan Yang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Lab for Marine Drugs and Byproducts of Pilot National Lab for Marine Science and Technology, Qingdao 266071, China; College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Weikang Zhang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Lab for Marine Drugs and Byproducts of Pilot National Lab for Marine Science and Technology, Qingdao 266071, China; College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Fang Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Lab for Marine Drugs and Byproducts of Pilot National Lab for Marine Science and Technology, Qingdao 266071, China; Key Laboratory of Sustainable Development of Polar Fisheries, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
| | - Dong Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Lab for Marine Drugs and Byproducts of Pilot National Lab for Marine Science and Technology, Qingdao 266071, China
| | - Xiangmin Meng
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiaochun Sun
- Marine Science Research Institute of Shandong Province (National Oceanographic Center, Qingdao), Qingdao 266104, China
| | - Jiakun Xu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Lab for Marine Drugs and Byproducts of Pilot National Lab for Marine Science and Technology, Qingdao 266071, China; Key Laboratory of Sustainable Development of Polar Fisheries, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China.
| |
Collapse
|
2
|
Bhosale S, Kandalkar S, Gilormini PA, Akintola O, Rowland R, Adabala PJP, King D, Deen MC, Chen X, Davies GJ, Vocadlo DJ, Bennet AJ. Development of Tunable Mechanism-Based Carbasugar Ligands that Stabilize Glycoside Hydrolases through the Formation of Transient Covalent Intermediates. ACS Catal 2024; 14:14769-14779. [PMID: 39386917 PMCID: PMC11459473 DOI: 10.1021/acscatal.4c04549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024]
Abstract
Mutations in many members of the set of human lysosomal glycoside hydrolases cause a wide range of lysosomal storage diseases. As a result, much effort has been directed toward identifying pharmacological chaperones of these lysosomal enzymes. The majority of the candidate chaperones are active site-directed competitive iminosugar inhibitors but these have met with limited success. As a first step toward an alternative class of pharmacological chaperones we explored the potential of small molecule mechanism-based reversible covalent inhibitors to form transient enzyme-inhibitor adducts. By serial synthesis and kinetic analysis of candidate molecules, we show that rational tuning of the chemical reactivity of glucose-configured carbasugars delivers cyclohexenyl-based allylic carbasugar that react with the lysosomal enzyme β-glucocerebrosidase (GCase) to form covalent enzyme-adducts with different half-lives. X-ray structural analysis of these compounds bound noncovalently to GCase, along with the structures of the covalent adducts of compounds that reacted with the catalytic nucleophile of GCase, reveal unexpected reactivities of these compounds. Using differential scanning fluorimetry, we show that formation of a transient covalent intermediate stabilizes the folded enzyme against thermal denaturation. In addition, these covalent adducts break down to liberate the active enzyme and a product that is no longer inhibitory. We further show that the one compound, which reacts through an unprecedented SN1'-like mechanism, exhibits exceptional reactivity-illustrated by this compound also covalently labeling an α-glucosidase. We anticipate that such carbasugar-based single turnover covalent ligands may serve as pharmacological chaperones for lysosomal glycoside hydrolases and other disease-associated retaining glycosidases. The unusual reactivity of these molecules should also open the door to creation of new chemical biology probes to explore the biology of this important superfamily of glycoside hydrolases.
Collapse
Affiliation(s)
- Sandeep Bhosale
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Sachin Kandalkar
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Pierre-André Gilormini
- Department
of Molecular Biology and Biochemistry, Simon
Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Oluwafemi Akintola
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Rhianna Rowland
- Department
of Chemistry, University of York, York YO10 5DD, U.K.
| | - Pal John Pal Adabala
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Dustin King
- Department
of Molecular Biology and Biochemistry, Simon
Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Matthew C. Deen
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Xi Chen
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Gideon J. Davies
- Department
of Chemistry, University of York, York YO10 5DD, U.K.
| | - David J. Vocadlo
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department
of Molecular Biology and Biochemistry, Simon
Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Andrew J. Bennet
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
3
|
Schulze MSED, Scholz D, Jnoff E, Hall A, Melin J, Sands ZA, Rodriguez E, Andre VM. Identification of ß-Glucocerebrosidase Activators for Glucosylceramide hydrolysis. ChemMedChem 2024; 19:e202300548. [PMID: 38381042 DOI: 10.1002/cmdc.202300548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/31/2024] [Indexed: 02/22/2024]
Abstract
Several novel chemical series were identified that modulate glucocerebrosidase (GCase). Compounds from these series are active on glucosylceramide, unlike other known GCase modulators. We obtained GCase crystal structures with two compounds that have distinct chemotypes. Positive allosteric modulators bind to a site on GCase and induce conformational changes, but also induce an equilibrium state between monomer and dimer.
Collapse
Affiliation(s)
| | - Diana Scholz
- UCB, Avenue de l'Industrie, Braine l'Alleud, 1420, Belgium
| | - Eric Jnoff
- UCB, Avenue de l'Industrie, Braine l'Alleud, 1420, Belgium
| | - Adrian Hall
- UCB, Avenue de l'Industrie, Braine l'Alleud, 1420, Belgium
| | - Jonathan Melin
- Present address: Grünenthal GmbH, 52099, Aachen, Germany
| | - Zara A Sands
- Present address: Eli Lilly, San Diego, CA92121, USA
| | | | | |
Collapse
|
4
|
Blumenreich S, Nehushtan T, Kupervaser M, Shalit T, Gabashvili A, Joseph T, Milenkovic I, Hardy J, Futerman AH. Large-scale proteomics analysis of five brain regions from Parkinson's disease patients with a GBA1 mutation. NPJ Parkinsons Dis 2024; 10:33. [PMID: 38331996 PMCID: PMC10853186 DOI: 10.1038/s41531-024-00645-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
Despite being the second most common neurodegenerative disorder, little is known about Parkinson's disease (PD) pathogenesis. A number of genetic factors predispose towards PD, among them mutations in GBA1, which encodes the lysosomal enzyme acid-β-glucosidase. We now perform non-targeted, mass spectrometry based quantitative proteomics on five brain regions from PD patients with a GBA1 mutation (PD-GBA) and compare to age- and sex-matched idiopathic PD patients (IPD) and controls. Two proteins were differentially-expressed in all five brain regions whereas significant differences were detected between the brain regions, with changes consistent with loss of dopaminergic signaling in the substantia nigra, and activation of a number of pathways in the cingulate gyrus, including ceramide synthesis. Mitochondrial oxidative phosphorylation was inactivated in PD samples in most brain regions and to a larger extent in PD-GBA. This study provides a comprehensive large-scale proteomics dataset for the study of PD-GBA.
Collapse
Affiliation(s)
| | | | - Meital Kupervaser
- Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Tali Shalit
- Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Alexandra Gabashvili
- Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Tammar Joseph
- Department of Biomolecular Sciences, Rehovot, 76100, Israel
| | - Ivan Milenkovic
- Department of Biomolecular Sciences, Rehovot, 76100, Israel
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - John Hardy
- Department of Neurogenerative Disease, UCL Dementia Research Institute, University College London, London, WC1N 3BG, UK
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Rehovot, 76100, Israel.
- The Joseph Meyerhof Professor of Biochemistry at the Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
5
|
Tax G, Guay KP, Pantalone L, Ceci M, Soldà T, Hitchman CJ, Hill JC, Vasiljević S, Lia A, Modenutti CP, Straatman KR, Santino A, Molinari M, Zitzmann N, Hebert DN, Roversi P, Trerotola M. Rescue of secretion of rare-disease-associated misfolded mutant glycoproteins in UGGT1 knock-out mammalian cells. Traffic 2024; 25:e12927. [PMID: 38272446 PMCID: PMC10832616 DOI: 10.1111/tra.12927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/02/2023] [Accepted: 12/05/2023] [Indexed: 01/27/2024]
Abstract
Endoplasmic reticulum (ER) retention of misfolded glycoproteins is mediated by the ER-localized eukaryotic glycoprotein secretion checkpoint, UDP-glucose glycoprotein glucosyl-transferase (UGGT). The enzyme recognizes a misfolded glycoprotein and flags it for ER retention by re-glucosylating one of its N-linked glycans. In the background of a congenital mutation in a secreted glycoprotein gene, UGGT-mediated ER retention can cause rare disease, even if the mutant glycoprotein retains activity ("responsive mutant"). Using confocal laser scanning microscopy, we investigated here the subcellular localization of the human Trop-2-Q118E, E227K and L186P mutants, which cause gelatinous drop-like corneal dystrophy (GDLD). Compared with the wild-type Trop-2, which is correctly localized at the plasma membrane, these Trop-2 mutants are retained in the ER. We studied fluorescent chimeras of the Trop-2 Q118E, E227K and L186P mutants in mammalian cells harboring CRISPR/Cas9-mediated inhibition of the UGGT1 and/or UGGT2 genes. The membrane localization of the Trop-2 Q118E, E227K and L186P mutants was successfully rescued in UGGT1-/- cells. UGGT1 also efficiently reglucosylated Trop-2-Q118E-EYFP in cellula. The study supports the hypothesis that UGGT1 modulation would constitute a novel therapeutic strategy for the treatment of pathological conditions associated to misfolded membrane glycoproteins (whenever the mutation impairs but does not abrogate function), and it encourages the testing of modulators of ER glycoprotein folding quality control as broad-spectrum rescue-of-secretion drugs in rare diseases caused by responsive secreted glycoprotein mutants.
Collapse
Affiliation(s)
- Gabor Tax
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Kevin P. Guay
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Ludovica Pantalone
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Italy
| | - Martina Ceci
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Italy
| | - Tatiana Soldà
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, UniversitàdellaSvizzeraItaliana (USI), Bellinzona, Switzerland
| | - Charlie J. Hitchman
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Johan C. Hill
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Snežana Vasiljević
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Andrea Lia
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Carlos P. Modenutti
- Departamento de QuímicaBiológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de QuímicaBiológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| | - Kees R. Straatman
- Core Biotechnology Services, University of Leicester, University Road, Leicester LE1 7RH, England, United Kingdom
| | - Angelo Santino
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Maurizio Molinari
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicole Zitzmann
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Daniel N. Hebert
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Pietro Roversi
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of AgriculturalBiology and Biotecnology, IBBA-CNR Unit of Milano, via Bassini 15, I-20133 Milano, Italy
| | - Marco Trerotola
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Italy
| |
Collapse
|
6
|
Tax G, Guay KP, Soldà T, Hitchman CJ, Hill JC, Vasiljević S, Lia A, Modenutti CP, Straatman KR, Santino A, Molinari M, Zitzmann N, Hebert DN, Roversi P, Trerotola M. Rescue of secretion of a rare-disease associated mis-folded mutant glycoprotein in UGGT1 knock-out mammalian cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542711. [PMID: 37398215 PMCID: PMC10312515 DOI: 10.1101/2023.05.30.542711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Endoplasmic reticulum (ER) retention of mis-folded glycoproteins is mediated by the ERlocalised eukaryotic glycoprotein secretion checkpoint, UDP-glucose glycoprotein glucosyl-transferase (UGGT). The enzyme recognises a mis-folded glycoprotein and flags it for ER retention by reglucosylating one of its N-linked glycans. In the background of a congenital mutation in a secreted glycoprotein gene, UGGT-mediated ER retention can cause rare disease even if the mutant glycoprotein retains activity ("responsive mutant"). Here, we investigated the subcellular localisation of the human Trop-2 Q118E variant, which causes gelatinous droplike corneal dystrophy (GDLD). Compared with the wild type Trop-2, which is correctly localised at the plasma membrane, the Trop-2-Q118E variant is found to be heavily retained in the ER. Using Trop-2-Q118E, we tested UGGT modulation as a rescue-of-secretion therapeutic strategy for congenital rare disease caused by responsive mutations in genes encoding secreted glycoproteins. We investigated secretion of a EYFP-fusion of Trop-2-Q118E by confocal laser scanning microscopy. As a limiting case of UGGT inhibition, mammalian cells harbouring CRISPR/Cas9-mediated inhibition of the UGGT1 and/or UGGT2 gene expressions were used. The membrane localisation of the Trop-2-Q118E-EYFP mutant was successfully rescued in UGGT1-/- and UGGT1/2-/- cells. UGGT1 also efficiently reglucosylated Trop-2-Q118E-EYFP in cellula. The study supports the hypothesis that UGGT1 modulation constitutes a novel therapeutic strategy for the treatment of Trop-2-Q118E associated GDLD, and it encourages the testing of modulators of ER glycoprotein folding Quality Control (ERQC) as broad-spectrum rescueof-secretion drugs in rare diseases caused by responsive secreted glycoprotein mutants.
Collapse
Affiliation(s)
- Gábor Tax
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Kevin P. Guay
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Tatiana Soldà
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Charlie J. Hitchman
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Johan C. Hill
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Snežana Vasiljević
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Andrea Lia
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Carlos P. Modenutti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| | - Kees R. Straatman
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Core Biotechnology Services, University of Leicester, University Road, Leicester LE1 7RH, England, United Kingdom
| | - Angelo Santino
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Maurizio Molinari
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicole Zitzmann
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Daniel N. Hebert
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Pietro Roversi
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of Agricultural Biology and Biotecnology, IBBACNR Unit of Milano, via Bassini 15, I-20133 Milano, Italy
| | - Marco Trerotola
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, Italy
| |
Collapse
|
7
|
Blumenreich S, Nehushtan T, Barav OB, Saville JT, Dingjan T, Hardy J, Fuller M, Futerman AH. Elevation of gangliosides in four brain regions from Parkinson's disease patients with a GBA mutation. NPJ Parkinsons Dis 2022; 8:99. [PMID: 35933559 PMCID: PMC9357011 DOI: 10.1038/s41531-022-00363-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
A number of genetic risk factors have been identified over the past decade for Parkinson's Disease (PD), with variants in GBA prominent among them. GBA encodes the lysosomal enzyme that degrades the glycosphingolipid, glucosylceramide (GlcCer), with the activity of this enzyme defective in Gaucher disease. Based on the ill-defined relationship between glycosphingolipid metabolism and PD, we now analyze levels of various lipids by liquid chromatography/electrospray ionization-tandem mass spectrometry in four brain regions from age- and sex-matched patient samples, including idiopathic PD, PD patients with a GBA mutation and compare both to control brains (n = 21 for each group) obtained from individuals who died from a cause unrelated to PD. Of all the glycerolipids, sterols, and (glyco)sphingolipids (251 lipids in total), the only lipid class which showed significant differences were the gangliosides (sialic acid-containing complex glycosphingolipids), which were elevated in 3 of the 4 PD-GBA brain regions. There was no clear correlation between levels of individual gangliosides and the genetic variant in Gaucher disease [9 samples of severe (neuronopathic), 4 samples of mild (non-neuronopathic) GBA variants, and 8 samples with low pathogenicity variants which have a higher risk for development of PD]. Most brain regions, i.e. occipital cortex, cingulate gyrus, and striatum, did not show a statistically significant elevation of GlcCer in PD-GBA. Only one region, the middle temporal gyrus, showed a small, but significant elevation in GlcCer concentration in PD-GBA. We conclude that changes in ganglioside, but not in GlcCer levels, may contribute to the association between PD and GBA mutations.
Collapse
Affiliation(s)
- Shani Blumenreich
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Tamar Nehushtan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Or B Barav
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Jennifer T Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital and Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tamir Dingjan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Dementia Research Institute, University College London, London, WC1N 3BG, UK
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital and Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel.
- The Joseph Meyerhof Professor of Biochemistry at the Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
8
|
GCase Enhancers: A Potential Therapeutic Option for Gaucher Disease and Other Neurological Disorders. Pharmaceuticals (Basel) 2022; 15:ph15070823. [PMID: 35890122 PMCID: PMC9325019 DOI: 10.3390/ph15070823] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 12/07/2022] Open
Abstract
Pharmaceutical chaperones (PCs) are small compounds able to bind and stabilize misfolded proteins, allowing them to recover their native folding and thus their biological activity. In particular, lysosomal storage disorders (LSDs), a class of metabolic disorders due to genetic mutations that result in misfolded lysosomal enzymes, can strongly benefit from the use of PCs able to facilitate their translocation to the lysosomes. This results in a recovery of their catalytic activity. No PC for the GCase enzyme (lysosomal acid-β-glucosidase, or glucocerebrosidase) has reached the market yet, despite the importance of this enzyme not only for Gaucher disease, the most common LSD, but also for neurological disorders, such as Parkinson’s disease. This review aims to describe the efforts made by the scientific community in the last 7 years (since 2015) in order to identify new PCs for the GCase enzyme, which have been mainly identified among glycomimetic-based compounds.
Collapse
|
9
|
Mechanistic Insight into the Mode of Action of Acid β-Glucosidase Enhancer Ambroxol. Int J Mol Sci 2022; 23:ijms23073536. [PMID: 35408914 PMCID: PMC8998264 DOI: 10.3390/ijms23073536] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 01/27/2023] Open
Abstract
Ambroxol (ABX) is a mucolytic agent used for the treatment of respiratory diseases. Bioactivity has been demonstrated as an enhancement effect on lysosomal acid β-glucosidase (β-Glu) activity in Gaucher disease (GD). The positive effects observed have been attributed to a mechanism of action similar to pharmacological chaperones (PCs), but an exact mechanistic description is still pending. The current study uses cell culture and in vitro assays to study the effects of ABX on β-Glu activity, processing, and stability upon ligand binding. Structural analogues bromohexine, 4-hydroxybromohexine, and norbromohexine were screened for chaperone efficacy, and in silico docking was performed. The sugar mimetic isofagomine (IFG) strongly inhibits β-Glu, while ABX exerts its inhibitory effect in the micromolar range. In GD patient fibroblasts, IFG and ABX increase mutant β-Glu activity to identical levels. However, the characteristics of the banding patterns of Endoglycosidase-H (Endo-H)-digested enzyme and a substantially lower half-life of ABX-treated β-Glu suggest different intracellular processing. In line with this observation, IFG efficiently stabilizes recombinant β-Glu against thermal denaturation in vitro, whereas ABX exerts no significant effect. Additional β-Glu enzyme activity testing using Bromohexine (BHX) and two related structures unexpectedly revealed that ABX alone can refunctionalize β-Glu in cellula. Taken together, our data indicate that ABX has little in vitro ability to act as PC, so the mode of action requires further clarification.
Collapse
|
10
|
Rowland RJ, Chen Y, Breen I, Wu L, Offen WA, Beenakker TJ, Su Q, van den Nieuwendijk AMCH, Aerts JMFG, Artola M, Overkleeft HS, Davies GJ. Design, Synthesis and Structural Analysis of Glucocerebrosidase Imaging Agents. Chemistry 2021; 27:16377-16388. [PMID: 34570911 PMCID: PMC9298352 DOI: 10.1002/chem.202102359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 12/15/2022]
Abstract
Gaucher disease (GD) is a lysosomal storage disorder caused by inherited deficiencies in β‐glucocerebrosidase (GBA). Current treatments require rapid disease diagnosis and a means of monitoring therapeutic efficacy, both of which may be supported by the use of GBA‐targeting activity‐based probes (ABPs). Here, we report the synthesis and structural analysis of a range of cyclophellitol epoxide and aziridine inhibitors and ABPs for GBA. We demonstrate their covalent mechanism‐based mode of action and uncover binding of the new N‐functionalised aziridines to the ligand binding cleft. These inhibitors became scaffolds for the development of ABPs; the O6‐fluorescent tags of which bind in an allosteric site at the dimer interface. Considering GBA's preference for O6‐ and N‐functionalised reagents, a bi‐functional aziridine ABP was synthesized as a potentially more powerful imaging agent. Whilst this ABP binds to two unique active site clefts of GBA, no further benefit in potency was achieved over our first generation ABPs. Nevertheless, such ABPs should serve useful in the study of GBA in relation to GD and inform the design of future probes.
Collapse
Affiliation(s)
- Rhianna J Rowland
- Department of Chemistry, York Structural Biology Laboratory (YSBL), University of York Heslington, York, YO10 5DD, UK
| | - Yurong Chen
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinwegg 55, 2300 RA, Leiden, Netherlands
| | - Imogen Breen
- Department of Chemistry, York Structural Biology Laboratory (YSBL), University of York Heslington, York, YO10 5DD, UK
| | - Liang Wu
- Department of Chemistry, York Structural Biology Laboratory (YSBL), University of York Heslington, York, YO10 5DD, UK
| | - Wendy A Offen
- Department of Chemistry, York Structural Biology Laboratory (YSBL), University of York Heslington, York, YO10 5DD, UK
| | - Thomas J Beenakker
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinwegg 55, 2300 RA, Leiden, Netherlands
| | - Qin Su
- Department of Medicinal Biochemistry, Leiden Institute of Chemistry, Leiden University, Einsteinwegg 55, 2300 RA, Leiden, Netherlands
| | | | - Johannes M F G Aerts
- Department of Medicinal Biochemistry, Leiden Institute of Chemistry, Leiden University, Einsteinwegg 55, 2300 RA, Leiden, Netherlands
| | - Marta Artola
- Department of Medicinal Biochemistry, Leiden Institute of Chemistry, Leiden University, Einsteinwegg 55, 2300 RA, Leiden, Netherlands
| | - Herman S Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinwegg 55, 2300 RA, Leiden, Netherlands
| | - Gideon J Davies
- Department of Chemistry, York Structural Biology Laboratory (YSBL), University of York Heslington, York, YO10 5DD, UK
| |
Collapse
|
11
|
van Smeden J, Al-Khakany H, Wang Y, Visscher D, Stephens N, Absalah S, Overkleeft HS, Aerts JMFG, Hovnanian A, Bouwstra JA. Skin barrier lipid enzyme activity in Netherton patients is associated with protease activity and ceramide abnormalities. J Lipid Res 2020; 61:859-869. [PMID: 32265319 PMCID: PMC7269766 DOI: 10.1194/jlr.ra120000639] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/31/2020] [Indexed: 12/17/2022] Open
Abstract
Individuals with Netherton syndrome (NTS) have increased serine protease activity, which strongly impacts the barrier function of the skin epidermis and leads to skin inflammation. Here, we investigated how serine protease activity in NTS correlates with changes in the stratum corneum (SC) ceramides, which are crucial components of the skin barrier. We examined two key enzymes involved in epidermal ceramide biosynthesis, β-glucocerebrosidase (GBA) and acid-sphingomyelinase (ASM). We compared in situ expression levels and activities of GBA and ASM between NTS patients and controls and correlated the expression and activities with i) SC ceramide profiles, ii) in situ serine protease activity, and iii) clinical presentation of patients. Using activity-based probe labeling, we visualized and localized active epidermal GBA, and a newly developed in situ zymography method enabled us to visualize and localize active ASM. Reduction in active GBA in NTS patients coincided with increased ASM activity, particularly in areas with increased serine protease activity. NTS patients with scaly erythroderma exhibited more pronounced anomalies in GBA and ASM activities than patients with ichthyosis linearis circumflexa. They also displayed a stronger increase in SC ceramides processed via ASM. We conclude that changes in the localization of active GBA and ASM correlate with i) altered SC ceramide composition in NTS patients, ii) local serine protease activity, and iii) the clinical manifestation of NTS.
Collapse
Affiliation(s)
- Jeroen van Smeden
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Centre for Human Drug Research, Leiden, The Netherlands
| | - Hanin Al-Khakany
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Yichen Wang
- INSERM UMR1163, Imagine Institute, Paris Descartes University, Paris, France
| | - Dani Visscher
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Nicole Stephens
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Samira Absalah
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Herman S Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Johannes M F G Aerts
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Alain Hovnanian
- INSERM UMR1163, Imagine Institute, Paris Descartes University, Paris, France; Department of Genetics Necker-Enfants Malades Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
| | - Joke A Bouwstra
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands. mailto:
| |
Collapse
|
12
|
Rowland RJ, Wu L, Liu F, Davies GJ. A baculoviral system for the production of human β-glucocerebrosidase enables atomic resolution analysis. Acta Crystallogr D Struct Biol 2020; 76:565-580. [PMID: 32496218 PMCID: PMC7271948 DOI: 10.1107/s205979832000501x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/09/2020] [Indexed: 11/18/2022] Open
Abstract
The lysosomal glycoside hydrolase β-glucocerebrosidase (GBA; sometimes called GBA1 or GCase) catalyses the hydrolysis of glycosphingolipids. Inherited deficiencies in GBA cause the lysosomal storage disorder Gaucher disease (GD). Consequently, GBA is of considerable medical interest, with continuous advances in the development of inhibitors, chaperones and activity-based probes. The development of new GBA inhibitors requires a source of active protein; however, the majority of structural and mechanistic studies of GBA today rely on clinical enzyme-replacement therapy (ERT) formulations, which are incredibly costly and are often difficult to obtain in adequate supply. Here, the production of active crystallizable GBA in insect cells using a baculovirus expression system is reported, providing a nonclinical source of recombinant GBA with comparable activity and biophysical properties to ERT preparations. Furthermore, a novel crystal form of GBA is described which diffracts to give a 0.98 Å resolution unliganded structure. A structure in complex with the inactivator 2,4-dinitrophenyl-2-deoxy-2-fluoro-β-D-glucopyranoside was also obtained, demonstrating the ability of this GBA formulation to be used in ligand-binding studies. In light of its purity, stability and activity, the GBA production protocol described here should circumvent the need for ERT formulations for structural and biochemical studies and serve to support GD research.
Collapse
Affiliation(s)
- Rhianna J. Rowland
- Department of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Liang Wu
- Department of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Feng Liu
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Gideon J. Davies
- Department of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10 5DD, United Kingdom
| |
Collapse
|
13
|
Glucocerebrosidase: Functions in and Beyond the Lysosome. J Clin Med 2020; 9:jcm9030736. [PMID: 32182893 PMCID: PMC7141376 DOI: 10.3390/jcm9030736] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023] Open
Abstract
Glucocerebrosidase (GCase) is a retaining β-glucosidase with acid pH optimum metabolizing the glycosphingolipid glucosylceramide (GlcCer) to ceramide and glucose. Inherited deficiency of GCase causes the lysosomal storage disorder named Gaucher disease (GD). In GCase-deficient GD patients the accumulation of GlcCer in lysosomes of tissue macrophages is prominent. Based on the above, the key function of GCase as lysosomal hydrolase is well recognized, however it has become apparent that GCase fulfills in the human body at least one other key function beyond lysosomes. Crucially, GCase generates ceramides from GlcCer molecules in the outer part of the skin, a process essential for optimal skin barrier property and survival. This review covers the functions of GCase in and beyond lysosomes and also pays attention to the increasing insight in hitherto unexpected catalytic versatility of the enzyme.
Collapse
|
14
|
Alméciga-Diaz CJ, Hidalgo OA, Olarte-Avellaneda S, Rodríguez-López A, Guzman E, Garzón R, Pimentel-Vera LN, Puentes-Tellez MA, Rojas-Rodriguez AF, Gorshkov K, Li R, Zheng W. Identification of Ezetimibe and Pranlukast as Pharmacological Chaperones for the Treatment of the Rare Disease Mucopolysaccharidosis Type IVA. J Med Chem 2019; 62:6175-6189. [PMID: 31188588 PMCID: PMC11292729 DOI: 10.1021/acs.jmedchem.9b00428] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mucopolysaccharidosis type IVA (MPS IVA) is a rare disease caused by mutations in the gene encoding the lysosomal enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS). We report here two GALNS pharmacological chaperones, ezetimibe and pranlukast, identified by molecular docking-based virtual screening. These compounds bound to the active cavity of GALNS and increased its thermal stability as well as the production of recombinant GALNS in bacteria, yeast, and HEK293 cells. MPS IVA fibroblasts treated with these chaperones exhibited increases in GALNS protein and enzyme activity and reduced the size of enlarged lysosomes. Abnormalities in autophagy markers p62 and LC3B-II were alleviated by ezetimibe and pranlukast. Combined treatment of recombinant GALNS with ezetimibe or pranlukast produced an additive effect. Altogether, the results demonstrate that ezetimibe and pranlukast can increase the yield of recombinant GALNS and be used as a monotherapy or combination therapy to improve the therapeutic efficacy of MPS IVA enzyme replacement therapy.
Collapse
Affiliation(s)
- Carlos J. Alméciga-Diaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - Oscar A. Hidalgo
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - Sergio Olarte-Avellaneda
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
- Pharmacy Department, Faculty of Science, Universidad Nacional de Colombia, Bogotá D.C., 111321, Colombia
| | - Alexander Rodríguez-López
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
- Chemistry Department, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - Esteban Guzman
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - Rafael Garzón
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - Luisa Natalia Pimentel-Vera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - María Alejandra Puentes-Tellez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - Andrés Felipe Rojas-Rodriguez
- Computational and Structural Biochemistry, Departamento de Nutrición y Bioquímica, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., 110231, Colombia
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rong Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
15
|
Zhou X, Paushter DH, Pagan MD, Kim D, Nunez Santos M, Lieberman RL, Overkleeft HS, Sun Y, Smolka MB, Hu F. Progranulin deficiency leads to reduced glucocerebrosidase activity. PLoS One 2019; 14:e0212382. [PMID: 31291241 PMCID: PMC6619604 DOI: 10.1371/journal.pone.0212382] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022] Open
Abstract
Mutation in the GRN gene, encoding the progranulin (PGRN) protein, shows a dose-dependent disease correlation, wherein haploinsufficiency results in frontotemporal lobar degeneration (FTLD) and complete loss results in neuronal ceroid lipofuscinosis (NCL). Although the exact function of PGRN is unknown, it has been increasingly implicated in lysosomal physiology. Here we report that PGRN interacts with the lysosomal enzyme, glucocerebrosidase (GCase), and is essential for proper GCase activity. GCase activity is significantly reduced in tissue lysates from PGRN-deficient mice. This is further evidence that reduced lysosomal hydrolase activity may be a pathological mechanism in cases of GRN-related FTLD and NCL.
Collapse
Affiliation(s)
- Xiaolai Zhou
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States of America
| | - Daniel H. Paushter
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States of America
| | - Mitchell D. Pagan
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States of America
| | - Dongsung Kim
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States of America
| | - Mariela Nunez Santos
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States of America
| | - Raquel L. Lieberman
- School of Chemistry and Biochemistry, Georgia Institute of Technology, NW, Atlanta, GA, United States of America
| | - Herman S. Overkleeft
- Leiden Institute of Chemistry, Leiden University, Gorlaeus Laboratories, RA Leiden, Netherlands
| | - Ying Sun
- Division of Human Genetics; Cincinnati Children's Hospital Medical Center and the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Marcus B. Smolka
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States of America
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States of America
- * E-mail:
| |
Collapse
|
16
|
Validation of anti-glucocerebrosidase antibodies for western blot analysis on protein lysates of murine and human cells. Biochem J 2019; 476:261-274. [DOI: 10.1042/bcj20180708] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/01/2023]
Abstract
Abstract
Gaucher disease (GD) is a rare lysosomal storage disorder caused by mutations in the GBA1 gene, encoding the lysosome-resident glucocerebrosidase enzyme involved in the hydrolysis of glucosylceramide. The discovery of an association between mutations in GBA1 and the development of synucleinopathies, including Parkinson disease, has directed attention to glucocerebrosidase as a potential therapeutic target for different synucleinopathies. These findings initiated an exponential growth in research and publications regarding the glucocerebrosidase enzyme. The use of various commercial and custom-made glucocerebrosidase antibodies has been reported, but standardized in-depth validation is still not available for many of these antibodies. This work details the evaluation of several previously reported glucocerebrosidase antibodies for western blot analysis, tested on protein lysates of murine gba+/+ and gba−/− immortalized neurons and primary human wild-type and type 2 GD fibroblasts.
Collapse
|
17
|
Ben Bdira F, Artola M, Overkleeft HS, Ubbink M, Aerts JMFG. Distinguishing the differences in β-glycosylceramidase folds, dynamics, and actions informs therapeutic uses. J Lipid Res 2018; 59:2262-2276. [PMID: 30279220 PMCID: PMC6277158 DOI: 10.1194/jlr.r086629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Glycosyl hydrolases (GHs) are carbohydrate-active enzymes that hydrolyze a specific β-glycosidic bond in glycoconjugate substrates; β-glucosidases degrade glucosylceramide, a ubiquitous glycosphingolipid. GHs are grouped into structurally similar families that themselves can be grouped into clans. GH1, GH5, and GH30 glycosidases belong to clan A hydrolases with a catalytic (β/α)8 TIM barrel domain, whereas GH116 belongs to clan O with a catalytic (α/α)6 domain. In humans, GH abnormalities underlie metabolic diseases. The lysosomal enzyme glucocerebrosidase (family GH30), deficient in Gaucher disease and implicated in Parkinson disease etiology, and the cytosol-facing membrane-bound glucosylceramidase (family GH116) remove the terminal glucose from the ceramide lipid moiety. Here, we compare enzyme differences in fold, action, dynamics, and catalytic domain stabilization by binding site occupancy. We also explore other glycosidases with reported glycosylceramidase activity, including human cytosolic β-glucosidase, intestinal lactase-phlorizin hydrolase, and lysosomal galactosylceramidase. Last, we describe the successful translation of research to practice: recombinant glycosidases and glucosylceramide metabolism modulators are approved drug products (enzyme replacement therapies). Activity-based probes now facilitate the diagnosis of enzyme deficiency and screening for compounds that interact with the catalytic pocket of glycosidases. Future research may deepen the understanding of the functional variety of these enzymes and their therapeutic potential.
Collapse
Affiliation(s)
- Fredj Ben Bdira
- Departments of Macromolecular Biochemistry,Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Marta Artola
- Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Herman S Overkleeft
- Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Marcellus Ubbink
- Departments of Macromolecular Biochemistry,Leiden Institute of Chemistry, Leiden, The Netherlands
| | | |
Collapse
|
18
|
Click chemistry in sphingolipid research. Chem Phys Lipids 2018; 215:71-83. [DOI: 10.1016/j.chemphyslip.2018.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 01/17/2023]
|
19
|
Kuo CL, van Meel E, Kytidou K, Kallemeijn WW, Witte M, Overkleeft HS, Artola ME, Aerts JM. Activity-Based Probes for Glycosidases: Profiling and Other Applications. Methods Enzymol 2017; 598:217-235. [PMID: 29306436 DOI: 10.1016/bs.mie.2017.06.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glycosidases mediate the fragmentation of glycoconjugates in the body, including the vital recycling of endogenous molecules. Several inherited diseases in man concern deficiencies in lysosomal glycosidases degrading glycosphingolipids. Prominent is Gaucher disease caused by an impaired lysosomal β-glucosidase (glucocerebrosidase, GBA) and resulting in pathological lysosomal storage of glucosylceramide (glucocerebroside) in tissue macrophages. GBA is a retaining glucosidase with a characteristic glycosyl-enzyme intermediate formed during catalysis. Using the natural suicide inhibitor cyclophellitol as a lead, we developed mechanism-based irreversible inhibitors of GBA equipped with a fluorescent reporter. These reagents covalently link to the catalytic nucleophile residue of GBA and permit specific and sensitive visualization of active enzyme molecules. The amphiphilic activity-based probes (ABPs) allow in situ detection of active GBA in cells and organisms. Furthermore, they may be used to biochemically confirm the diagnosis of Gaucher disease and they might assist in screening for small compounds interacting with the catalytic pocket. While the focus of this chapter is ABPs for β-glucosidases and Gaucher disease, the described concept has meanwhile been extended to other retaining glycosidases and related disease conditions as well.
Collapse
Affiliation(s)
- Chi-Lin Kuo
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Eline van Meel
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Kassiani Kytidou
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | | - Martin Witte
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | | - Marta Elena Artola
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
20
|
Gu X, Gupta V, Yang Y, Zhu JY, Carlson EJ, Kingsley C, Tash JS, Schönbrunn E, Hawkinson J, Georg GI. Structure-Activity Studies of N-Butyl-1-deoxynojirimycin (NB-DNJ) Analogues: Discovery of Potent and Selective Aminocyclopentitol Inhibitors of GBA1 and GBA2. ChemMedChem 2017; 12:1977-1984. [PMID: 28975712 PMCID: PMC5725710 DOI: 10.1002/cmdc.201700558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Indexed: 12/26/2022]
Abstract
Analogues of N‐butyl‐1‐deoxynojirimycin (NB‐DNJ) were prepared and assayed for inhibition of ceramide‐specific glucosyltransferase (CGT), non‐lysosomal β‐glucosidase 2 (GBA2) and the lysosomal β‐glucosidase 1 (GBA1). Compounds 5 a–6 f, which carry sterically demanding nitrogen substituents, and compound 13, devoid of the C3 and C5 hydroxy groups present in DNJ/NB‐DGJ (N‐butyldeoxygalactojirimycin) showed no inhibitory activity for CGT or GBA2. Inversion of stereochemistry at C4 of N‐(n‐butyl)‐ and N‐(n‐nonyl)‐DGJ (compounds 24) also led to a loss of activity in these assays. The aminocyclopentitols N‐(n‐butyl)‐ (35 a), N‐(n‐nonyl)‐4‐amino‐5‐(hydroxymethyl)cyclopentane‐ (35 b), and N‐(1‐(pentyloxy)methyl)adamantan‐1‐yl)‐1,2,3‐triol (35 f), were found to be selective inhibitors of GBA1 and GBA2 that did not inhibit CGT (>1 mm), with the exception of 35 f, which inhibited CGT with an IC50 value of 1 mm. The N‐butyl analogue 35 a was 100‐fold selective for inhibiting GBA1 over GBA2 (Ki values of 32 nm and 3.3 μm for GBA1 and GBA2, respectively). The N‐nonyl analogue 35 b displayed a Ki value of ≪14 nm for GBA1 inhibition and a Ki of 43 nm for GBA2. The N‐(1‐(pentyloxy)methyl)adamantan‐1‐yl) derivative 35 f had Ki values of ≈16 and 14 nm for GBA1 and GBA2, respectively. The related N‐bis‐substituted aminocyclopentitols were found to be significantly less potent inhibitors than their mono‐substituted analogues. The aminocyclopentitol scaffold should hold promise for further inhibitor development.
Collapse
Affiliation(s)
- Xingxian Gu
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, 66045, USA.,Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| | | | - Yan Yang
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Jin-Yi Zhu
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Erick J Carlson
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| | - Carolyn Kingsley
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| | - Joseph S Tash
- University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ernst Schönbrunn
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Jon Hawkinson
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| | - Gunda I Georg
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| |
Collapse
|
21
|
van Smeden J, Dijkhoff IM, Helder RWJ, Al-Khakany H, Boer DEC, Schreuder A, Kallemeijn WW, Absalah S, Overkleeft HS, Aerts JMFG, Bouwstra JA. In situ visualization of glucocerebrosidase in human skin tissue: zymography versus activity-based probe labeling. J Lipid Res 2017; 58:2299-2309. [PMID: 29025868 DOI: 10.1194/jlr.m079376] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/06/2017] [Indexed: 12/15/2022] Open
Abstract
Epidermal β-glucocerebrosidase (GBA1), an acid β-glucosidase normally located in lysosomes, converts (glucosyl)ceramides into ceramides, which is crucial to generate an optimal barrier function of the outermost skin layer, the stratum corneum (SC). Here we report on two developed in situ methods to localize active GBA in human epidermis: i) an optimized zymography method that is less labor intensive and visualizes enzymatic activity with higher resolution than currently reported methods using either substrate 4-methylumbelliferyl-β-D-glucopyranoside or resorufin-β-D-glucopyranoside; and ii) a novel technique to visualize active GBA1 molecules by their specific labeling with a fluorescent activity-based probe (ABP), MDW941. The latter method pro-ved to be more robust and sensitive, provided higher resolution microscopic images, and was less prone to sample preparation effects. Moreover, in contrast to the zymography substrates that react with various β-glucosidases, MDW941 specifically labeled GBA1. We demonstrate that active GBA1 in the epidermis is primarily located in the extracellular lipid matrix at the interface of the viable epidermis and the lower layers of the SC. With ABP-labeling, we observed reduced GBA1 activity in 3D-cultured skin models when supplemented with the reversible inhibitor, isofagomine, irrespective of GBA expression. This inhibition affected the SC ceramide composition: MS analysis revealed an inhibitor-dependent increase in the glucosylceramide:ceramide ratio.
Collapse
Affiliation(s)
- Jeroen van Smeden
- Division of Drug Delivery Technology, Cluster Biotherapeutics, Leiden Academic Centre for Drug Research Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Irini M Dijkhoff
- Division of Drug Delivery Technology, Cluster Biotherapeutics, Leiden Academic Centre for Drug Research Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Richard W J Helder
- Division of Drug Delivery Technology, Cluster Biotherapeutics, Leiden Academic Centre for Drug Research Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Hanin Al-Khakany
- Division of Drug Delivery Technology, Cluster Biotherapeutics, Leiden Academic Centre for Drug Research Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Daphne E C Boer
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Anne Schreuder
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Wouter W Kallemeijn
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Samira Absalah
- Division of Drug Delivery Technology, Cluster Biotherapeutics, Leiden Academic Centre for Drug Research Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Herman S Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Johannes M F G Aerts
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Joke A Bouwstra
- Division of Drug Delivery Technology, Cluster Biotherapeutics, Leiden Academic Centre for Drug Research Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| |
Collapse
|
22
|
Smith L, Mullin S, Schapira AHV. Insights into the structural biology of Gaucher disease. Exp Neurol 2017; 298:180-190. [PMID: 28923368 DOI: 10.1016/j.expneurol.2017.09.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/08/2017] [Accepted: 09/14/2017] [Indexed: 01/08/2023]
Abstract
Gaucher disease, the most common lysosomal storage disorder, is caused by mutations in the gene encoding the acid-β-glucosidase lysosomal hydrolase enzyme that cleaves glucocerebroside into glucose and ceramide. Reduced enzyme activity and impaired structural stability arise due to >300 known disease-causing mutations. Several of these mutations have also been associated with an increased risk of Parkinson disease (PD). Since the discovery of the acid-β-glucosidase X-ray structure, there have been major advances in our understanding of the structural properties of the protein. Analysis of specific residues has provided insight into their functional and structural importance and provided insight into the pathogenesis of Gaucher disease and the contribution to PD. Disease-causing mutations are positioned throughout the acid-β-glucosidase structure, with many located far from the active site and thus retaining some enzymatic activity however, thus far no clear relationship between mutation location and disease severity has been established. Here, we review the crystal structure of acid-β-glucosidase, while highlighting important structural aspects of the protein in detail. This review discusses the structural stability of acid-β-glucosidase, which can be altered by pH and glycosylation, and explores the relationship between known Gaucher disease and PD mutations, structural stability and disease severity.
Collapse
Affiliation(s)
- Laura Smith
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK
| | - Stephen Mullin
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK.
| |
Collapse
|