1
|
Iacono R, Paragliola FMP, Strazzulli A, Moracci M. A stable GH31 α-glucosidase as a model system for the study of mutations leading to human glycogen storage disease type II. J Enzyme Inhib Med Chem 2025; 40:2468859. [PMID: 39995088 PMCID: PMC11864002 DOI: 10.1080/14756366.2025.2468859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
GH31 glycosidases are widespread across organisms, but remarkably, less than 1% of them have been biochemically characterised to date. Among them, human lysosomal acid α-glucosidase (GAA) stands out due to its link to Pompe disease, a rare lysosomal storage disorder caused by its deficiency. This disease results in glycogen accumulation, severe cellular damage, motor impairment, and premature death. Structural and functional studies of GAA mutants are challenging due to their instability and lack of activity, hindering their expression and purification. The GH31 enzyme MalA from a hyperthermophilic archaeon is explored here as a stable homolog of GAA. MalA is highly expressible, easy to purify, and structurally characterised. The R400H mutant in MalA, corresponding to the pathogenic GAA R600H mutation, revealed here a 1200-fold drop in specificity constant and >8 °C reduction in thermal stability. We propose MalA's as a robust model for studying GAA mutations and developing therapeutic chaperones.
Collapse
Affiliation(s)
- Roberta Iacono
- Department of Biology, University of Naples “Federico II”, Naples, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | | | - Andrea Strazzulli
- Department of Biology, University of Naples “Federico II”, Naples, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- Task Force on Microbiome Studies, University of Naples “Federico II”, Naples, Italy
| | - Marco Moracci
- Department of Biology, University of Naples “Federico II”, Naples, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- Task Force on Microbiome Studies, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
2
|
Sutter PA, Dhari Z, Crocker SJ. Neuroimmunology in globoid cell leukodystrophy: A comprehensive review including treatments, models, and neuroimmune mechanisms underlying neuropathology. J Neuroimmunol 2025; 402:578573. [PMID: 40058166 DOI: 10.1016/j.jneuroim.2025.578573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 04/01/2025]
Abstract
Globoid cell leukodystrophy (GLD), or Krabbe's disease, is a fatal genetic demyelinating disease of the central nervous system (CNS) caused by loss-of-function mutations in galactosylceramidase (GALC). As a result of the loss of GALC enzymatic activity, there is an accumulation of a toxic lipid called galactosylsphingosine, or psychosine. Current treatments have focused on restoring GALC function as a means to reduce psychosine accumulation, which show promise, however, still have limited success at improving behavioral or cognitive deficits in infants with GLD. Recent studies have discovered a role for T cells in GLD, indicating that there is a previously understudied role for the adaptive immune system as a contributing factor to GLD pathophysiology. This review aims to provide a comprehensive discussion of the current field of GLD research including treatment advances and GLD pathophysiology, with a focus on the role of neuroimmunological mechanisms contributing to GLD.
Collapse
Affiliation(s)
- Pearl A Sutter
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Zaenab Dhari
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06032, USA; Mandell Center for Multiple Sclerosis, Mount Sinai Rehabilitation Hospital, Trinity Health of New England, Hartford, CT 06105, USA; Departemnt of Rehabilitative Medicine, Frank H. Netter MD School of Medicine, North Haven, CT 06473, USA
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06032, USA; Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06032, USA.
| |
Collapse
|
3
|
Katzy RE, van Neer RHP, Ferraz MJ, Nicolai K, Passioura T, Suga H, Jongkees SAK, Artola M. Development of selective nanomolar cyclic peptide ligands as GBA1 enzyme stabilisers. RSC Chem Biol 2025; 6:563-570. [PMID: 39936129 PMCID: PMC11808397 DOI: 10.1039/d4cb00218k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
The stabilisation of recombinant glycosidases by exogenous ligands, known as pharmacological chaperones or enzyme stabilisers, has recently garnered great clinical interest. This strategy can prevent enzyme degradation in the blood, reducing required dosages of recombinant enzyme and extending IV injection intervals, thereby reducing side effects, improving patient lifestyles and treatment costs. While this therapeutic approach has been successfully implemented for treating Pompe and Fabry diseases, clinical studies for Gaucher disease using chaperones alone or in combination with enzyme replacement therapy (ERT) have been limited, and no small molecule chaperones have yet been approved for this condition. Developing such therapies requires selective and effective reversible GBA1 ligands. Here, we describe the development of a new class of selective macrocyclic peptide GBA1 ligands using random nonstandard peptides integrated discovery (RaPID) technology, and demonstrate their ability to bind and stabilise rhGBA1 in plasma at nanomolar concentrations. These cyclic peptides do not inhibit endogenous GBA1 in cells due to poor cell permeability but can stabilise extracellular rhGBA1 in plasma, presenting significant potential as a combinatorial ERT-pharmacological chaperone therapy for Gaucher disease.
Collapse
Affiliation(s)
- Rebecca E Katzy
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University P.O. Box 9502 2300 RA Leiden The Netherlands
| | - Renier H P van Neer
- Department of Chemistry, Graduate School of Science, The University of Tokyo Tokyo Japan
| | - Maria J Ferraz
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University P.O. Box 9502 2300 RA Leiden The Netherlands
| | - Kim Nicolai
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University P.O. Box 9502 2300 RA Leiden The Netherlands
| | - Toby Passioura
- Department of Chemistry, Graduate School of Science, The University of Tokyo Tokyo Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo Tokyo Japan
| | - Seino A K Jongkees
- Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University Utrecht 3584 CG The Netherlands
| | - Marta Artola
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University P.O. Box 9502 2300 RA Leiden The Netherlands
| |
Collapse
|
4
|
İnci A, Dökmeci S. Extracellular chaperones in lysosomal storage diseases. Mol Genet Metab 2025; 145:109086. [PMID: 40106871 DOI: 10.1016/j.ymgme.2025.109086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Lysosomal storage disorders (LSDs) are a diverse group of inherited metabolic disorders characterized by the accumulation of undegraded substrates within lysosomes due to defective lysosomal function. Recent research has highlighted the pivotal role of extracellular chaperones in the pathophysiology of LSDs, revealing their crucial involvement in modulating disease progression. These chaperones aid in stabilizing and refolding misfolded lysosomal enzymes, enhancing their proper trafficking and function, which in turn reduces substrate accumulation. Furthermore, extracellular chaperones have emerged as promising biomarkers, with their levels in bodily fluids offering potential for disease diagnosis and monitoring. This review explores the current understanding of extracellular chaperones in the context of LSDs, examining their mechanisms of action, biomarker and therapeutic potential, and future directions in clinical application of LSDs.
Collapse
Affiliation(s)
- Aslı İnci
- Gazi University School of Medicine, Department of Pediatric Metabolism, Ankara, Turkey; Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey.
| | - Serap Dökmeci
- Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey
| |
Collapse
|
5
|
Chu W, Chen M, Lv X, Lu S, Wang C, Yin L, Qian L, Shi J. Status and frontiers of Fabre disease. Orphanet J Rare Dis 2025; 20:123. [PMID: 40075521 PMCID: PMC11905648 DOI: 10.1186/s13023-025-03646-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 03/01/2025] [Indexed: 03/14/2025] Open
Abstract
Fabry disease is characterized by an X sex chromosome gene mutation caused by α-galactosidase A deficiency, resulting in the accumulation of globotriaosylceramide and globotriaosylsphingosine in various organs, which induces end-organ lesions. In Fabry disease, enzymes with lost or decreased activity in the body are replaced by exogenous supplementation of normal-function α-galactosidase A. Currently, agalsidase α and agalsidase β are widely used for ERT therapy. However, this therapy has limitations such as high cost, short half-life, and production of neutralizing drug antibodies. The use of Migalastat as chaperone therapy has been approved in many countries, and it plays a therapeutic role by enhancing enzyme activity. However, companion therapy drugs are only suitable for patients with decreased enzyme activity, so the scope of their application is limited. In addition, there are several therapeutic drugs in development, including a new generation of ERT therapies, drugs resistant to neutralizing anti-drug antibody drugs, and substrate reduction therapy drugs. Due to the limitations of existing therapeutic drugs, researchers have begun to explore new therapeutic drugs for Fabry disease, so new pathogenic mechanisms and adjuvant therapeutic drugs have been continuously discovered, and the development of related drugs will contribute to disease control and treatment. This article summarizes the existing and potential drugs for treating Fabry disease to facilitate the selection of suitable and effective drugs for treatment.
Collapse
Affiliation(s)
- Wei Chu
- Department of Pharmacy, The First People's Hospital of Huzhou, The Directly Affiliated Hospital of Huzhou Teachers College, Huzhou, China
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Min Chen
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
- Department of Pharmacy, The First People's Hospital of Aksu District, Aksu, China
| | - Xiaoqin Lv
- Department of Drug Monitoring and Evaluation, Zhejiang Center for Drug and Cosmetic Evaluation, Hangzhou, China
| | - Sheng Lu
- Department of Pharmacy, The First People's Hospital of Huzhou, The Directly Affiliated Hospital of Huzhou Teachers College, Huzhou, China
| | - Changyan Wang
- Department of Clinical Laboratory, Huzhou Aishan Hospital of Integrated Chinese and Western Medicine, Huzhou, China
| | - Limin Yin
- Department of Pharmacy, First People's Hospital of Wenling, Wenling, China
| | - Linyan Qian
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
- Heart Center, Department of Cardiovascular Medicine, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China.
| | - Jiana Shi
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| |
Collapse
|
6
|
K M N, Karmakar S, Sahoo B, Mishrra N, Moitra P. Use of Quantum Dots as Nanotheranostic Agents: Emerging Applications in Rare Genetic Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407353. [PMID: 39828615 DOI: 10.1002/smll.202407353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Rare genetic diseases (RGDs) affect a small percentage of the global population but collectively have a substantial impact due to their diverse manifestations. Although the precise reasons behind these diseases remain unclear, roughly 80% of cases are genetically linked. Recent efforts focus on understanding pathology and developing new diagnostic and therapeutic approaches for RGDs. However, there persists a gap between fundamental research and clinical therapeutic approaches, where advancements in nanotechnology offer promising improvements. In this context, nanosized light-emitting quantum dots (QDs), ranging from 2-10 nm, are promising materials for diverse applications. Their size-tunable light emission, high quantum yield, and photostability allow for precise tracking of cargo. Additionally, QDs can be functionalized with therapeutic agents, antibodies, or peptides to target specific cellular pathways, enhancing treatment efficacy while minimizing side effects. By combining diagnostic and therapeutic capabilities in a single platform, QDs thus offer a versatile and powerful approach to tackle rare genetic disorders. Despite several reviews on various therapeutic applications of QDs, their utilization in the specific domain of RGDs is not well documented. This review highlight QDs' potential in diagnosing and treating certain RGDs and addresses the challenges limiting their application.
Collapse
Affiliation(s)
- Neethu K M
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Berhampur, Berhampur, Odisha, 760010, India
| | - Shyamal Karmakar
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Berhampur, Berhampur, Odisha, 760010, India
| | - Baishakhi Sahoo
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Berhampur, Berhampur, Odisha, 760010, India
| | - Navniet Mishrra
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Berhampur, Berhampur, Odisha, 760010, India
| | - Parikshit Moitra
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Berhampur, Berhampur, Odisha, 760010, India
| |
Collapse
|
7
|
Yin Y, Jiang J, Jin Y. A Real-World Data Analysis of Alglucosidase Alfa in the FDA Adverse Event Reporting System (FAERS) Database. Drugs R D 2025:10.1007/s40268-024-00502-5. [PMID: 39833603 DOI: 10.1007/s40268-024-00502-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Alglucosidase alfa for injection is used as an enzyme replacement therapy for the treatment of Pompe disease. The safety profile of alglucosidase alfa-associated adverse events requires a comprehensive evaluation. In this study, we aimed to identify drug safety alert signals and investigate the real-world safety of alglucosidase alfa to guide clinical decision making and optimize the risk-benefit balance. METHODS The adverse event reports from the first quarter of 2006 to the fourth quarter of 2023 were selected by exploring the Food and Drug Administration Adverse Event Reporting System (FAERS) database. The new and unexpected potential adverse event signals were detected using a disproportionality analysis, including the reporting odds ratio, the proportional reporting ratio, the Bayesian confidence propagation neural network, and the empirical Bayes geometric mean. Then, the Medical Dictionary for Regulatory Activities was used to systematically classify the results. RESULTS After analyzing 16,945,027 adverse event reports, a total of 4326 cases of adverse events related to alglucosidase alfa were identified, spanning 27 system organ classes. A total of 359 preferred terms of adverse events for glucosidase alpha were detected. Pyrexia ranked first, followed by pneumonia, dyspnea, respiratory failure, and disease progression according to occurrence frequency. The top three system organ classes were general disorders and administration-site conditions (n = 2466), respiratory, thoracic, and mediastinal disorders (n = 1749), and infections and infestations (n = 1551). In addition to adverse effects mentioned in the product label, our study also discovered rare but high signal intensity adverse events such as chronic recurrent multifocal osteomyelitis. CONCLUSIONS There are many adverse events associated with the clinical use of alglucosidase alfa, which should be closely monitored in the FAERS database. As the most effective enzyme replacement therapy for Pompe disease, it is crucial to closely monitor these adverse events. Ensuring patient safety while balancing drug effectiveness is particularly important.
Collapse
Affiliation(s)
- Yi Yin
- Department of Pediatric Intensive Care Unit, Shandong, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Ji'nan, Shandong, China.
| | - Jie Jiang
- Department of Pediatric Intensive Care Unit, Shandong, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Ji'nan, Shandong, China
| | - Youpeng Jin
- Department of Pediatric Intensive Care Unit, Shandong, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Ji'nan, Shandong, China.
| |
Collapse
|
8
|
Barroso M, Puchwein-Schwepcke A, Buettner L, Goebel I, Küchler K, Muntau AC, Delgado A, Garcia-Collazo AM, Martinell M, Barril X, Cubero E, Gersting SW. Use of the Novel Site-Directed Enzyme Enhancement Therapy (SEE-Tx) Drug Discovery Platform to Identify Pharmacological Chaperones for Glutaric Acidemia Type 1. J Med Chem 2024; 67:17087-17100. [PMID: 39312412 PMCID: PMC11472340 DOI: 10.1021/acs.jmedchem.4c00292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/30/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024]
Abstract
Allosteric regulators acting as pharmacological chaperones hold promise for innovative therapeutics since they target noncatalytic sites and stabilize the folded protein without competing with the natural substrate, resulting in a net gain of function. Exogenous allosteric regulators are typically more selective than active site inhibitors and can be more potent than competitive inhibitors when the natural substrate levels are high. To identify novel structure-targeted allosteric regulators (STARs) that bind to and stabilize the mitochondrial enzyme glutaryl-CoA dehydrogenase (GCDH), the computational site-directed enzyme enhancement therapy (SEE-Tx) technology was applied. SEE-Tx is an innovative drug discovery platform with the potential to identify drugs for treating protein misfolding disorders, such as glutaric acidemia type 1 (GA1) disease. Putative allosteric regulators were discovered using structure- and ligand-based virtual screening methods and validated using orthogonal biophysical and biochemical assays. The computational approach presented here could be used to discover allosteric regulators of other protein misfolding disorders.
Collapse
Affiliation(s)
- Madalena Barroso
- University
Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Alexandra Puchwein-Schwepcke
- Department
of Molecular Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, Munich 80337, Germany
- Department
of Pediatric Neurology and Developmental Medicine, University Children’s Hospital Basel, UKBB, Basel 4031, Switzerland
| | - Lars Buettner
- Pharmaceutical
Development Biologicals, Boehringer Ingelheim
Pharma GmbH & Co. KG, Biberach
an der Riss 88397, Germany
| | - Ingrid Goebel
- University
Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Katrin Küchler
- University
Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Ania C. Muntau
- University
Children’s Hospital, University Medical
Center Hamburg-Eppendorf, Hamburg 20246, Germany
- German
Center
for Child and Adolescent Health (DZKJ), Partner Site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Aida Delgado
- Gain
Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona 08028, Spain
| | - Ana M. Garcia-Collazo
- Gain
Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona 08028, Spain
| | - Marc Martinell
- Minoryx
Therapeutics S.L., Tecno
Campus Mataró-Maresme, Mataró, Barcelona 08302, Spain
| | - Xavier Barril
- Gain
Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona 08028, Spain
| | - Elena Cubero
- Gain
Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona 08028, Spain
| | - Søren W. Gersting
- University
Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- German
Center
for Child and Adolescent Health (DZKJ), Partner Site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
9
|
Leslie K, Berry SS, Miller GJ, Mahon CS. Sugar-Coated: Can Multivalent Glycoconjugates Improve upon Nature's Design? J Am Chem Soc 2024; 146:27215-27232. [PMID: 39340450 PMCID: PMC11467903 DOI: 10.1021/jacs.4c08818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Multivalent interactions between receptors and glycans play an important role in many different biological processes, including pathogen infection, self-recognition, and the immune response. The growth in the number of tools and techniques toward the assembly of multivalent glycoconjugates means it is possible to create synthetic systems that more and more closely resemble the diversity and complexity we observe in nature. In this Perspective we present the background to the recognition and binding enabled by multivalent interactions in nature, and discuss the strategies used to construct synthetic glycoconjugate equivalents. We highlight key discoveries and the current state of the art in their applications to glycan arrays, vaccines, and other therapeutic and diagnostic tools, with an outlook toward some areas we believe are of most interest for future work in this area.
Collapse
Affiliation(s)
- Kathryn
G. Leslie
- Department
of Chemistry, Durham University, Durham DH1 3LE, United Kingdom
| | - Sian S. Berry
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | - Gavin J. Miller
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | - Clare S. Mahon
- Department
of Chemistry, Durham University, Durham DH1 3LE, United Kingdom
| |
Collapse
|
10
|
Bhosale S, Kandalkar S, Gilormini PA, Akintola O, Rowland R, Adabala PJP, King D, Deen MC, Chen X, Davies GJ, Vocadlo DJ, Bennet AJ. Development of Tunable Mechanism-Based Carbasugar Ligands that Stabilize Glycoside Hydrolases through the Formation of Transient Covalent Intermediates. ACS Catal 2024; 14:14769-14779. [PMID: 39386917 PMCID: PMC11459473 DOI: 10.1021/acscatal.4c04549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024]
Abstract
Mutations in many members of the set of human lysosomal glycoside hydrolases cause a wide range of lysosomal storage diseases. As a result, much effort has been directed toward identifying pharmacological chaperones of these lysosomal enzymes. The majority of the candidate chaperones are active site-directed competitive iminosugar inhibitors but these have met with limited success. As a first step toward an alternative class of pharmacological chaperones we explored the potential of small molecule mechanism-based reversible covalent inhibitors to form transient enzyme-inhibitor adducts. By serial synthesis and kinetic analysis of candidate molecules, we show that rational tuning of the chemical reactivity of glucose-configured carbasugars delivers cyclohexenyl-based allylic carbasugar that react with the lysosomal enzyme β-glucocerebrosidase (GCase) to form covalent enzyme-adducts with different half-lives. X-ray structural analysis of these compounds bound noncovalently to GCase, along with the structures of the covalent adducts of compounds that reacted with the catalytic nucleophile of GCase, reveal unexpected reactivities of these compounds. Using differential scanning fluorimetry, we show that formation of a transient covalent intermediate stabilizes the folded enzyme against thermal denaturation. In addition, these covalent adducts break down to liberate the active enzyme and a product that is no longer inhibitory. We further show that the one compound, which reacts through an unprecedented SN1'-like mechanism, exhibits exceptional reactivity-illustrated by this compound also covalently labeling an α-glucosidase. We anticipate that such carbasugar-based single turnover covalent ligands may serve as pharmacological chaperones for lysosomal glycoside hydrolases and other disease-associated retaining glycosidases. The unusual reactivity of these molecules should also open the door to creation of new chemical biology probes to explore the biology of this important superfamily of glycoside hydrolases.
Collapse
Affiliation(s)
- Sandeep Bhosale
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Sachin Kandalkar
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Pierre-André Gilormini
- Department
of Molecular Biology and Biochemistry, Simon
Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Oluwafemi Akintola
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Rhianna Rowland
- Department
of Chemistry, University of York, York YO10 5DD, U.K.
| | - Pal John Pal Adabala
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Dustin King
- Department
of Molecular Biology and Biochemistry, Simon
Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Matthew C. Deen
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Xi Chen
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Gideon J. Davies
- Department
of Chemistry, University of York, York YO10 5DD, U.K.
| | - David J. Vocadlo
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department
of Molecular Biology and Biochemistry, Simon
Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Andrew J. Bennet
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
11
|
Wang YJ, Seibert H, Ahn LY, Schaffer AE, Mu TW. Pharmacological chaperones restore proteostasis of epilepsy-associated GABA A receptor variants. Pharmacol Res 2024; 208:107356. [PMID: 39216838 PMCID: PMC11457296 DOI: 10.1016/j.phrs.2024.107356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Recent advances in genetic diagnosis identified variants in genes encoding GABAA receptors as causative for genetic epilepsy. Here, we selected eight disease-associated variants in the α1 subunit of GABAA receptors causing mild to severe clinical phenotypes and showed that they are loss of function, mainly by reducing the folding and surface trafficking of the α1 protein. Furthermore, we sought client protein-specific pharmacological chaperones to restore the function of pathogenic receptors. Applications of positive allosteric modulators, including Hispidulin and TP003, increase the functional surface expression of the α1 variants. Mechanism of action study demonstrated that they enhance the folding, assembly, and trafficking and reduce the degradation of GABAA variants without activating the unfolded protein response in HEK293T cells and human iPSC-derived neurons. Since these compounds cross the blood-brain barrier, such a pharmacological chaperoning strategy holds great promise to treat genetic epilepsy in a GABAA receptor-specific manner.
Collapse
Affiliation(s)
- Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Hailey Seibert
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Lucie Y Ahn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ashleigh E Schaffer
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
12
|
Chaudhary R, Rehman M, Agarwal V, Kumar A, Kaushik AS, Srivastava S, Srivastava S, Verma R, Rajinikanth PS, Mishra V. Terra incognita of glial cell dynamics in the etiology of leukodystrophies: Broadening disease and therapeutic perspectives. Life Sci 2024; 354:122953. [PMID: 39122110 DOI: 10.1016/j.lfs.2024.122953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
13
|
Martins AC, Oshiro MY, Albericio F, de la Torre BG. Food and Drug Administration (FDA) Approvals of Biological Drugs in 2023. Biomedicines 2024; 12:1992. [PMID: 39335511 PMCID: PMC11428688 DOI: 10.3390/biomedicines12091992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
An increase in total drug (small molecules and biologics) approvals by the Food and Drug Administration (FDA) was seen in 2023 compared with the previous year. Cancer remained the disease most targeted by monoclonal antibodies (mAbs), followed by autoimmune conditions. Our data reveal the prevalence of approvals for biologics even during years when the total number of authorizations was low, such as in 2022. Over half the drugs that received the green light in 2023 benefited from expedited programs, as the incidence of many diseases increased. In addition, over half of the biologics approved received Orphan Drug Designation from the FDA. This narrative review delves into details of the most significant approvals in 2023, including mAbs, enzymes, and proteins, explaining their mechanisms of action, differences from previous drugs, placebo, and standards of care, and outcomes in clinical trials. Given the varying number of drugs authorized annually by the U.S. health authority, this review also examines the limits of external influences over the FDA's decisions and independence regarding drug approvals and withdrawals.
Collapse
Affiliation(s)
- Alexander C Martins
- School of Health Sciences, UAM, Universidade Anhembi-Morumbi, São Paulo 03101-001, Brazil
- Medical Information Department, Thermo Fisher Scientific, São Paulo 4542011, Brazil
| | - Mariana Y Oshiro
- School of Health Sciences, UAM, Universidade Anhembi-Morumbi, São Paulo 03101-001, Brazil
| | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
| | - Beatriz G de la Torre
- KRISP, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| |
Collapse
|
14
|
Pisani A, Wilson KM, Batista JL, Kantola I, Ortiz A, Politei J, Al-Shaar L, Maski M, Crespo A, Ponce E, Linhart A. Clinical outcomes in patients switching from agalsidase beta to migalastat: A Fabry Registry analysis. J Inherit Metab Dis 2024; 47:1080-1095. [PMID: 38961737 DOI: 10.1002/jimd.12773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
Fabry Registry data were analyzed among 83 agalsidase beta-treated patients with Fabry disease who switched to migalastat. Outcomes (estimated glomerular filtration rate [eGFR], urine protein-creatinine ratio [UPCR], plasma globotriaosylceramide [GL-3], plasma globotriaosylsphingosine [lyso-GL-3], interventricular septal wall thickness [IVST], left posterior wall thickness [LPWT], left ventricular mass index [LVMI]) were assessed using linear mixed models to estimate annual change over time in the pre- and postswitch periods. eGFR decreased throughout both periods (preswitch: -0.85 mL/min/1.73 m2/year; postswitch: -1.96 mL/min/1.73 m2/year; both p < 0.0001), with steeper decline postswitch (ppre/post = 0.01) in both classic and late-onset patients. UPCR increased significantly postswitch (ppre/post = 0.003) among classic patients and was stable in both periods among late-onset patients. GL-3 trajectories worsened postswitch across phenotypes (ppre/post = 0.0005 classic, 0.02 late-onset). LPWT was stable preswitch (0.07 mm/year, p = 0.25) and decreased postswitch (-0.51 mm/year, p = 0.0005; ppre/post = 0.0009), primarily among late-onset patients. IVST and LVMI slopes varied significantly by phenotype. Among classic patients, IVST and LVMI were stable and decreasing, respectively preswitch and increasing postswitch (ppre/post = 0.02 IVST, 0.01 LVMI). Among late-onset patients, IVST significantly decreased postswitch (ppre/post = 0.0003); LVMI was stable over time (ppre/post = 0.89). Ultimately, eGFR and GL-3 trajectories worsened postswitch across phenotypes, while UPCR and cardiac measures worsened among classic and stabilized/improved among late-onset patients. These findings indicate variability in long-term outcomes after switching from ERT to migalastat, underscoring the importance of careful monitoring.
Collapse
Affiliation(s)
- Antonio Pisani
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | | | - Ilkka Kantola
- Division of Medicine, Turku University Hospital, Turku University, Turku, Finland
| | - Alberto Ortiz
- Jiménez Díaz Foundation University Hospital and IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Politei
- Neurology Department, Fundación SPINE, Buenos Aires, Argentina
| | | | | | | | | | - Aleš Linhart
- Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
- General University Hospital, Prague, Czech Republic
| |
Collapse
|
15
|
Darie-Ion L, Petre BA. An update on multiplexed mass spectrometry-based lysosomal storage disease diagnosis. MASS SPECTROMETRY REVIEWS 2024; 43:1135-1149. [PMID: 37584312 DOI: 10.1002/mas.21864] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/17/2023]
Abstract
Lysosomal storage disorders (LSDs) are a type of inherited metabolic disorders in which biomolecules, accumulate as a specific substrate in lysosomes due to specific individual enzyme deficiencies. Despite the fact that LSDs are incurable, various approaches, including enzyme replacement therapy, hematopoietic stem cell transplantation, or gene therapy are now available. Therefore, a timely diagnosis is a critical initial step in patient treatment. The-state-of-the-art in LSD diagnostic uses, in the first stage, enzymatic activity determination by fluorimetry or by mass spectrometry (MS) with the aid of dry blood spots, based on different enzymatic substrate structures. Due to its sensitivity, high precision, and ability to screen for an unprecedented number of diseases in a single assay, multiplexed tandem MS-based enzyme activity assays for the screening of LSDs in newborns have recently received a lot of attention. Here, (i) we review the current approaches used for simultaneous enzymatic activity determination of LSDs in dried blood spots using multiplex-LC-MS/MS; (ii) we explore the need for designing novel enzymatic substrates that generate different enzymatic products with distinct molecular masses in multiplexed-MS studies; and (iii) we give examples of the relevance of affinity-MS technique as a basis for reversing undesirable immune-reactivity in enzyme replacement therapy.
Collapse
Affiliation(s)
- Laura Darie-Ion
- Group of Biochemistry, Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, Iaşi, Romania
| | - Brînduşa Alina Petre
- Group of Biochemistry, Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, Iaşi, Romania
- Laboratory of Proteomics, Center for Fundamental Research and Experimental Development in Translation Medicine-TRANSCEND, Regional Institute of Oncology, Iaşi, Romania
| |
Collapse
|
16
|
Beraza-Millor M, Rodríguez-Castejón J, Del Pozo-Rodríguez A, Rodríguez-Gascón A, Solinís MÁ. Systematic Review of Genetic Substrate Reduction Therapy in Lysosomal Storage Diseases: Opportunities, Challenges and Delivery Systems. BioDrugs 2024; 38:657-680. [PMID: 39177875 PMCID: PMC11358353 DOI: 10.1007/s40259-024-00674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Genetic substrate reduction therapy (gSRT), which involves the use of nucleic acids to downregulate the genes involved in the biosynthesis of storage substances, has been investigated in the treatment of lysosomal storage diseases (LSDs). OBJECTIVE To analyze the application of gSRT to the treatment of LSDs, identifying the silencing tools and delivery systems used, and the main challenges for its development and clinical translation, highlighting the contribution of nanotechnology to overcome them. METHODS A systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) reporting guidelines was performed. PubMed, Scopus, and Web of Science databases were used for searching terms related to LSDs and gene-silencing strategies and tools. RESULTS Fabry, Gaucher, and Pompe diseases and mucopolysaccharidoses I and III are the only LSDs for which gSRT has been studied, siRNA and lipid nanoparticles being the silencing strategy and the delivery system most frequently employed, respectively. Only in one recently published study was CRISPR/Cas9 applied to treat Fabry disease. Specific tissue targeting, availability of relevant cell and animal LSD models, and the rare disease condition are the main challenges with gSRT for the treatment of these diseases. Out of the 11 studies identified, only two gSRT studies were evaluated in animal models. CONCLUSIONS Nucleic acid therapies are expanding the clinical tools and therapies currently available for LSDs. Recent advances in CRISPR/Cas9 technology and the growing impact of nanotechnology are expected to boost the clinical translation of gSRT in the near future, and not only for LSDs.
Collapse
Affiliation(s)
- Marina Beraza-Millor
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Julen Rodríguez-Castejón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country, UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain.
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Spain.
| |
Collapse
|
17
|
Ketata I, Ellouz E. From pathological mechanisms in Krabbe disease to cutting-edge therapy: A comprehensive review. Neuropathology 2024; 44:255-277. [PMID: 38444347 DOI: 10.1111/neup.12967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/07/2024]
Abstract
Since its initial documentation by Knud Krabbe in 1916, numerous studies have scrutinized the characteristics of Krabbe disease (KD) until the identification of the mutation in the GALC gene. In alignment with that, we investigated the natural history of KD spanning eight decades to gain a deeper understanding of the evolutionary trajectory of its mechanisms. Through our comprehensive analysis, we unearthed additional novel elements in molecular biology involving the micropathological mechanism of the disease. This review offers an updated perspective on the metabolic disorder that defines KD. Recently, extracellular vesicles (EVs), autophagy impairment, and α-synuclein have emerged as pivotal players in the neuropathological processes. EVs might serve as a cellular mechanism to avoid or alleviate the detrimental impacts of excessive toxic psychosine levels, and extracting EVs could contribute to synapse dysfunction. Autophagy impairment was found to be independent of psychosine and reliant on AKT and B-cell lymphoma 2. Additionally, α-synuclein has been recognized for inducing cellular death and dysfunction in common biological pathways. Our objective is to assess the effectiveness of advanced therapies in addressing this particular condition. While hematopoietic stem cells have been a primary treatment, its administration proves challenging, particularly in the presymptomatic phase. In this review, we have compiled information from over 10 therapy trials, comparing them based on their benefits and disadvantage.
Collapse
Affiliation(s)
- Imen Ketata
- Neurology Department, University Hospital of Gabes, Gabes, Tunisia
- Sfax University, Sfax Faculty of Medicine, Sfax, Tunisia
| | - Emna Ellouz
- Neurology Department, University Hospital of Gabes, Gabes, Tunisia
- Sfax University, Sfax Faculty of Medicine, Sfax, Tunisia
| |
Collapse
|
18
|
Losada JC, Triana H, Vanegas E, Caro A, Rodríguez-López A, Espejo-Mojica AJ, Alméciga-Diaz CJ. Identification of Orthosteric and Allosteric Pharmacological Chaperones for Mucopolysaccharidosis Type IIIB. Chembiochem 2024; 25:e202400081. [PMID: 38830828 DOI: 10.1002/cbic.202400081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/05/2024]
Abstract
Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal inherited disease caused by mutations in gene encoding the lysosomal enzyme N-acetyl-alpha-glucosaminidase (NAGLU). These mutations result in reduced NAGLU activity, preventing it from catalyzing the hydrolysis of the glycosaminoglycan heparan sulfate (HS). There are currently no approved treatments for MPS IIIB. A novel approach in the treatment of lysosomal storage diseases is the use of pharmacological chaperones (PC). In this study, we used a drug repurposing approach to identify and characterize novel potential PCs for NAGLU enzyme. We modeled the interaction of natural and artificial substrates within the active cavity of NAGLU (orthosteric site) and predicted potential allosteric sites. We performed a virtual screening for both the orthosteric and the predicted allosteric site against a curated database of human tested molecules. Considering the binding affinity and predicted blood-brain barrier permeability and gastrointestinal absorption, we selected atovaquone and piperaquine as orthosteric and allosteric PCs. The PCs were evaluated by their capacity to bind NAGLU and the ability to restore the enzymatic activity in human MPS IIIB fibroblasts These results represent novel PCs described for MPS IIIB and demonstrate the potential to develop novel therapeutic alternatives for this and other protein deficiency diseases.
Collapse
Affiliation(s)
- Juan Camilo Losada
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Lab 305 A., Bogotá D.C., 110231, Colombia
| | - Heidy Triana
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Lab 305 A., Bogotá D.C., 110231, Colombia
| | - Egdda Vanegas
- Chemistry Department, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 52, Room 110 305 A., Bogotá D.C., 110231, Colombia
| | - Angela Caro
- Chemistry Department, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 52, Room 110 305 A., Bogotá D.C., 110231, Colombia
| | - Alexander Rodríguez-López
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Lab 305 A., Bogotá D.C., 110231, Colombia
- Dogma Biotech, Cr 13 A No. 127 A-84, Bogotá D.C., 110111, Colombia
| | - Angela Johana Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Lab 305 A., Bogotá D.C., 110231, Colombia
| | - Carlos Javier Alméciga-Diaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Lab 305 A., Bogotá D.C., 110231, Colombia
| |
Collapse
|
19
|
Monticelli M, Hay Mele B, Wright DM, Guerriero S, Andreotti G, Cubellis MV. Exploring ligand interactions with human phosphomannomutases using recombinant bacterial thermal shift assay and biochemical validation. Biochimie 2024; 222:123-131. [PMID: 38458414 DOI: 10.1016/j.biochi.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/26/2024] [Indexed: 03/10/2024]
Abstract
PMM2-CDG, a disease caused by mutations in phosphomannomutase-2, is the most common congenital disorder of glycosylation. Yet, it still lacks a cure. Targeting phosphomannomutase-2 with pharmacological chaperones or inhibiting the phosphatase activity of phosphomannomutase-1 to enhance intracellular glucose-1,6-bisphosphate have been proposed as therapeutical approaches. We used Recombinant Bacterial Thermal Shift Assay to assess the binding of a substrate analog to phosphomannomutase-2 and the specific binding to phosphomannomutase-1 of an FDA-approved drug - clodronate. We also deepened the clodronate binding by enzyme activity assays and in silico docking. Our results confirmed the selective binding of clodronate to phosphomannomutase-1 and shed light on such binding.
Collapse
Affiliation(s)
- Maria Monticelli
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078, Pozzuoli, Italy; Department of Biology, University of Napoli "Federico II", Complesso Universitario Monte Sant'Angelo, Via Cinthia, 80126, Napoli, Italy
| | - Bruno Hay Mele
- Department of Biology, University of Napoli "Federico II", Complesso Universitario Monte Sant'Angelo, Via Cinthia, 80126, Napoli, Italy
| | - Demi Marie Wright
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078, Pozzuoli, Italy; Institute of Chemistry and Biochemistry, Freie Universität Berlin, D-14195, Berlin, Germany
| | - Simone Guerriero
- Department of Biology, University of Napoli "Federico II", Complesso Universitario Monte Sant'Angelo, Via Cinthia, 80126, Napoli, Italy
| | - Giuseppina Andreotti
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078, Pozzuoli, Italy.
| | - Maria Vittoria Cubellis
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078, Pozzuoli, Italy; Department of Biology, University of Napoli "Federico II", Complesso Universitario Monte Sant'Angelo, Via Cinthia, 80126, Napoli, Italy; Stazione Zoologica "Anton Dohrn", Villa Comunale, Naples, Italy
| |
Collapse
|
20
|
Schoser B, Raben N, Varfaj F, Walzer M, Toscano A. Acid α-glucosidase (GAA) activity and glycogen content in muscle biopsy specimens of patients with Pompe disease: A systematic review. Mol Genet Metab Rep 2024; 39:101085. [PMID: 38698877 PMCID: PMC11064613 DOI: 10.1016/j.ymgmr.2024.101085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Pompe disease is a rare genetic disorder characterized by a deficiency of acid α-glucosidase (GAA), leading to the accumulation of glycogen in various tissues, especially in skeletal muscles. The disease manifests as a large spectrum of phenotypes from infantile-onset Pompe disease (IOPD) to late-onset Pompe disease (LOPD), depending on the age of symptoms onset. Quantifying GAA activity and glycogen content in skeletal muscle provides important information about the disease severity. However, the distribution of GAA and glycogen levels in skeletal muscles from healthy individuals and those impacted by Pompe disease remains poorly understood, and there is currently no universally accepted standard assay for GAA activity measurement. This systematic literature review aims to provide an overview of the available information on GAA activity and glycogen content levels in skeletal muscle biopsies from patients with Pompe disease. A structured review of PubMed and Google Scholar literature (with the latter used to check that no additional publications were identified) was conducted to identify peer-reviewed publications on glycogen storage disease type II [MeSH term] + GAA, protein human (supplementary concept), Pompe, muscle; and muscle, acid alpha-glucosidase. A limit of English language was applied. Results were grouped by methodologies used to quantify GAA activity and glycogen content in skeletal muscle. The search and selection strategy were devised and carried out in line with Preferred Reporting of Items in Systematic Reviews and Meta-Analysis guidelines and documented using a flowchart. Bibliographies of papers included in the analysis were reviewed and applicable publications not already identified in the search were included. Of the 158 articles retrieved, 24 (comprising >100 muscle biopsies from >100 patients) were included in the analysis, with four different assays. Analysis revealed that patients with IOPD exhibited markedly lower GAA activity in skeletal muscles than those with LOPD, regardless of the measurement method employed. Additionally, patients with IOPD had notably higher glycogen content levels in skeletal muscles than those with LOPD. In general, however, it was difficult to fully characterize GAA activity because of the different methods used. The findings underscore the challenges in the interpretation and comparison of the results across studies because of the substantial methodological variations. There is a need to establish standardized reference ranges of GAA activity and glycogen content in healthy individuals and in Pompe disease patients based on globally standardized methods to improve comparability and reliability in assessing this rare disease.
Collapse
Affiliation(s)
- Benedikt Schoser
- Friedrich-Baur-Institute, Department of Neurology, LMU Klinikum, Ludwig-Maximilians University, Munich, Germany
| | | | | | - Mark Walzer
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Antonio Toscano
- ERN-NMD Center of Messina for Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
21
|
Cubero E, Ruano A, Delgado A, Barril X, Morales S, Trapero A, Leoni L, Bellotto M, Maj R, Guzmán BCF, Pérez-Carmona N, Garcia-Collazo AM. Discovery of allosteric regulators with clinical potential to stabilize alpha-L-iduronidase in mucopolysaccharidosis type I. PLoS One 2024; 19:e0303789. [PMID: 38768102 PMCID: PMC11104609 DOI: 10.1371/journal.pone.0303789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is an inherited lysosomal disease caused by lowered activity of the enzyme alpha-L-iduronidase (IDUA). Current therapeutic options show limited efficacy and do not treat some important aspects of the disease. Therefore, it may be advantageous to identify strategies that could improve the efficacy of existing treatments. Pharmacological chaperones are small molecules that protect proteins from degradation, and their use in combination with enzyme replacement therapy (ERT) has been proposed as an alternative therapeutic strategy. Using the SEE-Tx® proprietary computational drug discovery platform, a new allosteric ligand binding cavity in IDUA was identified distal from the active site. Virtual high-throughput screening of approximately 5 million compounds using the SEE-Tx® docking platform identified a subset of small molecules that bound to the druggable cavity and functioned as novel allosteric chaperones of IDUA. Experimental validation by differential scanning fluorimetry showed an overall hit rate of 11.4%. Biophysical studies showed that one exemplary hit molecule GT-01803 bound to (Kd = 22 μM) and stabilized recombinant human IDUA (rhIDUA) in a dose-dependent manner. Co-administration of rhIDUA and GT-01803 increased IDUA activity in patient-derived fibroblasts. Preliminary in vivo studies have shown that GT-01803 improved the pharmacokinetic (PK) profile of rhIDUA, increasing plasma levels in a dose-dependent manner. Furthermore, GT-01803 also increased IDUA enzymatic activity in bone marrow tissue, which benefits least from standard ERT. Oral bioavailability of GT-01803 was found to be good (50%). Overall, the discovery and validation of a novel allosteric chaperone for rhIDUA presents a promising strategy to enhance the efficacy of existing treatments for MPS I. The compound's ability to increase rhIDUA activity in patient-derived fibroblasts and its good oral bioavailability underscore its potential as a potent adjunct to ERT, particularly for addressing aspects of the disease less responsive to standard treatment.
Collapse
Affiliation(s)
- Elena Cubero
- Gain Therapeutics Sucursal en España, Barcelona Science Park, Barcelona, Spain
| | - Ana Ruano
- Gain Therapeutics Sucursal en España, Barcelona Science Park, Barcelona, Spain
| | - Aida Delgado
- Gain Therapeutics Sucursal en España, Barcelona Science Park, Barcelona, Spain
| | - Xavier Barril
- Gain Therapeutics Sucursal en España, Barcelona Science Park, Barcelona, Spain
- Facultat de Farmacia, IBUB & IQTC, Universitat de Barcelona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Sara Morales
- Gain Therapeutics Sucursal en España, Barcelona Science Park, Barcelona, Spain
| | - Ana Trapero
- Gain Therapeutics Sucursal en España, Barcelona Science Park, Barcelona, Spain
| | | | | | - Roberto Maj
- GT Gain Therapeutics SA, Lugano, Switzerland
| | | | | | | |
Collapse
|
22
|
Didiasova M, Cesaro S, Feldhoff S, Bettin I, Tiegel N, Füssgen V, Bertoldi M, Tikkanen R. Functional Characterization of a Spectrum of Genetic Variants in a Family with Succinic Semialdehyde Dehydrogenase Deficiency. Int J Mol Sci 2024; 25:5237. [PMID: 38791277 PMCID: PMC11121183 DOI: 10.3390/ijms25105237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Succinic semialdehyde dehydrogenase (SSADH) is a mitochondrial enzyme involved in the catabolism of the neurotransmitter γ-amino butyric acid. Pathogenic variants in the gene encoding this enzyme cause SSADH deficiency, a developmental disease that manifests as hypotonia, autism, and epilepsy. SSADH deficiency patients usually have family-specific gene variants. Here, we describe a family exhibiting four different SSADH variants: Val90Ala, Cys93Phe, and His180Tyr/Asn255Asp (a double variant). We provide a structural and functional characterization of these variants and show that Cys93Phe and Asn255Asp are pathogenic variants that affect the stability of the SSADH protein. Due to the impairment of the cofactor NAD+ binding, these variants show a highly reduced enzyme activity. However, Val90Ala and His180Tyr exhibit normal activity and expression. The His180Tyr/Asn255Asp variant exhibits a highly reduced activity as a recombinant species, is inactive, and shows a very low expression in eukaryotic cells. A treatment with substances that support protein folding by either increasing chaperone protein expression or by chemical means did not increase the expression of the pathogenic variants of the SSADH deficiency patient. However, stabilization of the folding of pathogenic SSADH variants by other substances may provide a treatment option for this disease.
Collapse
Affiliation(s)
- Miroslava Didiasova
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, DE-35390 Giessen, Germany; (M.D.); (S.F.)
| | - Samuele Cesaro
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy; (S.C.); (I.B.); (M.B.)
| | - Simon Feldhoff
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, DE-35390 Giessen, Germany; (M.D.); (S.F.)
| | - Ilaria Bettin
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy; (S.C.); (I.B.); (M.B.)
| | - Nana Tiegel
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, DE-35390 Giessen, Germany; (M.D.); (S.F.)
| | - Vera Füssgen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, DE-35390 Giessen, Germany; (M.D.); (S.F.)
| | - Mariarita Bertoldi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy; (S.C.); (I.B.); (M.B.)
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, DE-35390 Giessen, Germany; (M.D.); (S.F.)
| |
Collapse
|
23
|
Didiasova M, Banning A, Tikkanen R. Development of precision therapies for rare inborn errors of metabolism: Functional investigations in cell culture models. J Inherit Metab Dis 2024; 47:509-516. [PMID: 37606592 DOI: 10.1002/jimd.12674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
Due to the low number of patients, rare genetic diseases are a special challenge for the development of therapies, especially for diseases that result from numerous, patient-specific pathogenic variants. Precision medicine makes use of various kinds of molecular information about a specific variant, so that the possibilities for an effective therapy based on the molecular features of the variants can be elucidated. The attention to personalized precision therapies has increased among scientists and clinicians, since the "single drug for all patients" approach does not allow the classification of individuals in subgroups according to the differences in the disease genotype or phenotype. This review article summarizes some approaches of personalized precision medicine that can be used for a cost-effective and fast development of therapies, even for single patients. We have focused on specific examples on inborn errors of metabolism, with special attention on drug repurposing. Furthermore, we provide an overview of cell culture models that are suitable for precision medicine approaches.
Collapse
Affiliation(s)
- Miroslava Didiasova
- Medical Faculty, Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Antje Banning
- Medical Faculty, Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Ritva Tikkanen
- Medical Faculty, Institute of Biochemistry, University of Giessen, Giessen, Germany
| |
Collapse
|
24
|
Alyazidi AS, Muthaffar OY, Baaishrah LS, Shawli MK, Jambi AT, Aljezani MA, Almaghrabi MA. Current Concepts in the Management of Sanfilippo Syndrome (MPS III): A Narrative Review. Cureus 2024; 16:e58023. [PMID: 38738088 PMCID: PMC11087936 DOI: 10.7759/cureus.58023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Sanfilippo syndrome is a childhood-onset (1-4 years) autosomal recessive lysosomal storage disease that presents as a neurodegenerative disease by targeting the brain and spinal cord. It is also known as mucopolysaccharidosis III. Mucopolysaccharidosis III is divided into four subtypes (A, B, C, or D). It can cause delayed speech, behavior problems, and features of autism spectrum disorder. Sanfilippo syndrome is of a higher prevalence within consanguineous families that carry its gene alteration. If both parents have a nonfunctional copy of a gene linked to this condition, their children will have a 25% (1 in 4) chance of developing the disease. In Saudi Arabia, the incidence rate is estimated at 2 per 100,000 live births. Recent research focused on promising treatment approaches, such as gene therapy, modified enzyme replacement therapy, and stem cells. These approaches work by exogenous administration of the proper version of the mutant enzyme (enzyme replacement therapy), cleaning the defective enzyme in individuals with glycolipid storage disorders (substrate reduction therapy), or using a pharmacological chaperone to target improperly folded proteins. However, there is currently no approved curative medication for Sanfilippo syndrome that can effectively halt or reverse the disorder.
Collapse
Affiliation(s)
- Anas S Alyazidi
- Pediatrics, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Osama Y Muthaffar
- Pediatrics, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Layan S Baaishrah
- Faculty of Pharmacy, King Abdulaziz University Hospital, Jeddah, SAU
| | - Mohammed K Shawli
- Medicine, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Abdulaziz T Jambi
- Medicine, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Maram A Aljezani
- Pediatric Neurology, King Abdulaziz University Hospital, Jeddah, SAU
| | | |
Collapse
|
25
|
Feng S, Rcheulishvili N, Jiang X, Zhu P, Pan X, Wei M, Wang PG, Ji Y, Papukashvili D. A review on Gaucher disease: therapeutic potential of β-glucocerebrosidase-targeted mRNA/saRNA approach. Int J Biol Sci 2024; 20:2111-2129. [PMID: 38617529 PMCID: PMC11008270 DOI: 10.7150/ijbs.87741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 03/07/2024] [Indexed: 04/16/2024] Open
Abstract
Gaucher disease (GD), a rare hereditary lysosomal storage disorder, occurs due to a deficiency in the enzyme β-glucocerebrosidase (GCase). This deficiency leads to the buildup of substrate glucosylceramide (GlcCer) in macrophages, eventually resulting in various complications. Among its three types, GD2 is particularly severe with neurological involvements. Current treatments, such as enzyme replacement therapy (ERT), are not effective for GD2 and GD3 due to their inability to cross the blood-brain barrier (BBB). Other treatment approaches, such as gene or chaperone therapies are still in experimental stages. Additionally, GD treatments are costly and can have certain side effects. The successful use of messenger RNA (mRNA)-based vaccines for COVID-19 in 2020 has sparked interest in nucleic acid-based therapies. Remarkably, mRNA technology also offers a novel approach for protein replacement purposes. Additionally, self-amplifying RNA (saRNA) technology shows promise, potentially producing more protein at lower doses. This review aims to explore the potential of a cost-effective mRNA/saRNA-based approach for GD therapy. The use of GCase-mRNA/saRNA as a protein replacement therapy could offer a new and promising direction for improving the quality of life and extending the lifespan of individuals with GD.
Collapse
Affiliation(s)
- Shunping Feng
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
| | - Nino Rcheulishvili
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | | | - Pan Zhu
- Cheerland Biomedicine, Shenzhen, China
| | - Xuehua Pan
- Shenzhen Pengbo Biotech Co. Ltd, Shenzhen, China
| | - Meilan Wei
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
| | - Peng George Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
| | - Yang Ji
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
| | - Dimitri Papukashvili
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
26
|
Mohamed FE, Al-Jasmi F. Exploring the efficacy and safety of Ambroxol in Gaucher disease: an overview of clinical studies. Front Pharmacol 2024; 15:1335058. [PMID: 38414738 PMCID: PMC10896849 DOI: 10.3389/fphar.2024.1335058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/17/2024] [Indexed: 02/29/2024] Open
Abstract
Gaucher disease (GD) is mainly caused by glucocerebrosidase (GCase) enzyme deficiency due to genetic variations in the GBA1 gene leading to the toxic accumulation of sphingolipids in various organs, which causes symptoms such as anemia, thrombocytopenia, hepatosplenomegaly, and neurological manifestations. GD is clinically classified into the non-neuronopathic type 1, and the acute and chronic neuronopathic forms, types 2 and 3, respectively. In addition to the current approved GD medications, the repurposing of Ambroxol (ABX) has emerged as a prospective enzyme enhancement therapy option showing its potential to enhance mutated GCase activity and reduce glucosylceramide accumulation in GD-affected tissues of different GBA1 genotypes. The variability in response to ABX varies across different variants, highlighting the diversity in patients' therapeutic outcomes. Its oral availability and safety profile make it an attractive option, particularly for patients with neurological manifestations. Clinical trials are essential to explore further ABX's potential as a therapeutic medication for GD to encourage pharmaceutical companies' investment in its development. This review highlights the potential of ABX as a pharmacological chaperone therapy for GD and stresses the importance of addressing response variability in clinical studies to improve the management of this rare and complex disorder.
Collapse
Affiliation(s)
- Feda E. Mohamed
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Fatma Al-Jasmi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Abu Dhabi, United Arab Emirates
- Department of Pediatrics, Tawam Hospital, Al Ain, United Arab Emirates
| |
Collapse
|
27
|
Uribe-Carretero E, Rey V, Fuentes JM, Tamargo-Gómez I. Lysosomal Dysfunction: Connecting the Dots in the Landscape of Human Diseases. BIOLOGY 2024; 13:34. [PMID: 38248465 PMCID: PMC10813815 DOI: 10.3390/biology13010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Lysosomes are the main organelles responsible for the degradation of macromolecules in eukaryotic cells. Beyond their fundamental role in degradation, lysosomes are involved in different physiological processes such as autophagy, nutrient sensing, and intracellular signaling. In some circumstances, lysosomal abnormalities underlie several human pathologies with different etiologies known as known as lysosomal storage disorders (LSDs). These disorders can result from deficiencies in primary lysosomal enzymes, dysfunction of lysosomal enzyme activators, alterations in modifiers that impact lysosomal function, or changes in membrane-associated proteins, among other factors. The clinical phenotype observed in affected patients hinges on the type and location of the accumulating substrate, influenced by genetic mutations and residual enzyme activity. In this context, the scientific community is dedicated to exploring potential therapeutic approaches, striving not only to extend lifespan but also to enhance the overall quality of life for individuals afflicted with LSDs. This review provides insights into lysosomal dysfunction from a molecular perspective, particularly in the context of human diseases, and highlights recent advancements and breakthroughs in this field.
Collapse
Affiliation(s)
- Elisabet Uribe-Carretero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (E.U.-C.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Verónica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jose Manuel Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (E.U.-C.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Isaac Tamargo-Gómez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
28
|
Tax G, Guay KP, Pantalone L, Ceci M, Soldà T, Hitchman CJ, Hill JC, Vasiljević S, Lia A, Modenutti CP, Straatman KR, Santino A, Molinari M, Zitzmann N, Hebert DN, Roversi P, Trerotola M. Rescue of secretion of rare-disease-associated misfolded mutant glycoproteins in UGGT1 knock-out mammalian cells. Traffic 2024; 25:e12927. [PMID: 38272446 PMCID: PMC10832616 DOI: 10.1111/tra.12927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/02/2023] [Accepted: 12/05/2023] [Indexed: 01/27/2024]
Abstract
Endoplasmic reticulum (ER) retention of misfolded glycoproteins is mediated by the ER-localized eukaryotic glycoprotein secretion checkpoint, UDP-glucose glycoprotein glucosyl-transferase (UGGT). The enzyme recognizes a misfolded glycoprotein and flags it for ER retention by re-glucosylating one of its N-linked glycans. In the background of a congenital mutation in a secreted glycoprotein gene, UGGT-mediated ER retention can cause rare disease, even if the mutant glycoprotein retains activity ("responsive mutant"). Using confocal laser scanning microscopy, we investigated here the subcellular localization of the human Trop-2-Q118E, E227K and L186P mutants, which cause gelatinous drop-like corneal dystrophy (GDLD). Compared with the wild-type Trop-2, which is correctly localized at the plasma membrane, these Trop-2 mutants are retained in the ER. We studied fluorescent chimeras of the Trop-2 Q118E, E227K and L186P mutants in mammalian cells harboring CRISPR/Cas9-mediated inhibition of the UGGT1 and/or UGGT2 genes. The membrane localization of the Trop-2 Q118E, E227K and L186P mutants was successfully rescued in UGGT1-/- cells. UGGT1 also efficiently reglucosylated Trop-2-Q118E-EYFP in cellula. The study supports the hypothesis that UGGT1 modulation would constitute a novel therapeutic strategy for the treatment of pathological conditions associated to misfolded membrane glycoproteins (whenever the mutation impairs but does not abrogate function), and it encourages the testing of modulators of ER glycoprotein folding quality control as broad-spectrum rescue-of-secretion drugs in rare diseases caused by responsive secreted glycoprotein mutants.
Collapse
Affiliation(s)
- Gabor Tax
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Kevin P. Guay
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Ludovica Pantalone
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Italy
| | - Martina Ceci
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Italy
| | - Tatiana Soldà
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, UniversitàdellaSvizzeraItaliana (USI), Bellinzona, Switzerland
| | - Charlie J. Hitchman
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Johan C. Hill
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Snežana Vasiljević
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Andrea Lia
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Carlos P. Modenutti
- Departamento de QuímicaBiológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de QuímicaBiológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| | - Kees R. Straatman
- Core Biotechnology Services, University of Leicester, University Road, Leicester LE1 7RH, England, United Kingdom
| | - Angelo Santino
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Maurizio Molinari
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicole Zitzmann
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Daniel N. Hebert
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Pietro Roversi
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of AgriculturalBiology and Biotecnology, IBBA-CNR Unit of Milano, via Bassini 15, I-20133 Milano, Italy
| | - Marco Trerotola
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Italy
| |
Collapse
|
29
|
Anding A, Kinton S, Baranowski K, Brezzani A, De Busser H, Dufault MR, Finn P, Keefe K, Tetrault T, Li Y, Qiu W, Raes K, Vitse O, Zhang M, Ziegler R, Sardi SP, Hunter B, George K. Increasing Enzyme Mannose-6-Phosphate Levels but Not Miglustat Coadministration Enhances the Efficacy of Enzyme Replacement Therapy in Pompe Mice. J Pharmacol Exp Ther 2023; 387:188-203. [PMID: 37679046 DOI: 10.1124/jpet.123.001593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/17/2023] [Accepted: 06/01/2023] [Indexed: 09/09/2023] Open
Abstract
Pompe disease is a rare glycogen storage disorder caused by a deficiency in the lysosomal enzyme acid α-glucosidase, which leads to muscle weakness, cardiac and respiratory failure, and early mortality. Alglucosidase alfa, a recombinant human acid α-glucosidase, was the first approved treatment of Pompe disease, but its uptake into skeletal muscle via the cation-independent mannose-6-phosphate (M6P) receptor (CIMPR) is limited. Avalglucosidase alfa has received marketing authorization in several countries for infantile-onset and/or late-onset Pompe disease. This recently approved enzyme replacement therapy (ERT) was glycoengineered to maximize CIMPR binding through high-affinity interactions with ∼7 bis-M6P moieties. Recently, small molecules like the glucosylceramide synthase inhibitor miglustat were reported to increase the stability of recombinant human acid α-glucosidase, and it was suggested that an increased serum half-life would result in better glycogen clearance. Here, the effects of miglustat on alglucosidase alfa and avalglucosidase alfa stability, activity, and efficacy in Pompe mice were evaluated. Although miglustat increased the stability of both enzymes in fluorescent protein thermal shift assays and when incubated in neutral pH buffer over time, it reduced their enzymatic activity by ∼50%. Improvement in tissue glycogen clearance and transcriptional dysregulation in Pompe mice correlated with M6P levels but not with miglustat coadministration. These results further substantiate the crucial role of CIMPR binding in lysosomal targeting of ERTs. SIGNIFICANCE STATEMENT: This work describes important new insights into the treatment of Pompe disease using currently approved enzyme replacement therapies (ERTs) coadministered with miglustat. Although miglustat increased the stability of ERTs in vitro, there was no positive impact to glycogen clearance and transcriptional correction in Pompe mice. However, increasing mannose-6-phosphate levels resulted in increased cell uptake in vitro and increased glycogen clearance and transcriptional correction in Pompe mice, further underscoring the crucial role of cation-independent mannose-6-phosphate receptor-mediated lysosomal targeting for ERTs.
Collapse
Affiliation(s)
- Allyson Anding
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Sofia Kinton
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Kaitlyn Baranowski
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Alexander Brezzani
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Hilde De Busser
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Michael R Dufault
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Patrick Finn
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Kelly Keefe
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Tanya Tetrault
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Yi Li
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Weiliang Qiu
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Katrien Raes
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Olivier Vitse
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Mindy Zhang
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Robin Ziegler
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - S Pablo Sardi
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Bridge Hunter
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Kelly George
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| |
Collapse
|
30
|
Kim Y, Li H, Choi J, Boo J, Jo H, Hyun JY, Shin I. Glycosidase-targeting small molecules for biological and therapeutic applications. Chem Soc Rev 2023; 52:7036-7070. [PMID: 37671645 DOI: 10.1039/d3cs00032j] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Glycosidases are ubiquitous enzymes that catalyze the hydrolysis of glycosidic linkages in oligosaccharides and glycoconjugates. These enzymes play a vital role in a wide variety of biological events, such as digestion of nutritional carbohydrates, lysosomal catabolism of glycoconjugates, and posttranslational modifications of glycoproteins. Abnormal glycosidase activities are associated with a variety of diseases, particularly cancer and lysosomal storage disorders. Owing to the physiological and pathological significance of glycosidases, the development of small molecules that target these enzymes is an active area in glycoscience and medicinal chemistry. Research efforts carried out thus far have led to the discovery of numerous glycosidase-targeting small molecules that have been utilized to elucidate biological processes as well as to develop effective chemotherapeutic agents. In this review, we describe the results of research studies reported since 2018, giving particular emphasis to the use of fluorescent probes for detection and imaging of glycosidases, activity-based probes for covalent labelling of these enzymes, glycosidase inhibitors, and glycosidase-activatable prodrugs.
Collapse
Affiliation(s)
- Yujun Kim
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Hui Li
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Joohee Choi
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Jihyeon Boo
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Hyemi Jo
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| | - Ji Young Hyun
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| | - Injae Shin
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| |
Collapse
|
31
|
Keyzor I, Shohet S, Castelli J, Sitaraman S, Veleva-Rotse B, Weimer JM, Fox B, Willer T, Tuske S, Crathorne L, Belzar KJ. Therapeutic Role of Pharmacological Chaperones in Lysosomal Storage Disorders: A Review of the Evidence and Informed Approach to Reclassification. Biomolecules 2023; 13:1227. [PMID: 37627292 PMCID: PMC10452329 DOI: 10.3390/biom13081227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
The treatment landscape for lysosomal storage disorders (LSDs) is rapidly evolving. An increase in the number of preclinical and clinical studies in the last decade has demonstrated that pharmacological chaperones are a feasible alternative to enzyme replacement therapy (ERT) for individuals with LSDs. A systematic search was performed to retrieve and critically assess the evidence from preclinical and clinical applications of pharmacological chaperones in the treatment of LSDs and to elucidate the mechanisms by which they could be effective in clinical practice. Publications were screened according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) reporting guidelines. Fifty-two articles evaluating 12 small molecules for the treatment of seven LSDs are included in this review. Overall, a substantial amount of preclinical and clinical data support the potential of pharmacological chaperones as treatments for Fabry disease, Gaucher disease, and Pompe disease. Most of the available clinical evidence evaluated migalastat for the treatment of Fabry disease. There was a lack of consistency in the terminology used to describe pharmacological chaperones in the literature. Therefore, the new small molecule chaperone (SMC) classification system is proposed to inform a standardized approach for new, emerging small molecule therapies in LSDs.
Collapse
Affiliation(s)
- Ian Keyzor
- Amicus Therapeutics Ltd., Marlow SL7 1HZ, UK
| | | | | | | | | | | | - Brian Fox
- Amicus Therapeutics Inc., Princeton, NJ 08542, USA
| | - Tobias Willer
- Amicus Therapeutics Inc., Philadelphia, PA 19104, USA
| | - Steve Tuske
- Amicus Therapeutics Inc., Philadelphia, PA 19104, USA
| | - Louise Crathorne
- Prescript Communications Ltd., Letchworth Garden City SG6 3TA, UK
| | - Klara J. Belzar
- Prescript Communications Ltd., Letchworth Garden City SG6 3TA, UK
| |
Collapse
|
32
|
Monda E, Bakalakos A, Rubino M, Verrillo F, Diana G, De Michele G, Altobelli I, Lioncino M, Perna A, Falco L, Palmiero G, Elliott PM, Limongelli G. Targeted Therapies in Pediatric and Adult Patients With Hypertrophic Heart Disease: From Molecular Pathophysiology to Personalized Medicine. Circ Heart Fail 2023; 16:e010687. [PMID: 37477018 DOI: 10.1161/circheartfailure.123.010687] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/06/2023] [Indexed: 07/22/2023]
Abstract
Hypertrophic cardiomyopathy is a myocardial disease defined by an increased left ventricular wall thickness not solely explained by abnormal loading conditions. It is often genetically determined, with sarcomeric gene mutations accounting for around 50% of cases. Several conditions, including syndromic, metabolic, infiltrative, and neuromuscular diseases, may present with left ventricular hypertrophy, mimicking the hypertrophic cardiomyopathy phenotype but showing a different pathophysiology, clinical course, and outcome. Despite being rare, they are collectively responsible for a large proportion of patients presenting with hypertrophic heart disease, and their timely diagnosis can significantly impact patients' management. The understanding of disease pathophysiology has advanced over the last few years, and several therapeutic targets have been identified, leading to a new era of tailored treatments applying to different etiologies associated with left ventricular hypertrophy. This review aims to provide an overview of the existing and emerging therapies for the principal causes of hypertrophic heart disease, discussing the potential impact on patients' management and clinical outcome.
Collapse
Affiliation(s)
- Emanuele Monda
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
- Institute of Cardiovascular Sciences, University College London, United Kingdom (E.M., A.B., P.M.E., G.L.)
| | - Athanasios Bakalakos
- Institute of Cardiovascular Sciences, University College London, United Kingdom (E.M., A.B., P.M.E., G.L.)
| | - Marta Rubino
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Federica Verrillo
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Gaetano Diana
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Gianantonio De Michele
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Ippolita Altobelli
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Michele Lioncino
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Alessia Perna
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Luigi Falco
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Giuseppe Palmiero
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
| | - Perry M Elliott
- Institute of Cardiovascular Sciences, University College London, United Kingdom (E.M., A.B., P.M.E., G.L.)
| | - Giuseppe Limongelli
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy (E.M., M.R., F.V., G.D., G.D.M., I.A., M.L., A.P., L.F., G.P., G.L.)
- Institute of Cardiovascular Sciences, University College London, United Kingdom (E.M., A.B., P.M.E., G.L.)
| |
Collapse
|
33
|
Tax G, Guay KP, Soldà T, Hitchman CJ, Hill JC, Vasiljević S, Lia A, Modenutti CP, Straatman KR, Santino A, Molinari M, Zitzmann N, Hebert DN, Roversi P, Trerotola M. Rescue of secretion of a rare-disease associated mis-folded mutant glycoprotein in UGGT1 knock-out mammalian cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542711. [PMID: 37398215 PMCID: PMC10312515 DOI: 10.1101/2023.05.30.542711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Endoplasmic reticulum (ER) retention of mis-folded glycoproteins is mediated by the ERlocalised eukaryotic glycoprotein secretion checkpoint, UDP-glucose glycoprotein glucosyl-transferase (UGGT). The enzyme recognises a mis-folded glycoprotein and flags it for ER retention by reglucosylating one of its N-linked glycans. In the background of a congenital mutation in a secreted glycoprotein gene, UGGT-mediated ER retention can cause rare disease even if the mutant glycoprotein retains activity ("responsive mutant"). Here, we investigated the subcellular localisation of the human Trop-2 Q118E variant, which causes gelatinous droplike corneal dystrophy (GDLD). Compared with the wild type Trop-2, which is correctly localised at the plasma membrane, the Trop-2-Q118E variant is found to be heavily retained in the ER. Using Trop-2-Q118E, we tested UGGT modulation as a rescue-of-secretion therapeutic strategy for congenital rare disease caused by responsive mutations in genes encoding secreted glycoproteins. We investigated secretion of a EYFP-fusion of Trop-2-Q118E by confocal laser scanning microscopy. As a limiting case of UGGT inhibition, mammalian cells harbouring CRISPR/Cas9-mediated inhibition of the UGGT1 and/or UGGT2 gene expressions were used. The membrane localisation of the Trop-2-Q118E-EYFP mutant was successfully rescued in UGGT1-/- and UGGT1/2-/- cells. UGGT1 also efficiently reglucosylated Trop-2-Q118E-EYFP in cellula. The study supports the hypothesis that UGGT1 modulation constitutes a novel therapeutic strategy for the treatment of Trop-2-Q118E associated GDLD, and it encourages the testing of modulators of ER glycoprotein folding Quality Control (ERQC) as broad-spectrum rescueof-secretion drugs in rare diseases caused by responsive secreted glycoprotein mutants.
Collapse
Affiliation(s)
- Gábor Tax
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Kevin P. Guay
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Tatiana Soldà
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Charlie J. Hitchman
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
| | - Johan C. Hill
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Snežana Vasiljević
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Andrea Lia
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Carlos P. Modenutti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| | - Kees R. Straatman
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Core Biotechnology Services, University of Leicester, University Road, Leicester LE1 7RH, England, United Kingdom
| | - Angelo Santino
- Institute of Sciences of Food Production, ISPA-CNR Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Maurizio Molinari
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicole Zitzmann
- Institute of Glycobiology, Department of Biochemistry, South Parks Road, Oxford OX1 3RQ, United Kingdom
| | - Daniel N. Hebert
- Department of Biochemistry and Molecular Biology, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, United States
| | - Pietro Roversi
- Leicester Institute of Chemical and Structural Biology and Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7HR, England, United Kingdom
- Institute of Agricultural Biology and Biotecnology, IBBACNR Unit of Milano, via Bassini 15, I-20133 Milano, Italy
| | - Marco Trerotola
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Italy; Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, Italy
| |
Collapse
|
34
|
Wang YJ, Seibert H, Ahn LY, Schaffer AE, Mu TW. Pharmacological chaperones restore proteostasis of epilepsy-associated GABA A receptor variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537383. [PMID: 37131660 PMCID: PMC10153171 DOI: 10.1101/2023.04.18.537383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Recent advances in genetic diagnosis identified variants in genes encoding GABAA receptors as causative for genetic epilepsy. Here, we selected eight disease-associated variants in the α 1 subunit of GABAA receptors causing mild to severe clinical phenotypes and showed that they are loss of function, mainly by reducing the folding and surface trafficking of the α 1 protein. Furthermore, we sought client protein-specific pharmacological chaperones to restore the function of pathogenic receptors. Applications of positive allosteric modulators, including Hispidulin and TP003, increase the functional surface expression of the α 1 variants. Mechanism of action study demonstrated that they enhance the folding and assembly and reduce the degradation of GABAA variants without activating the unfolded protein response in HEK293T cells and human iPSC-derived neurons. Since these compounds cross the blood-brain barrier, such a pharmacological chaperoning strategy holds great promise to treat genetic epilepsy in a GABAA receptor-specific manner.
Collapse
Affiliation(s)
- Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Hailey Seibert
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Lucie Y. Ahn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Ashleigh E. Schaffer
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| |
Collapse
|
35
|
Gehrlein A, Udayar V, Anastasi N, Morella ML, Ruf I, Brugger D, von der Mark S, Thoma R, Rufer A, Heer D, Pfahler N, Jochner A, Niewoehner J, Wolf L, Fueth M, Ebeling M, Villaseñor R, Zhu Y, Deen MC, Shan X, Ehsaei Z, Taylor V, Sidransky E, Vocadlo DJ, Freskgård PO, Jagasia R. Targeting neuronal lysosomal dysfunction caused by β-glucocerebrosidase deficiency with an enzyme-based brain shuttle construct. Nat Commun 2023; 14:2057. [PMID: 37045813 PMCID: PMC10097658 DOI: 10.1038/s41467-023-37632-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
Mutations in glucocerebrosidase cause the lysosomal storage disorder Gaucher's disease and are the most common risk factor for Parkinson's disease. Therapies to restore the enzyme's function in the brain hold great promise for treating the neurological implications. Thus, we developed blood-brain barrier penetrant therapeutic molecules by fusing transferrin receptor-binding moieties to β-glucocerebrosidase (referred to as GCase-BS). We demonstrate that these fusion proteins show significantly increased uptake and lysosomal efficiency compared to the enzyme alone. In a cellular disease model, GCase-BS rapidly rescues the lysosomal proteome and lipid accumulations beyond known substrates. In a mouse disease model, intravenous injection of GCase-BS leads to a sustained reduction of glucosylsphingosine and can lower neurofilament-light chain plasma levels. Collectively, these findings demonstrate the potential of GCase-BS for treating GBA1-associated lysosomal dysfunction, provide insight into candidate biomarkers, and may ultimately open a promising treatment paradigm for lysosomal storage diseases extending beyond the central nervous system.
Collapse
Affiliation(s)
- Alexandra Gehrlein
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Vinod Udayar
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Nadia Anastasi
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Martino L Morella
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Department of Anatomy and Neurosciences, Amsterdam University Medical Center | VUmc, Amsterdam, Netherlands
| | - Iris Ruf
- Roche Pharma Research and Early Development, Therapeutic Modalities, Lead Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Doris Brugger
- Roche Pharma Research and Early Development, Therapeutic Modalities, Lead Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Sophia von der Mark
- Roche Pharma Research and Early Development, Therapeutic Modalities, Lead Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Ralf Thoma
- Roche Pharma Research and Early Development, Therapeutic Modalities, Lead Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Arne Rufer
- Roche Pharma Research and Early Development, Therapeutic Modalities, Lead Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Dominik Heer
- Roche Pharma Research and Early Development, Therapeutic Modalities, Lead Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Nina Pfahler
- Roche Pharma Research and Early Development, Therapeutic Modalities, Lead Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Interfaculty Institute of Biochemistry & Structural Biology Biochemistry (IFIB), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Anton Jochner
- Roche Pharma Research and Early Development, Therapeutic Modalities Large Molecule Research, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Jens Niewoehner
- Roche Pharma Research and Early Development, Therapeutic Modalities Large Molecule Research, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Luise Wolf
- Roche Pharma Research and Early Development, Data & Analytics, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Matthias Fueth
- Roche Pharma Research and Early Development, Pharmaceutical Science, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Martin Ebeling
- Roche Pharma Research and Early Development, Pharmaceutical Science, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Roberto Villaseñor
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yanping Zhu
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Matthew C Deen
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Xiaoyang Shan
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Zahra Ehsaei
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ellen Sidransky
- Molecular Neurogenetics Section, National Human Genome Research Institute, Bethesda, MD, USA
| | - David J Vocadlo
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Per-Ola Freskgård
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- BioArctic AB, Stockholm, Sweden
| | - Ravi Jagasia
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
36
|
Grasso D, Galderisi S, Santucci A, Bernini A. Pharmacological Chaperones and Protein Conformational Diseases: Approaches of Computational Structural Biology. Int J Mol Sci 2023; 24:ijms24065819. [PMID: 36982893 PMCID: PMC10054308 DOI: 10.3390/ijms24065819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/09/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Whenever a protein fails to fold into its native structure, a profound detrimental effect is likely to occur, and a disease is often developed. Protein conformational disorders arise when proteins adopt abnormal conformations due to a pathological gene variant that turns into gain/loss of function or improper localization/degradation. Pharmacological chaperones are small molecules restoring the correct folding of a protein suitable for treating conformational diseases. Small molecules like these bind poorly folded proteins similarly to physiological chaperones, bridging non-covalent interactions (hydrogen bonds, electrostatic interactions, and van der Waals contacts) loosened or lost due to mutations. Pharmacological chaperone development involves, among other things, structural biology investigation of the target protein and its misfolding and refolding. Such research can take advantage of computational methods at many stages. Here, we present an up-to-date review of the computational structural biology tools and approaches regarding protein stability evaluation, binding pocket discovery and druggability, drug repurposing, and virtual ligand screening. The tools are presented as organized in an ideal workflow oriented at pharmacological chaperones' rational design, also with the treatment of rare diseases in mind.
Collapse
Affiliation(s)
- Daniela Grasso
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Silvia Galderisi
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Andrea Bernini
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, 53100 Siena, Italy
| |
Collapse
|
37
|
Critchley BJ, Gaspar HB, Benedetti S. Targeting the central nervous system in lysosomal storage diseases: Strategies to deliver therapeutics across the blood-brain barrier. Mol Ther 2023; 31:657-675. [PMID: 36457248 PMCID: PMC10014236 DOI: 10.1016/j.ymthe.2022.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are multisystem inherited metabolic disorders caused by dysfunctional lysosomal activity, resulting in the accumulation of undegraded macromolecules in a variety of organs/tissues, including the central nervous system (CNS). Treatments include enzyme replacement therapy, stem/progenitor cell transplantation, and in vivo gene therapy. However, these treatments are not fully effective in treating the CNS as neither enzymes, stem cells, nor viral vectors efficiently cross the blood-brain barrier. Here, we review the latest advancements in improving delivery of different therapeutic agents to the CNS and comment upon outstanding questions in the field of neurological LSDs.
Collapse
Affiliation(s)
- Bethan J Critchley
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK
| | - H Bobby Gaspar
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; Orchard Therapeutics Ltd., London EC4N 6EU, UK
| | - Sara Benedetti
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London WC1N 1DZ, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
38
|
Monteiro C, Mesgarzadeh JS, Anselmo J, Fernandes J, Novais M, Rodrigues C, Powers DL, Powers ET, Coelho T, Kelly JW. Tafamidis polyneuropathy amelioration requires modest increases in transthyretin stability even though increases in plasma native TTR and decreases in non-native TTR do not predict response. Amyloid 2023; 30:81-95. [PMID: 36178172 PMCID: PMC9992127 DOI: 10.1080/13506129.2022.2126308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/23/2022] [Accepted: 09/13/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND TTR aggregation causes hereditary transthyretin (TTR) polyneuropathy (ATTRv-PN) in individuals with destabilised TTR variants. ATTRv-PN can be treated with ligands that bind TTR and prevent aggregation. One such ligand, tafamidis, is widely approved to treat ATTRv-PN. We explore how TTR stabilisation markers relate to clinical efficacy in 210 ATTRv-PN patients taking tafamidis. METHODS TTR concentration in patient plasma was measured before and after tafamidis treatment using assays for native or combined native + non-native TTR. TTR tetramer dissociation kinetics, which are slowed by tafamidis binding, were also measured. RESULTS Native TTR levels increased by 56.8% while combined native + non-native TTR levels increased by 3.1% after 24 months of tafamidis treatment, implying that non-native TTR decreased. Accordingly, the fraction of native TTR increased from 0.54 to 0.71 with tafamidis administration. Changes in native and non-native TTR levels were uncorrelated with clinical response to tafamidis. TTR tetramer dissociation generally slowed to an extent consistent with ∼40% of TTR being tafamidis-bound. Male non-responders had a lower extent of binding. CONCLUSIONS Native and non-native TTR concentration changes cannot be used as surrogate measures for therapeutic efficacy. Also, successful tafamidis therapy requires only moderate TTR stabilisation. Male patients may benefit from higher tafamidis doses.
Collapse
Affiliation(s)
- Cecília Monteiro
- Department of Chemistry, The Scripps Research Institute,
10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Jaleh S. Mesgarzadeh
- Department of Chemistry, The Scripps Research Institute,
10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - João Anselmo
- Unidade Corino de Andrade, Centro Hospitalar do Porto,
Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
| | - Joana Fernandes
- Unidade Corino de Andrade, Centro Hospitalar do Porto,
Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
| | - Marta Novais
- Unidade Corino de Andrade, Centro Hospitalar do Porto,
Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
| | - Carla Rodrigues
- Unidade Corino de Andrade, Centro Hospitalar do Porto,
Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
| | - David L. Powers
- Department of Mathematics, Clarkson University, Potsdam, NY
13676, USA
| | - Evan T. Powers
- Department of Chemistry, The Scripps Research Institute,
10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Teresa Coelho
- Unidade Corino de Andrade, Centro Hospitalar do Porto,
Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
- Department of Neurophysiology, Centro Hospitalar do Porto,
Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
| | - Jeffery W. Kelly
- Department of Chemistry, The Scripps Research Institute,
10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
39
|
Fabry Disease: Switch from Enzyme Replacement Therapy to Oral Chaperone Migalastat: What Do We Know Today? Healthcare (Basel) 2023; 11:healthcare11040449. [PMID: 36832983 PMCID: PMC9957019 DOI: 10.3390/healthcare11040449] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/13/2023] [Accepted: 01/22/2023] [Indexed: 02/08/2023] Open
Abstract
Fabry disease is a lysosomal storage disorder caused by the deficiency of the α-galactosidase-A enzyme. The result is the progressive accumulation of complex glycosphingolipids and cellular dysfunction. Cardiac, renal, and neurological involvement significantly reduces life expectancy. Currently, there is increasing evidence that clinical response to treatment improves with early and timely initiation. Until a few years ago, treatment options for Fabry disease were limited to enzyme replacement therapy with agalsidase alfa or beta administered by intravenous infusion every 2 weeks. Migalastat (Galafold®) is an oral pharmacological chaperone that increases the enzyme activity of "amenable" mutations. The safety and efficacy of migalastat were supported in the phase III FACETS and ATTRACT studies, compared to available enzyme replacement therapies, showing a reduction in left ventricular mass, and stabilization of kidney function and plasma Lyso-Gb3. Similar results were confirmed in subsequent extension publications, both in patients who started migalastat as their first treatment and in patients who were previously on enzyme replacement therapy and switched to migalastat. In this review we describe the safety and efficacy of switching from enzyme replacement therapy to migalastat in patients with Fabry disease and "amenable" mutations, referring to publications available to date.
Collapse
|
40
|
Kleynerman A, Rybova J, Faber ML, McKillop WM, Levade T, Medin JA. Acid Ceramidase Deficiency: Bridging Gaps between Clinical Presentation, Mouse Models, and Future Therapeutic Interventions. Biomolecules 2023; 13:biom13020274. [PMID: 36830643 PMCID: PMC9953133 DOI: 10.3390/biom13020274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Farber disease (FD) and spinal muscular atrophy with progressive myoclonic epilepsy (SMA-PME) are ultra-rare, autosomal-recessive, acid ceramidase (ACDase) deficiency disorders caused by ASAH1 gene mutations. Currently, 73 different mutations in the ASAH1 gene have been described in humans. These mutations lead to reduced ACDase activity and ceramide (Cer) accumulation in many tissues. Presenting as divergent clinical phenotypes, the symptoms of FD vary depending on central nervous system (CNS) involvement and severity. Classic signs of FD include, but are not limited to, a hoarse voice, distended joints, and lipogranulomas found subcutaneously and in other tissues. Patients with SMA-PME lack the most prominent clinical signs seen in FD. Instead, they demonstrate muscle weakness, tremors, and myoclonic epilepsy. Several ACDase-deficient mouse models have been developed to help elucidate the complex consequences of Cer accumulation. In this review, we compare clinical reports on FD patients and experimental descriptions of ACDase-deficient mouse models. We also discuss clinical presentations, potential therapeutic strategies, and future directions for the study of FD and SMA-PME.
Collapse
Affiliation(s)
- Annie Kleynerman
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jitka Rybova
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary L. Faber
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - William M. McKillop
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, CHU Toulouse, and INSERM U1037, Centre de Recherches en Cancérologie de Toulouse, Université Paul Sabatier, 31062 Toulouse, France
| | - Jeffrey A. Medin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: ; Tel.: +1-414-955-4118
| |
Collapse
|
41
|
Hoshikawa T, Watanabe T, Kotake M, Tiberghien N, Woo CK, Lewis S, Briston T, Koglin M, Staddon JM, Powney B, Schapira AHV, Takle AK. Identification of pyrimidinyl piperazines as non-iminosugar glucocerebrosidase (GCase) pharmacological chaperones. Bioorg Med Chem Lett 2023; 81:129130. [PMID: 36640928 DOI: 10.1016/j.bmcl.2023.129130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Glucocerebrosidase (GCase) is a lysosomal enzyme encoded by the GBA1 gene, loss of function variants of which cause an autosomal recessive lysosomal storage disorder, Gaucher disease (GD). Heterozygous variants of GBA1 are also known as the strongest common genetic risk factor for Parkinson's disease (PD). Restoration of GCase enzymatic function using a pharmacological chaperone strategy is considered a promising therapeutic approach for PD and GD. We identified compound 4 as a GCase pharmacological chaperone with sub-micromolar activity from a high-throughput screening (HTS) campaign. Compound 4 was further optimised to ER-001230194 (compound 25). ER-001230194 shows improved ADME and physicochemical properties and therefore represents a novel pharmacological chaperone with which to investigate GCase pharmacology further.
Collapse
Affiliation(s)
- Tamaki Hoshikawa
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom.
| | - Toru Watanabe
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| | - Makoto Kotake
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| | - Nathalie Tiberghien
- Charles River Laboratories, 7-9 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Chi-Kit Woo
- Charles River Laboratories, 7-9 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Sian Lewis
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| | - Thomas Briston
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| | - Mumta Koglin
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| | - James M Staddon
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| | - Ben Powney
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Andrew K Takle
- Hatfield Research Laboratories, Eisai Ltd., Hatfield AL10 9SN, United Kingdom
| |
Collapse
|
42
|
Clemente F, Davighi MG, Matassini C, Cardona F, Goti A, Morrone A, Paoli P, Tejero T, Merino P, Cacciarini M. Light-Triggered Control of Glucocerebrosidase Inhibitors: Towards Photoswitchable Pharmacological Chaperones. Chemistry 2023; 29:e202203841. [PMID: 36598148 DOI: 10.1002/chem.202203841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/05/2023]
Abstract
Piperidine-based photoswitchable derivatives have been developed as putative pharmacological chaperones for glucocerebrosidase (GCase), the defective enzyme in Gaucher disease (GD). The structure-activity study revealed that both the iminosugar and the light-sensitive azobenzene are essential features to exert inhibitory activity towards human GCase and a system with the correct inhibition trend (IC50 of the light-activated form lower than IC50 of the dark form) was identified. Kinetic analyses showed that all compounds are non-competitive inhibitors (mixed or pure) of GCase and the enzyme allosteric site involved in the interaction was identified by means of MD simulations. A moderate activity enhancement of mutant GCase assessed in GD patients' fibroblasts (ex vivo experiments) carrying the most common mutation was recorded. This promising observation paves the way for further studies to improve the benefit of the light-to-dark thermal conversion for chaperoning activity.
Collapse
Affiliation(s)
- Francesca Clemente
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy
| | - Maria Giulia Davighi
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy
| | - Camilla Matassini
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy
| | - Francesca Cardona
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy.,Associated with LENS, Via N. Carrara 1, 50019, Sesto F.no, FI, Italy
| | - Andrea Goti
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy.,Associated with LENS, Via N. Carrara 1, 50019, Sesto F.no, FI, Italy
| | - Amelia Morrone
- Laboratory of Molecular Biology of Neurometabolic Diseases, Neuroscience Department, Meyer Children's Hospital, Viale Pieraccini 24, 50139, Firenze, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 24, 50139, Firenze, Italy
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Firenze, Italy
| | - Tomás Tejero
- Institute of Chemical Synthesis and Homogeneous Catalysis. (ISQCH), University of Zaragoza, Campus San Francisco, Zaragoza, 50009, Spain
| | - Pedro Merino
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Campus San Francisco, Zaragoza, 50009, Spain
| | - Martina Cacciarini
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy.,Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100, Copenhagen Ø, Denmark
| |
Collapse
|
43
|
Palaiodimou L, Kokotis P, Zompola C, Papagiannopoulou G, Bakola E, Papadopoulou M, Zouvelou V, Petras D, Vlachopoulos C, Tsivgoulis G. Fabry Disease: Current and Novel Therapeutic Strategies. A Narrative Review. Curr Neuropharmacol 2023; 21:440-456. [PMID: 35652398 PMCID: PMC10207921 DOI: 10.2174/1570159x20666220601124117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/11/2022] [Accepted: 05/20/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Fabry disease (FD) is an inherited lysosomal storage disorder, leading to multisystemic manifestations and causing significant morbidity and mortality. OBJECTIVE The aim of this narrative review is to present the current and novel therapeutic strategies in FD, including symptomatic and specific treatment options. METHODS A systematic literature search was conducted to identify relevant studies, including completed and ongoing randomized-controlled clinical trials (RCTs), prospective or retrospective cohort studies, case series and case reports that provided clinical data regarding FD treatment. RESULTS A multidisciplinary symptomatic treatment is recommended for FD patients, personalized according to disease manifestations and their severity. During the last two decades, FD-specific treatments, including two enzyme-replacement-therapies (agalsidase alfa and agalsidase beta) and chaperone treatment with migalastat have been approved for use and allowed for symptoms' stabilization or even disease burden reduction. More therapeutic agents are currently under investigation. Substrate reduction therapies, including lucerastat and venglustat, have shown promising results in RCTs and may be used either as monotherapy or as complementary therapy to established enzymereplacement- therapies. More stable enzyme-replacement-therapy molecules that are associated with less adverse events and lower likelihood of neutralizing antibodies formation have also been developed. Ex-vivo and in-vivo gene therapy is being tested in animal models and pilot human clinical trials, with preliminary results showing a favorable safety and efficacy profile. CONCLUSION The therapeutic landscape in FD appears to be actively expanding with more treatment options expected to become available in the near future, allowing for a more personalized approach in FD patients.
Collapse
Affiliation(s)
- Lina Palaiodimou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Kokotis
- First Department of Neurology, National and Kapodistrian University of Athens, School of Medicine, Eginition Hospital, Athens, Greece
| | - Christina Zompola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Papagiannopoulou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Bakola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianna Papadopoulou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Zouvelou
- First Department of Neurology, National and Kapodistrian University of Athens, School of Medicine, Eginition Hospital, Athens, Greece
| | - Dimitrios Petras
- Nephrology Department, Hippokration General Hospital, Athens, Greece
| | | | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
44
|
Mächtel R, Boros FA, Dobert JP, Arnold P, Zunke F. From Lysosomal Storage Disorders to Parkinson's Disease - Challenges and Opportunities. J Mol Biol 2022:167932. [PMID: 36572237 DOI: 10.1016/j.jmb.2022.167932] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Lysosomes are specialized organelles with an acidic pH that act as recycling hubs for intracellular and extracellular components. They harbour numerous different hydrolytic enzymes to degrade substrates like proteins, peptides, and glycolipids. Reduced catalytic activity of lysosomal enzymes can cause the accumulation of these substrates and loss of lysosomal integrity, resulting in lysosomal dysfunction and lysosomal storage disorders (LSDs). Post-mitotic cells, such as neurons, seem to be highly sensitive to damages induced by lysosomal dysfunction, thus LSDs often manifest with neurological symptoms. Interestingly, some LSDs and Parkinson's disease (PD) share common cellular pathomechanisms, suggesting convergence of aetiology of the two disease types. This is further underlined by genetic associations of several lysosomal genes involved in LSDs with PD. The increasing number of lysosome-associated genetic risk factors for PD makes it necessary to understand functions and interactions of lysosomal proteins/enzymes both in health and disease, thereby holding the potential to identify new therapeutic targets. In this review, we highlight genetic and mechanistic interactions between the complex lysosomal network, LSDs and PD, and elaborate on methodical challenges in lysosomal research.
Collapse
Affiliation(s)
- Rebecca Mächtel
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | | | - Jan Philipp Dobert
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Friederike Zunke
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany.
| |
Collapse
|
45
|
Gil-Martínez J, Bernardo-Seisdedos G, Mato JM, Millet O. The use of pharmacological chaperones in rare diseases caused by reduced protein stability. Proteomics 2022; 22:e2200222. [PMID: 36205620 DOI: 10.1002/pmic.202200222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022]
Abstract
Rare diseases are most often caused by inherited genetic disorders that, after translation, will result in a protein with altered function. Decreased protein stability is the most frequent mechanism associated with a congenital pathogenic missense mutation and it implies the destabilization of the folded conformation in favour of unfolded or misfolded states. In the cellular context and when experimental data is available, a mutant protein with altered thermodynamic stability often also results in impaired homeostasis, with the deleterious accumulation of protein aggregates, metabolites and/or metabolic by-products. In the last decades, a significant effort has enabled the characterization of rare diseases associated to protein stability defects and triggered the development of innovative therapeutic intervention lines, say, the use of pharmacological chaperones to correct the intracellular impaired homeostasis. Here, we review the current knowledge on rare diseases caused by reduced protein stability, paying special attention to the thermodynamic aspects of the protein destabilization, also focusing on some examples where pharmacological chaperones are being tested.
Collapse
Affiliation(s)
- Jon Gil-Martínez
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia, Spain
| | | | - José M Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia, Spain.,ATLAS Molecular Pharma, Bizkaia, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
46
|
Li X, Ren X, Zhang Y, Ding L, Huo M, Li Q. Fabry disease: Mechanism and therapeutics strategies. Front Pharmacol 2022; 13:1025740. [PMID: 36386210 PMCID: PMC9643830 DOI: 10.3389/fphar.2022.1025740] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Fabry disease is a monogenic disease characterized by a deficiency or loss of the α-galactosidase A (GLA). The resulting impairment in lysosomal GLA enzymatic activity leads to the pathogenic accumulation of enzymatic substrate and, consequently, the progressive appearance of clinical symptoms in target organs, including the heart, kidney, and brain. However, the mechanisms involved in Fabry disease-mediated organ damage are largely ambiguous and poorly understood, which hinders the development of therapeutic strategies for the treatment of this disorder. Although currently available clinical approaches have shown some efficiency in the treatment of Fabry disease, they all exhibit limitations that need to be overcome. In this review, we first introduce current mechanistic knowledge of Fabry disease and discuss potential therapeutic strategies for its treatment. We then systemically summarize and discuss advances in research on therapeutic approaches, including enzyme replacement therapy (ERT), gene therapy, and chaperone therapy, as well as strategies targeting subcellular compartments, such as lysosomes, the endoplasmic reticulum, and the nucleus. Finally, the future development of potential therapeutic strategies is discussed based on the results of mechanistic studies and the limitations associated with these therapeutic approaches.
Collapse
Affiliation(s)
- Xi Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiangyi Ren
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Yabing Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Lin Ding
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Minfeng Huo
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| |
Collapse
|
47
|
MPSI Manifestations and Treatment Outcome: Skeletal Focus. Int J Mol Sci 2022; 23:ijms231911168. [PMID: 36232472 PMCID: PMC9569890 DOI: 10.3390/ijms231911168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/23/2022] Open
Abstract
Mucopolysaccharidosis type I (MPSI) (OMIM #252800) is an autosomal recessive disorder caused by pathogenic variants in the IDUA gene encoding for the lysosomal alpha-L-iduronidase enzyme. The deficiency of this enzyme causes systemic accumulation of glycosaminoglycans (GAGs). Although disease manifestations are typically not apparent at birth, they can present early in life, are progressive, and include a wide spectrum of phenotypic findings. Among these, the storage of GAGs within the lysosomes disrupts cell function and metabolism in the cartilage, thus impairing normal bone development and ossification. Skeletal manifestations of MPSI are often refractory to treatment and severely affect patients’ quality of life. This review discusses the pathological and molecular processes leading to impaired endochondral ossification in MPSI patients and the limitations of current therapeutic approaches. Understanding the underlying mechanisms responsible for the skeletal phenotype in MPSI patients is crucial, as it could lead to the development of new therapeutic strategies targeting the skeletal abnormalities of MPSI in the early stages of the disease.
Collapse
|
48
|
Meek RW, Brockerman J, Fordwour OB, Zandberg WF, Davies GJ, Vocadlo DJ. The primary familial brain calcification-associated protein MYORG is an α-galactosidase with restricted substrate specificity. PLoS Biol 2022; 20:e3001764. [PMID: 36129849 PMCID: PMC9491548 DOI: 10.1371/journal.pbio.3001764] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Primary familial brain calcification (PFBC) is characterised by abnormal deposits of calcium phosphate within various regions of the brain that are associated with severe cognitive impairments, psychiatric conditions, and movement disorders. Recent studies in diverse populations have shown a link between mutations in myogenesis-regulating glycosidase (MYORG) and the development of this disease. MYORG is a member of glycoside hydrolase (GH) family 31 (GH31) and, like the other mammalian GH31 enzyme α-glucosidase II, this enzyme is found in the lumen of the endoplasmic reticulum (ER). Though presumed to act as an α-glucosidase due to its localization and sequence relatedness to α-glucosidase II, MYORG has never been shown to exhibit catalytic activity. Here, we show that MYORG is an α-galactosidase and present the high-resolution crystal structure of MYORG in complex with substrate and inhibitor. Using these structures, we map detrimental mutations that are associated with MYORG-associated brain calcification and define how these mutations may drive disease progression through loss of enzymatic activity. Finally, we also detail the thermal stabilisation of MYORG afforded by a clinically approved small molecule ligand, opening the possibility of using pharmacological chaperones to enhance the activity of mutant forms of MYORG. MYORG is an enzyme genetically linked to primary familial brain calcification that has historically been presumed to act as an α-glucosidase. This study describes the crystal structure of dimeric MYORG and, surprisingly, reveals it to be an α-galactosidase with restricted specificity.
Collapse
Affiliation(s)
- Richard W. Meek
- Department of Chemistry. University of York, York, United Kingdom
| | - Jacob Brockerman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Osei B. Fordwour
- Department of Chemistry, Irving K. Barber Faculty of Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Wesley F. Zandberg
- Department of Chemistry, Irving K. Barber Faculty of Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Gideon J. Davies
- Department of Chemistry. University of York, York, United Kingdom
- * E-mail: (GJD); (DJV)
| | - David J. Vocadlo
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- * E-mail: (GJD); (DJV)
| |
Collapse
|
49
|
Mucopolysaccharidoses and the blood-brain barrier. Fluids Barriers CNS 2022; 19:76. [PMID: 36117162 PMCID: PMC9484072 DOI: 10.1186/s12987-022-00373-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022] Open
Abstract
Mucopolysaccharidoses comprise a set of genetic diseases marked by an enzymatic dysfunction in the degradation of glycosaminoglycans in lysosomes. There are eight clinically distinct types of mucopolysaccharidosis, some with various subtypes, based on which lysosomal enzyme is deficient and symptom severity. Patients with mucopolysaccharidosis can present with a variety of symptoms, including cognitive dysfunction, hepatosplenomegaly, skeletal abnormalities, and cardiopulmonary issues. Additionally, the onset and severity of symptoms can vary depending on the specific disorder, with symptoms typically arising during early childhood. While there is currently no cure for mucopolysaccharidosis, there are clinically approved therapies for the management of clinical symptoms, such as enzyme replacement therapy. Enzyme replacement therapy is typically administered intravenously, which allows for the systemic delivery of the deficient enzymes to peripheral organ sites. However, crossing the blood-brain barrier (BBB) to ameliorate the neurological symptoms of mucopolysaccharidosis continues to remain a challenge for these large macromolecules. In this review, we discuss the transport mechanisms for the delivery of lysosomal enzymes across the BBB. Additionally, we discuss the several therapeutic approaches, both preclinical and clinical, for the treatment of mucopolysaccharidoses.
Collapse
|
50
|
Santana AG, Robinson K, Vickers C, Deen MC, Chen H, Zhou S, Dai B, Fuller M, Boraston AB, Vocadlo DJ, Clarke LA, Withers SG. Pharmacological Chaperones for GCase that Switch Conformation with pH Enhance Enzyme Levels in Gaucher Animal Models. Angew Chem Int Ed Engl 2022; 61:e202207974. [DOI: 10.1002/anie.202207974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Andrés G. Santana
- Dept. of Chemistry University of British Columbia Vancouver BC, V6T 1Z1 Canada
| | - Kyle Robinson
- Dept. of Chemistry University of British Columbia Vancouver BC, V6T 1Z1 Canada
| | - Chelsea Vickers
- Dept. of Biochemistry and Microbiology University of Victoria Victoria BC, V8W 3P6 Canada
| | - Matthew C. Deen
- Dept. of Chemistry and Dept. of Mol. Biology and Biochemistry Simon Fraser University Burnaby BC, V5A 1S6 Canada
| | - Hong‐Ming Chen
- Dept. of Chemistry University of British Columbia Vancouver BC, V6T 1Z1 Canada
| | - Stephen Zhou
- Dept. of Medical Genetics University of British Columbia Women's Hospital & Health Centre Vancouver BC, V6H 3N1 Canada
| | - Ben Dai
- Dept. of Medical Genetics University of British Columbia Women's Hospital & Health Centre Vancouver BC, V6H 3N1 Canada
| | - Maria Fuller
- Genetics and Molecular Pathology SA Pathology at Women's and Children's Hospital N. Adelaide South Australia 5006 Australia
| | - Alisdair B. Boraston
- Dept. of Biochemistry and Microbiology University of Victoria Victoria BC, V8W 3P6 Canada
| | - David J. Vocadlo
- Dept. of Chemistry and Dept. of Mol. Biology and Biochemistry Simon Fraser University Burnaby BC, V5A 1S6 Canada
| | - Lorne A. Clarke
- Dept. of Medical Genetics University of British Columbia Women's Hospital & Health Centre Vancouver BC, V6H 3N1 Canada
| | - Stephen G. Withers
- Dept. of Chemistry University of British Columbia Vancouver BC, V6T 1Z1 Canada
| |
Collapse
|