Development of Tunable Mechanism-Based Carbasugar Ligands that Stabilize Glycoside Hydrolases through the Formation of Transient Covalent IntermediatesClick to copy article linkArticle link copied!
- Sandeep BhosaleSandeep BhosaleDepartment of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Sandeep Bhosale
- Sachin KandalkarSachin KandalkarDepartment of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Sachin Kandalkar
- Pierre-André GilorminiPierre-André GilorminiDepartment of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Pierre-André Gilormini
- Oluwafemi AkintolaOluwafemi AkintolaDepartment of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Oluwafemi Akintola
- Rhianna RowlandRhianna RowlandDepartment of Chemistry, University of York, York YO10 5DD, U.K.More by Rhianna Rowland
- Pal John Pal AdabalaPal John Pal AdabalaDepartment of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Pal John Pal Adabala
- Dustin KingDustin KingDepartment of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Dustin King
- Matthew C. DeenMatthew C. DeenDepartment of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Matthew C. Deen
- Xi ChenXi ChenDepartment of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Xi Chen
- Gideon J. DaviesGideon J. DaviesDepartment of Chemistry, University of York, York YO10 5DD, U.K.More by Gideon J. Davies
- David J. Vocadlo*David J. Vocadlo*Email: [email protected]Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaDepartment of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by David J. Vocadlo
- Andrew J. Bennet*Andrew J. Bennet*Email: [email protected]Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, CanadaMore by Andrew J. Bennet
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.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Figure 1
Figure 1. Small molecule covalent inhibitors and the mechanism of action of retaining glycosidases. (a) Mechanism used by glucocerebrosidase for hydrolysis of its natural β-glucoside substrate. (b) Kinetic scheme for GCase-catalyzed turnover of β-d-glucopyranoside substrates. (c) Mechanism of action for carbasugar covalent inhibitors. (d) Kinetic scheme for covalent carbasugar inhibitors.
Results
Synthesis and Testing of First-Generation Allylic Carbasugar Covalent Inhibitors
Figure 2
Figure 2. Structures of carbasugars used in this study and their in vitro characterization. Carbasugar covalent inhibitors used in the current study. The pseudoanomeric carbons (shown by the asterisk on 1) are drawn in identical positions for all compounds.
Synthesis and Testing of Second-Generation Allylic Carbasugar Covalent Inhibitors
Scheme 1
aReagents: (i) NaIO4, H2O, rt; (ii) Ph3P+CH3, BuLi, 0 °C; (iii) dioxane/water, cat H+, 90 °C; (iv) CH2CHMgBr, THF, 0 °C; (v) Grubbs’ 2nd generation, CH2Cl2, heat; (vi) CH3COCOCH3, HC(OCH3)3, cat H+; (vii) DAST, diglyme, 0 °C; (viii) TFA, CH2Cl2 (for 4 and 5 from 20 and 21, respectively); (ix) Dess-Martin periodinane, CH2Cl2; (x) NaBD4 (MeOD); (xi) TFA, CH2Cl2, H2O; (xii) BCl3, CH2Cl2, −78 °C (for 6, 7, 6-D, and 7-D from 26 to 29, respectively). Abbreviations: DAST, (diethylamino)sulfur trifluoride; TFA, trifluoroacetic acid.
Figure 3
Figure 3. In vitro characterization of carbasugars used in this study. (a) Nonlinear least-squares fit of kapp values to a modified Michaelis–Menten expression (eq S3) for the approach to steady-state GCase activity on incubation with d-carbaxylosyl fluoride 4. (b) Linear fit of kapp values for the approach to steady state for covalent inhibition of GCase by d-carbaxylosyl chloride 6 (blue) and l-carbaxylosyl chloride 7 (red). (c) Stabilization of folded GCase measured using differential scanning fluorimetry (DSF) in the presence of d-carbaxylosyl chloride 6 (blue), l-carbaxylosyl chloride 7 (red), or vehicle along (black). All lines are the best nonlinear least-squares fit to the appropriate equation. All experiments were carried out at T = 25 °C in McIlvaine buffer pH 5.2 containing 0.1% Triton X-100, 0.24% sodium taurodeoxycholate, and 0.1% BSA. (d) Mechanisms for the GCase pseudoglycosylation step by single turnover chaperones 6 and 7, which after diffusion of chloride out of the active site give identical allylic cation intermediates that are trapped by the enzymatic nucleophile (Glu340) to give the same covalent intermediate (E-αCarb).
Synthesis and Testing of Third-Generation Allylic Carbasugar Covalent Inhibitors
inhibitor | kinact/Ki (M–1 s–1) | kreact (s–1) | t1/2react (min) |
---|---|---|---|
4 | 0.59 ± 0.27a | ||
5 | N.I.b | ||
6 | 6.9 ± 0.1 | (2.2 ± 0.2) × 10–4 | 52.5 ± 4.8 |
7 | 41 ± 2 | (2.1 ± 0.1) × 10–4 | 55.0 ± 2.6 |
kinact = (2.0 ± 0.3) × 10–3 s–1, Ki = 3.4 ± 1.5 mM.
N.I. no time-dependent inhibition observed at 6 mM.
Conditions: T = 25 °C in McIlvaine buffer pH 5.2 containing 0.1% Triton X-100, 0.24% sodium taurodeoxycholate, and 0.1% BSA.
X-ray Structural Analysis of Michaelis Complexes and Covalent Intermediates of GCase
Figure 4
Figure 4. Structural characterization of covalent intermediates and Michaelis complex between GCase and carbasugars. (a) Crystal structure of the GCase-6 complex, showing two orientations of the covalent complex in which 6 adopts an envelope (2E) conformation. The maximum-likelihood σA-weighted 2Fo – Fc electron density map is contoured to 1 σ (0.37 e/Å3). (b) Crystal structure of the GCase-7 complex, showing two orientations of the covalent complex in which, the carbasugar adopts an envelope (2E) conformation, numbering of the ring starts at the covalent attachment to the enzyme. The maximum-likelihood σA-weighted 2Fo – Fc electron density map is contoured to 1 σ (0.36 e/Å3). Carbasugar average b-factor = 20 Å2. (c) Crystal structure of the GCase-5 complex, showing three orientations of l-carbasugar fluoride bound noncovalently in the active site in a half-chair (4H3) conformation. Green atom indicates fluorine. The maximum-likelihood σA-weighted 2Fo – Fc electron density map is contoured to 1 σ (0.37 e/Å3).
Reactivity of Carbaxylosyl Chlorides with Other Glycoside Hydrolases
inhibitor | kinact/Ki (M–1 s–1) | kreact (s–1) | t1/2react (h) |
---|---|---|---|
7 | 7.70 ± 0.07 | (2.35 ± 0.17) × 10–6 | 81.8 ± 5.9 |
Conditions: experiments were carried out at T = 37 °C in 50 mM sodium phosphate buffer, pH 7.0 containing 0.1% BSA.
Figure 5
Figure 5. pH-rate profile for the logarithm of the inactivation second-order rate constants (kinact/Ki) for yeast α-glucosidase by 7. All experiments were performed at T = 37 °C in 50 mM buffer (acetic acid-sodium acetate buffer for pH 5.0–5.5, sodium phosphate buffer for pH 6.0–7.5, TAPS buffer for pH 7.9) containing 0.1% BSA. The line through the points is the best fit to a model where two protonation events (low pH) lead to an inactive enzyme, while a single deprotonation at higher pH values gives inactive enzyme.
Discussion
(i) | Both experimental (38−40) and theoretical (41) studies indicate that an SN2 displacement has a significantly lower TS free energy than the corresponding SN2′ counterpart. As a result, if covalent labeling involved SN2 and SN2′ mechanisms inhibitor 6 should covalently label GCase more rapidly than does 7, a situation that is contrary to our observations | ||||
(ii) | Contrary to the commonly held view, fluoride is a viable leaving group in bona fide SN1 reactions performed in polar H-bonding environments. For instance, the difference in SN1 reactivity of 1-adamantyl chloride (1-AdCl) (42) and 1-adamantyl fluoride (1-AdF) (43) in trifluoroethanol is approximately 760-fold at 25 °C. Moreover, this ratio is almost independent of temperature as the major difference in the corresponding activation parameters is a larger negative entropy (ΔS⧧) for the solvolysis of 1-AdF, likely caused by the obligatory strong H-bonding that is needed to assist fluoride ion departure for formation of the 1-adamantyl carbocation (44,45) | ||||
(iii) | The ratio of the second order rate constants (kinact/Ki) governing the GCase-catalyzed reactions of 6 and 4 is 12 ± 5, a ratio that is consistent with SN1 reactions in which the presence of an optimally positioned H-bonding donor only assists departure of fluoride to give enzyme-bound allylic cations, (34) and | ||||
(iv) | l-Carbaxylosyl chloride 7 undergoes pseudoglycosylation faster than does the d-enantiomer 6 because 7 binds in the conformation of the transition state for GCase-catalyzed cleavage of natural substrate glucosidic bonds (4H3), a conformation that enables the most efficient stabilization of positive charge development at the allylic cation-like transition state (Figure 3d). |
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscatal.4c04549.
General methods and reagents, eqs S1–S3, Tables S1–S3, Scheme S1, Figures S1–S78 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
The authors are grateful for support from the Canadian Institutes of Health Research (A.J.B.: grant-173228) and the Natural Sciences and Engineering Council of Canada (A.J.B.: RGPIN-04910 and CRDPJ-480335, and D.J.V.: RGPIN-05426). We thank Diamond Light Source for access to beamline I03 and I04 (proposal numbers mx18598 and mx24948). G.J.D. thanks the Royal Society for the Ken Murray Research Professorship and RJR thanks the BBSRC for White Rose Doctoral Training Partnership funding (BB/M011151/1). D.J.V. thanks the Canada Research Chairs program for support as a Tier I Canada Research Chair in Chemical Biology.
References
This article references 46 other publications.
- 1Platt, F. M.; d’Azzo, A.; Davidson, B. L.; Neufeld, E. F.; Tifft, C. J. Lysosomal storage diseases. Nat. Rev. Dis. Primers 2018, 4, 27, DOI: 10.1038/s41572-018-0025-4Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3czksl2ntg%253D%253D&md5=9a52212427e380c3dc78006560c800ceLysosomal storage diseasesPlatt Frances M; d'Azzo Alessandra; Davidson Beverly L; Davidson Beverly L; Neufeld Elizabeth F; Tifft Cynthia JNature reviews. Disease primers (2018), 4 (1), 27 ISSN:.Lysosomal storage diseases (LSDs) are a group of over 70 diseases that are characterized by lysosomal dysfunction, most of which are inherited as autosomal recessive traits. These disorders are individually rare but collectively affect 1 in 5,000 live births. LSDs typically present in infancy and childhood, although adult-onset forms also occur. Most LSDs have a progressive neurodegenerative clinical course, although symptoms in other organ systems are frequent. LSD-associated genes encode different lysosomal proteins, including lysosomal enzymes and lysosomal membrane proteins. The lysosome is the key cellular hub for macromolecule catabolism, recycling and signalling, and defects that impair any of these functions cause the accumulation of undigested or partially digested macromolecules in lysosomes (that is, 'storage') or impair the transport of molecules, which can result in cellular damage. Consequently, the cellular pathogenesis of these diseases is complex and is currently incompletely understood. Several LSDs can be treated with approved, disease-specific therapies that are mostly based on enzyme replacement. However, small-molecule therapies, including substrate reduction and chaperone therapies, have also been developed and are approved for some LSDs, whereas gene therapy and genome editing are at advanced preclinical stages and, for a few disorders, have already progressed to the clinic.
- 2Varki, A. Essentials of Glycobiology, 4th ed.; Cold Spring Harbor Laboratory Press: Cold Spring Harbor: Cold Spring Harbor, NY, 2022.Google ScholarThere is no corresponding record for this reference.
- 3Selnick, H. G.; Hess, J. F.; Tang, C. Y.; Liu, K.; Schachter, J. B.; Ballard, J. E.; Marcus, J.; Klein, D. J.; Wang, X. H.; Pearson, M.; Savage, M. J.; Kaul, R.; Li, T. S.; Vocadlo, D. J.; Zhou, Y. X.; Zhu, Y. B.; Mu, C. W.; Wang, Y. D.; Wei, Z. Y.; Bai, C.; Duffy, J. L.; McEachern, E. J. Discovery of MK-8719, a potent O-GlcNAcase inhibitor as a potential treatment for tauopathies. J. Med. Chem. 2019, 62, 10062– 10097, DOI: 10.1021/acs.jmedchem.9b01090Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslWms73F&md5=79e26635e124c90c86d4c1beb8b63e23Discovery of MK-8719, a potent O-GlcNAcase inhibitor as a potential treatment for tauopathiesSelnick, Harold G.; Hess, J. Fred; Tang, Cuyue; Liu, Kun; Schachter, Joel B.; Ballard, Jeanine E.; Marcus, Jacob; Klein, Daniel J.; Wang, Xiaohai; Pearson, Michelle; Savage, Mary J.; Kaul, Ramesh; Li, Tong-Shuang; Vocadlo, David J.; Zhou, Yuanxi; Zhu, Yongbao; Mu, Changwei; Wang, Yaode; Wei, Zhongyong; Bai, Chang; Duffy, Joseph L.; McEachern, Ernest J.Journal of Medicinal Chemistry (2019), 62 (22), 10062-10097CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Inhibition of O-GlcNAcase (OGA) has emerged as a promising therapeutic approach to treat tau pathol. in neurodegenerative diseases such as Alzheimer's disease and progressive supranuclear palsy. Beginning with carbohydrate-based lead mols., we pursued an optimization strategy of reducing polar surface area to align the desired drug-like properties of potency, selectivity, high central nervous system (CNS) exposure, metabolic stability, favorable pharmacokinetics, and robust in vivo pharmacodynamic response. Herein, we describe the medicinal chem. and pharmacol. studies that led to the identification of (3aR,5S,6S,7R,7aR)-5-(difluoromethyl)-2-(ethylamino)-3a,6,7,7a-tetrahydro-5H-pyrano[3,2-d]thiazole-6,7-diol 42 (MK-8719), a highly potent and selective OGA inhibitor with excellent CNS penetration that has been advanced to first-in-human phase I clin. trials.
- 4den Heijer, J. M.; Kruithof, A. C.; van Amerongen, G.; de Kam, M. L.; Thijssen, E.; Grievink, H. W.; Moerland, M.; Walker, M.; Been, K.; Skerlj, R.; Justman, C.; Dudgeon, L.; Lansbury, P.; Cullen, V. C.; Hilt, D. C.; Groeneveld, G. J. A randomized single and multiple ascending dose study in healthy volunteers of LTI-291, a centrally penetrant glucocerebrosidase activator. Br. J. Clin. Pharmacol. 2021, 87, 3561– 3573, DOI: 10.1111/bcp.14772Google ScholarThere is no corresponding record for this reference.
- 5Patterson, M. C.; Vecchio, D.; Prady, H.; Abel, L.; Wraith, J. E. Miglustat for treatment of Niemann-Pick C disease: a randomised controlled study. Lancet Neurol. 2007, 6, 765– 772, DOI: 10.1016/S1474-4422(07)70194-1Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVyrsr3F&md5=d402dde48fc19092735a4844a2d874b7Miglustat for treatment of Niemann-Pick C disease: a randomised controlled studyPatterson, Marc C.; Vecchio, Darleen; Prady, Helena; Abel, Larry; Wraith, James E.Lancet Neurology (2007), 6 (9), 765-772CODEN: LNAEAM; ISSN:1474-4422. (Elsevier Ltd.)Background: Niemann-Pick type C disease (NPC) is an inherited neurodegenerative disorder characterized by an intracellular lipid-trafficking defect with secondary accumulation of glycosphingolipids. Miglustat, a small iminosugar, reversibly inhibits glucosylceramide synthase, which catalyzes the first committed step of glycosphingolipid synthesis. Miglustat is able to cross the blood-brain barrier, and is thus a potential therapy for neurol. diseases. We aimed to establish the effect of miglustat on several markers of NPC severity. Methods: Patients aged 12 years or older who had NPC (n = 29) were randomly assigned to receive either miglustat 200 mg three times a day (n = 20) or std. care (n = 9) for 12 mo. 12 Children younger than 12 years of age were included in an addnl. cohort; all received miglustat at a dose adjusted for body surface area. All participants were then treated with miglustat for an addnl. year in an extension study. The primary endpoint was horizontal saccadic eye movement (HSEM) velocity, based on its correlation with disease progression. This study is registered as an International Std. Randomised Controlled Trial, no. ISRCTN26761144. Findings: At 12 mo, HSEM velocity had improved in patients treated with miglustat vs. those receiving std. care; results were significant when patients taking benzodiazepines were excluded (p = 0·028). Children showed an improvement in HSEM velocity of similar size at 12 mo. Improvement in swallowing capacity, stable auditory acuity, and a slower deterioration in ambulatory index were also seen in treated patients older than 12 years. The safety and tolerability of miglustat 200 mg three times a day in study participants was consistent with previous trials in type I Gaucher disease, where half this dose was used. Interpretation: Miglustat improves or stabilizes several clin. relevant markers of NPC. This is the first agent studied in NPC for which there is both animal and clin. data supporting a disease modifying benefit.
- 6Hughes, D. A.; Nicholls, K.; Shankar, S. P.; Sunder-Plassmann, G.; Koeller, D.; Nedd, K.; Vockley, G.; Hamazaki, T.; Lachmann, R.; Ohashi, T. Oral pharmacological chaperone migalastat compared with enzyme replacement therapy in Fabry disease: 18-month results from the randomised phase III ATTRACT study. J. Med. Gen. 2017, 54, 288– 296, DOI: 10.1136/jmedgenet-2016-104178Google ScholarThere is no corresponding record for this reference.
- 7Fan, J. Q.; Ishii, S.; Asano, N.; Suzuki, Y. Accelerated transport and maturation of lysosomal α-galactosidase A in Fabry lymphoblasts by an enzyme inhibitor. Nat. Med. 1999, 5, 112– 115, DOI: 10.1038/4801Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXmtF2muw%253D%253D&md5=dbc82fd0fae5c7e33f99add6f956bb98Accelerated transport and maturation of lysosomal α-galactosidase A in Fabry lymphoblasts by an enzyme inhibitorFan, Jian-Qiang; Ishii, Satoshi; Asano, Naoki; Suzuki, YoshiyukiNature Medicine (New York) (1999), 5 (1), 112-115CODEN: NAMEFI; ISSN:1078-8956. (Nature America)Fabry disease is a disorder of glycosphingolipid metab. caused by deficiency of lysosomal α-galactosidase A (α-Gal A), resulting in renal failure along with premature myocardial infarction and strokes. No effective treatment of this disorder is available at present. Studies of residual activities of mutant enzymes in many Fabry patients showed that some of them had kinetic properties similar to those for normal α-Gal A, but were significantly less stable, esp. in conditions of neutral pH. The biosynthetic processing was delayed in cultured fibroblasts of a Fabry patient, and the mutant protein formed an aggregate in endoplasmic reticulum, indicating that the enzyme deficiency in some mutants was mainly caused by abortive exit from the endoplasmic reticulum, leading to excessive degrdn. of the enzyme. We report here that 1-deoxy-galactonojirimycin (DGJ), a potent competitive inhibitor of α-Gal A, effectively enhanced α-Gal A activity in Fabry lymphoblasts, when administrated at concns. lower than that usually required for intracellular inhibition of the enzyme. DGJ seemed to accelerate transport and maturation of the mutant enzyme. Oral administration of DGJ to transgenic mice overexpressing a mutant α-Gal A substantially elevated the enzyme activity in some organs. We propose a new mol. therapeutic strategy for genetic metabolic diseases of administering competitive inhibitors as chem. chaperons' at sub-inhibitory intracellular concns.
- 8Parenti, G.; Andria, G.; Valenzano, K. J. Pharmacological chaperone therapy: Preclinical development, clinical translation, and prospects for the treatment of lysosomal storage disorders. Mol. Ther. 2015, 23, 1138– 1148, DOI: 10.1038/mt.2015.62Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1ygsb%252FO&md5=8d3c0c1dada373b9868927777d72d3cfPharmacological Chaperone Therapy: Preclinical Development, Clinical Translation, and Prospects for the Treatment of Lysosomal Storage DisordersParenti, Giancarlo; Andria, Generoso; Valenzano, Kenneth J.Molecular Therapy (2015), 23 (7), 1138-1148CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)A review. Lysosomal storage disorders (LSDs) are a group of inborn metabolic diseases caused by mutations in genes that encode proteins involved in different lysosomal functions, in most instances acidic hydrolases. Different therapeutic approaches have been developed to treat these disorders. Pharmacol. chaperone therapy (PCT) is an emerging approach based on small-mol. ligands that selectively bind and stabilize mutant enzymes, increase their cellular levels, and improve lysosomal trafficking and activity. Compared to other approaches, PCT shows advantages, particularly in terms of oral administration, broad biodistribution, and pos. impact on patients' quality of life. After preclin. in vitro and in vivo studies, PCT is now being translated in the first clin. trials, either as monotherapy or in combination with enzyme replacement therapy, for some of the most prevalent LSDs. For some LSDs, the results of the first clin. trials are encouraging and warrant further development. Future research in the field of PCT will be directed toward the identification of novel chaperones, including new allosteric drugs, and the exploitation of synergies between chaperone treatment and other therapeutic approaches.
- 9Witte, M. D.; Kallemeijn, W. W.; Aten, J.; Li, K. Y.; Strijland, A.; Donker-Koopman, W. E.; van den Nieuwendijk, A.; Bleijlevens, B.; Kramer, G.; Florea, B. I.; Hooibrink, B.; Hollak, C. E. M.; Ottenhoff, R.; Boot, R. G.; van der Marel, G. A.; Overkleeft, H. S.; Aerts, J. Ultrasensitive in situ visualization of active glucocerebrosidase molecules. Nat. Chem. Biol. 2010, 6, 907– 913, DOI: 10.1038/nchembio.466Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVansb7N&md5=e9ead74400f958f1d4d2d5f5402d5c2fUltrasensitive in situ visualization of active glucocerebrosidase moleculesWitte, Martin D.; Kallemeijn, Wouter W.; Aten, Jan; Li, Kah-Yee; Strijland, Anneke; Donker-Koopman, Wilma E.; van den Nieuwendijk, Adrianus M. C. H.; Bleijlevens, Boris; Kramer, Gertjan; Florea, Bogdan I.; Hooibrink, Berend; Hollak, Carla E. M.; Ottenhoff, Roelof; Boot, Rolf G.; van der Marel, Gijsbert A.; Overkleeft, Herman S.; Aerts, Johannes M. F. G.Nature Chemical Biology (2010), 6 (12), 907-913CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Deficiency of glucocerebrosidase (GBA) underlies Gaucher disease, a common lysosomal storage disorder. Carriership for Gaucher disease has recently been identified as major risk for parkinsonism. Presently, no method exists to visualize active GBA mols. in situ. The authors here report the design, synthesis and application of two fluorescent activity-based probes allowing highly specific labeling of active GBA mols. in vitro and in cultured cells and mice in vivo. Detection of in vitro labeled recombinant GBA on slab gels after electrophoresis is in the low attomolar range. Using cell or tissue lysates, the authors obtained exclusive labeling of GBA mols. The authors present evidence from fluorescence-activated cell sorting anal., fluorescence microscopy and pulse-chase expts. of highly efficient labeling of GBA mols. in intact cells as well as tissues of mice. In addn., the authors illustrate the use of the fluorescent probes to study inhibitors and tentative chaperones in living cells.
- 10Scherer, M.; Santana, A. G.; Robinson, K.; Zhou, S.; Overkleeft, H. S.; Clarke, L.; Withers, S. G. Lipid-mimicking phosphorus-based glycosidase inactivators as pharmacological chaperones for the treatment of Gaucher’s disease. Chem. Sci. 2021, 12, 13909– 13913, DOI: 10.1039/D1SC03831AGoogle Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVKhsb%252FP&md5=d6e35d888c911d870832a1e2bf111db7Lipid-mimicking phosphorus-based glycosidase inactivators as pharmacological chaperones for the treatment of Gaucher's diseaseScherer, Manuel; Santana, Andres G.; Robinson, Kyle; Zhou, Steven; Overkleeft, Hermen S.; Clarke, Lorne; Withers, Stephen G.Chemical Science (2021), 12 (41), 13909-13913CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Gaucher's disease, the most prevalent lysosomal storage disorder, is caused by missense mutation of the GBA gene, ultimately resulting in deficient GCase activity, hence the excessive build-up of cellular glucosylceramide. Among different therapeutic strategies, pharmacol. chaperoning of mutant GCase represents an attractive approach that relies on small org. mols. acting as protein stabilizers. Herein, we expand upon a new class of transient GCase inactivators based on a reactive 2-deoxy-2-fluoro-β-D-glucoside tethered to an array of lipid-mimicking phosphorus-based aglycons, which not only improve the selectivity and inactivation efficiency, but also the stability of these compds. in aq. media. This hypothesis was further validated with kinetic and cellular studies confirming restoration of catalytic activity in Gaucher cells after treatment with these pharmacol. chaperones.
- 11Adams, B. T.; Niccoli, S.; Chowdhury, M. A.; Esarik, A. N. K.; Lees, S. J.; Rempel, B. P.; Phenix, C. P. N-Alkylated aziridines are easily-prepared, potent, specific and cell-permeable covalent inhibitors of human β-glucocerebrosidase. Chem. Commun. 2015, 51, 11390– 11393, DOI: 10.1039/C5CC03828FGoogle Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVCmu7nI&md5=c5f088dbe0d16ac976f5070113e6f74bN-Alkylated aziridines are easily-prepared, potent, specific and cell-permeable covalent inhibitors of human β-glucocerebrosidaseAdams, B. T.; Niccoli, S.; Chowdhury, M. A.; Esarik, A. N. K.; Lees, S. J.; Rempel, B. P.; Phenix, C. P.Chemical Communications (Cambridge, United Kingdom) (2015), 51 (57), 11390-11393CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)β-Glucocerebrosidase deficiency leads to Gaucher disease and is a potential marker of Parkinson's disease. The authors have identified N-octyl conduritol aziridine as a potent and specific covalent inactivator of GBA1 in living cells. This compd. is a promising lead towards a positron emission tomog. probe intended to image GBA1 activity.
- 12Kallemeijn, W. W.; Li, K. Y.; Witte, M. D.; Marques, A. R.; Aten, J.; Scheij, S.; Jiang, J.; Willems, L. I.; Voorn-Brouwer, T. M.; van Roomen, C. P. A. A.; Ottenhoff, R.; Boot, R. G.; van den Elst, H.; Walvoort, M. T.; Florea, B. I.; Codee, J. D.; van der Marel, G. A.; Aerts, J. M.; Overkleeft, H. S. Novel Activity-Based Probes for Broad-Spectrum Profiling of Retaining β-Exoglucosidases In Situ and In Vivo. Angew. Chem. Int. Ed., Engl. 2012, 51, 12529– 12533, DOI: 10.1002/anie.201207771Google ScholarThere is no corresponding record for this reference.
- 13Do, J.; McKinney, C.; Sharma, P.; Sidransky, E. Glucocerebrosidase and its relevance to Parkinson disease. Mol. Neurodegener. 2019, 14, 36, DOI: 10.1186/s13024-019-0336-2Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MrjtFCksQ%253D%253D&md5=7777e87956e7a0fae7f677e49267cc55Glucocerebrosidase and its relevance to Parkinson diseaseDo Jenny; McKinney Cindy; Sharma Pankaj; Sidransky EllenMolecular neurodegeneration (2019), 14 (1), 36 ISSN:.Mutations in GBA1, the gene encoding the lysosomal enzyme glucocerebrosidase, are among the most common known genetic risk factors for the development of Parkinson disease and related synucleinopathies. A great deal is known about GBA1, as mutations in GBA1 are causal for the rare autosomal storage disorder Gaucher disease. Over the past decades, significant progress has been made in understanding the genetics and cell biology of glucocerebrosidase. A least 495 different mutations, found throughout the 11 exons of the gene are reported, including both common and rare variants. Mutations in GBA1 may lead to degradation of the protein, disruptions in lysosomal targeting and diminished performance of the enzyme in the lysosome.Gaucher disease is phenotypically diverse and has both neuronopathic and non-neuronopathic forms. Both patients with Gaucher disease and heterozygous carriers are at increased risk of developing Parkinson disease and Dementia with Lewy Bodies, although our understanding of the mechanism for this association remains incomplete. There appears to be an inverse relationship between glucocerebrosidase and α-synuclein levels, and even patients with sporadic Parkinson disease have decreased glucocerebrosidase. Glucocerebrosidase may interact with α-synuclein to maintain basic cellular functions, or impaired glucocerebrosidase could contribute to Parkinson pathogenesis by disrupting lysosomal homeostasis, enhancing endoplasmic reticulum stress or contributing to mitochondrial impairment. However, the majority of patients with GBA1 mutations never develop parkinsonism, so clearly other risk factors play a role. Treatments for Gaucher disease have been developed that increase visceral glucocerebrosidase levels and decrease lipid storage, although they have yet to properly address the neurological defects associated with impaired glucocerebrosidase. Mouse and induced pluripotent stem cell derived models have improved our understanding of glucocerebrosidase function and the consequences of its deficiency. These models have been used to test novel therapies including chaperone proteins, histone deacetylase inhibitors, and gene therapy approaches that enhance glucocerebrosidase levels and could prove efficacious in the treatment of forms of parkinsonism. Consequently, this rare monogenic disorder, Gaucher disease, provides unique insights directly applicable to our understanding and treatment of Parkinson disease, a common and complex neurodegenerative disorder.
- 14Ben Bdira, F.; Kallemeijn, W. W.; Oussoren, S. V.; Scheij, S.; Bleijlevens, B.; Florea, B. I.; van Roomen, C.; Ottenhoff, R.; van Kooten, M.; Walvoort, M. T. C.; Witte, M. D.; Boot, R. G.; Ubbink, M.; Overkleeft, H. S.; Aerts, J. Stabilization of Glucocerebrosidase by Active Site Occupancy. ACS Chem. Biol. 2017, 12, 1830– 1841, DOI: 10.1021/acschembio.7b00276Google ScholarThere is no corresponding record for this reference.
- 15Vocadlo, D. J.; Davies, G. J. Mechanistic insights into glycosidase chemistry. Curr. Opin. Chem. Biol. 2008, 12, 539– 555, DOI: 10.1016/j.cbpa.2008.05.010Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1KgsbnF&md5=dc269026d21f0e114c25b120b3c1e9b9Mechanistic insights into glycosidase chemistryVocadlo, David J.; Davies, Gideon J.Current Opinion in Chemical Biology (2008), 12 (5), 539-555CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. The enzymic hydrolysis of the glycosidic bond continues to gain importance, reflecting the critically important roles complex glycans play in health and disease as well as the rekindled interest in enzymic biomass conversion. Recent advances include the broadening of our understanding of enzyme reaction coordinates, through both computational and structural studies, improved understanding of enzyme inhibition through transition state mimicry and fascinating insights into mechanism yielded by phys. org. chem. approaches.
- 16Adamson, C.; Pengelly, R.; Shamsi Kazem Abadi, S.; Chakladar, S.; Draper, J.; Britton, R.; Gloster, T.; Bennet, A. J. Structural snapshots for mechanism-based inactivation of a glycoside hydrolase by cyclopropyl-carbasugars. Angew. Chem., Int. Ed. 2016, 55, 14978– 14982, DOI: 10.1002/anie.201607431Google ScholarThere is no corresponding record for this reference.
- 17Shamsi Kazem Abadi, S.; Tran, M.; Yadav, A. K.; Adabala, P. J. P.; Chakladar, S.; Bennet, A. J. New class of glycoside hydrolase mechanism-based covalent inhibitors: Glycosylation transition state conformations. J. Am. Chem. Soc. 2017, 139, 10625– 10628, DOI: 10.1021/jacs.7b05065Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Sht7fJ&md5=67e2df7e82c721ccf61901087fb373a7New Class of Glycoside Hydrolase Mechanism-Based Covalent Inhibitors: Glycosylation Transition State ConformationsShamsi Kazem Abadi, Saeideh; Tran, Michael; Yadav, Anuj K.; Adabala, Pal John Pal; Chakladar, Saswati; Bennet, Andrew J.Journal of the American Chemical Society (2017), 139 (31), 10625-10628CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The design of covalent inhibitors in glycoscience research is important for the development of chem. biol. probes. Here we report the synthesis of a new carbocyclic mechanism-based covalent inhibitor of an α-glucosidase. The enzyme efficiently catalyzes its alkylation via either an allylic cation or a cationic transition state. We show this allylic covalent inhibitor has different catalytic proficiencies for pseudoglycosylation and deglycosylation. Such inhibitors have the potential to be useful chem. biol. tools.
- 18Danby, P. M.; Withers, S. G. Glycosyl cations versus allylic cations in spontaneous and enzymatic hydrolysis. J. Am. Chem. Soc. 2017, 139, 10629– 10632, DOI: 10.1021/jacs.7b05628Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1WmtL%252FF&md5=b2be051f5f2a039d46667e7ef4e7c4dbGlycosyl cations versus allylic cations in spontaneous and enzymatic hydrolysisDanby, Phillip M.; Withers, Stephen G.Journal of the American Chemical Society (2017), 139 (31), 10629-10632CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Enzymic prenyl and glycosyl transfer are seemingly unrelated reactions that yield mols. and protein modifications with disparate biol. functions. However, both reactions employ diphosphate-activated donors and each proceed via cationic species: allylic cations and oxocarbenium ions, resp. Here, we explored the relation between these processes by prepg. valienyl ethers to serve as glycoside mimics that were capable of allylic rather than oxocarbenium cation stabilization. Rate consts. for the spontaneous hydrolysis of aryl glycosides and their analogous valienyl ethers were found to be almost identical, as were the corresponding activation enthalpies and entropies. This close similarity extended to the assocd. secondary kinetic isotope effects (KIEs), indicating very similar transition state stabilities and structures. Screening a library of >100 β-glucosidases identified a no. of enzymes that catalyzed the hydrolysis of these valienyl ethers with kcat values of ≤20 s-1. Detailed anal. of one such enzyme showed that ether hydrolysis occurred via the analogous mechanisms found for glycosides, and through a very similar transition state. This suggested that the generally lower rates of enzymic cleavage of the cyclitol ethers reflects evolutionary specialization of these enzymes toward glycosides rather than inherent reactivity differences.
- 19Parenti, G. Treating lysosomal storage diseases with pharmacological chaperones: from concept to clinics. EMBO Mol. Med. 2009, 1, 268– 279, DOI: 10.1002/emmm.200900036Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1CjurrF&md5=c897e238ada0121bd4fd8d9a663aed0aTreating lysosomal storage diseases with pharmacological chaperones: from concept to clinicsParenti, GiancarloEMBO Molecular Medicine (2009), 1 (5), 268-279CODEN: EMMMAM; ISSN:1757-4684. (John Wiley & Sons Ltd.)A review. Lysosomal storage diseases (LSDs) are a group of genetic disorders due to defects in any aspect of lysosomal biol. During the past two decades, different approaches have been introduced for the treatment of these conditions. Among them, enzyme replacement therapy (ERT) represented a major advance and is used successfully in the treatment of some of these disorders. However, ERT has limitations such as insufficient biodistribution of recombinant enzymes and high costs. An emerging strategy for the treatment of LSDs is pharmacol. chaperone therapy (PCT), based on the use of chaperone mols. that assist the folding of mutated enzymes and improve their stability and lysosomal trafficking. After proof-of-concept studies, PCT is now being translated into clin. applications for Fabry, Gaucher and Pompe disease. This approach, however, can only be applied to patients carrying chaperone-responsive mutations. The recent demonstration of a synergistic effect of chaperones and ERT expands the applications of PCT and prompts a re-evaluation of their therapeutic use and potential. This review discusses the strengths and drawbacks of the potential therapies available for LSDs and proposes that future research should be directed towards the development of treatment protocols based on the combination of different therapies to improve the clin. outcome of LSD patients.
- 20Sidransky, E.; Lopez, G. The link between the GBA gene and parkinsonism. Lancet Neurol. 2012, 11, 986– 998, DOI: 10.1016/S1474-4422(12)70190-4Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFCqtbnJ&md5=522c2d24e1e4f8e826a211991fe35df8The link between the GBA gene and parkinsonismSidransky, Ellen; Lopez, GriselLancet Neurology (2012), 11 (11), 986-998CODEN: LNAEAM; ISSN:1474-4422. (Elsevier Ltd.)A review. Summary: Mutations in the glucocerebrosidase (GBA) gene, which encodes the lysosomal enzyme that is deficient in Gaucher's disease, are important and common risk factors for Parkinson's disease and related disorders. This assocn. was first recognized in the clinic, where parkinsonism was noted, albeit rarely, in patients with Gaucher's disease and more frequently in relatives who were obligate carriers. Subsequently, findings from large studies showed that patients with Parkinson's disease and assocd. Lewy body disorders had an increased frequency of GBA mutations when compared with control individuals. Patients with GBA-assocd. parkinsonism exhibit varying parkinsonian phenotypes but tend to have an earlier age of onset and more assocd. cognitive changes than patients with parkinsonism without GBA mutations. Hypotheses proposed to explain this assocn. include a gain-of-function due to mutations in glucocerebrosidase that promotes α-synuclein aggregation; substrate accumulation due to enzymic loss-of-function, which affects α-synuclein processing and clearance; and a bidirectional feedback loop. Identification of the pathol. mechanisms underlying GBA-assocd. parkinsonism will improve our understanding of the genetics, pathophysiol., and treatment for both rare and common neurol. diseases.
- 21Sinnott, M. L.; Souchard, I. J. The mechanism of action of β-galactosidase. Effect of aglycone nature and α-deuterium substitution on the hydrolysis of aryl galactosides. Biochem. J. 1973, 133, 89– 98, DOI: 10.1042/bj1330089Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE3sXks1eitrY%253D&md5=968441696aad79c9d0b7a549ed86c381Mechanism of action of β-galactosidase. Effect of aglycone nature and α-deuterium substitution on the hydrolysis of aryl galactosidesSinnott, Michael L.; Souchard, Jan J. L.Biochemical Journal (1973), 133 (1), 89-98CODEN: BIJOAK; ISSN:0264-6021.Steady-state kinetic parameters for the hydrolysis of 13 aryl β-D-galactopyranosides catalyzed by β-galactosidase (I) showed no simple dependence on aglycone acidity. The α-deuterium kinetic isotope effects (kH/kD) for 7 substrates and the effects of methanolysis of galactosyl-I in 1.5M MeOH on kH/kD were incompatible with a 2-step mechanism. A conformational change, liberation of a galactopyranosyl cation in an intimate ion-pair, nonproductive but preferential collapse of the ion pair to a covalent species, and reaction of the galactosyl-I through the ion-paired form was proposed as a reaction mechanism for β-galactosidase.
- 22Kempton, J. B.; Withers, S. G. Mechanism of Agrobacterium .beta.-glucosidase: kinetic studies. Biochemistry 1992, 31, 9961– 9969, DOI: 10.1021/bi00156a015Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XmtVOrsbg%253D&md5=645c6f00c3d37e0b98ec1d67a8a740d4Mechanism of Agrobacterium β-glucosidase: kinetic studiesKempton, Julie B.; Withers, Stephen G.Biochemistry (1992), 31 (41), 9961-9CODEN: BICHAW; ISSN:0006-2960.β-Glucosidase from A. faecalis (previously Alcaligenes faecalis) was subjected to a detailed kinetic investigation with a range of substrates to probe its specificity and mechanism. It had a relatively broad specificity for the substrate sugar moiety and exhibited a classical pH dependence for its kinetic parameters with 3 different substrates and an identical pH dependence for its inactivation by a mechanism-based inactivator, cyclophellitol. Measurement of kcat and Km values for a series of aryl glucoside substrates allowed construction of a Broensted plot, the concave-downward shape of which was consistent with the anticipated 2-step mechanism involving a glucosyl-enzyme intermediate which is formed and hydrolyzed via oxocarbonium ion-like transition states. The slope of the leaving group-dependent portion of the Broensted plot (β1g = -0.7) indicated a large degree of bond cleavage at the transition state. Secondary deuterium kinetic isotope effects measured for 5 different aryl glucosides were also consistent with this mechanism and further suggested that the transition state for formation of the glucosyl-enzyme intermediate, probed with the slower substrates for which kH/kD = 1.06, is more SN2-like than that for its hydrolysis (for which kH/kD - 1.11). Reasons for this difference are proposed, and values of Ki for several ground-state and transition-state analog inhibitors are presented which support the concept of sp2-hybridized transition states.
- 23Breen, I. Z.; Artola, M.; Wu, L.; Beenakker, T. J. M.; Offen, W. A.; Aerts, J. M. F. G.; Davies, G. J.; Overkleeft, H. S. Competitive and covalent inhibitors of human lysosomal retaining exoglucosidases. eLS; Wiley & Sons Ltd.: Chichester, 2018.Google ScholarThere is no corresponding record for this reference.
- 24Segel, I. H. Enzyme Kinetics: Behavior and Analysis of Rapid Equilibrium and Steady State Enzyme Systems; Wiley: New York, 1975.Google ScholarThere is no corresponding record for this reference.
- 25Jones, C. C.; Sinnott, M. L. Leaving ability and basicity of leaving groups attached by first-row elements. J. Chem. Soc., Chem. Commun. 1977, 767– 768, DOI: 10.1039/c39770000767Google ScholarThere is no corresponding record for this reference.
- 26Farren-Dai, M.; Sannikova, N.; Świderek, K.; Moliner, V.; Bennet, A. J. Fundamental insight into glycoside hydrolase-catalyzed hydrolysis of the universal Koshland substrates–glycopyranosyl fluorides. ACS Catal. 2021, 11, 10383– 10393, DOI: 10.1021/acscatal.1c01918Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslWksLvL&md5=99d92a35ade39a31092971fd351c9081Fundamental Insight into Glycoside Hydrolase-Catalyzed Hydrolysis of the Universal Koshland Substrates-Glycopyranosyl FluoridesFarren-Dai, Marco; Sannikova, Natalia; Swiderek, Katarzyna; Moliner, Vicent; Bennet, Andrew J.ACS Catalysis (2021), 11 (16), 10383-10393CODEN: ACCACS; ISSN:2155-5435. (American Chemical Society)The authors measured a panel of five kinetic isotope effects [KIEs; three secondary deuterium (1-, 2-, and 5-2H) and two primary (1-13C and 5-18O) effects] for the catalyzed hydrolysis of α-D-glucopyranosyl fluoride by two inverting glycoside hydrolases (GHs). The exptl. KIEs were compared to av. values computed with multiscale QM/MM methods for the hydrolysis of α-D-glucopyranosyl fluoride promoted by an inverting α-glucosidase (Aspergillus niger, GH family 15) to give β-D-glucopyranose, which the authors explored by the generation of free energy surfaces. The authors also measured the same KIEs for catalysis of α-D-glucopyranosyl fluoride by the Trichoderma virens GH55 inverting β-glucosidase using the authors' panel of isotopologues; this reaction occurs via the "Hehre resynthesis-hydrolysis mechanism" to give, by two inversions of configuration, the hydrolysis product α-D-glucopyranose. The transition states for both enzymic reactions are essentially identical with fluoride ion departure occurring within a glycoside hydrolase active site that stabilizes pyranosylium ion-like TSs, and with catalysis driven solely by H-bonding assistance from an enzymic carboxylic acid residue. That is, no assistance is required from the bound nucleophile, which in these two cases is either a water mol. (GH15) or a sugar hydroxyl group (GH55).
- 27Boer, D. E.; Mirzaian, M.; Ferraz, M. J.; Zwiers, K. C.; Baks, M. V.; Hazeu, M. D.; Ottenhoff, R.; Marques, A. R. A.; Meijer, R.; Roos, J. C. P.; Cox, T. M.; Boot, R. G.; Pannu, N.; Overkleeft, H. S.; Artola, M.; Aerts, J. M. Human glucocerebrosidase mediates formation of xylosyl-cholesterol by β-xylosidase and transxylosidase reactions. J. Lipid Res. 2021, 62, 100018, DOI: 10.1194/jlr.RA120001043Google ScholarThere is no corresponding record for this reference.
- 28Beauhaire, J.; Ducrot, P. H. An epoxide derived from D-glucose as the key intermediate for penaresidine and sphingolipids synthesis. Synth. Commun. 1998, 28, 2443– 2456, DOI: 10.1080/00397919808004296Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktFCitrk%253D&md5=80b7b7320718c1c3e7e70efab3b2295fAn epoxide derived from D-glucose as the key intermediate for penaresidine and sphingolipids synthesisBeauhaire, Josiane; Ducrot, Paul-HenriSynthetic Communications (1998), 28 (13), 2443-2456CODEN: SYNCAV; ISSN:0039-7911. (Marcel Dekker, Inc.)A multigram-scale synthesis of 3R,4R,5R-3,5-dibenzyloxy-4-p-methoxybenzyl-1,2-epoxypentane and its use as intermediate for sphingolipids, penazeridine and penazetidine synthesis are described.
- 29Ley, S. V.; Baeschlin, D. K.; Dixon, D. J.; Foster, A. C.; Ince, S. J.; Priepke, W. M.; Reynolds, D. J. 1,2-Diacetals: A new opportunity for organic synthesis. Chem. Rev. 2001, 101, 53– 80, DOI: 10.1021/cr990101jGoogle Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXos1Ckt70%253D&md5=08dc53ff1be3a0f022e812efd4013d631,2-Diacetals: A New Opportunity for Organic SynthesisLey, Steven V.; Baeschlin, Daniel K.; Dixon, Darren J.; Foster, Alison C.; Ince, Stuart J.; Priepke, Henning W. M.; Reynolds, Dominic J.Chemical Reviews (Washington, D. C.) (2001), 101 (1), 53-80CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review with 127 refs. 1,2-Diacetals offer diol protection.
- 30Shih, T. L.; Liao, W. Y.; Yen, W. C. Regioselective fluorination in synthesis of deoxyfluoro quercitols from D-(−)-quinic acid. Tetrahedron 2014, 70, 9621– 9627, DOI: 10.1016/j.tet.2014.11.001Google ScholarThere is no corresponding record for this reference.
- 31Baici, A.; Schenker, P.; Wachter, M.; Ruedi, P. 3-Fluoro-2,4-dioxa-3-phosphadecalins as inhibitors of acetylcholinesterase. A reappraisal of kinetic mechanisms and diagnostic methods. Chem. Biodivers. 2009, 6, 261– 282, DOI: 10.1002/cbdv.200800334Google ScholarThere is no corresponding record for this reference.
- 32McCarter, J. D.; Withers, S. G. 5-Fluoro Glycosides: A New Class of Mechanism-Based Inhibitors of Both α- and β-Glucosidases. J. Am. Chem. Soc. 1996, 118, 241– 242, DOI: 10.1021/ja952732aGoogle Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XjsVKrtA%253D%253D&md5=051a0d0bd5ad03a5af007d1137be78235-Fluoro Glycosides: A New Class of Mechanism-Based Inhibitors of Both α- and β-GlucosidasesMcCarter, John D.; Withers, Stephen G.Journal of the American Chemical Society (1996), 118 (1), 241-2CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Mechanism-based inhibitors of retaining glycosidases are of considerable interest, both academically and as potential therapeutics. 5-Fluoroglycosyl fluorides I represent a new class of such inhibitors, functioning via the formation of a stabilized 5-fluoroglycosyl-enzyme intermediate that turns over only very slowly. Synthesis is achieved via radical bromination of the corresponding protected glycosyl fluoride, displacement of the bromide by fluoride and deprotonation. 5-Fluoro-β-glucosyl fluoride functions as a time-dependent inactivator of Agrobacterium faecalis β-glucosidase with a second order rate consts. for inactivation of ki/Ki = 660 min-1 mM-1. Turnover of the 5-fluoro α-glucosyl-enzyme intermediate occurs with a half life of 8.5 min. 5-Fluoro α-glucosyl fluoride functions similarly as an inactivator of yeast α-glucosidase, but inactivation is too rapid to allow detn. of kinetic parameters for the inactivation process. Turnover is also more rapid, occurring with a rate const. of 6.6 min-1. Steady state apparent Ki values of 0.3 μM and 1.4 μM are detd. for β-glucosidase and α-glucosidase resp.
- 33Ren, W.; Farren-Dai, M.; Sannikova, N.; Świderek, K.; Wang, Y.; Akintola, O.; Britton, R.; Moliner, V.; Bennet, A. J. Glycoside hydrolase stabilization of transition state charge: New directions for inhibitor design. Chem. Sci. 2020, 11, 10488– 10495, DOI: 10.1039/D0SC04401FGoogle Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVClurbP&md5=becf2cf359a256beee092f5945e647e0Glycoside hydrolase stabilization of transition state charge: new directions for inhibitor designRen, Weiwu; Farren-Dai, Marco; Sannikova, Natalia; Swiderek, Katarzyna; Wang, Yang; Akintola, Oluwafemi; Britton, Robert; Moliner, Vicent; Bennet, Andrew J.Chemical Science (2020), 11 (38), 10488-10495CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Carbasugars are structural mimics of naturally occurring carbohydrates that can interact with and inhibit enzymes involved in carbohydrate processing. In particular, carbasugars have attracted attention as inhibitors of glycoside hydrolases (GHs) and as therapeutic leads in several disease areas. However, it is unclear how the carbasugars are recognized and processed by GHs. Here, we report the synthesis of three carbasugar isotopologues and provide a detailed transition state (TS) anal. for the formation of the initial GH-carbasugar covalent intermediate, as well as for hydrolysis of this intermediate, using a combination of exptl. measured kinetic isotope effects and hybrid QM/MM calcns. We find that the α-galactosidase from Thermotoga maritima effectively stabilizes TS charge development on a remote C5-allylic center acting in concert with the reacting carbasugar, and catalysis proceeds via an exploded, or loose, SN2 transition state with no discrete enzyme-bound cationic intermediate. We conclude that, in complement to what we know about the TS structures of enzyme-natural substrate complexes, knowledge of the TS structures of enzymes reacting with non-natural carbasugar substrates shows that GHs can stabilize a wider range of pos. charged TS structures than previously thought. Furthermore, this enhanced understanding will enable the design of new carbasugar GH transition state analogs to be used as, for example, chem. biol. tools and pharmaceutical lead compds.
- 34Akintola, O.; Farren-Dai, M.; Ren, W.; Bhosale, S.; Britton, R.; Świderek, K.; Moliner, V.; Bennet, A. J. Glycoside hydrolase catalysis: Do substrates and mechanism-based covalent inhibitors react via matching transition states?. ACS Catal. 2022, 12, 14667– 14678, DOI: 10.1021/acscatal.2c04027Google ScholarThere is no corresponding record for this reference.
- 35Rowland, R. J.; Wu, L.; Liu, F.; Davies, G. J. A baculoviral system for the production of human β-glucocerebrosidase enables atomic resolution analysis. Acta Crystallogr. Sect. D-Struct. Biol. 2020, 76, 565– 580, DOI: 10.1107/S205979832000501XGoogle ScholarThere is no corresponding record for this reference.
- 36Speciale, G.; Thompson, A. J.; Davies, G. J.; Williams, S. J. Dissecting conformational contributions to glycosidase catalysis and inhibition. Curr. Opin. Struct. Biol. 2014, 28, 1– 13, DOI: 10.1016/j.sbi.2014.06.003Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlSqurbE&md5=80834f55b372466d07e64e1c7d7c7555Dissecting conformational contributions to glycosidase catalysis and inhibitionSpeciale, Gaetano; Thompson, Andrew J.; Davies, Gideon J.; Williams, Spencer J.Current Opinion in Structural Biology (2014), 28 (), 1-13CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Glycoside hydrolases (GHs) are classified into >100 sequence-based families. These enzymes process a wide variety of complex carbohydrates with varying stereochem. at the anomeric and other ring positions. The shapes that these sugars adopt upon binding to their cognate GHs, and the conformational changes that occur along the catalysis reaction coordinate is termed the conformational itinerary. Efforts to define the conformational itineraries of GHs have focussed upon the crit. points of the reaction: substrate-bound (Michaelis), transition state, intermediate (if relevant) and product-bound. Recent approaches to defining conformational itineraries that marry X-ray crystallog. of enzymes bound to ligands that mimic the crit. points, along with advanced computational methods and kinetic isotope effects are discussed.
- 37Matsusaka, K.; Chiba, S.; Shimomura, T. Purification and substrate specificity of brewer’s yeast .ALPHA.-glucosidase. Agric. Biol. Chem. 1977, 41, 1917– 1923, DOI: 10.1271/bbb1961.41.1917Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE1cXhvVWjtw%253D%253D&md5=b7be52fbe741da0dfd1c16676dfd13a3Purification and substrate specificity of Brewers' yeast α-glucosidaseMatsusaka, Kouetsu; Chiba, Seiya; Shimomura, TokujiAgricultural and Biological Chemistry (1977), 41 (10), 1917-23CODEN: ABCHA6; ISSN:0002-1369.Three kinds of α-glucosidase (I, II and III) were isolated from brewers' yeast. α-Glucosidases I and II were homogeneous in disc electrophoresis, but α-glucosidase III was not. Their pH optima were 6.3-7.1. Each of them was a typical α-glucosidase showing a preferential activity on Ph α-glucoside (I). α-Glucosidase I was isomaltase, which can hydrolyze isomaltose but not maltose. α-Glucosidases II and III showed no activity on isomaltose. The ratio of velocity of hydrolysis for I and isomaltose of α-glucosidase I was 100:9, and those for I and maltose of α-glucosidases II and III were 100:17 and 100:18, resp.
- 38Bordwell, F. G. Are nucleophilic bimolecular concerted reactions involving four or more bonds a myth?. Acc. Chem. Res. 1970, 3, 281– 290, DOI: 10.1021/ar50033a001Google ScholarThere is no corresponding record for this reference.
- 39Bordwell, F. G.; Mecca, T. G. Nucleophilic substitutions in allylic systems. Further evidence against existence of concerted SN2’ mechanism. J. Am. Chem. Soc. 1972, 94, 5829– 5837, DOI: 10.1021/ja00771a048Google ScholarThere is no corresponding record for this reference.
- 40Kantner, S. S.; Humski, K.; Goering, H. L. On the solvolysis of 2-cyclohexenyl 3,5-dinitrobenzoate and p-nitrobenzoate in aqueous acetone. Introduction of acyl-oxygen cleavage by basic buffer systems. J. Am. Chem. Soc. 1982, 104, 1693– 1697, DOI: 10.1021/ja00370a040Google ScholarThere is no corresponding record for this reference.
- 41Streitwieser, A.; Jayasree, E. G.; Hasanayn, F.; Leung, S. S. H. A theoretical study of SN2’ reactions of allylic halides: Role of ion pairs. J. Org. Chem. 2008, 73, 9426– 9434, DOI: 10.1021/jo8020743Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlWqu7vJ&md5=5d733dd816b847c74d270de5d2131da8A Theoretical Study of SN2' Reactions of Allylic Halides: Role of Ion PairsStreitwieser, A.; Jayasree, E. G.; Hasanayn, F.; Leung, S. S.-H.Journal of Organic Chemistry (2008), 73 (23), 9426-9434CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)Various disparate exptl. results are explained by the hypothesis that reactions of anionic nucleophiles with allylic halides are generally SN2. The SN2' reactions that do occur proceed generally with anti stereochem. Reactions with ion pair nucleophiles occur preferentially as SN2' reactions with syn stereochem. This hypothesis is consistent with a variety of computations at the HF, B3LYP, mPW1PW91 and MP2 levels with the 6-31+G(d) basis set of reactions of Li and Na fluoride and chloride with allyl halides and 4-halo-2-pentenes. Solvation is considered by a combination of coordination of di-Me ether to the lithium and sodium cations and "dielec. solvation" with a polarized continuum model.
- 42Bentley, T. W.; Carter, G. E. The SN2-SN1 spectrum. 4. The SN2 (intermediate) mechanism for aolvolysis of tert-butyl chloride: A revised Y scale of solvent ionizing power based on solvolysis of 1-adamantyl chloride. J. Am. Chem. Soc. 1982, 104, 5741– 5747, DOI: 10.1021/ja00385a031Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL38Xls1ajsr8%253D&md5=354c3dde00ddb693959c9ba57876c315The SN2-SN1 spectrum. 4. The SN2 (intermediate) mechanism for solvolyses of tert-butyl chloride: a revised Y scale of solvent ionizing power based on solvolyses of 1-adamantyl chlorideBentley, T. William; Carter, Gillian E.Journal of the American Chemical Society (1982), 104 (21), 5741-7CODEN: JACSAT; ISSN:0002-7863.New kinetic data for solvolysis of 1-adamantyl chloride and bromide were detd. in several solvents. The reliability of the results is supported by the consistent activation parameters and by bromide-chloride rate ratios, which closely parallel the corresponding ratios obtained for solvolysis of tert-Bu halides. The effects of added salts on solvolyzes are given. A new scale of solvent ionizing power for chlorides (YCl) was defined as log (k/k0) = mYCl (k = solvolysis rate const. for 1-adamantyl chloride in any solvent at 25°; k0 = same for 80 vol.% aq. EtOH; m = 1); an analogous scale is defined using data for 1-adamantyl bromide. These Y values were used to correlate rate data for tert-Bu and 1-adamantyl halides.
- 43Ohga, Y.; Munakata, M.; Kitagawa, T.; Kinoshita, T.; Takeuchi, K.; Oishi, Y.; Fujimoto, H. Solvolyses of bicyclo[2.2.2]oct-1-yl and 1-adamantyl systems containing an ethylidene substituent on the 2-position: typical examples of rate enhancements ascribed to relief of F-strain. J. Org. Chem. 1994, 59, 4056– 4067, DOI: 10.1021/jo00094a012Google ScholarThere is no corresponding record for this reference.
- 44Chan, J.; Tang, A.; Bennet, A. J. A stepwise solvent-promoted SNi reaction of α-D-glucopyranosyl fluoride: Mechanistic implications for retaining glycosyltransferases. J. Am. Chem. Soc. 2012, 134, 1212– 1220, DOI: 10.1021/ja209339jGoogle Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFylur7O&md5=719b679e96b9141d5c98b35c021afa8dA Stepwise Solvent-Promoted SNi Reaction of α-D-Glucopyranosyl Fluoride: Mechanistic Implications for Retaining GlycosyltransferasesChan, Jefferson; Tang, Ariel; Bennet, Andrew J.Journal of the American Chemical Society (2012), 134 (2), 1212-1220CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The solvolysis of α-d-glucopyranosyl fluoride in hexafluoro-2-propanol gives two products, 1,1,1,3,3,3-hexafluoropropan-2-yl α-d-glucopyranoside and 1,6-anhydro-β-d-glucopyranose. The ratio of these two products is essentially unchanged for reactions that are performed between 56 and 100 °C. The activation parameters for the solvolysis reaction are as follows: ΔH⧺ = 81.4 ± 1.7 kJ mol-1, and ΔS⧺ = -90.3 ± 4.6 J mol-1 K-1. To characterize, by use of multiple kinetic isotope effect (KIE) measurements, the TS for the solvolysis reaction in hexafluoro-2-propanol, we synthesized a series of isotopically labeled α-d-glucopyranosyl fluorides. The measured KIEs for the C1 deuterium, C2 deuterium, C5 deuterium, anomeric carbon, ring oxygen, O6, and solvent deuterium are 1.185 ± 0.006, 1.080 ± 0.010, 0.987 ± 0.007, 1.008 ± 0.007, 0.997 ± 0.006, 1.003 ± 0.007, and 1.68 ± 0.07, resp. The transition state for the solvolysis reaction was modeled computationally using the exptl. KIE values as constraints. Taken together, the reported data are consistent with the retained solvolysis product being formed in an SNi (DN⧺*ANss) reaction with a late transition state in which cleavage of the glycosidic bond is coupled to the transfer of a proton from a solvating hexafluoro-2-propanol mol. In comparison, the inverted product, 1,6-anhydro-β-d-glucopyranose, is formed by intramol. capture of a solvent-equilibrated glucopyranosylium ion, which results from dissocn. of the solvent-sepd. ion pair formed in the rate-limiting ionization reaction (DN⧺ + AN). The implications that this model reaction have for the mode of action of retaining glycosyltransferases are discussed.
- 45Sinnott, M. L.; Jencks, W. P. Solvolysis of D-glucopyranosyl derivatives in mixtures of ethanol and 2,2,2-trifluoroethanol. J. Am. Chem. Soc. 1980, 102, 2026– 2032, DOI: 10.1021/ja00526a043Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3cXktVygu7c%253D&md5=c9c081fe94897789fa673d211344f1c7Solvolysis of D-glucopyranosyl derivatives in mixtures of ethanol and 2,2,2-trifluoroethanolSinnott, Michael L.; Jencks, William P.Journal of the American Chemical Society (1980), 102 (6), 2026-32CODEN: JACSAT; ISSN:0002-7863.The products of solvolysis of α- and β-D-glucopyranosyl fluorides, 2,4-dinitrophenyl β-D-glucopyranoside, and the trifluoromethanesulfonates of the β-D-glucopyranosyl 3-bromopyridinium and α-D-glucopyranosyl 4-methylpyridinium ions in an equimolar mixt. of ethanol and trifluoroethanol buffered with ∼2 equiv of 2,6-lutidine have been examd. by gas-liq. chromatog. of their trimethylsilyl ethers. The initial products of the solvolyses of Ph α- and β-D-glucopyranosides catalyzed by CF3SO3H in an equimolar mixt. of ethanol and trifluoroethanol, and the products of uncatalyzed solvolysis of β-D-glucopyranosyl-p-nitrophenyltriazene, have been likewise examd. The compn. of the medium for solvolysis of the glucosyl fluorides has also been systematically varied from pure ethanol to pure trifluoroethanol. The percentage of products with the same anomeric configuration as the starting material is in the range 8.1-88.5%; change of leaving group, at const. anomeric configuration, or of anomeric configuration, at const. leaving group, yields different product distributions. Therefore the transition state of the product-detg. step contains the leaving group. The preference for attack by ethanol as compared with trifluoroethanol varies from 0.9 to 20 in a way which shows no general systematic distinction between pathways for retention or inversion. The nucleophilic selectivity for retention is lowered by anionic leaving groups, esp. fluoride, which preferentially stabilize the transition state contg. trifluoroethanol by H bonding. Nucleophilic attack at the α face is preferred over nucleophilic attack at the β face, and exhibits a lower selectivity: this is ascribed to H bonding between the O atom of the 2-hydroxyl group and the hydroxyl group of the approaching alc. A model for solvolysis involving a reversibly formed ion pair or encounter complex is incompatible with the selectivities still obsd. with leaving groups less nucleophilic than the solvent components: a model involving selection between the components of a pool of solvent mols. by an irreversibly formed ion pair or encounter complex requires an implausibly large pool to explain obsd. specificities. The obsd. selectivities are a consequence of the facilitation of the departure of the leaving group by the solvent, from either side of the reaction center.
- 46Wan, Q.; Parks, J. M.; Hanson, B. L.; Fisher, S. Z.; Ostermann, A.; Schrader, T. E.; Graham, D. E.; Coates, L.; Langan, P.; Kovalevsky, A. Direct determination of protonation states and visualization of hydrogen bonding in a glycoside hydrolase with neutron crystallography. Proc. Natl. Acad. Sci. U.S.A. 2015, 112, 12384– 12389, DOI: 10.1073/pnas.1504986112Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFamsLrK&md5=783870c0386e3f3a42b3451fa7335762Direct determination of protonation states and visualization of hydrogen bonding in a glycoside hydrolase with neutron crystallographyWan, Qun; Parks, Jerry M.; Hanson, B. Leif; Fisher, Suzanne Zoe; Ostermann, Andreas; Schrader, Tobias E.; Graham, David E.; Coates, Leighton; Langan, Paul; Kovalevsky, AndreyProceedings of the National Academy of Sciences of the United States of America (2015), 112 (40), 12384-12389CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Glycoside hydrolases (GHs) apply acid/base chem. to catalyze the decompn. of complex carbohydrates. These ubiquitous enzymes accept protons from solvent and donate them to substrates at close to neutral pH by modulating the pKa values of key side-chains during catalysis. However, it is not known how the catalytic acid residue acquires a proton and transfers it efficiently to the substrate. To better understand GH chem., we used macromol. neutron crystallog. to directly det. protonation and ionization states of the active site residues of a family 11 GH, Trichoderma reesei xylanase II (XynII), at multiple pD (pD = pH + 0.4) values. The general acid (Glu residue) cycled between 2 conformations, upward and downward, but was protonated only in the downward orientation. The authors performed continuum electrostatics calcns. to est. the pKa values of the catalytic Glu residues in both the apoenzyme and substrate-bound states of XynII. The calcd. pKa of the Glu residue increased substantially when the side-chain moved down. The energy barrier required to rotate the catalytic Glu residue back to the upward conformation, where it could protonate the glycosidic O atom of the substrate, was 4.3 kcal/mol according to free energy simulations. These findings shed light on the initial stage of the glycoside hydrolysis reaction in which mol. motion enables the general acid catalyst to obtain a proton from the bulk solvent and deliver it to the glycosidic O atom.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 1 publications.
- Oluwafemi Akintola, Sandeep Bhosale, Andrew J. Bennet. Mechanism-Based Allylic Carbasugar Chlorides That Form Covalent Intermediates with α- and β-Galactosidases. Molecules 2024, 29
(20)
, 4870. https://doi.org/10.3390/molecules29204870
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Small molecule covalent inhibitors and the mechanism of action of retaining glycosidases. (a) Mechanism used by glucocerebrosidase for hydrolysis of its natural β-glucoside substrate. (b) Kinetic scheme for GCase-catalyzed turnover of β-d-glucopyranoside substrates. (c) Mechanism of action for carbasugar covalent inhibitors. (d) Kinetic scheme for covalent carbasugar inhibitors.
Figure 2
Figure 2. Structures of carbasugars used in this study and their in vitro characterization. Carbasugar covalent inhibitors used in the current study. The pseudoanomeric carbons (shown by the asterisk on 1) are drawn in identical positions for all compounds.
Scheme 1
Scheme 1. Synthesis of d- and l-Carbaxylopyranosyl Halides; (a) Synthesis of Intermediates 14 and 15, (b) Synthesis of Intermediates 16 and 23, (c) Synthesis of Target CarbasugarsaaReagents: (i) NaIO4, H2O, rt; (ii) Ph3P+CH3, BuLi, 0 °C; (iii) dioxane/water, cat H+, 90 °C; (iv) CH2CHMgBr, THF, 0 °C; (v) Grubbs’ 2nd generation, CH2Cl2, heat; (vi) CH3COCOCH3, HC(OCH3)3, cat H+; (vii) DAST, diglyme, 0 °C; (viii) TFA, CH2Cl2 (for 4 and 5 from 20 and 21, respectively); (ix) Dess-Martin periodinane, CH2Cl2; (x) NaBD4 (MeOD); (xi) TFA, CH2Cl2, H2O; (xii) BCl3, CH2Cl2, −78 °C (for 6, 7, 6-D, and 7-D from 26 to 29, respectively). Abbreviations: DAST, (diethylamino)sulfur trifluoride; TFA, trifluoroacetic acid.
Figure 3
Figure 3. In vitro characterization of carbasugars used in this study. (a) Nonlinear least-squares fit of kapp values to a modified Michaelis–Menten expression (eq S3) for the approach to steady-state GCase activity on incubation with d-carbaxylosyl fluoride 4. (b) Linear fit of kapp values for the approach to steady state for covalent inhibition of GCase by d-carbaxylosyl chloride 6 (blue) and l-carbaxylosyl chloride 7 (red). (c) Stabilization of folded GCase measured using differential scanning fluorimetry (DSF) in the presence of d-carbaxylosyl chloride 6 (blue), l-carbaxylosyl chloride 7 (red), or vehicle along (black). All lines are the best nonlinear least-squares fit to the appropriate equation. All experiments were carried out at T = 25 °C in McIlvaine buffer pH 5.2 containing 0.1% Triton X-100, 0.24% sodium taurodeoxycholate, and 0.1% BSA. (d) Mechanisms for the GCase pseudoglycosylation step by single turnover chaperones 6 and 7, which after diffusion of chloride out of the active site give identical allylic cation intermediates that are trapped by the enzymatic nucleophile (Glu340) to give the same covalent intermediate (E-αCarb).
Figure 4
Figure 4. Structural characterization of covalent intermediates and Michaelis complex between GCase and carbasugars. (a) Crystal structure of the GCase-6 complex, showing two orientations of the covalent complex in which 6 adopts an envelope (2E) conformation. The maximum-likelihood σA-weighted 2Fo – Fc electron density map is contoured to 1 σ (0.37 e/Å3). (b) Crystal structure of the GCase-7 complex, showing two orientations of the covalent complex in which, the carbasugar adopts an envelope (2E) conformation, numbering of the ring starts at the covalent attachment to the enzyme. The maximum-likelihood σA-weighted 2Fo – Fc electron density map is contoured to 1 σ (0.36 e/Å3). Carbasugar average b-factor = 20 Å2. (c) Crystal structure of the GCase-5 complex, showing three orientations of l-carbasugar fluoride bound noncovalently in the active site in a half-chair (4H3) conformation. Green atom indicates fluorine. The maximum-likelihood σA-weighted 2Fo – Fc electron density map is contoured to 1 σ (0.37 e/Å3).
Figure 5
Figure 5. pH-rate profile for the logarithm of the inactivation second-order rate constants (kinact/Ki) for yeast α-glucosidase by 7. All experiments were performed at T = 37 °C in 50 mM buffer (acetic acid-sodium acetate buffer for pH 5.0–5.5, sodium phosphate buffer for pH 6.0–7.5, TAPS buffer for pH 7.9) containing 0.1% BSA. The line through the points is the best fit to a model where two protonation events (low pH) lead to an inactive enzyme, while a single deprotonation at higher pH values gives inactive enzyme.
References
This article references 46 other publications.
- 1Platt, F. M.; d’Azzo, A.; Davidson, B. L.; Neufeld, E. F.; Tifft, C. J. Lysosomal storage diseases. Nat. Rev. Dis. Primers 2018, 4, 27, DOI: 10.1038/s41572-018-0025-41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3czksl2ntg%253D%253D&md5=9a52212427e380c3dc78006560c800ceLysosomal storage diseasesPlatt Frances M; d'Azzo Alessandra; Davidson Beverly L; Davidson Beverly L; Neufeld Elizabeth F; Tifft Cynthia JNature reviews. Disease primers (2018), 4 (1), 27 ISSN:.Lysosomal storage diseases (LSDs) are a group of over 70 diseases that are characterized by lysosomal dysfunction, most of which are inherited as autosomal recessive traits. These disorders are individually rare but collectively affect 1 in 5,000 live births. LSDs typically present in infancy and childhood, although adult-onset forms also occur. Most LSDs have a progressive neurodegenerative clinical course, although symptoms in other organ systems are frequent. LSD-associated genes encode different lysosomal proteins, including lysosomal enzymes and lysosomal membrane proteins. The lysosome is the key cellular hub for macromolecule catabolism, recycling and signalling, and defects that impair any of these functions cause the accumulation of undigested or partially digested macromolecules in lysosomes (that is, 'storage') or impair the transport of molecules, which can result in cellular damage. Consequently, the cellular pathogenesis of these diseases is complex and is currently incompletely understood. Several LSDs can be treated with approved, disease-specific therapies that are mostly based on enzyme replacement. However, small-molecule therapies, including substrate reduction and chaperone therapies, have also been developed and are approved for some LSDs, whereas gene therapy and genome editing are at advanced preclinical stages and, for a few disorders, have already progressed to the clinic.
- 2Varki, A. Essentials of Glycobiology, 4th ed.; Cold Spring Harbor Laboratory Press: Cold Spring Harbor: Cold Spring Harbor, NY, 2022.There is no corresponding record for this reference.
- 3Selnick, H. G.; Hess, J. F.; Tang, C. Y.; Liu, K.; Schachter, J. B.; Ballard, J. E.; Marcus, J.; Klein, D. J.; Wang, X. H.; Pearson, M.; Savage, M. J.; Kaul, R.; Li, T. S.; Vocadlo, D. J.; Zhou, Y. X.; Zhu, Y. B.; Mu, C. W.; Wang, Y. D.; Wei, Z. Y.; Bai, C.; Duffy, J. L.; McEachern, E. J. Discovery of MK-8719, a potent O-GlcNAcase inhibitor as a potential treatment for tauopathies. J. Med. Chem. 2019, 62, 10062– 10097, DOI: 10.1021/acs.jmedchem.9b010903https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslWms73F&md5=79e26635e124c90c86d4c1beb8b63e23Discovery of MK-8719, a potent O-GlcNAcase inhibitor as a potential treatment for tauopathiesSelnick, Harold G.; Hess, J. Fred; Tang, Cuyue; Liu, Kun; Schachter, Joel B.; Ballard, Jeanine E.; Marcus, Jacob; Klein, Daniel J.; Wang, Xiaohai; Pearson, Michelle; Savage, Mary J.; Kaul, Ramesh; Li, Tong-Shuang; Vocadlo, David J.; Zhou, Yuanxi; Zhu, Yongbao; Mu, Changwei; Wang, Yaode; Wei, Zhongyong; Bai, Chang; Duffy, Joseph L.; McEachern, Ernest J.Journal of Medicinal Chemistry (2019), 62 (22), 10062-10097CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Inhibition of O-GlcNAcase (OGA) has emerged as a promising therapeutic approach to treat tau pathol. in neurodegenerative diseases such as Alzheimer's disease and progressive supranuclear palsy. Beginning with carbohydrate-based lead mols., we pursued an optimization strategy of reducing polar surface area to align the desired drug-like properties of potency, selectivity, high central nervous system (CNS) exposure, metabolic stability, favorable pharmacokinetics, and robust in vivo pharmacodynamic response. Herein, we describe the medicinal chem. and pharmacol. studies that led to the identification of (3aR,5S,6S,7R,7aR)-5-(difluoromethyl)-2-(ethylamino)-3a,6,7,7a-tetrahydro-5H-pyrano[3,2-d]thiazole-6,7-diol 42 (MK-8719), a highly potent and selective OGA inhibitor with excellent CNS penetration that has been advanced to first-in-human phase I clin. trials.
- 4den Heijer, J. M.; Kruithof, A. C.; van Amerongen, G.; de Kam, M. L.; Thijssen, E.; Grievink, H. W.; Moerland, M.; Walker, M.; Been, K.; Skerlj, R.; Justman, C.; Dudgeon, L.; Lansbury, P.; Cullen, V. C.; Hilt, D. C.; Groeneveld, G. J. A randomized single and multiple ascending dose study in healthy volunteers of LTI-291, a centrally penetrant glucocerebrosidase activator. Br. J. Clin. Pharmacol. 2021, 87, 3561– 3573, DOI: 10.1111/bcp.14772There is no corresponding record for this reference.
- 5Patterson, M. C.; Vecchio, D.; Prady, H.; Abel, L.; Wraith, J. E. Miglustat for treatment of Niemann-Pick C disease: a randomised controlled study. Lancet Neurol. 2007, 6, 765– 772, DOI: 10.1016/S1474-4422(07)70194-15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVyrsr3F&md5=d402dde48fc19092735a4844a2d874b7Miglustat for treatment of Niemann-Pick C disease: a randomised controlled studyPatterson, Marc C.; Vecchio, Darleen; Prady, Helena; Abel, Larry; Wraith, James E.Lancet Neurology (2007), 6 (9), 765-772CODEN: LNAEAM; ISSN:1474-4422. (Elsevier Ltd.)Background: Niemann-Pick type C disease (NPC) is an inherited neurodegenerative disorder characterized by an intracellular lipid-trafficking defect with secondary accumulation of glycosphingolipids. Miglustat, a small iminosugar, reversibly inhibits glucosylceramide synthase, which catalyzes the first committed step of glycosphingolipid synthesis. Miglustat is able to cross the blood-brain barrier, and is thus a potential therapy for neurol. diseases. We aimed to establish the effect of miglustat on several markers of NPC severity. Methods: Patients aged 12 years or older who had NPC (n = 29) were randomly assigned to receive either miglustat 200 mg three times a day (n = 20) or std. care (n = 9) for 12 mo. 12 Children younger than 12 years of age were included in an addnl. cohort; all received miglustat at a dose adjusted for body surface area. All participants were then treated with miglustat for an addnl. year in an extension study. The primary endpoint was horizontal saccadic eye movement (HSEM) velocity, based on its correlation with disease progression. This study is registered as an International Std. Randomised Controlled Trial, no. ISRCTN26761144. Findings: At 12 mo, HSEM velocity had improved in patients treated with miglustat vs. those receiving std. care; results were significant when patients taking benzodiazepines were excluded (p = 0·028). Children showed an improvement in HSEM velocity of similar size at 12 mo. Improvement in swallowing capacity, stable auditory acuity, and a slower deterioration in ambulatory index were also seen in treated patients older than 12 years. The safety and tolerability of miglustat 200 mg three times a day in study participants was consistent with previous trials in type I Gaucher disease, where half this dose was used. Interpretation: Miglustat improves or stabilizes several clin. relevant markers of NPC. This is the first agent studied in NPC for which there is both animal and clin. data supporting a disease modifying benefit.
- 6Hughes, D. A.; Nicholls, K.; Shankar, S. P.; Sunder-Plassmann, G.; Koeller, D.; Nedd, K.; Vockley, G.; Hamazaki, T.; Lachmann, R.; Ohashi, T. Oral pharmacological chaperone migalastat compared with enzyme replacement therapy in Fabry disease: 18-month results from the randomised phase III ATTRACT study. J. Med. Gen. 2017, 54, 288– 296, DOI: 10.1136/jmedgenet-2016-104178There is no corresponding record for this reference.
- 7Fan, J. Q.; Ishii, S.; Asano, N.; Suzuki, Y. Accelerated transport and maturation of lysosomal α-galactosidase A in Fabry lymphoblasts by an enzyme inhibitor. Nat. Med. 1999, 5, 112– 115, DOI: 10.1038/48017https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXmtF2muw%253D%253D&md5=dbc82fd0fae5c7e33f99add6f956bb98Accelerated transport and maturation of lysosomal α-galactosidase A in Fabry lymphoblasts by an enzyme inhibitorFan, Jian-Qiang; Ishii, Satoshi; Asano, Naoki; Suzuki, YoshiyukiNature Medicine (New York) (1999), 5 (1), 112-115CODEN: NAMEFI; ISSN:1078-8956. (Nature America)Fabry disease is a disorder of glycosphingolipid metab. caused by deficiency of lysosomal α-galactosidase A (α-Gal A), resulting in renal failure along with premature myocardial infarction and strokes. No effective treatment of this disorder is available at present. Studies of residual activities of mutant enzymes in many Fabry patients showed that some of them had kinetic properties similar to those for normal α-Gal A, but were significantly less stable, esp. in conditions of neutral pH. The biosynthetic processing was delayed in cultured fibroblasts of a Fabry patient, and the mutant protein formed an aggregate in endoplasmic reticulum, indicating that the enzyme deficiency in some mutants was mainly caused by abortive exit from the endoplasmic reticulum, leading to excessive degrdn. of the enzyme. We report here that 1-deoxy-galactonojirimycin (DGJ), a potent competitive inhibitor of α-Gal A, effectively enhanced α-Gal A activity in Fabry lymphoblasts, when administrated at concns. lower than that usually required for intracellular inhibition of the enzyme. DGJ seemed to accelerate transport and maturation of the mutant enzyme. Oral administration of DGJ to transgenic mice overexpressing a mutant α-Gal A substantially elevated the enzyme activity in some organs. We propose a new mol. therapeutic strategy for genetic metabolic diseases of administering competitive inhibitors as chem. chaperons' at sub-inhibitory intracellular concns.
- 8Parenti, G.; Andria, G.; Valenzano, K. J. Pharmacological chaperone therapy: Preclinical development, clinical translation, and prospects for the treatment of lysosomal storage disorders. Mol. Ther. 2015, 23, 1138– 1148, DOI: 10.1038/mt.2015.628https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1ygsb%252FO&md5=8d3c0c1dada373b9868927777d72d3cfPharmacological Chaperone Therapy: Preclinical Development, Clinical Translation, and Prospects for the Treatment of Lysosomal Storage DisordersParenti, Giancarlo; Andria, Generoso; Valenzano, Kenneth J.Molecular Therapy (2015), 23 (7), 1138-1148CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)A review. Lysosomal storage disorders (LSDs) are a group of inborn metabolic diseases caused by mutations in genes that encode proteins involved in different lysosomal functions, in most instances acidic hydrolases. Different therapeutic approaches have been developed to treat these disorders. Pharmacol. chaperone therapy (PCT) is an emerging approach based on small-mol. ligands that selectively bind and stabilize mutant enzymes, increase their cellular levels, and improve lysosomal trafficking and activity. Compared to other approaches, PCT shows advantages, particularly in terms of oral administration, broad biodistribution, and pos. impact on patients' quality of life. After preclin. in vitro and in vivo studies, PCT is now being translated in the first clin. trials, either as monotherapy or in combination with enzyme replacement therapy, for some of the most prevalent LSDs. For some LSDs, the results of the first clin. trials are encouraging and warrant further development. Future research in the field of PCT will be directed toward the identification of novel chaperones, including new allosteric drugs, and the exploitation of synergies between chaperone treatment and other therapeutic approaches.
- 9Witte, M. D.; Kallemeijn, W. W.; Aten, J.; Li, K. Y.; Strijland, A.; Donker-Koopman, W. E.; van den Nieuwendijk, A.; Bleijlevens, B.; Kramer, G.; Florea, B. I.; Hooibrink, B.; Hollak, C. E. M.; Ottenhoff, R.; Boot, R. G.; van der Marel, G. A.; Overkleeft, H. S.; Aerts, J. Ultrasensitive in situ visualization of active glucocerebrosidase molecules. Nat. Chem. Biol. 2010, 6, 907– 913, DOI: 10.1038/nchembio.4669https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVansb7N&md5=e9ead74400f958f1d4d2d5f5402d5c2fUltrasensitive in situ visualization of active glucocerebrosidase moleculesWitte, Martin D.; Kallemeijn, Wouter W.; Aten, Jan; Li, Kah-Yee; Strijland, Anneke; Donker-Koopman, Wilma E.; van den Nieuwendijk, Adrianus M. C. H.; Bleijlevens, Boris; Kramer, Gertjan; Florea, Bogdan I.; Hooibrink, Berend; Hollak, Carla E. M.; Ottenhoff, Roelof; Boot, Rolf G.; van der Marel, Gijsbert A.; Overkleeft, Herman S.; Aerts, Johannes M. F. G.Nature Chemical Biology (2010), 6 (12), 907-913CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Deficiency of glucocerebrosidase (GBA) underlies Gaucher disease, a common lysosomal storage disorder. Carriership for Gaucher disease has recently been identified as major risk for parkinsonism. Presently, no method exists to visualize active GBA mols. in situ. The authors here report the design, synthesis and application of two fluorescent activity-based probes allowing highly specific labeling of active GBA mols. in vitro and in cultured cells and mice in vivo. Detection of in vitro labeled recombinant GBA on slab gels after electrophoresis is in the low attomolar range. Using cell or tissue lysates, the authors obtained exclusive labeling of GBA mols. The authors present evidence from fluorescence-activated cell sorting anal., fluorescence microscopy and pulse-chase expts. of highly efficient labeling of GBA mols. in intact cells as well as tissues of mice. In addn., the authors illustrate the use of the fluorescent probes to study inhibitors and tentative chaperones in living cells.
- 10Scherer, M.; Santana, A. G.; Robinson, K.; Zhou, S.; Overkleeft, H. S.; Clarke, L.; Withers, S. G. Lipid-mimicking phosphorus-based glycosidase inactivators as pharmacological chaperones for the treatment of Gaucher’s disease. Chem. Sci. 2021, 12, 13909– 13913, DOI: 10.1039/D1SC03831A10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVKhsb%252FP&md5=d6e35d888c911d870832a1e2bf111db7Lipid-mimicking phosphorus-based glycosidase inactivators as pharmacological chaperones for the treatment of Gaucher's diseaseScherer, Manuel; Santana, Andres G.; Robinson, Kyle; Zhou, Steven; Overkleeft, Hermen S.; Clarke, Lorne; Withers, Stephen G.Chemical Science (2021), 12 (41), 13909-13913CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Gaucher's disease, the most prevalent lysosomal storage disorder, is caused by missense mutation of the GBA gene, ultimately resulting in deficient GCase activity, hence the excessive build-up of cellular glucosylceramide. Among different therapeutic strategies, pharmacol. chaperoning of mutant GCase represents an attractive approach that relies on small org. mols. acting as protein stabilizers. Herein, we expand upon a new class of transient GCase inactivators based on a reactive 2-deoxy-2-fluoro-β-D-glucoside tethered to an array of lipid-mimicking phosphorus-based aglycons, which not only improve the selectivity and inactivation efficiency, but also the stability of these compds. in aq. media. This hypothesis was further validated with kinetic and cellular studies confirming restoration of catalytic activity in Gaucher cells after treatment with these pharmacol. chaperones.
- 11Adams, B. T.; Niccoli, S.; Chowdhury, M. A.; Esarik, A. N. K.; Lees, S. J.; Rempel, B. P.; Phenix, C. P. N-Alkylated aziridines are easily-prepared, potent, specific and cell-permeable covalent inhibitors of human β-glucocerebrosidase. Chem. Commun. 2015, 51, 11390– 11393, DOI: 10.1039/C5CC03828F11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVCmu7nI&md5=c5f088dbe0d16ac976f5070113e6f74bN-Alkylated aziridines are easily-prepared, potent, specific and cell-permeable covalent inhibitors of human β-glucocerebrosidaseAdams, B. T.; Niccoli, S.; Chowdhury, M. A.; Esarik, A. N. K.; Lees, S. J.; Rempel, B. P.; Phenix, C. P.Chemical Communications (Cambridge, United Kingdom) (2015), 51 (57), 11390-11393CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)β-Glucocerebrosidase deficiency leads to Gaucher disease and is a potential marker of Parkinson's disease. The authors have identified N-octyl conduritol aziridine as a potent and specific covalent inactivator of GBA1 in living cells. This compd. is a promising lead towards a positron emission tomog. probe intended to image GBA1 activity.
- 12Kallemeijn, W. W.; Li, K. Y.; Witte, M. D.; Marques, A. R.; Aten, J.; Scheij, S.; Jiang, J.; Willems, L. I.; Voorn-Brouwer, T. M.; van Roomen, C. P. A. A.; Ottenhoff, R.; Boot, R. G.; van den Elst, H.; Walvoort, M. T.; Florea, B. I.; Codee, J. D.; van der Marel, G. A.; Aerts, J. M.; Overkleeft, H. S. Novel Activity-Based Probes for Broad-Spectrum Profiling of Retaining β-Exoglucosidases In Situ and In Vivo. Angew. Chem. Int. Ed., Engl. 2012, 51, 12529– 12533, DOI: 10.1002/anie.201207771There is no corresponding record for this reference.
- 13Do, J.; McKinney, C.; Sharma, P.; Sidransky, E. Glucocerebrosidase and its relevance to Parkinson disease. Mol. Neurodegener. 2019, 14, 36, DOI: 10.1186/s13024-019-0336-213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MrjtFCksQ%253D%253D&md5=7777e87956e7a0fae7f677e49267cc55Glucocerebrosidase and its relevance to Parkinson diseaseDo Jenny; McKinney Cindy; Sharma Pankaj; Sidransky EllenMolecular neurodegeneration (2019), 14 (1), 36 ISSN:.Mutations in GBA1, the gene encoding the lysosomal enzyme glucocerebrosidase, are among the most common known genetic risk factors for the development of Parkinson disease and related synucleinopathies. A great deal is known about GBA1, as mutations in GBA1 are causal for the rare autosomal storage disorder Gaucher disease. Over the past decades, significant progress has been made in understanding the genetics and cell biology of glucocerebrosidase. A least 495 different mutations, found throughout the 11 exons of the gene are reported, including both common and rare variants. Mutations in GBA1 may lead to degradation of the protein, disruptions in lysosomal targeting and diminished performance of the enzyme in the lysosome.Gaucher disease is phenotypically diverse and has both neuronopathic and non-neuronopathic forms. Both patients with Gaucher disease and heterozygous carriers are at increased risk of developing Parkinson disease and Dementia with Lewy Bodies, although our understanding of the mechanism for this association remains incomplete. There appears to be an inverse relationship between glucocerebrosidase and α-synuclein levels, and even patients with sporadic Parkinson disease have decreased glucocerebrosidase. Glucocerebrosidase may interact with α-synuclein to maintain basic cellular functions, or impaired glucocerebrosidase could contribute to Parkinson pathogenesis by disrupting lysosomal homeostasis, enhancing endoplasmic reticulum stress or contributing to mitochondrial impairment. However, the majority of patients with GBA1 mutations never develop parkinsonism, so clearly other risk factors play a role. Treatments for Gaucher disease have been developed that increase visceral glucocerebrosidase levels and decrease lipid storage, although they have yet to properly address the neurological defects associated with impaired glucocerebrosidase. Mouse and induced pluripotent stem cell derived models have improved our understanding of glucocerebrosidase function and the consequences of its deficiency. These models have been used to test novel therapies including chaperone proteins, histone deacetylase inhibitors, and gene therapy approaches that enhance glucocerebrosidase levels and could prove efficacious in the treatment of forms of parkinsonism. Consequently, this rare monogenic disorder, Gaucher disease, provides unique insights directly applicable to our understanding and treatment of Parkinson disease, a common and complex neurodegenerative disorder.
- 14Ben Bdira, F.; Kallemeijn, W. W.; Oussoren, S. V.; Scheij, S.; Bleijlevens, B.; Florea, B. I.; van Roomen, C.; Ottenhoff, R.; van Kooten, M.; Walvoort, M. T. C.; Witte, M. D.; Boot, R. G.; Ubbink, M.; Overkleeft, H. S.; Aerts, J. Stabilization of Glucocerebrosidase by Active Site Occupancy. ACS Chem. Biol. 2017, 12, 1830– 1841, DOI: 10.1021/acschembio.7b00276There is no corresponding record for this reference.
- 15Vocadlo, D. J.; Davies, G. J. Mechanistic insights into glycosidase chemistry. Curr. Opin. Chem. Biol. 2008, 12, 539– 555, DOI: 10.1016/j.cbpa.2008.05.01015https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1KgsbnF&md5=dc269026d21f0e114c25b120b3c1e9b9Mechanistic insights into glycosidase chemistryVocadlo, David J.; Davies, Gideon J.Current Opinion in Chemical Biology (2008), 12 (5), 539-555CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. The enzymic hydrolysis of the glycosidic bond continues to gain importance, reflecting the critically important roles complex glycans play in health and disease as well as the rekindled interest in enzymic biomass conversion. Recent advances include the broadening of our understanding of enzyme reaction coordinates, through both computational and structural studies, improved understanding of enzyme inhibition through transition state mimicry and fascinating insights into mechanism yielded by phys. org. chem. approaches.
- 16Adamson, C.; Pengelly, R.; Shamsi Kazem Abadi, S.; Chakladar, S.; Draper, J.; Britton, R.; Gloster, T.; Bennet, A. J. Structural snapshots for mechanism-based inactivation of a glycoside hydrolase by cyclopropyl-carbasugars. Angew. Chem., Int. Ed. 2016, 55, 14978– 14982, DOI: 10.1002/anie.201607431There is no corresponding record for this reference.
- 17Shamsi Kazem Abadi, S.; Tran, M.; Yadav, A. K.; Adabala, P. J. P.; Chakladar, S.; Bennet, A. J. New class of glycoside hydrolase mechanism-based covalent inhibitors: Glycosylation transition state conformations. J. Am. Chem. Soc. 2017, 139, 10625– 10628, DOI: 10.1021/jacs.7b0506517https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Sht7fJ&md5=67e2df7e82c721ccf61901087fb373a7New Class of Glycoside Hydrolase Mechanism-Based Covalent Inhibitors: Glycosylation Transition State ConformationsShamsi Kazem Abadi, Saeideh; Tran, Michael; Yadav, Anuj K.; Adabala, Pal John Pal; Chakladar, Saswati; Bennet, Andrew J.Journal of the American Chemical Society (2017), 139 (31), 10625-10628CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The design of covalent inhibitors in glycoscience research is important for the development of chem. biol. probes. Here we report the synthesis of a new carbocyclic mechanism-based covalent inhibitor of an α-glucosidase. The enzyme efficiently catalyzes its alkylation via either an allylic cation or a cationic transition state. We show this allylic covalent inhibitor has different catalytic proficiencies for pseudoglycosylation and deglycosylation. Such inhibitors have the potential to be useful chem. biol. tools.
- 18Danby, P. M.; Withers, S. G. Glycosyl cations versus allylic cations in spontaneous and enzymatic hydrolysis. J. Am. Chem. Soc. 2017, 139, 10629– 10632, DOI: 10.1021/jacs.7b0562818https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1WmtL%252FF&md5=b2be051f5f2a039d46667e7ef4e7c4dbGlycosyl cations versus allylic cations in spontaneous and enzymatic hydrolysisDanby, Phillip M.; Withers, Stephen G.Journal of the American Chemical Society (2017), 139 (31), 10629-10632CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Enzymic prenyl and glycosyl transfer are seemingly unrelated reactions that yield mols. and protein modifications with disparate biol. functions. However, both reactions employ diphosphate-activated donors and each proceed via cationic species: allylic cations and oxocarbenium ions, resp. Here, we explored the relation between these processes by prepg. valienyl ethers to serve as glycoside mimics that were capable of allylic rather than oxocarbenium cation stabilization. Rate consts. for the spontaneous hydrolysis of aryl glycosides and their analogous valienyl ethers were found to be almost identical, as were the corresponding activation enthalpies and entropies. This close similarity extended to the assocd. secondary kinetic isotope effects (KIEs), indicating very similar transition state stabilities and structures. Screening a library of >100 β-glucosidases identified a no. of enzymes that catalyzed the hydrolysis of these valienyl ethers with kcat values of ≤20 s-1. Detailed anal. of one such enzyme showed that ether hydrolysis occurred via the analogous mechanisms found for glycosides, and through a very similar transition state. This suggested that the generally lower rates of enzymic cleavage of the cyclitol ethers reflects evolutionary specialization of these enzymes toward glycosides rather than inherent reactivity differences.
- 19Parenti, G. Treating lysosomal storage diseases with pharmacological chaperones: from concept to clinics. EMBO Mol. Med. 2009, 1, 268– 279, DOI: 10.1002/emmm.20090003619https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1CjurrF&md5=c897e238ada0121bd4fd8d9a663aed0aTreating lysosomal storage diseases with pharmacological chaperones: from concept to clinicsParenti, GiancarloEMBO Molecular Medicine (2009), 1 (5), 268-279CODEN: EMMMAM; ISSN:1757-4684. (John Wiley & Sons Ltd.)A review. Lysosomal storage diseases (LSDs) are a group of genetic disorders due to defects in any aspect of lysosomal biol. During the past two decades, different approaches have been introduced for the treatment of these conditions. Among them, enzyme replacement therapy (ERT) represented a major advance and is used successfully in the treatment of some of these disorders. However, ERT has limitations such as insufficient biodistribution of recombinant enzymes and high costs. An emerging strategy for the treatment of LSDs is pharmacol. chaperone therapy (PCT), based on the use of chaperone mols. that assist the folding of mutated enzymes and improve their stability and lysosomal trafficking. After proof-of-concept studies, PCT is now being translated into clin. applications for Fabry, Gaucher and Pompe disease. This approach, however, can only be applied to patients carrying chaperone-responsive mutations. The recent demonstration of a synergistic effect of chaperones and ERT expands the applications of PCT and prompts a re-evaluation of their therapeutic use and potential. This review discusses the strengths and drawbacks of the potential therapies available for LSDs and proposes that future research should be directed towards the development of treatment protocols based on the combination of different therapies to improve the clin. outcome of LSD patients.
- 20Sidransky, E.; Lopez, G. The link between the GBA gene and parkinsonism. Lancet Neurol. 2012, 11, 986– 998, DOI: 10.1016/S1474-4422(12)70190-420https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFCqtbnJ&md5=522c2d24e1e4f8e826a211991fe35df8The link between the GBA gene and parkinsonismSidransky, Ellen; Lopez, GriselLancet Neurology (2012), 11 (11), 986-998CODEN: LNAEAM; ISSN:1474-4422. (Elsevier Ltd.)A review. Summary: Mutations in the glucocerebrosidase (GBA) gene, which encodes the lysosomal enzyme that is deficient in Gaucher's disease, are important and common risk factors for Parkinson's disease and related disorders. This assocn. was first recognized in the clinic, where parkinsonism was noted, albeit rarely, in patients with Gaucher's disease and more frequently in relatives who were obligate carriers. Subsequently, findings from large studies showed that patients with Parkinson's disease and assocd. Lewy body disorders had an increased frequency of GBA mutations when compared with control individuals. Patients with GBA-assocd. parkinsonism exhibit varying parkinsonian phenotypes but tend to have an earlier age of onset and more assocd. cognitive changes than patients with parkinsonism without GBA mutations. Hypotheses proposed to explain this assocn. include a gain-of-function due to mutations in glucocerebrosidase that promotes α-synuclein aggregation; substrate accumulation due to enzymic loss-of-function, which affects α-synuclein processing and clearance; and a bidirectional feedback loop. Identification of the pathol. mechanisms underlying GBA-assocd. parkinsonism will improve our understanding of the genetics, pathophysiol., and treatment for both rare and common neurol. diseases.
- 21Sinnott, M. L.; Souchard, I. J. The mechanism of action of β-galactosidase. Effect of aglycone nature and α-deuterium substitution on the hydrolysis of aryl galactosides. Biochem. J. 1973, 133, 89– 98, DOI: 10.1042/bj133008921https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE3sXks1eitrY%253D&md5=968441696aad79c9d0b7a549ed86c381Mechanism of action of β-galactosidase. Effect of aglycone nature and α-deuterium substitution on the hydrolysis of aryl galactosidesSinnott, Michael L.; Souchard, Jan J. L.Biochemical Journal (1973), 133 (1), 89-98CODEN: BIJOAK; ISSN:0264-6021.Steady-state kinetic parameters for the hydrolysis of 13 aryl β-D-galactopyranosides catalyzed by β-galactosidase (I) showed no simple dependence on aglycone acidity. The α-deuterium kinetic isotope effects (kH/kD) for 7 substrates and the effects of methanolysis of galactosyl-I in 1.5M MeOH on kH/kD were incompatible with a 2-step mechanism. A conformational change, liberation of a galactopyranosyl cation in an intimate ion-pair, nonproductive but preferential collapse of the ion pair to a covalent species, and reaction of the galactosyl-I through the ion-paired form was proposed as a reaction mechanism for β-galactosidase.
- 22Kempton, J. B.; Withers, S. G. Mechanism of Agrobacterium .beta.-glucosidase: kinetic studies. Biochemistry 1992, 31, 9961– 9969, DOI: 10.1021/bi00156a01522https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XmtVOrsbg%253D&md5=645c6f00c3d37e0b98ec1d67a8a740d4Mechanism of Agrobacterium β-glucosidase: kinetic studiesKempton, Julie B.; Withers, Stephen G.Biochemistry (1992), 31 (41), 9961-9CODEN: BICHAW; ISSN:0006-2960.β-Glucosidase from A. faecalis (previously Alcaligenes faecalis) was subjected to a detailed kinetic investigation with a range of substrates to probe its specificity and mechanism. It had a relatively broad specificity for the substrate sugar moiety and exhibited a classical pH dependence for its kinetic parameters with 3 different substrates and an identical pH dependence for its inactivation by a mechanism-based inactivator, cyclophellitol. Measurement of kcat and Km values for a series of aryl glucoside substrates allowed construction of a Broensted plot, the concave-downward shape of which was consistent with the anticipated 2-step mechanism involving a glucosyl-enzyme intermediate which is formed and hydrolyzed via oxocarbonium ion-like transition states. The slope of the leaving group-dependent portion of the Broensted plot (β1g = -0.7) indicated a large degree of bond cleavage at the transition state. Secondary deuterium kinetic isotope effects measured for 5 different aryl glucosides were also consistent with this mechanism and further suggested that the transition state for formation of the glucosyl-enzyme intermediate, probed with the slower substrates for which kH/kD = 1.06, is more SN2-like than that for its hydrolysis (for which kH/kD - 1.11). Reasons for this difference are proposed, and values of Ki for several ground-state and transition-state analog inhibitors are presented which support the concept of sp2-hybridized transition states.
- 23Breen, I. Z.; Artola, M.; Wu, L.; Beenakker, T. J. M.; Offen, W. A.; Aerts, J. M. F. G.; Davies, G. J.; Overkleeft, H. S. Competitive and covalent inhibitors of human lysosomal retaining exoglucosidases. eLS; Wiley & Sons Ltd.: Chichester, 2018.There is no corresponding record for this reference.
- 24Segel, I. H. Enzyme Kinetics: Behavior and Analysis of Rapid Equilibrium and Steady State Enzyme Systems; Wiley: New York, 1975.There is no corresponding record for this reference.
- 25Jones, C. C.; Sinnott, M. L. Leaving ability and basicity of leaving groups attached by first-row elements. J. Chem. Soc., Chem. Commun. 1977, 767– 768, DOI: 10.1039/c39770000767There is no corresponding record for this reference.
- 26Farren-Dai, M.; Sannikova, N.; Świderek, K.; Moliner, V.; Bennet, A. J. Fundamental insight into glycoside hydrolase-catalyzed hydrolysis of the universal Koshland substrates–glycopyranosyl fluorides. ACS Catal. 2021, 11, 10383– 10393, DOI: 10.1021/acscatal.1c0191826https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslWksLvL&md5=99d92a35ade39a31092971fd351c9081Fundamental Insight into Glycoside Hydrolase-Catalyzed Hydrolysis of the Universal Koshland Substrates-Glycopyranosyl FluoridesFarren-Dai, Marco; Sannikova, Natalia; Swiderek, Katarzyna; Moliner, Vicent; Bennet, Andrew J.ACS Catalysis (2021), 11 (16), 10383-10393CODEN: ACCACS; ISSN:2155-5435. (American Chemical Society)The authors measured a panel of five kinetic isotope effects [KIEs; three secondary deuterium (1-, 2-, and 5-2H) and two primary (1-13C and 5-18O) effects] for the catalyzed hydrolysis of α-D-glucopyranosyl fluoride by two inverting glycoside hydrolases (GHs). The exptl. KIEs were compared to av. values computed with multiscale QM/MM methods for the hydrolysis of α-D-glucopyranosyl fluoride promoted by an inverting α-glucosidase (Aspergillus niger, GH family 15) to give β-D-glucopyranose, which the authors explored by the generation of free energy surfaces. The authors also measured the same KIEs for catalysis of α-D-glucopyranosyl fluoride by the Trichoderma virens GH55 inverting β-glucosidase using the authors' panel of isotopologues; this reaction occurs via the "Hehre resynthesis-hydrolysis mechanism" to give, by two inversions of configuration, the hydrolysis product α-D-glucopyranose. The transition states for both enzymic reactions are essentially identical with fluoride ion departure occurring within a glycoside hydrolase active site that stabilizes pyranosylium ion-like TSs, and with catalysis driven solely by H-bonding assistance from an enzymic carboxylic acid residue. That is, no assistance is required from the bound nucleophile, which in these two cases is either a water mol. (GH15) or a sugar hydroxyl group (GH55).
- 27Boer, D. E.; Mirzaian, M.; Ferraz, M. J.; Zwiers, K. C.; Baks, M. V.; Hazeu, M. D.; Ottenhoff, R.; Marques, A. R. A.; Meijer, R.; Roos, J. C. P.; Cox, T. M.; Boot, R. G.; Pannu, N.; Overkleeft, H. S.; Artola, M.; Aerts, J. M. Human glucocerebrosidase mediates formation of xylosyl-cholesterol by β-xylosidase and transxylosidase reactions. J. Lipid Res. 2021, 62, 100018, DOI: 10.1194/jlr.RA120001043There is no corresponding record for this reference.
- 28Beauhaire, J.; Ducrot, P. H. An epoxide derived from D-glucose as the key intermediate for penaresidine and sphingolipids synthesis. Synth. Commun. 1998, 28, 2443– 2456, DOI: 10.1080/0039791980800429628https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktFCitrk%253D&md5=80b7b7320718c1c3e7e70efab3b2295fAn epoxide derived from D-glucose as the key intermediate for penaresidine and sphingolipids synthesisBeauhaire, Josiane; Ducrot, Paul-HenriSynthetic Communications (1998), 28 (13), 2443-2456CODEN: SYNCAV; ISSN:0039-7911. (Marcel Dekker, Inc.)A multigram-scale synthesis of 3R,4R,5R-3,5-dibenzyloxy-4-p-methoxybenzyl-1,2-epoxypentane and its use as intermediate for sphingolipids, penazeridine and penazetidine synthesis are described.
- 29Ley, S. V.; Baeschlin, D. K.; Dixon, D. J.; Foster, A. C.; Ince, S. J.; Priepke, W. M.; Reynolds, D. J. 1,2-Diacetals: A new opportunity for organic synthesis. Chem. Rev. 2001, 101, 53– 80, DOI: 10.1021/cr990101j29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXos1Ckt70%253D&md5=08dc53ff1be3a0f022e812efd4013d631,2-Diacetals: A New Opportunity for Organic SynthesisLey, Steven V.; Baeschlin, Daniel K.; Dixon, Darren J.; Foster, Alison C.; Ince, Stuart J.; Priepke, Henning W. M.; Reynolds, Dominic J.Chemical Reviews (Washington, D. C.) (2001), 101 (1), 53-80CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review with 127 refs. 1,2-Diacetals offer diol protection.
- 30Shih, T. L.; Liao, W. Y.; Yen, W. C. Regioselective fluorination in synthesis of deoxyfluoro quercitols from D-(−)-quinic acid. Tetrahedron 2014, 70, 9621– 9627, DOI: 10.1016/j.tet.2014.11.001There is no corresponding record for this reference.
- 31Baici, A.; Schenker, P.; Wachter, M.; Ruedi, P. 3-Fluoro-2,4-dioxa-3-phosphadecalins as inhibitors of acetylcholinesterase. A reappraisal of kinetic mechanisms and diagnostic methods. Chem. Biodivers. 2009, 6, 261– 282, DOI: 10.1002/cbdv.200800334There is no corresponding record for this reference.
- 32McCarter, J. D.; Withers, S. G. 5-Fluoro Glycosides: A New Class of Mechanism-Based Inhibitors of Both α- and β-Glucosidases. J. Am. Chem. Soc. 1996, 118, 241– 242, DOI: 10.1021/ja952732a32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XjsVKrtA%253D%253D&md5=051a0d0bd5ad03a5af007d1137be78235-Fluoro Glycosides: A New Class of Mechanism-Based Inhibitors of Both α- and β-GlucosidasesMcCarter, John D.; Withers, Stephen G.Journal of the American Chemical Society (1996), 118 (1), 241-2CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Mechanism-based inhibitors of retaining glycosidases are of considerable interest, both academically and as potential therapeutics. 5-Fluoroglycosyl fluorides I represent a new class of such inhibitors, functioning via the formation of a stabilized 5-fluoroglycosyl-enzyme intermediate that turns over only very slowly. Synthesis is achieved via radical bromination of the corresponding protected glycosyl fluoride, displacement of the bromide by fluoride and deprotonation. 5-Fluoro-β-glucosyl fluoride functions as a time-dependent inactivator of Agrobacterium faecalis β-glucosidase with a second order rate consts. for inactivation of ki/Ki = 660 min-1 mM-1. Turnover of the 5-fluoro α-glucosyl-enzyme intermediate occurs with a half life of 8.5 min. 5-Fluoro α-glucosyl fluoride functions similarly as an inactivator of yeast α-glucosidase, but inactivation is too rapid to allow detn. of kinetic parameters for the inactivation process. Turnover is also more rapid, occurring with a rate const. of 6.6 min-1. Steady state apparent Ki values of 0.3 μM and 1.4 μM are detd. for β-glucosidase and α-glucosidase resp.
- 33Ren, W.; Farren-Dai, M.; Sannikova, N.; Świderek, K.; Wang, Y.; Akintola, O.; Britton, R.; Moliner, V.; Bennet, A. J. Glycoside hydrolase stabilization of transition state charge: New directions for inhibitor design. Chem. Sci. 2020, 11, 10488– 10495, DOI: 10.1039/D0SC04401F33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVClurbP&md5=becf2cf359a256beee092f5945e647e0Glycoside hydrolase stabilization of transition state charge: new directions for inhibitor designRen, Weiwu; Farren-Dai, Marco; Sannikova, Natalia; Swiderek, Katarzyna; Wang, Yang; Akintola, Oluwafemi; Britton, Robert; Moliner, Vicent; Bennet, Andrew J.Chemical Science (2020), 11 (38), 10488-10495CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Carbasugars are structural mimics of naturally occurring carbohydrates that can interact with and inhibit enzymes involved in carbohydrate processing. In particular, carbasugars have attracted attention as inhibitors of glycoside hydrolases (GHs) and as therapeutic leads in several disease areas. However, it is unclear how the carbasugars are recognized and processed by GHs. Here, we report the synthesis of three carbasugar isotopologues and provide a detailed transition state (TS) anal. for the formation of the initial GH-carbasugar covalent intermediate, as well as for hydrolysis of this intermediate, using a combination of exptl. measured kinetic isotope effects and hybrid QM/MM calcns. We find that the α-galactosidase from Thermotoga maritima effectively stabilizes TS charge development on a remote C5-allylic center acting in concert with the reacting carbasugar, and catalysis proceeds via an exploded, or loose, SN2 transition state with no discrete enzyme-bound cationic intermediate. We conclude that, in complement to what we know about the TS structures of enzyme-natural substrate complexes, knowledge of the TS structures of enzymes reacting with non-natural carbasugar substrates shows that GHs can stabilize a wider range of pos. charged TS structures than previously thought. Furthermore, this enhanced understanding will enable the design of new carbasugar GH transition state analogs to be used as, for example, chem. biol. tools and pharmaceutical lead compds.
- 34Akintola, O.; Farren-Dai, M.; Ren, W.; Bhosale, S.; Britton, R.; Świderek, K.; Moliner, V.; Bennet, A. J. Glycoside hydrolase catalysis: Do substrates and mechanism-based covalent inhibitors react via matching transition states?. ACS Catal. 2022, 12, 14667– 14678, DOI: 10.1021/acscatal.2c04027There is no corresponding record for this reference.
- 35Rowland, R. J.; Wu, L.; Liu, F.; Davies, G. J. A baculoviral system for the production of human β-glucocerebrosidase enables atomic resolution analysis. Acta Crystallogr. Sect. D-Struct. Biol. 2020, 76, 565– 580, DOI: 10.1107/S205979832000501XThere is no corresponding record for this reference.
- 36Speciale, G.; Thompson, A. J.; Davies, G. J.; Williams, S. J. Dissecting conformational contributions to glycosidase catalysis and inhibition. Curr. Opin. Struct. Biol. 2014, 28, 1– 13, DOI: 10.1016/j.sbi.2014.06.00336https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlSqurbE&md5=80834f55b372466d07e64e1c7d7c7555Dissecting conformational contributions to glycosidase catalysis and inhibitionSpeciale, Gaetano; Thompson, Andrew J.; Davies, Gideon J.; Williams, Spencer J.Current Opinion in Structural Biology (2014), 28 (), 1-13CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Glycoside hydrolases (GHs) are classified into >100 sequence-based families. These enzymes process a wide variety of complex carbohydrates with varying stereochem. at the anomeric and other ring positions. The shapes that these sugars adopt upon binding to their cognate GHs, and the conformational changes that occur along the catalysis reaction coordinate is termed the conformational itinerary. Efforts to define the conformational itineraries of GHs have focussed upon the crit. points of the reaction: substrate-bound (Michaelis), transition state, intermediate (if relevant) and product-bound. Recent approaches to defining conformational itineraries that marry X-ray crystallog. of enzymes bound to ligands that mimic the crit. points, along with advanced computational methods and kinetic isotope effects are discussed.
- 37Matsusaka, K.; Chiba, S.; Shimomura, T. Purification and substrate specificity of brewer’s yeast .ALPHA.-glucosidase. Agric. Biol. Chem. 1977, 41, 1917– 1923, DOI: 10.1271/bbb1961.41.191737https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE1cXhvVWjtw%253D%253D&md5=b7be52fbe741da0dfd1c16676dfd13a3Purification and substrate specificity of Brewers' yeast α-glucosidaseMatsusaka, Kouetsu; Chiba, Seiya; Shimomura, TokujiAgricultural and Biological Chemistry (1977), 41 (10), 1917-23CODEN: ABCHA6; ISSN:0002-1369.Three kinds of α-glucosidase (I, II and III) were isolated from brewers' yeast. α-Glucosidases I and II were homogeneous in disc electrophoresis, but α-glucosidase III was not. Their pH optima were 6.3-7.1. Each of them was a typical α-glucosidase showing a preferential activity on Ph α-glucoside (I). α-Glucosidase I was isomaltase, which can hydrolyze isomaltose but not maltose. α-Glucosidases II and III showed no activity on isomaltose. The ratio of velocity of hydrolysis for I and isomaltose of α-glucosidase I was 100:9, and those for I and maltose of α-glucosidases II and III were 100:17 and 100:18, resp.
- 38Bordwell, F. G. Are nucleophilic bimolecular concerted reactions involving four or more bonds a myth?. Acc. Chem. Res. 1970, 3, 281– 290, DOI: 10.1021/ar50033a001There is no corresponding record for this reference.
- 39Bordwell, F. G.; Mecca, T. G. Nucleophilic substitutions in allylic systems. Further evidence against existence of concerted SN2’ mechanism. J. Am. Chem. Soc. 1972, 94, 5829– 5837, DOI: 10.1021/ja00771a048There is no corresponding record for this reference.
- 40Kantner, S. S.; Humski, K.; Goering, H. L. On the solvolysis of 2-cyclohexenyl 3,5-dinitrobenzoate and p-nitrobenzoate in aqueous acetone. Introduction of acyl-oxygen cleavage by basic buffer systems. J. Am. Chem. Soc. 1982, 104, 1693– 1697, DOI: 10.1021/ja00370a040There is no corresponding record for this reference.
- 41Streitwieser, A.; Jayasree, E. G.; Hasanayn, F.; Leung, S. S. H. A theoretical study of SN2’ reactions of allylic halides: Role of ion pairs. J. Org. Chem. 2008, 73, 9426– 9434, DOI: 10.1021/jo802074341https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlWqu7vJ&md5=5d733dd816b847c74d270de5d2131da8A Theoretical Study of SN2' Reactions of Allylic Halides: Role of Ion PairsStreitwieser, A.; Jayasree, E. G.; Hasanayn, F.; Leung, S. S.-H.Journal of Organic Chemistry (2008), 73 (23), 9426-9434CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)Various disparate exptl. results are explained by the hypothesis that reactions of anionic nucleophiles with allylic halides are generally SN2. The SN2' reactions that do occur proceed generally with anti stereochem. Reactions with ion pair nucleophiles occur preferentially as SN2' reactions with syn stereochem. This hypothesis is consistent with a variety of computations at the HF, B3LYP, mPW1PW91 and MP2 levels with the 6-31+G(d) basis set of reactions of Li and Na fluoride and chloride with allyl halides and 4-halo-2-pentenes. Solvation is considered by a combination of coordination of di-Me ether to the lithium and sodium cations and "dielec. solvation" with a polarized continuum model.
- 42Bentley, T. W.; Carter, G. E. The SN2-SN1 spectrum. 4. The SN2 (intermediate) mechanism for aolvolysis of tert-butyl chloride: A revised Y scale of solvent ionizing power based on solvolysis of 1-adamantyl chloride. J. Am. Chem. Soc. 1982, 104, 5741– 5747, DOI: 10.1021/ja00385a03142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL38Xls1ajsr8%253D&md5=354c3dde00ddb693959c9ba57876c315The SN2-SN1 spectrum. 4. The SN2 (intermediate) mechanism for solvolyses of tert-butyl chloride: a revised Y scale of solvent ionizing power based on solvolyses of 1-adamantyl chlorideBentley, T. William; Carter, Gillian E.Journal of the American Chemical Society (1982), 104 (21), 5741-7CODEN: JACSAT; ISSN:0002-7863.New kinetic data for solvolysis of 1-adamantyl chloride and bromide were detd. in several solvents. The reliability of the results is supported by the consistent activation parameters and by bromide-chloride rate ratios, which closely parallel the corresponding ratios obtained for solvolysis of tert-Bu halides. The effects of added salts on solvolyzes are given. A new scale of solvent ionizing power for chlorides (YCl) was defined as log (k/k0) = mYCl (k = solvolysis rate const. for 1-adamantyl chloride in any solvent at 25°; k0 = same for 80 vol.% aq. EtOH; m = 1); an analogous scale is defined using data for 1-adamantyl bromide. These Y values were used to correlate rate data for tert-Bu and 1-adamantyl halides.
- 43Ohga, Y.; Munakata, M.; Kitagawa, T.; Kinoshita, T.; Takeuchi, K.; Oishi, Y.; Fujimoto, H. Solvolyses of bicyclo[2.2.2]oct-1-yl and 1-adamantyl systems containing an ethylidene substituent on the 2-position: typical examples of rate enhancements ascribed to relief of F-strain. J. Org. Chem. 1994, 59, 4056– 4067, DOI: 10.1021/jo00094a012There is no corresponding record for this reference.
- 44Chan, J.; Tang, A.; Bennet, A. J. A stepwise solvent-promoted SNi reaction of α-D-glucopyranosyl fluoride: Mechanistic implications for retaining glycosyltransferases. J. Am. Chem. Soc. 2012, 134, 1212– 1220, DOI: 10.1021/ja209339j44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFylur7O&md5=719b679e96b9141d5c98b35c021afa8dA Stepwise Solvent-Promoted SNi Reaction of α-D-Glucopyranosyl Fluoride: Mechanistic Implications for Retaining GlycosyltransferasesChan, Jefferson; Tang, Ariel; Bennet, Andrew J.Journal of the American Chemical Society (2012), 134 (2), 1212-1220CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The solvolysis of α-d-glucopyranosyl fluoride in hexafluoro-2-propanol gives two products, 1,1,1,3,3,3-hexafluoropropan-2-yl α-d-glucopyranoside and 1,6-anhydro-β-d-glucopyranose. The ratio of these two products is essentially unchanged for reactions that are performed between 56 and 100 °C. The activation parameters for the solvolysis reaction are as follows: ΔH⧺ = 81.4 ± 1.7 kJ mol-1, and ΔS⧺ = -90.3 ± 4.6 J mol-1 K-1. To characterize, by use of multiple kinetic isotope effect (KIE) measurements, the TS for the solvolysis reaction in hexafluoro-2-propanol, we synthesized a series of isotopically labeled α-d-glucopyranosyl fluorides. The measured KIEs for the C1 deuterium, C2 deuterium, C5 deuterium, anomeric carbon, ring oxygen, O6, and solvent deuterium are 1.185 ± 0.006, 1.080 ± 0.010, 0.987 ± 0.007, 1.008 ± 0.007, 0.997 ± 0.006, 1.003 ± 0.007, and 1.68 ± 0.07, resp. The transition state for the solvolysis reaction was modeled computationally using the exptl. KIE values as constraints. Taken together, the reported data are consistent with the retained solvolysis product being formed in an SNi (DN⧺*ANss) reaction with a late transition state in which cleavage of the glycosidic bond is coupled to the transfer of a proton from a solvating hexafluoro-2-propanol mol. In comparison, the inverted product, 1,6-anhydro-β-d-glucopyranose, is formed by intramol. capture of a solvent-equilibrated glucopyranosylium ion, which results from dissocn. of the solvent-sepd. ion pair formed in the rate-limiting ionization reaction (DN⧺ + AN). The implications that this model reaction have for the mode of action of retaining glycosyltransferases are discussed.
- 45Sinnott, M. L.; Jencks, W. P. Solvolysis of D-glucopyranosyl derivatives in mixtures of ethanol and 2,2,2-trifluoroethanol. J. Am. Chem. Soc. 1980, 102, 2026– 2032, DOI: 10.1021/ja00526a04345https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3cXktVygu7c%253D&md5=c9c081fe94897789fa673d211344f1c7Solvolysis of D-glucopyranosyl derivatives in mixtures of ethanol and 2,2,2-trifluoroethanolSinnott, Michael L.; Jencks, William P.Journal of the American Chemical Society (1980), 102 (6), 2026-32CODEN: JACSAT; ISSN:0002-7863.The products of solvolysis of α- and β-D-glucopyranosyl fluorides, 2,4-dinitrophenyl β-D-glucopyranoside, and the trifluoromethanesulfonates of the β-D-glucopyranosyl 3-bromopyridinium and α-D-glucopyranosyl 4-methylpyridinium ions in an equimolar mixt. of ethanol and trifluoroethanol buffered with ∼2 equiv of 2,6-lutidine have been examd. by gas-liq. chromatog. of their trimethylsilyl ethers. The initial products of the solvolyses of Ph α- and β-D-glucopyranosides catalyzed by CF3SO3H in an equimolar mixt. of ethanol and trifluoroethanol, and the products of uncatalyzed solvolysis of β-D-glucopyranosyl-p-nitrophenyltriazene, have been likewise examd. The compn. of the medium for solvolysis of the glucosyl fluorides has also been systematically varied from pure ethanol to pure trifluoroethanol. The percentage of products with the same anomeric configuration as the starting material is in the range 8.1-88.5%; change of leaving group, at const. anomeric configuration, or of anomeric configuration, at const. leaving group, yields different product distributions. Therefore the transition state of the product-detg. step contains the leaving group. The preference for attack by ethanol as compared with trifluoroethanol varies from 0.9 to 20 in a way which shows no general systematic distinction between pathways for retention or inversion. The nucleophilic selectivity for retention is lowered by anionic leaving groups, esp. fluoride, which preferentially stabilize the transition state contg. trifluoroethanol by H bonding. Nucleophilic attack at the α face is preferred over nucleophilic attack at the β face, and exhibits a lower selectivity: this is ascribed to H bonding between the O atom of the 2-hydroxyl group and the hydroxyl group of the approaching alc. A model for solvolysis involving a reversibly formed ion pair or encounter complex is incompatible with the selectivities still obsd. with leaving groups less nucleophilic than the solvent components: a model involving selection between the components of a pool of solvent mols. by an irreversibly formed ion pair or encounter complex requires an implausibly large pool to explain obsd. specificities. The obsd. selectivities are a consequence of the facilitation of the departure of the leaving group by the solvent, from either side of the reaction center.
- 46Wan, Q.; Parks, J. M.; Hanson, B. L.; Fisher, S. Z.; Ostermann, A.; Schrader, T. E.; Graham, D. E.; Coates, L.; Langan, P.; Kovalevsky, A. Direct determination of protonation states and visualization of hydrogen bonding in a glycoside hydrolase with neutron crystallography. Proc. Natl. Acad. Sci. U.S.A. 2015, 112, 12384– 12389, DOI: 10.1073/pnas.150498611246https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFamsLrK&md5=783870c0386e3f3a42b3451fa7335762Direct determination of protonation states and visualization of hydrogen bonding in a glycoside hydrolase with neutron crystallographyWan, Qun; Parks, Jerry M.; Hanson, B. Leif; Fisher, Suzanne Zoe; Ostermann, Andreas; Schrader, Tobias E.; Graham, David E.; Coates, Leighton; Langan, Paul; Kovalevsky, AndreyProceedings of the National Academy of Sciences of the United States of America (2015), 112 (40), 12384-12389CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Glycoside hydrolases (GHs) apply acid/base chem. to catalyze the decompn. of complex carbohydrates. These ubiquitous enzymes accept protons from solvent and donate them to substrates at close to neutral pH by modulating the pKa values of key side-chains during catalysis. However, it is not known how the catalytic acid residue acquires a proton and transfers it efficiently to the substrate. To better understand GH chem., we used macromol. neutron crystallog. to directly det. protonation and ionization states of the active site residues of a family 11 GH, Trichoderma reesei xylanase II (XynII), at multiple pD (pD = pH + 0.4) values. The general acid (Glu residue) cycled between 2 conformations, upward and downward, but was protonated only in the downward orientation. The authors performed continuum electrostatics calcns. to est. the pKa values of the catalytic Glu residues in both the apoenzyme and substrate-bound states of XynII. The calcd. pKa of the Glu residue increased substantially when the side-chain moved down. The energy barrier required to rotate the catalytic Glu residue back to the upward conformation, where it could protonate the glycosidic O atom of the substrate, was 4.3 kcal/mol according to free energy simulations. These findings shed light on the initial stage of the glycoside hydrolysis reaction in which mol. motion enables the general acid catalyst to obtain a proton from the bulk solvent and deliver it to the glycosidic O atom.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscatal.4c04549.
General methods and reagents, eqs S1–S3, Tables S1–S3, Scheme S1, Figures S1–S78 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.