1
|
Li H, Doray B, Jennings BC, Lee WS, Liu L, Kornfeld S, Li H. Structure of a truncated human GlcNAc-1-phosphotransferase variant reveals the basis for its hyperactivity. J Biol Chem 2024; 300:107706. [PMID: 39178950 PMCID: PMC11418123 DOI: 10.1016/j.jbc.2024.107706] [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: 05/02/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
Mutations that cause loss of function of GlcNAc-1-phosphotransferase (PTase) lead to the lysosomal storage disorder mucolipidosis II. PTase is the key enzyme of the mannose 6-phosphate (M6P) targeting system that is responsible for tagging lysosomal hydrolases with the M6P moiety for their delivery to the lysosome. We had previously generated a truncated hyperactive form of PTase termed S1S3 which was shown to notably increase the phosphorylation level of secreted lysosomal enzymes and enhance their uptake by cells. Here, we report the 3.4 Å cryo-EM structure of soluble S1S3 lacking both transmembrane domains and cytosolic tails. The structure reveals a high degree of conservation of the catalytic core to full-length PTase. In this dimeric structure, the EF-hand of one protomer is observed interacting with the conserved region four of the other. In addition, we present a high-quality EM 3D map of the UDP-GlcNAc bound form of the full-length soluble protein showing the key molecular interactions between the nucleotide sugar donor and side chain amino acids of the protein. Finally, although the domain organization of S1S3 is very similar to that of the Drosophila melanogaster (fruit fly) PTase homolog, we establish that the latter does not act on lysosomal hydrolases.
Collapse
Affiliation(s)
- Hua Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Balraj Doray
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Benjamin C Jennings
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Wang-Sik Lee
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lin Liu
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stuart Kornfeld
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA.
| |
Collapse
|
2
|
Young DJ, Edwards AJ, Quiroz Caceda KG, Liberzon E, Barrientos J, Hong S, Turner J, Choyke PL, Arlauckas S, Lazorchak AS, Morgan RA, Sato N, Dunbar CE. In vivo tracking of ex vivo generated 89 Zr-oxine labeled plasma cells by PET in a non-human primate model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595782. [PMID: 38903108 PMCID: PMC11188104 DOI: 10.1101/2024.05.24.595782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
B cells are an attractive platform for engineering to produce protein-based biologics absent in genetic disorders, and potentially for the treatment of metabolic diseases and cancer. As part of pre-clinical development of B cell medicines, we demonstrate a method to collect, ex vivo expand, differentiate, radioactively label, and track adoptively transferred non-human primate (NHP) B cells. These cells underwent 10- to 15-fold expansion, initiated IgG class switching, and differentiated into antibody secreting cells. Zirconium-89-oxine labeled cells were infused into autologous donors without any preconditioning and tracked by PET/CT imaging. Within 24 hours of infusion, 20% of the initial dose homed to the bone marrow and spleen and distributed stably and equally between the two. Interestingly, approximately half of the dose homed to the liver. Image analysis of the bone marrow demonstrated inhomogeneous distribution of the cells. The subjects experienced no clinically significant side effects or laboratory abnormalities. A second infusion of B cells into one of the subjects resulted in an almost identical distribution of cells, suggesting a non-limiting engraftment niche and feasibility of repeated infusions. This work supports the NHP as a valuable model to assess the potential of B cell medicines as potential treatment for human diseases.
Collapse
|
3
|
Rocamora F, Peralta AG, Shin S, Sorrentino J, Wu MYM, Toth EA, Fuerst TR, Lewis NE. Glycosylation shapes the efficacy and safety of diverse protein, gene and cell therapies. Biotechnol Adv 2023; 67:108206. [PMID: 37354999 PMCID: PMC11168894 DOI: 10.1016/j.biotechadv.2023.108206] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Over recent decades, therapeutic proteins have had widespread success in treating a myriad of diseases. Glycosylation, a near universal feature of this class of drugs, is a critical quality attribute that significantly influences the physical properties, safety profile and biological activity of therapeutic proteins. Optimizing protein glycosylation, therefore, offers an important avenue to developing more efficacious therapies. In this review, we discuss specific examples of how variations in glycan structure and glycoengineering impacts the stability, safety, and clinical efficacy of protein-based drugs that are already in the market as well as those that are still in preclinical development. We also highlight the impact of glycosylation on next generation biologics such as T cell-based cancer therapy and gene therapy.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Angelo G Peralta
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seunghyeon Shin
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - James Sorrentino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mina Ying Min Wu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
4
|
Novel Gene-Correction-Based Therapeutic Modalities for Monogenic Liver Disorders. Bioengineering (Basel) 2022; 9:bioengineering9080392. [PMID: 36004917 PMCID: PMC9404740 DOI: 10.3390/bioengineering9080392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
The majority of monogenic liver diseases are autosomal recessive disorders, with few being sex-related or co-dominant. Although orthotopic liver transplantation (LT) is currently the sole therapeutic option for end-stage patients, such an invasive surgical approach is severely restricted by the lack of donors and post-transplant complications, mainly associated with life-long immunosuppressive regimens. Therefore, the last decade has witnessed efforts for innovative cellular or gene-based therapeutic strategies. Gene therapy is a promising approach for treatment of many hereditary disorders, such as monogenic inborn errors. The liver is an organ characterized by unique features, making it an attractive target for in vivo and ex vivo gene transfer. The current genetic approaches for hereditary liver diseases are mediated by viral or non-viral vectors, with promising results generated by gene-editing tools, such as CRISPR-Cas9 technology. Despite massive progress in experimental gene-correction technologies, limitations in validated approaches for monogenic liver disorders have encouraged researchers to refine promising gene therapy protocols. Herein, we highlighted the most common monogenetic liver disorders, followed by proposed genetic engineering approaches, offered as promising therapeutic modalities.
Collapse
|
5
|
Zhang H, Nagree MS, Liu H, Pan X, Medin JA, Lipinski DM. rAAV-mediated over-expression of acid ceramidase prevents retinopathy in a mouse model of Farber lipogranulomatosis. Gene Ther 2022; 30:297-308. [PMID: 35902747 DOI: 10.1038/s41434-022-00359-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/09/2022]
Abstract
Farber disease (FD) is a rare monogenic lysosomal storage disorder caused by mutations in ASAH1 that results in a deficiency of acid ceramidase (ACDase) activity and the abnormal systemic accumulation of ceramide species, leading to multi-system organ failure involving neurological decline and retinopathy. Here we describe the effects of rAAV-mediated ASAH1 over-expression on the progression of retinopathy in a mouse model of FD (Asah1P361R/P361R) and its littermate controls (Asah1+/+ and Asah1+/P361R). Using a combination of non-invasive multimodal imaging, electrophysiology, post-mortem histology and mass spectrometry we demonstrate that ASAH1 over-expression significantly reduces central retinal thickening, ceramide accumulation, macrophage activation and limits fundus hyper-reflectivity and auto-fluorescence in FD mice, indicating rAAV-mediated over-expression of biologically active ACDase protein is able to rescue the anatomical retinal phenotype of Farber disease. Unexpectedly, ACDase over-expression in Asah1+/+ and Asah1+/P361R control eyes was observed to induce abnormal fundus hyper-reflectivity, auto-fluorescence and retinal thickening that closely resembles a FD phenotype. This study represents the first evidence of a gene therapy for Farber disease-related retinopathy. Importantly, the described gene therapy approach could be used to preserve vision in FD patients synergistically with broader enzyme replacement strategies aimed at preserving life.
Collapse
Affiliation(s)
- Hanmeng Zhang
- Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Murtaza S Nagree
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Department of Pediatrics: Section of Hematology/Oncology/BMT, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Haoyuan Liu
- Department of Mathematics, Shanghai Normal University, Shanghai, China
| | - Xiaoqing Pan
- Department of Mathematics, Shanghai Normal University, Shanghai, China
| | - Jeffrey A Medin
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Department of Pediatrics: Section of Hematology/Oncology/BMT, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Daniel M Lipinski
- Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA. .,Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
6
|
Del Grosso A, Parlanti G, Mezzena R, Cecchini M. Current treatment options and novel nanotechnology-driven enzyme replacement strategies for lysosomal storage disorders. Adv Drug Deliv Rev 2022; 188:114464. [PMID: 35878795 DOI: 10.1016/j.addr.2022.114464] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/26/2022] [Accepted: 07/19/2022] [Indexed: 11/01/2022]
Abstract
Lysosomal storage disorders (LSDs) are a vast group of more than 50 clinically identified metabolic diseases. They are singly rare, but they affect collectively 1 on 5,000 live births. They result in most of the cases from an enzymatic defect within lysosomes, which causes the subsequent augmentation of unwanted substrates. This accumulation process leads to plenty of clinical signs, determined by the specific substrate and accumulation area. The majority of LSDs present a broad organ and tissue engagement. Brain, connective tissues, viscera and bones are usually afflicted. Among them, brain disease is markedly frequent (two-thirds of LSDs). The most clinically employed approach to treat LSDs is enzyme replacement therapy (ERT), which is practiced by administering systemically the missed or defective enzyme. It represents a healthful strategy for 11 LSDs at the moment, but it solves the pathology only in the case of Gaucher disease. This approach, in fact, is not efficacious in the case of LSDs that have an effect on the central nervous system (CNS) due to the existence of the blood-brain barrier (BBB). Additionally, ERT suffers from several other weak points, such as low penetration of the exogenously administered enzyme to poorly vascularized areas, the development of immunogenicity and infusion-associated reactions (IARs), and, last but not least, the very high cost and lifelong needed. To ameliorate these weaknesses lot of efforts have been recently spent around the development of innovative nanotechnology-driven ERT strategies. They may boost the power of ERT and minimize adverse reactions by loading enzymes into biodegradable nanomaterials. Enzyme encapsulation into biocompatible liposomes, micelles, and polymeric nanoparticles, for example, can protect enzymatic activity, eliminating immunologic reactions and premature enzyme degradation. It can also permit a controlled release of the payload, ameliorating pharmacokinetics and pharmacodynamics of the drug. Additionally, the potential to functionalize the surface of the nanocarrier with targeting agents (antibodies or peptides), could promote the passage through biological barriers. In this review we examined the clinically applied ERTs, highlighting limitations that do not allow to completely cure the specific LSD. Later, we critically consider the nanotechnology-based ERT strategies that have beenin-vitroand/orin-vivotested to improve ERT efficacy.
Collapse
Affiliation(s)
- Ambra Del Grosso
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Gabriele Parlanti
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Roberta Mezzena
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
7
|
Joosten MDW, Clabbers JMK, Jonca N, Mazereeuw-Hautier J, Gostyński AH. New developments in the molecular treatment of ichthyosis: review of the literature. Orphanet J Rare Dis 2022; 17:269. [PMID: 35840979 PMCID: PMC9287901 DOI: 10.1186/s13023-022-02430-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/30/2022] [Indexed: 11/24/2022] Open
Abstract
Ichthyosis covers a wide spectrum of diseases affecting the cornification of the skin. In recent years, new advances in understanding the pathophysiology of ichthyosis have been made. This knowledge, combined with constant development of pathogenesis-based therapies, such as protein replacement therapy and gene therapy, are rather promising for patients with inherited skin diseases. Several ongoing trials are investigating the potency of these new approaches and various studies have already been published. Furthermore, a lot of case series report that biological therapeutics are effective treatment options, mainly for Netherton syndrome and autosomal recessive congenital ichthyosis. It is expected that some of these new therapies will prove their efficacy and will be incorporated in the treatment of ichthyosis.
Collapse
Affiliation(s)
- M D W Joosten
- Department of Dermatology, The Netherlands and European Reference Network - Skin, Maastricht University Medical Center, Maastricht, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P. Debyelaan 25, 6229HX, Maastricht, The Netherlands
| | - J M K Clabbers
- Department of Dermatology, The Netherlands and European Reference Network - Skin, Maastricht University Medical Center, Maastricht, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P. Debyelaan 25, 6229HX, Maastricht, The Netherlands.,Department of Dermatology, Haga Hospital, The Hague, The Netherlands
| | - N Jonca
- Cell Biology and Cytology Laboratory, CNRS, Inserm, UPS, European Reference Network - Skin, University Hospital Center of Toulouse and Infinity, Federal Biology Institute, Toulouse University, Toulouse, France
| | - J Mazereeuw-Hautier
- Department of Dermatology, European Reference Network - Skin, University Hospital Center of Toulouse, Toulouse, France
| | - A H Gostyński
- Department of Dermatology, The Netherlands and European Reference Network - Skin, Maastricht University Medical Center, Maastricht, The Netherlands. .,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P. Debyelaan 25, 6229HX, Maastricht, The Netherlands.
| |
Collapse
|
8
|
Fernández-Pereira C, San Millán-Tejado B, Gallardo-Gómez M, Pérez-Márquez T, Alves-Villar M, Melcón-Crespo C, Fernández-Martín J, Ortolano S. Therapeutic Approaches in Lysosomal Storage Diseases. Biomolecules 2021; 11:biom11121775. [PMID: 34944420 PMCID: PMC8698519 DOI: 10.3390/biom11121775] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 02/07/2023] Open
Abstract
Lysosomal Storage Diseases are multisystemic disorders determined by genetic variants, which affect the proteins involved in lysosomal function and cellular metabolism. Different therapeutic approaches, which are based on the physiologic mechanisms that regulate lysosomal function, have been proposed for these diseases. Currently, enzyme replacement therapy, gene therapy, or small molecules have been approved or are under clinical development to treat lysosomal storage disorders. The present article reviews the main therapeutic strategies that have been proposed so far, highlighting possible limitations and future perspectives.
Collapse
Affiliation(s)
- Carlos Fernández-Pereira
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
| | - Beatriz San Millán-Tejado
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
| | - María Gallardo-Gómez
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
| | - Tania Pérez-Márquez
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
| | - Marta Alves-Villar
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
| | - Cristina Melcón-Crespo
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
- Department of Pediatrics, Hospital Álvaro Cunqueiro, SERGAS, 36213 Vigo, Spain
| | - Julián Fernández-Martín
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
- Department of Internal Medicine, Hospital Álvaro Cunqueiro, SERGAS, 36213 Vigo, Spain
| | - Saida Ortolano
- Rare Disease and Pediatric Medicine Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (C.F.-P.); (B.S.M.-T.); (M.G.-G.); (T.P.-M.); (M.A.-V.); (C.M.-C.); (J.F.-M.)
- Correspondence: ; Tel.: +34-986217466
| |
Collapse
|
9
|
Stütz AE, Thonhofer M, Weber P, Wolfsgruber A, Wrodnigg TM. Pharmacological Chaperones for β-Galactosidase Related to G M1 -Gangliosidosis and Morquio B: Recent Advances. CHEM REC 2021; 21:2980-2989. [PMID: 34816592 DOI: 10.1002/tcr.202100269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
A short survey on selected β-galactosidase inhibitors as potential pharmacological chaperones for GM1 -gangliosidosis and Morquio B associated mutants of human lysosomal β-galactosidase is provided highlighting recent developments in this particular area of lysosomal storage disorders and orphan diseases.
Collapse
Affiliation(s)
- Arnold E Stütz
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010, Graz, Austria
| | - Martin Thonhofer
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010, Graz, Austria
| | - Patrick Weber
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010, Graz, Austria
| | - Andreas Wolfsgruber
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010, Graz, Austria
| | - Tanja M Wrodnigg
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010, Graz, Austria
| |
Collapse
|
10
|
Magalhães A, Vilares-Morgado R, Cunha AM, Leão-Teles E, Falcão M, Carneiro Â, Falcão-Reis F. Increased Choroidal Thickness in Morquio Syndrome. Case Rep Ophthalmol 2021; 12:816-823. [PMID: 34720983 PMCID: PMC8543289 DOI: 10.1159/000518443] [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: 03/07/2021] [Accepted: 07/11/2021] [Indexed: 12/02/2022] Open
Abstract
The purpose of this clinical case report is to describe a case of mucopolysaccharidosis type IVA (MPS IVA), or Morquio syndrome, with increased choroidal thickness in enhanced-depth imaging optical coherence tomography (EDI-OCT) which can represent choroidal deposition of glycosaminoglycans (GAGs). A 21-year-old male with genetically confirmed diagnosis of MPS IVA was examined at our Pediatric Ophthalmology clinic as part of our follow-up protocol for MPS patients. His best-corrected visual acuity was 4/10 in his right eye (OD) and 6/10 in the left eye (OS). Mild diffuse corneal opacification was evident. Intraocular pressure was within normal range. Fundus examination and color fundus photography revealed no abnormalities. EDI-OCT revealed significantly increased choroidal thickness in his right eye and in his left eye, suggesting the presence of choroidal deposition of GAGs, despite absence of retinal or optic disc GAG deposition or other chorioretinal involvement. To our knowledge, this is the first case of MPS IVA described in the literature with suspected choroidal deposition of GAGs. With improved control of systemic features of MPS IVA, life expectancy of these patients has increased, allowing for more ocular manifestations to develop. The parallel development of technology in ophthalmology, such as the EDI-OCT, further contributes to the detection of these unprecedented ocular features in MPSs.
Collapse
Affiliation(s)
- Augusto Magalhães
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Rodrigo Vilares-Morgado
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine of University of Porto, Porto, Portugal
| | - Ana Maria Cunha
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Elisa Leão-Teles
- Department of Pediatrics, Reference Centre of Inherited Metabolic Diseases, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Manuel Falcão
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine of University of Porto, Porto, Portugal
| | - Ângela Carneiro
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine of University of Porto, Porto, Portugal
| | - Fernando Falcão-Reis
- Department of Ophthalmology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine of University of Porto, Porto, Portugal
| |
Collapse
|
11
|
Zhang X, Liu H, Meena N, Li C, Zong G, Raben N, Puertollano R, Wang LX. Chemoenzymatic glycan-selective remodeling of a therapeutic lysosomal enzyme with high-affinity M6P-glycan ligands. Enzyme substrate specificity is the name of the game. Chem Sci 2021; 12:12451-12462. [PMID: 34603676 PMCID: PMC8480326 DOI: 10.1039/d1sc03188k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/13/2021] [Indexed: 11/21/2022] Open
Abstract
Functionalization of therapeutic lysosomal enzymes with mannose-6-phosphate (M6P) glycan ligands represents a major strategy for enhancing the cation-independent M6P receptor (CI-MPR)-mediated cellular uptake, thus improving the overall therapeutic efficacy of the enzymes. However, the minimal high-affinity M6P-containing N-glycan ligands remain to be identified and their efficient and site-selective conjugation to therapeutic lysosomal enzymes is a challenging task. We report here the chemical synthesis of truncated M6P-glycan oxazolines and their use for enzymatic glycan remodeling of recombinant human acid α-glucosidase (rhGAA), an enzyme used for treatment of Pompe disease which is a disorder caused by a deficiency of the glycogen-degrading lysosomal enzyme. Structure-activity relationship studies identified M6P tetrasaccharide oxazoline as the minimal substrate for enzymatic transglycosylation yielding high-affinity M6P glycan ligands for the CI-MPR. Taking advantage of the substrate specificity of endoglycosidases Endo-A and Endo-F3, we found that Endo-A and Endo-F3 could efficiently deglycosylate the respective high-mannose and complex type N-glycans in rhGAA and site-selectively transfer the synthetic M6P N-glycan to the deglycosylated rhGAA without product hydrolysis. This discovery enabled a highly efficient one-pot deglycosylation/transglycosylation strategy for site-selective M6P-glycan remodeling of rhGAA to obtain a more homogeneous product. The Endo-A and Endo-F3 remodeled rhGAAs maintained full enzyme activity and demonstrated 6- and 20-fold enhanced binding affinities for CI-MPR receptor, respectively. Using an in vitro cell model system for Pompe disease, we demonstrated that the M6P-glycan remodeled rhGAA greatly outperformed the commercial rhGAA (Lumizyme) and resulted in the reversal of cellular pathology. This study provides a general and efficient method for site-selective M6P-glycan remodeling of recombinant lysosomal enzymes to achieve enhanced M6P receptor binding and cellular uptake, which could lead to improved overall therapeutic efficacy of enzyme replacement therapy.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Chemistry and Biochemistry, University of Maryland 8051 Regents Drive College Park Maryland 20742 USA
| | - Huiying Liu
- Department of Chemistry and Biochemistry, University of Maryland 8051 Regents Drive College Park Maryland 20742 USA
| | - Naresh Meena
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH Bethesda Maryland 20892 USA
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland 8051 Regents Drive College Park Maryland 20742 USA
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland 8051 Regents Drive College Park Maryland 20742 USA
| | - Nina Raben
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH Bethesda Maryland 20892 USA
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH Bethesda Maryland 20892 USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland 8051 Regents Drive College Park Maryland 20742 USA
| |
Collapse
|
12
|
Enzyme Therapy: Current Challenges and Future Perspectives. Int J Mol Sci 2021; 22:ijms22179181. [PMID: 34502086 PMCID: PMC8431097 DOI: 10.3390/ijms22179181] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
In recent years, enzymes have risen as promising therapeutic tools for different pathologies, from metabolic deficiencies, such as fibrosis conditions, ocular pathologies or joint problems, to cancer or cardiovascular diseases. Treatments based on the catalytic activity of enzymes are able to convert a wide range of target molecules to restore the correct physiological metabolism. These treatments present several advantages compared to established therapeutic approaches thanks to their affinity and specificity properties. However, enzymes present some challenges, such as short in vivo half-life, lack of targeted action and, in particular, patient immune system reaction against the enzyme. For this reason, it is important to monitor serum immune response during treatment. This can be achieved by conventional techniques (ELISA) but also by new promising tools such as microarrays. These assays have gained popularity due to their high-throughput analysis capacity, their simplicity, and their potential to monitor the immune response of patients during enzyme therapies. In this growing field, research is still ongoing to solve current health problems such as COVID-19. Currently, promising therapeutic alternatives using the angiotensin-converting enzyme 2 (ACE2) are being studied to treat COVID-19.
Collapse
|
13
|
Recapture Lysosomal Enzyme Deficiency via Targeted Gene Disruption in the Human Near-Haploid Cell Line HAP1. Genes (Basel) 2021; 12:genes12071076. [PMID: 34356092 PMCID: PMC8308024 DOI: 10.3390/genes12071076] [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: 06/04/2021] [Revised: 07/04/2021] [Accepted: 07/12/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Advancement in genome engineering enables rapid and targeted disruption of any coding sequences to study gene functions or establish human disease models. We explored whether this approach can be used to study Gaucher disease, one of the most common types of lysosomal storage diseases (LSDs) in a near-haploid human cell line (HAP1). RESULTS CRISPR-Cas9 targeting to coding sequences of β-glucocerebrosidase (GBA), the causative gene of Gaucher disease, resulted in an insertional mutation and premature termination of GBA. We confirmed the GBA knockout at both the gene and enzyme levels by genotyping and GBA enzymatic assay. Characterization of the knockout line showed no significant changes in cell morphology and growth. Lysosomal staining revealed more granular lysosomes in the cytosol of the GBA-knockout line compared to its parental control. Flow cytometry analysis further confirmed that more lysosomes accumulated in the cytosol of the knockout line, recapturing the disease phenotype. Finally, we showed that this knockout cell line could be used to evaluate a replacement therapy by recombinant human GBA. CONCLUSIONS Targeted gene disruption in human HAP1 cells enables rapid establishment of the Gaucher model to capture the key pathology and to test replacement therapy. We expect that this streamlined method can be used to generate human disease models of other LSDs, most of which are still lacking both appropriate human disease models and specific treatments to date.
Collapse
|
14
|
Hendrickx G, Danyukova T, Baranowsky A, Rolvien T, Angermann A, Schweizer M, Keller J, Schröder J, Meyer-Schwesinger C, Muschol N, Paganini C, Rossi A, Amling M, Pohl S, Schinke T. Enzyme replacement therapy in mice lacking arylsulfatase B targets bone-remodeling cells, but not chondrocytes. Hum Mol Genet 2021; 29:803-816. [PMID: 31943020 PMCID: PMC7104678 DOI: 10.1093/hmg/ddaa006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 12/27/2022] Open
Abstract
Mucopolysaccharidosis type VI (MPS-VI), caused by mutational inactivation of the glycosaminoglycan-degrading enzyme arylsulfatase B (Arsb), is a lysosomal storage disorder primarily affecting the skeleton. We have previously reported that Arsb-deficient mice display high trabecular bone mass and impaired skeletal growth. In the present study, we treated them by weekly injection of recombinant human ARSB (rhARSB) to analyze the impact of enzyme replacement therapy (ERT) on skeletal growth and bone remodeling. We found that all bone-remodeling abnormalities of Arsb-deficient mice were prevented by ERT, whereas chondrocyte defects were not. Likewise, histologic analysis of the surgically removed femoral head from an ERT-treated MPS-VI patient revealed that only chondrocytes were pathologically affected. Remarkably, a side-by-side comparison with other cell types demonstrated that chondrocytes have substantially reduced capacity to endocytose rhARSB, together with low expression of the mannose receptor. We finally took advantage of Arsb-deficient mice to establish quantification of chondroitin sulfation for treatment monitoring. Our data demonstrate that bone-remodeling cell types are accessible to systemically delivered rhARSB, whereas the uptake into chondrocytes is inefficient.
Collapse
Affiliation(s)
- Gretl Hendrickx
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tatyana Danyukova
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anke Baranowsky
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Alexandra Angermann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michaela Schweizer
- Department of Electron Microscopy, Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Johannes Keller
- Center for Musculoskeletal Surgery, Charité University Medicine, 10117 Berlin, Germany
| | - Jörg Schröder
- Center for Musculoskeletal Surgery, Charité University Medicine, 10117 Berlin, Germany
| | - Catherine Meyer-Schwesinger
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nicole Muschol
- International Center for Lysosomal Diseases, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Chiara Paganini
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Antonio Rossi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sandra Pohl
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
15
|
Marchetti M, Faggiano S, Mozzarelli A. Enzyme Replacement Therapy for Genetic Disorders Associated with Enzyme Deficiency. Curr Med Chem 2021; 29:489-525. [PMID: 34042028 DOI: 10.2174/0929867328666210526144654] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/23/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
Mutations in human genes might lead to loss of functional proteins, causing diseases. Among these genetic disorders, a large class is associated with the deficiency in metabolic enzymes, resulting in both an increase in the concentration of substrates and a loss in the metabolites produced by the catalyzed reactions. The identification of therapeutic actions based on small molecules represents a challenge to medicinal chemists because the target is missing. Alternative approaches are biology-based, ranging from gene and stem cell therapy, CRISPR/Cas9 technology, distinct types of RNAs, and enzyme replacement therapy (ERT). This review will focus on the latter approach that since the 1990s has been successfully applied to cure many rare diseases, most of them being lysosomal storage diseases or metabolic diseases. So far, a dozen enzymes have been approved by FDA/EMA for lysosome storage disorders and only a few for metabolic diseases. Enzymes for replacement therapy are mainly produced in mammalian cells and some in plant cells and yeasts and are further processed to obtain active, highly bioavailable, less degradable products. Issues still under investigation for the increase in ERT efficacy are the optimization of enzymes interaction with cell membrane and internalization, the reduction in immunogenicity, and the overcoming of blood-brain barrier limitations when neuronal cells need to be targeted. Overall, ERT has demonstrated its efficacy and safety in the treatment of many genetic rare diseases, both saving newborn lives and improving patients' life quality, and represents a very successful example of targeted biologics.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Biopharmanet-TEC Interdepartmental Center, University of Parma, Parco Area delle Scienze, Bldg 33., 43124, Parma, Italy
| | - Serena Faggiano
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Andrea Mozzarelli
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
16
|
Weber P, Thonhofer M, Averill S, Davies GJ, Santana AG, Müller P, Nasseri SA, Offen WA, Pabst BM, Paschke E, Schalli M, Torvisco A, Tschernutter M, Tysoe C, Windischhofer W, Withers SG, Wolfsgruber A, Wrodnigg TM, Stütz AE. Mechanistic Insights into the Chaperoning of Human Lysosomal-Galactosidase Activity: Highly Functionalized Aminocyclopentanes and C-5a-Substituted Derivatives of 4- epi-Isofagomine. Molecules 2020; 25:molecules25174025. [PMID: 32899288 PMCID: PMC7504770 DOI: 10.3390/molecules25174025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/16/2022] Open
Abstract
Glycosidase inhibitors have shown great potential as pharmacological chaperones for lysosomal storage diseases. In light of this, a series of new cyclopentanoid β-galactosidase inhibitors were prepared and their inhibitory and pharmacological chaperoning activities determined and compared with those of lipophilic analogs of the potent β-d-galactosidase inhibitor 4-epi-isofagomine. Structure-activity relationships were investigated by X-ray crystallography as well as by alterations in the cyclopentane moiety such as deoxygenation and replacement by fluorine of a “strategic” hydroxyl group. New compounds have revealed highly promising activities with a range of β-galactosidase-compromised human cell lines and may serve as leads towards new pharmacological chaperones for GM1-gangliosidosis and Morquio B disease.
Collapse
Affiliation(s)
- Patrick Weber
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.W.); (M.T.); (S.A.); (M.S.); (A.W.); (T.M.W.)
| | - Martin Thonhofer
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.W.); (M.T.); (S.A.); (M.S.); (A.W.); (T.M.W.)
| | - Summer Averill
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.W.); (M.T.); (S.A.); (M.S.); (A.W.); (T.M.W.)
| | - Gideon J. Davies
- Department of Chemistry, University of York, Heslington, York YO10 5DD, North Yorkshire, UK; (G.J.D.); (W.A.O.)
| | - Andres Gonzalez Santana
- Chemistry Department, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada; (A.G.S.); (S.A.N.); (C.T.); (S.G.W.)
| | - Philipp Müller
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.M.); (A.T.)
| | - Seyed A. Nasseri
- Chemistry Department, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada; (A.G.S.); (S.A.N.); (C.T.); (S.G.W.)
| | - Wendy A. Offen
- Department of Chemistry, University of York, Heslington, York YO10 5DD, North Yorkshire, UK; (G.J.D.); (W.A.O.)
| | - Bettina M. Pabst
- Laboratory of Metabolic Diseases, Department of Pediatrics, MedUni Graz, Auenbruggerplatz 30, A-8036 Graz, Austria; (B.M.P.); (E.P.); (M.T.); (W.W.)
| | - Eduard Paschke
- Laboratory of Metabolic Diseases, Department of Pediatrics, MedUni Graz, Auenbruggerplatz 30, A-8036 Graz, Austria; (B.M.P.); (E.P.); (M.T.); (W.W.)
| | - Michael Schalli
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.W.); (M.T.); (S.A.); (M.S.); (A.W.); (T.M.W.)
| | - Ana Torvisco
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.M.); (A.T.)
| | - Marion Tschernutter
- Laboratory of Metabolic Diseases, Department of Pediatrics, MedUni Graz, Auenbruggerplatz 30, A-8036 Graz, Austria; (B.M.P.); (E.P.); (M.T.); (W.W.)
| | - Christina Tysoe
- Chemistry Department, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada; (A.G.S.); (S.A.N.); (C.T.); (S.G.W.)
| | - Werner Windischhofer
- Laboratory of Metabolic Diseases, Department of Pediatrics, MedUni Graz, Auenbruggerplatz 30, A-8036 Graz, Austria; (B.M.P.); (E.P.); (M.T.); (W.W.)
| | - Stephen G. Withers
- Chemistry Department, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada; (A.G.S.); (S.A.N.); (C.T.); (S.G.W.)
| | - Andreas Wolfsgruber
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.W.); (M.T.); (S.A.); (M.S.); (A.W.); (T.M.W.)
| | - Tanja M. Wrodnigg
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.W.); (M.T.); (S.A.); (M.S.); (A.W.); (T.M.W.)
| | - Arnold E. Stütz
- Glycogroup, Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria; (P.W.); (M.T.); (S.A.); (M.S.); (A.W.); (T.M.W.)
- Correspondence: ; Tel.: +43-316-873-32079
| |
Collapse
|
17
|
La Cognata V, Guarnaccia M, Polizzi A, Ruggieri M, Cavallaro S. Highlights on Genomics Applications for Lysosomal Storage Diseases. Cells 2020; 9:E1902. [PMID: 32824006 PMCID: PMC7465195 DOI: 10.3390/cells9081902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a heterogeneous group of rare multisystem genetic disorders occurring mostly in infancy and childhood, characterized by a gradual accumulation of non-degraded substrates inside the lysosome. Although the cellular pathogenesis of LSDs is complex and still not fully understood, the approval of disease-specific therapies and the rapid emergence of novel diagnostic methods led to the implementation of extensive national newborn screening (NBS) programs in several countries. In the near future, this will help the development of standardized workflows aimed to more timely diagnose these conditions. Hereby, we report an overview of LSD diagnostic process and treatment strategies, provide an update on the worldwide NBS programs, and discuss the opportunities and challenges arising from genomics applications in screening, diagnosis, and research.
Collapse
Affiliation(s)
- Valentina La Cognata
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, 95126 Catania, Italy; (V.L.C.); (M.G.)
| | - Maria Guarnaccia
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, 95126 Catania, Italy; (V.L.C.); (M.G.)
| | - Agata Polizzi
- Chair of Pediatrics, Department of Educational Sciences, University of Catania, Via Casa Nutrizione, 39, 95124 Catania, Italy;
| | - Martino Ruggieri
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, AOU “Policlinico”, PO “G. Rodolico”, Via S. Sofia, 78, 95123 Catania, Italy;
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, 95126 Catania, Italy; (V.L.C.); (M.G.)
| |
Collapse
|
18
|
Sun Y, Liou B, Chu Z, Fannin V, Blackwood R, Peng Y, Grabowski GA, Davis HW, Qi X. Systemic enzyme delivery by blood-brain barrier-penetrating SapC-DOPS nanovesicles for treatment of neuronopathic Gaucher disease. EBioMedicine 2020; 55:102735. [PMID: 32279952 PMCID: PMC7251241 DOI: 10.1016/j.ebiom.2020.102735] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022] Open
Abstract
Background Enzyme replacement therapy (ERT) can positively affect the visceral manifestations of lysosomal storage diseases (LSDs). However, the exclusion of the intravenous ERT agents from the central nervous system (CNS) prevents direct therapeutic effects. Methods Using a neuronopathic Gaucher disease (nGD) mouse model, CNS-ERT was created using a systemic, non-invasive, and CNS-selective delivery system based on nanovesicles of saposin C (SapC) and dioleoylphosphatidylserine (DOPS) to deliver to CNS cells and tissues the corrective, functional acid β-glucosidase (GCase). Findings Compared to free GCase, human GCase formulated with SapC-DOPS nanovesicles (SapC-DOPS-GCase) was more stable in serum, taken up into cells, mostly by a mannose receptor-independent pathway, and resulted in higher activity in GCase-deficient cells. In contrast to free GCase, SapC-DOPS-GCase nanovesicles penetrated through the blood-brain barrier into the CNS. The CNS targeting was mediated by surface phosphatidylserine (PS) of blood vessel and brain cells. Increased GCase activity and reduced GCase substrate levels were found in the CNS of SapC-DOPS-GCase-treated nGD mice, which showed profound improvement in brain inflammation and neurological phenotypes. Interpretation This first-in-class CNS-ERT approach provides considerable promise of therapeutic benefits for neurodegenerative diseases. Funding This study was supported by the National Institutes of Health grants R21NS 095047 to XQ and YS, R01NS 086134 and UH2NS092981 in part to YS; Cincinnati Children's Hospital Medical Center Research Innovation/Pilot award to YS and XQ; Gardner Neuroscience Institute/Neurobiology Research Center Pilot award to XQ and YS, Hematology-Oncology Programmatic Support from University of Cincinnati and New Drug State Key Project grant 009ZX09102-205 to XQ.
Collapse
Affiliation(s)
- Ying Sun
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Benjamin Liou
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhengtao Chu
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Venette Fannin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rachel Blackwood
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yanyan Peng
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gregory A Grabowski
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Harold W Davis
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Xiaoyang Qi
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
19
|
Poletto E, Baldo G, Gomez-Ospina N. Genome Editing for Mucopolysaccharidoses. Int J Mol Sci 2020; 21:E500. [PMID: 31941077 PMCID: PMC7014411 DOI: 10.3390/ijms21020500] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Genome editing holds the promise of one-off and potentially curative therapies for many patients with genetic diseases. This is especially true for patients affected by mucopolysaccharidoses as the disease pathophysiology is amenable to correction using multiple approaches. Ex vivo and in vivo genome editing platforms have been tested primarily on MSPI and MPSII, with in vivo approaches having reached clinical testing in both diseases. Though we still await proof of efficacy in humans, the therapeutic tools established for these two diseases should pave the way for other mucopolysaccharidoses. Herein, we review the current preclinical and clinical development studies, using genome editing as a therapeutic approach for these diseases. The development of new genome editing platforms and the variety of genetic modifications possible with each tool provide potential applications of genome editing for mucopolysaccharidoses, which vastly exceed the potential of current approaches. We expect that in a not-so-distant future, more genome editing-based strategies will be established, and individual diseases will be treated through multiple approaches.
Collapse
Affiliation(s)
- Edina Poletto
- Gene Therapy Center, Hospital de Clinicas de Porto Alegre, Porto Alegre 90035-007, Brazil; (E.P.); (G.B.)
- Post-Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Guilherme Baldo
- Gene Therapy Center, Hospital de Clinicas de Porto Alegre, Porto Alegre 90035-007, Brazil; (E.P.); (G.B.)
- Post-Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | | |
Collapse
|
20
|
Lachmann RH. Treating lysosomal storage disorders: What have we learnt? J Inherit Metab Dis 2020; 43:125-132. [PMID: 31140601 DOI: 10.1002/jimd.12131] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/26/2019] [Accepted: 05/27/2019] [Indexed: 12/19/2022]
Abstract
The first enzyme replacement therapy (ERT) for a lysosomal storage disorder (LSD) was approved in 1991 and we now have more than 25 years of experience of treating patients with type 1 Gaucher disease. Because of the remarkable success of this therapy, enormous effort and resource has gone into developing other ERTs, for Gaucher (where three different enzyme preparations have now been approved) and for other LSDs. We now have more than 10 years of clinical experience in using ERT to treat Gaucher, Fabry, Pompe and MPS I, II, and VI. This article aims to assess the real-life experience of a selection of these innovative and expensive treatments to see if they have met the high expectations which were set for them when they launched.
Collapse
Affiliation(s)
- Robin H Lachmann
- Charles Dent Metabolic Unit, National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
21
|
Do MA, Levy D, Brown A, Marriott G, Lu B. Targeted delivery of lysosomal enzymes to the endocytic compartment in human cells using engineered extracellular vesicles. Sci Rep 2019; 9:17274. [PMID: 31754156 PMCID: PMC6872767 DOI: 10.1038/s41598-019-53844-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022] Open
Abstract
Targeted delivery of lysosomal enzymes to the endocytic compartment of human cells represents a transformative technology for treating a large family of lysosomal storage diseases (LSDs). Gaucher disease is one of the most common types of LSDs caused by mutations to the lysosomal β-glucocerebrosidase (GBA). Here, we describe a genetic strategy to produce engineered exosomes loaded with GBA in two different spatial configurations for targeted delivery to the endocytic compartment of recipient cells. By fusing human GBA to an exosome-anchoring protein: vesicular stomatitis virus glycoprotein (VSVG), we demonstrate that the chimeric proteins were successfully integrated into exosomes which were secreted as extracellular vesicles (EVs) by producer cells. Isolation and molecular characterization of EVs confirmed that the fusion proteins were loaded onto exosomes without altering their surface markers, particle size or distribution. Further, enzyme-loaded exosomes/EVs added to cultured medium were taken up by recipient cells. Further, the endocytosed exosomes/EVs targeted to endocytic compartments exhibited a significant increase in GBA activity. Together, we have developed a novel method for targeting and delivery of lysosomal enzymes to their natural location: the endocytic compartment of recipient cells. Since exosomes/EVs have an intrinsic ability to cross the blood-brain-barrier, our technology may provide a new approach to treat severe types of LSDs, including Gaucher disease with neurological complications.
Collapse
Affiliation(s)
- Mai Anh Do
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California, 95053, USA
| | - Daniel Levy
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California, 95053, USA
| | - Annie Brown
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California, 95053, USA
| | - Gerard Marriott
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA94720, USA
| | - Biao Lu
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California, 95053, USA.
| |
Collapse
|
22
|
Del Grosso A, Galliani M, Angella L, Santi M, Tonazzini I, Parlanti G, Signore G, Cecchini M. Brain-targeted enzyme-loaded nanoparticles: A breach through the blood-brain barrier for enzyme replacement therapy in Krabbe disease. SCIENCE ADVANCES 2019; 5:eaax7462. [PMID: 31799395 PMCID: PMC6867879 DOI: 10.1126/sciadv.aax7462] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/19/2019] [Indexed: 05/31/2023]
Abstract
Lysosomal storage disorders (LSDs) result from an enzyme deficiency within lysosomes. The systemic administration of the missing enzyme, however, is not effective in the case of LSDs with central nervous system (CNS)-involvement. Here, an enzyme delivery system based on the encapsulation of cross-linked enzyme aggregates (CLEAs) into poly-(lactide-co-glycolide) (PLGA) nanoparticles (NPs) functionalized with brain targeting peptides (Ang2, g7 or Tf2) is demonstrated for Krabbe disease, a neurodegenerative LSD caused by galactosylceramidase (GALC) deficiency. We first synthesize and characterize Ang2-, g7- and Tf2-targeted GALC CLEA NPs. We study NP cell trafficking and capability to reinstate enzymatic activity in vitro. Then, we successfully test our formulations in the Twitcher mouse. We report enzymatic activity measurements in the nervous system and in accumulation districts upon intraperitoneal injections, demonstrating activity recovery in the brain up to the unaffected mice level. Together, these results open new therapeutic perspectives for all LSDs with major CNS-involvement.
Collapse
Affiliation(s)
- Ambra Del Grosso
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Marianna Galliani
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
- Center for Nanotechnology Innovation@NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Lucia Angella
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Melissa Santi
- Center for Nanotechnology Innovation@NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Ilaria Tonazzini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Gabriele Parlanti
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Giovanni Signore
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
- Fondazione Pisana per la Scienza ONLUS, 56017 Pisa, Italy
| | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
23
|
Balakrishnan B, An D, Nguyen V, DeAntonis C, Martini PGV, Lai K. Novel mRNA-Based Therapy Reduces Toxic Galactose Metabolites and Overcomes Galactose Sensitivity in a Mouse Model of Classic Galactosemia. Mol Ther 2019; 28:304-312. [PMID: 31604675 DOI: 10.1016/j.ymthe.2019.09.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/04/2019] [Accepted: 09/12/2019] [Indexed: 11/19/2022] Open
Abstract
Classic galactosemia (CG) is a potentially lethal inborn error of galactose metabolism that results from deleterious mutations in the human galactose-1 phosphate uridylyltransferase (GALT) gene. Previously, we constructed a GalT-/- (GalT-deficient) mouse model that exhibits galactose sensitivity in the newborn mutant pups, reduced fertility in adult females, impaired motor functions, and growth restriction in both sexes. In this study, we tested whether restoration of hepatic GALT activity alone could decrease galactose-1 phosphate (gal-1P) and plasma galactose in the mouse model. The administration of different doses of mouse GalT (mGalT) mRNA resulted in a dose-dependent increase in mGalT protein expression and enzyme activity in the liver of GalT-deficient mice. Single intravenous (i.v.) dose of human GALT (hGALT) mRNA decreased gal-1P in mutant mouse liver and red blood cells (RBCs) within 24 h with low levels maintained for over a week. Repeated i.v. injections increased hepatic GalT expression, nearly normalized gal-1P levels in liver, and decreased gal-1P levels in RBCs and peripheral tissues throughout all doses. Moreover, repeated dosing reduced plasma galactose by 60% or more throughout all four doses. Additionally, a single intraperitoneal dose of hGALT mRNA overcame the galactose sensitivity and promoted the growth in a GalT-/- newborn pup.
Collapse
Affiliation(s)
- Bijina Balakrishnan
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT 84108, USA
| | - Ding An
- Moderna, Inc., Cambridge, MA 02139, USA
| | - Vi Nguyen
- Moderna, Inc., Cambridge, MA 02139, USA
| | | | | | - Kent Lai
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT 84108, USA.
| |
Collapse
|
24
|
Gigliobianco MR, Di Martino P, Deng S, Casadidio C, Censi R. New Advanced Strategies for the Treatment of Lysosomal Diseases Affecting the Central Nervous System. Curr Pharm Des 2019; 25:1933-1950. [DOI: 10.2174/1381612825666190708213159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 11/22/2022]
Abstract
Lysosomal Storage Disorders (LSDs), also known as lysosomal diseases (LDs) are a group of serious genetic diseases characterized by not only the accumulation of non-catabolized compounds in the lysosomes due to the deficiency of specific enzymes which usually eliminate these compounds, but also by trafficking, calcium changes and acidification. LDs mainly affect the central nervous system (CNS), which is difficult to reach for drugs and biological molecules due to the presence of the blood-brain barrier (BBB). While some therapies have proven highly effective in treating peripheral disorders in LD patients, they fail to overcome the BBB. Researchers have developed many strategies to circumvent this problem, for example, by creating carriers for enzyme delivery, which improve the enzyme’s half-life and the overexpression of receptors and transporters in the luminal or abluminal membranes of the BBB. This review aims to successfully examine the strategies developed during the last decade for the treatment of LDs, which mainly affect the CNS. Among the LD treatments, enzyme-replacement therapy (ERT) and gene therapy have proven effective, while nanoparticle, fusion protein, and small molecule-based therapies seem to offer considerable promise to treat the CNS pathology. This work also analyzed the challenges of the study to design new drug delivery systems for the effective treatment of LDs. Polymeric nanoparticles and liposomes are explored from their technological point of view and for the most relevant preclinical studies showing that they are excellent choices to protect active molecules and transport them through the BBB to target specific brain substrates for the treatment of LDs.
Collapse
Affiliation(s)
- Maria R. Gigliobianco
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Piera Di Martino
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Siyuan Deng
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Cristina Casadidio
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| | - Roberta Censi
- School of Pharmacy, University of Camerino, Via A. D'Accoiso, 16, 62032, Camerino MC, Italy
| |
Collapse
|
25
|
Pohl S, Angermann A, Jeschke A, Hendrickx G, Yorgan TA, Makrypidi-Fraune G, Steigert A, Kuehn SC, Rolvien T, Schweizer M, Koehne T, Neven M, Winter O, Velho RV, Albers J, Streichert T, Pestka JM, Baldauf C, Breyer S, Stuecker R, Muschol N, Cox TM, Saftig P, Paganini C, Rossi A, Amling M, Braulke T, Schinke T. The Lysosomal Protein Arylsulfatase B Is a Key Enzyme Involved in Skeletal Turnover. J Bone Miner Res 2018; 33:2186-2201. [PMID: 30075049 DOI: 10.1002/jbmr.3563] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/10/2018] [Accepted: 06/20/2018] [Indexed: 12/24/2022]
Abstract
Skeletal pathologies are frequently observed in lysosomal storage disorders, yet the relevance of specific lysosomal enzymes in bone remodeling cell types is poorly defined. Two lysosomal enzymes, ie, cathepsin K (Ctsk) and Acp5 (also known as tartrate-resistant acid phosphatase), have long been known as molecular marker proteins of differentiated osteoclasts. However, whereas the cysteine protease Ctsk is directly involved in the degradation of bone matrix proteins, the molecular function of Acp5 in osteoclasts is still unknown. Here we show that Acp5, in concert with Acp2 (lysosomal acid phosphatase), is required for dephosphorylation of the lysosomal mannose 6-phosphate targeting signal to promote the activity of specific lysosomal enzymes. Using an unbiased approach we identified the glycosaminoglycan-degrading enzyme arylsulfatase B (Arsb), mutated in mucopolysaccharidosis type VI (MPS-VI), as an osteoclast marker, whose activity depends on dephosphorylation by Acp2 and Acp5. Similar to Acp2/Acp5-/- mice, Arsb-deficient mice display lysosomal storage accumulation in osteoclasts, impaired osteoclast activity, and high trabecular bone mass. Of note, the most prominent lysosomal storage accumulation was observed in osteocytes from Arsb-deficient mice, yet this pathology did not impair production of sclerostin (Sost) and Fgf23. Because the influence of enzyme replacement therapy (ERT) on bone remodeling in MPS-VI is still unknown, we additionally treated Arsb-deficient mice by weekly injection of recombinant human ARSB from 12 to 24 weeks of age. We found that the high bone mass phenotype of Arsb-deficient mice and the underlying bone cell deficits were fully corrected by ERT in the trabecular compartment. Taken together, our results do not only show that the function of Acp5 in osteoclasts is linked to dephosphorylation and activation of lysosomal enzymes, they also provide an important proof-of-principle for the feasibility of ERT to correct bone cell pathologies in lysosomal storage disorders. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Sandra Pohl
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandra Angermann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Jeschke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gretl Hendrickx
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georgia Makrypidi-Fraune
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Steigert
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja C Kuehn
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Department of Electron Microscopy, Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Koehne
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Orthodontics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olga Winter
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renata Voltolini Velho
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joachim Albers
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Streichert
- Department of Clinical Chemistry, University Hospital Cologne, Cologne, Germany
| | - Jan M Pestka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Baldauf
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Breyer
- Department of Orthopedics, Children's Hospital Hamburg-Altona, Hamburg, Germany
| | - Ralf Stuecker
- Department of Orthopedics, Children's Hospital Hamburg-Altona, Hamburg, Germany
| | - Nicole Muschol
- Department of Electron Microscopy, Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timothy M Cox
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Chiara Paganini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Antonio Rossi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Braulke
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
26
|
Wissner R, Steinauer A, Knox SL, Thompson AD, Schepartz A. Fluorescence Correlation Spectroscopy Reveals Efficient Cytosolic Delivery of Protein Cargo by Cell-Permeant Miniature Proteins. ACS CENTRAL SCIENCE 2018; 4:1379-1393. [PMID: 30410976 PMCID: PMC6202653 DOI: 10.1021/acscentsci.8b00446] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Indexed: 05/21/2023]
Abstract
New methods for delivering proteins into the cytosol of mammalian cells are being reported at a rapid pace. Differentiating between these methods in a quantitative manner is difficult, however, as most assays for evaluating cytosolic protein delivery are qualitative and indirect and thus often misleading. Here we make use of fluorescence correlation spectroscopy (FCS) to determine with precision and accuracy the relative efficiencies with which seven different previously reported "cell-penetrating peptides" (CPPs) transport a model protein cargo-the self-labeling enzyme SNAP-tag-beyond endosomal membranes and into the cytosol. Using FCS, we discovered that the miniature protein ZF5.3 is an exceptional vehicle for delivering SNAP-tag to the cytosol. When delivered by ZF5.3, SNAP-tag can achieve a cytosolic concentration as high as 250 nM, generally at least 2-fold and as much as 6-fold higher than any other CPP evaluated. Additionally, we show that ZF5.3 can be fused to a second enzyme cargo-the engineered peroxidase APEX2-and reliably delivers the active enzyme to the cell interior. As FCS allows one to realistically assess the relative merits of protein transduction domains, we anticipate that it will greatly accelerate the identification, evaluation, and optimization of strategies to deliver large, intact proteins to intracellular locales.
Collapse
Affiliation(s)
- Rebecca
F. Wissner
- Department
of Chemistry, and Department of Molecular, Cellular, and Developmental
Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Angela Steinauer
- Department
of Chemistry, and Department of Molecular, Cellular, and Developmental
Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Susan L. Knox
- Department
of Chemistry, and Department of Molecular, Cellular, and Developmental
Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Alexander D. Thompson
- Department
of Chemistry, and Department of Molecular, Cellular, and Developmental
Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Alanna Schepartz
- Department
of Chemistry, and Department of Molecular, Cellular, and Developmental
Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| |
Collapse
|
27
|
Abstract
Glycosylation is one of the most prevalent posttranslational modifications that profoundly affects the structure and functions of proteins in a wide variety of biological recognition events. However, the structural complexity and heterogeneity of glycoproteins, usually resulting from the variations of glycan components and/or the sites of glycosylation, often complicates detailed structure-function relationship studies and hampers the therapeutic applications of glycoproteins. To address these challenges, various chemical and biological strategies have been developed for producing glycan-defined homogeneous glycoproteins. This review highlights recent advances in the development of chemoenzymatic methods for synthesizing homogeneous glycoproteins, including the generation of various glycosynthases for synthetic purposes, endoglycosidase-catalyzed glycoprotein synthesis and glycan remodeling, and direct enzymatic glycosylation of polypeptides and proteins. The scope, limitation, and future directions of each method are discussed.
Collapse
Affiliation(s)
- Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
28
|
Meng B, Wang J, Wang Q, Serianni AS, Pan Q. Synthesis of high-mannose oligosaccharides containing mannose-6-phosphate residues using regioselective glycosylation. Carbohydr Res 2018; 467:23-32. [PMID: 30075362 PMCID: PMC6121786 DOI: 10.1016/j.carres.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 11/23/2022]
Abstract
Molecular recognition of mannose-6-phosphate (M6P)-modified oligosaccharides by transmembrane M6P receptors is a key signaling event in lysosomal protein trafficking in vivo. Access to M6P-containing high-mannose N-glycans is essential to achieving a thorough understanding of the M6P ligand-receptor recognition process. Herein we report the application of a versatile and reliable chemical strategy to prepare asymmetric di-antennary M6P-tagged high-mannose oligosaccharides in >20% overall yield and in high purity (>98%). Regioselective chemical glycosylation coupled with effective phosphorylation and product purification protocols were applied to rapidly assemble these oligosaccharides. The development of this synthetic strategy simplifies the preparation of M6P-tagged high-mannose oligosaccharides, which will improve access to these compounds to study their structures and biological functions.
Collapse
Affiliation(s)
- Bo Meng
- Omicron Biochemicals, Inc., 115 South Hill Street, South Bend, IN, 46617-2701, USA
| | - Jun Wang
- Omicron Biochemicals, Inc., 115 South Hill Street, South Bend, IN, 46617-2701, USA
| | - Quanli Wang
- Omicron Biochemicals, Inc., 115 South Hill Street, South Bend, IN, 46617-2701, USA
| | - Anthony S Serianni
- Omicron Biochemicals, Inc., 115 South Hill Street, South Bend, IN, 46617-2701, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556-5670, USA
| | - Qingfeng Pan
- Omicron Biochemicals, Inc., 115 South Hill Street, South Bend, IN, 46617-2701, USA.
| |
Collapse
|
29
|
Abstract
Lysosomal storage diseases (LSDs) are a heterogeneous group of genetic disorders caused by defects in lysosomal function that lead to multiorgan system damage. Due to wide clinical variability within even a single disorder, making a diagnosis can be difficult and identification may be delayed. Enzyme replacement therapy (ERT) was first approved as a treatment for the LSD Gaucher disease in 1991. ERT development for other LSDs followed, and ERT is currently approved for eight LSDs in the United States. ERT may help slow progression and improve clinical symptoms, but it cannot affect neurologic features due to its inability to cross the blood-brain barrier. Additional therapies for LSDs that have been investigated include stem cell transplants, gene therapy, small molecule approaches, and genome editing. Although newer approaches seem promising, there is no "cure" for any LSDs, and management remains focused on early diagnosis and treatment. [Pediatr Ann. 2018;47(5):e191-e197.].
Collapse
|
30
|
Abstract
Enzymes are attractive as immunotherapeutics because they can catalyze shifts in the local availability of immunostimulatory and immunosuppressive signals. Clinical success of enzyme immunotherapeutics frequently hinges upon achieving sustained biocatalysis over relevant time scales. The time scale and location of biocatalysis are often dictated by the location of the substrate. For example, therapeutic enzymes that convert substrates distributed systemically are typically designed to have a long half-life in circulation, whereas enzymes that convert substrates localized to a specific tissue or cell population can be more effective when designed to accumulate at the target site. This Topical Review surveys approaches to improve enzyme immunotherapeutic efficacy via chemical modification, encapsulation, and immobilization that increases enzyme accumulation at target sites or extends enzyme half-life in circulation. Examples provided illustrate "replacement therapies" to restore deficient enzyme function, as well as "enhancement therapies" that augment native enzyme function via supraphysiologic doses. Existing FDA-approved enzyme immunotherapies are highlighted, followed by discussion of emerging experimental strategies such as those designed to enhance antitumor immunity or resolve inflammation.
Collapse
Affiliation(s)
- Shaheen A Farhadi
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Evelyn Bracho-Sanchez
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Sabrina L Freeman
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| |
Collapse
|
31
|
Shemesh E, Deroma L, Hendriksz CJ, Hollak C, Krishan A. Enzyme replacement therapy for mucopolysaccharidosis type IV (Morquio syndrome). Hippokratia 2018. [DOI: 10.1002/14651858.cd012961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Elad Shemesh
- Institute of Child Health, University of Liverpool; c/o Cochrane Cystic Fibrosis & Genetic Disorders Review Group; Alder Hey Children's NHS Foundation Trust Eaton Road Liverpool UK L12 2AP
| | - Laura Deroma
- University Hospital "Santa Maria della Misericordia"; Regional Coordinator Centre for Rare Diseases; Piazzale Santa Maria della Misericordia 15 Udine Italy 33100
| | - Christian J Hendriksz
- Salford Royal NHS Foundation Trust; Adult Inherited Metabolic Disorders, The Mark Holland Metabolic Unit; Stott Lane Salford UK M6 8HD
| | - Carla Hollak
- Academic Medical Center; Department of Internal Medicine, Division of Endocrinology & Metabolism; Postbus 22660 Amsterdam Netherlands 1100 DD
| | - Ashma Krishan
- University of Edinburgh; Edinburgh Clinical Trials Unit; Outpatients Building, 2nd Floor Western General Hospital, Crewe Road South Edinburgh UK EH4 2XU
| |
Collapse
|
32
|
Jin ZC, Kitajima T, Dong W, Huang YF, Ren WW, Guan F, Chiba Y, Gao XD, Fujita M. Genetic disruption of multiple α1,2-mannosidases generates mammalian cells producing recombinant proteins with high-mannose-type N-glycans. J Biol Chem 2018; 293:5572-5584. [PMID: 29475941 DOI: 10.1074/jbc.m117.813030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/04/2018] [Indexed: 12/27/2022] Open
Abstract
Recombinant therapeutic proteins are becoming very important pharmaceutical agents for treating intractable diseases. Most biopharmaceutical proteins are produced in mammalian cells because this ensures correct folding and glycosylation for protein stability and function. However, protein production in mammalian cells has several drawbacks, including heterogeneity of glycans attached to the produced protein. In this study, we established cell lines with high-mannose-type N-linked, low-complexity glycans. We first knocked out two genes encoding Golgi mannosidases (MAN1A1 and MAN1A2) in HEK293 cells. Single knockout (KO) cells did not exhibit changes in N-glycan structures, whereas double KO cells displayed increased high-mannose-type and decreased complex-type glycans. In our effort to eliminate the remaining complex-type glycans, we found that knocking out a gene encoding the endoplasmic reticulum mannosidase I (MAN1B1) in the double KO cells reduced most of the complex-type glycans. In triple KO (MAN1A1, MAN1A2, and MAN1B1) cells, Man9GlcNAc2 and Man8GlcNAc2 were the major N-glycan structures. Therefore, we expressed two lysosomal enzymes, α-galactosidase-A and lysosomal acid lipase, in the triple KO cells and found that the glycans on these enzymes were sensitive to endoglycosidase H treatment. The N-glycan structures on recombinant proteins expressed in triple KO cells were simplified and changed from complex types to high-mannose types at the protein level. Our results indicate that the triple KO HEK293 cells are suitable for producing recombinant proteins, including lysosomal enzymes with high-mannose-type N-glycans.
Collapse
Affiliation(s)
- Ze-Cheng Jin
- From the Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Toshihiko Kitajima
- From the Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Weijie Dong
- the College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning, China, and
| | - Yi-Fan Huang
- From the Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei-Wei Ren
- From the Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Feng Guan
- From the Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yasunori Chiba
- the Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Central 2, 1-1-1 Umezono, Tsukuba, Ibaraki 305-8568, Japan
| | - Xiao-Dong Gao
- From the Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China,
| | - Morihisa Fujita
- From the Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China,
| |
Collapse
|
33
|
Park K. Efficient therapy of Pompe disease by an acid α-glucosidase conjugate. J Control Release 2018; 269:441-442. [PMID: 29290222 DOI: 10.1016/j.jconrel.2017.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kinam Park
- Purdue University Biomedical Engineering and PharmaceuticsWest Lafayette, IN 47907, USA.
| |
Collapse
|
34
|
Tong W, Dwyer CA, Thacker BE, Glass CA, Brown JR, Hamill K, Moremen KW, Sarrazin S, Gordts PLSM, Dozier LE, Patrick GN, Tor Y, Esko JD. Guanidinylated Neomycin Conjugation Enhances Intranasal Enzyme Replacement in the Brain. Mol Ther 2017; 25:2743-2752. [PMID: 28958576 PMCID: PMC5768556 DOI: 10.1016/j.ymthe.2017.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/20/2022] Open
Abstract
Iduronidase (IDUA)-deficient mice accumulate glycosaminoglycans in cells and tissues and exhibit many of the same neuropathological symptoms of patients suffering from Mucopolysaccharidosis I. Intravenous enzyme-replacement therapy for Mucopolysaccharidosis I ameliorates glycosaminoglycan storage and many of the somatic aspects of the disease but fails to treat neurological symptoms due to poor transport across the blood-brain barrier. In this study, we examined the delivery of IDUA conjugated to guanidinoneomycin (GNeo), a molecular transporter. GNeo-IDUA and IDUA injected intravenously resulted in reduced hepatic glycosaminoglycan accumulation but had no effect in the brain due to fast clearance from the circulation. In contrast, intranasally administered GNeo-IDUA entered the brain rapidly. Repetitive intranasal treatment with GNeo-IDUA reduced glycosaminoglycan storage, lysosome size and number, and neurodegenerative astrogliosis in the olfactory bulb and primary somatosensory cortex, whereas IDUA was less effective. The enhanced efficacy of GNeo-IDUA was not the result of increased nose-to-brain delivery or enzyme stability, but rather due to more efficient uptake into neurons and astrocytes. GNeo conjugation also enhanced glycosaminoglycan clearance by intranasally delivered sulfamidase to the brain of sulfamidase-deficient mice, a model of Mucopolysaccharidosis IIIA. These findings suggest the general utility of the guanidinoglycoside-based delivery system for restoring missing lysosomal enzymes in the brain.
Collapse
Affiliation(s)
- Wenyong Tong
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Chrissa A Dwyer
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Bryan E Thacker
- TEGA Therapeutics, Inc., 9500 Gilman Drive, La Jolla, CA 92093-0713, USA
| | - Charles A Glass
- TEGA Therapeutics, Inc., 9500 Gilman Drive, La Jolla, CA 92093-0713, USA
| | - Jillian R Brown
- TEGA Therapeutics, Inc., 9500 Gilman Drive, La Jolla, CA 92093-0713, USA
| | - Kristina Hamill
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, USA
| | - Kelley W Moremen
- Department of Biochemistry, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Stéphane Sarrazin
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Philip L S M Gordts
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Lara E Dozier
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0366 USA
| | - Gentry N Patrick
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0366 USA
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0687, USA.
| |
Collapse
|
35
|
Platt FM. Emptying the stores: lysosomal diseases and therapeutic strategies. Nat Rev Drug Discov 2017; 17:133-150. [PMID: 29147032 DOI: 10.1038/nrd.2017.214] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lysosomal storage disorders (LSDs) - designated as 'orphan' diseases - are inborn errors of metabolism caused by defects in genes that encode proteins involved in various aspects of lysosomal homeostasis. For many years, LSDs were viewed as unattractive targets for the development of therapies owing to their low prevalence. However, the development and success of the first commercial biologic therapy for an LSD - enzyme replacement therapy for type 1 Gaucher disease - coupled with regulatory incentives rapidly catalysed commercial interest in therapeutically targeting LSDs. Despite ongoing challenges, various therapeutic strategies for LSDs now exist, with many agents approved, undergoing clinical trials or in preclinical development.
Collapse
Affiliation(s)
- Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
36
|
Kelly JM, Gross AL, Martin DR, Byrne ME. Polyethylene glycol-b-poly(lactic acid) polymersomes as vehicles for enzyme replacement therapy. Nanomedicine (Lond) 2017; 12:2591-2606. [PMID: 29111890 DOI: 10.2217/nnm-2017-0221] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM Polymersomes are created to deliver an enzyme-based therapy to the brain in lysosomal storage disease patients. MATERIALS & METHODS Polymersomes are formed via the injection method using poly(ethylene glycol)-b-poly(lactic acid) (PEGPLA) and bound to apolipoprotein E, to create a brain-targeted delivery vehicle. RESULTS Polymersomes have a smallest average diameter of 145 ± 21 nm and encapsulate β-galactosidase at 72.0 ± 12.2% efficiency. PEGPLA polymersomes demonstrate limited release at physiologic pH (7.4), with a burst release at the acidic pH (4.8) of the lysosome. PEGPLA polymersomes facilitate delivery of active β-galactosidase to an in vitro model of GM1 gangliosidosis. CONCLUSION The foundation has been laid for testing of PEGPLA polymersomes to deliver enzymatic treatments to the brain in lysosomal storage disorders for the first time.
Collapse
Affiliation(s)
- Jessica M Kelly
- Biomimetic & Biohybrid Materials, Biomedical Devices, & Drug Delivery Laboratories, Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL 36849, USA.,Scott Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.,US Department of Education GAANN Graduate Fellowship Program in Biological & Pharmaceutical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Amanda L Gross
- Scott Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.,Department of Anatomy, Physiology, & Pharmacology, Scott Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Douglas R Martin
- Scott Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.,US Department of Education GAANN Graduate Fellowship Program in Biological & Pharmaceutical Engineering, Auburn University, Auburn, AL 36849, USA.,Department of Anatomy, Physiology, & Pharmacology, Scott Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Mark E Byrne
- Biomimetic & Biohybrid Materials, Biomedical Devices, & Drug Delivery Laboratories, Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL 36849, USA.,US Department of Education GAANN Graduate Fellowship Program in Biological & Pharmaceutical Engineering, Auburn University, Auburn, AL 36849, USA.,Biomimetic & Biohybrid Materials, Biomedical Devices, & Drug Delivery Laboratories, Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
37
|
Schielen PCJI, Kemper EA, Gelb MH. Newborn Screening for Lysosomal Storage Diseases: A Concise Review of the Literature on Screening Methods, Therapeutic Possibilities and Regional Programs. Int J Neonatal Screen 2017; 3:6. [PMID: 28730181 PMCID: PMC5515486 DOI: 10.3390/ijns3020006] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Newborn screening for lysosomal storage diseases (LSDs) is increasingly being considered as an option. The development of analytical screening methods, of second-tier methods, and of therapeutic possibilities, are paving the way for routine screening for LSDs in the coming years. Here, we give a brief description of the current status quo, what screening methods are currently available or are in the pipeline, what is the current status of therapeutic possibilities for LSDs, what LSDs are the most obvious candidates for introduction in screening programs, and what LSDs are already part of regional or national pilot or routine screening programs worldwide.
Collapse
Affiliation(s)
- Peter C. J. I. Schielen
- Reference Laboratory for Neonatal Screening, Centre for Infectious Diseases Research, Diagnostics and Screening, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
- Correspondence: ; Tel.: +31-30-274-3534
| | - Evelien A. Kemper
- Department of Clinical Chemistry, IJsselland Hospital, 2906 ZC Capelle ad IJssel, The Netherlands
| | - Michael H. Gelb
- Departments of Chemistry and Biochemistry, University of Washington, Seattle, WD 98195, USA
| |
Collapse
|
38
|
Emerging therapies for neuropathic lysosomal storage disorders. Prog Neurobiol 2017; 152:166-180. [DOI: 10.1016/j.pneurobio.2016.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 09/29/2016] [Accepted: 10/02/2016] [Indexed: 12/18/2022]
|
39
|
Wang Y, MacDonald RG, Thinakaran G, Kar S. Insulin-Like Growth Factor-II/Cation-Independent Mannose 6-Phosphate Receptor in Neurodegenerative Diseases. Mol Neurobiol 2017; 54:2636-2658. [PMID: 26993302 PMCID: PMC5901910 DOI: 10.1007/s12035-016-9849-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/09/2016] [Indexed: 12/11/2022]
Abstract
The insulin-like growth factor II/mannose 6-phosphate (IGF-II/M6P) receptor is a multifunctional single transmembrane glycoprotein. Recent studies have advanced our understanding of the structure, ligand-binding properties, and trafficking of the IGF-II/M6P receptor. This receptor has been implicated in a variety of important cellular processes including growth and development, clearance of IGF-II, proteolytic activation of enzymes, and growth factor precursors, in addition to its well-known role in the delivery of lysosomal enzymes. The IGF-II/M6P receptor, distributed widely in the central nervous system, has additional roles in mediating neurotransmitter release and memory enhancement/consolidation, possibly through activating IGF-II-related intracellular signaling pathways. Recent studies suggest that overexpression of the IGF-II/M6P receptor may have an important role in regulating the levels of transcripts and proteins involved in the development of Alzheimer's disease (AD)-the prevalent cause of dementia affecting the elderly population in our society. It is reported that IGF-II/M6P receptor overexpression can increase the levels/processing of amyloid precursor protein leading to the generation of β-amyloid peptide, which is associated with degeneration of neurons and subsequent development of AD pathology. Given the significance of the receptor in mediating the transport and functioning of the lysosomal enzymes, it is being considered for therapeutic delivery of enzymes to the lysosomes to treat lysosomal storage disorders. Notwithstanding these results, additional studies are required to validate and fully characterize the function of the IGF-II/M6P receptor in the normal brain and its involvement in various neurodegenerative disorders including AD. It is also critical to understand the interaction between the IGF-II/M6P receptor and lysosomal enzymes in neurodegenerative processes, which may shed some light on developing approaches to detect and prevent neurodegeneration through the dysfunction of the receptor and the endosomal-lysosomal system.
Collapse
Affiliation(s)
- Y Wang
- Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2M8, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - R G MacDonald
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - G Thinakaran
- Departments of Neurobiology, Neurology, and Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - S Kar
- Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2M8, Canada.
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada.
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB, T6G 2M8, Canada.
| |
Collapse
|
40
|
Liu L, Lee WS, Doray B, Kornfeld S. Engineering of GlcNAc-1-Phosphotransferase for Production of Highly Phosphorylated Lysosomal Enzymes for Enzyme Replacement Therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 5:59-65. [PMID: 28480305 PMCID: PMC5415318 DOI: 10.1016/j.omtm.2017.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/22/2017] [Indexed: 12/27/2022]
Abstract
Several lysosomal enzymes currently used for enzyme replacement therapy in patients with lysosomal storage diseases contain very low levels of mannose 6-phosphate, limiting their uptake via mannose 6-phosphate receptors on the surface of the deficient cells. These enzymes are produced at high levels by mammalian cells and depend on endogenous GlcNAc-1-phosphotransferase α/β precursor to phosphorylate the mannose residues on their glycan chains. We show that co-expression of an engineered truncated GlcNAc-1-phosphotransferase α/β precursor and the lysosomal enzyme of interest in the producing cells resulted in markedly increased phosphorylation and cellular uptake of the secreted lysosomal enzyme. This method also results in the production of highly phosphorylated acid β-glucocerebrosidase, a lysosomal enzyme that normally has just trace amounts of this modification.
Collapse
Affiliation(s)
- Lin Liu
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wang-Sik Lee
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Balraj Doray
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stuart Kornfeld
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
41
|
Aguisanda F, Thorne N, Zheng W. Targeting Wolman Disease and Cholesteryl Ester Storage Disease: Disease Pathogenesis and Therapeutic Development. Curr Chem Genom Transl Med 2017; 11:1-18. [PMID: 28401034 PMCID: PMC5362971 DOI: 10.2174/2213988501711010001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 09/20/2016] [Accepted: 11/23/2016] [Indexed: 12/13/2022] Open
Abstract
Wolman disease (WD) and cholesteryl ester storage disease (CESD) are lysosomal storage diseases (LSDs) caused by a deficiency in lysosomal acid lipase (LAL) due to mutations in the LIPA gene. This enzyme is critical to the proper degradation of cholesterol in the lysosome. LAL function is completely lost in WD while some residual activity remains in CESD. Both are rare diseases with an incidence rate of less than 1/100,000 births for WD and approximate 2.5/100,000 births for CESD. Clinical manifestation of WD includes hepatosplenomegaly, calcified adrenal glands, severe malabsorption and a failure to thrive. As in CESD, histological analysis of WD tissues reveals the accumulation of triglycerides (TGs) and esterified cholesterol (EC) in cellular lysosomes. However, the clinical presentation of CESD is less severe and more variable than WD. This review is to provide an overview of the disease pathophysiology and the current state of therapeutic development for both of WD and CESD. The review will also discuss the application of patient derived iPSCs for further drug discovery.
Collapse
Affiliation(s)
- Francis Aguisanda
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3370, USA
| | - Natasha Thorne
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3370, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3370, USA
| |
Collapse
|
42
|
Kim JH, Chi YH, Kim GH, Yoo HW, Lee JH. Long-term clinical course of a patient with mucopolysaccharidosis type IIIB. KOREAN JOURNAL OF PEDIATRICS 2016; 59:S37-S40. [PMID: 28018442 PMCID: PMC5177708 DOI: 10.3345/kjp.2016.59.11.s37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 02/02/2015] [Accepted: 02/24/2015] [Indexed: 01/14/2023]
Abstract
Mucopolysaccharidosis type III (MPS III) is a rare genetic disorder caused by lysosomal storage of heparan sulfate. MPS IIIB results from a deficiency in the enzyme alpha-N-acetyl-D-glucosaminidase (NAGLU). Affected patients begin showing behavioral changes, progressive profound mental retardation, and severe disability from the age of 2 to 6 years. We report a patient with MPS IIIB with a long-term follow-up duration. He showed normal development until 3 years. Subsequently, he presented behavioral changes, sleep disturbance, and progressive motor dysfunction. He had been hospitalized owing to recurrent pneumonia and epilepsy with severe cognitive dysfunction. The patient had compound heterozygous c.1444C>T (p.R482W) and c.1675G>T (p.D559Y) variants of NAGLU. Considering that individuals with MPS IIIB have less prominent facial features and skeletal changes, evaluation of long-term clinical course is important for diagnosis. Although no effective therapies for MPS IIIB have been developed yet, early and accurate diagnosis can provide important information for family planning in families at risk of the disorder.
Collapse
Affiliation(s)
- Ja Hye Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Yang Hyun Chi
- Department of Pediatrics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Gu-Hwan Kim
- Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea.; Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Jun Hwa Lee
- Department of Pediatrics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| |
Collapse
|
43
|
Roncador A, Oppici E, Talelli M, Pariente AN, Donini M, Dusi S, Voltattorni CB, Vicent MJ, Cellini B. Use of polymer conjugates for the intraperoxisomal delivery of engineered human alanine:glyoxylate aminotransferase as a protein therapy for primary hyperoxaluria type I. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:897-907. [PMID: 27993722 DOI: 10.1016/j.nano.2016.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/25/2016] [Accepted: 12/07/2016] [Indexed: 01/22/2023]
Abstract
Alanine:glyoxylate aminotransferase (AGT) is a liver peroxisomal enzyme whose deficit causes the rare disorder Primary Hyperoxaluria Type I (PH1). We now describe the conjugation of poly(ethylene glycol)-co-poly(L-glutamic acid) (PEG-PGA) block-co-polymer to AGT via the formation of disulfide bonds between the polymer and solvent-exposed cysteine residues of the enzyme. PEG-PGA conjugation did not affect AGT structural/functional properties and allowed the enzyme to be internalized in a cellular model of PH1 and to restore glyoxylate-detoxification. The insertion of the C387S/K390S amino acid substitutions, known to favor interaction with the peroxisomal import machinery, reduced conjugation efficiency, but endowed conjugates with the ability to reach the peroxisomal compartment. These results, along with the finding that conjugates are hemocompatible, stable in plasma, and non-immunogenic, hold promise for the development of polypeptide-based AGT conjugates as a therapeutic option for PH1 patients and represent the base for applications to other diseases related to deficits in peroxisomal proteins.
Collapse
Affiliation(s)
- Alessandro Roncador
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy
| | - Elisa Oppici
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy
| | - Marina Talelli
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Amaya Niño Pariente
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Marta Donini
- Department of Medicine, Section of General Pathology, University of Verona, Verona (VR), Italy
| | - Stefano Dusi
- Department of Medicine, Section of General Pathology, University of Verona, Verona (VR), Italy
| | - Carla Borri Voltattorni
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy
| | - María J Vicent
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain.
| | - Barbara Cellini
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy.
| |
Collapse
|
44
|
Stütz AE, Wrodnigg TM. Carbohydrate-Processing Enzymes of the Lysosome: Diseases Caused by Misfolded Mutants and Sugar Mimetics as Correcting Pharmacological Chaperones. Adv Carbohydr Chem Biochem 2016; 73:225-302. [PMID: 27816107 DOI: 10.1016/bs.accb.2016.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lysosomal storage diseases are hereditary disorders caused by mutations on genes encoding for one of the more than fifty lysosomal enzymes involved in the highly ordered degradation cascades of glycans, glycoconjugates, and other complex biomolecules in the lysosome. Several of these metabolic disorders are associated with the absence or the lack of activity of carbohydrate-processing enzymes in this cell compartment. In a recently introduced therapy concept, for susceptible mutants, small substrate-related molecules (so-called pharmacological chaperones), such as reversible inhibitors of these enzymes, may serve as templates for the correct folding and transport of the respective protein mutant, thus improving its concentration and, consequently, its enzymatic activity in the lysosome. Carbohydrate-processing enzymes in the lysosome, related lysosomal diseases, and the scope and limitations of reported reversible inhibitors as pharmacological chaperones are discussed with a view to possibly extending and improving research efforts in this area of orphan diseases.
Collapse
Affiliation(s)
- Arnold E Stütz
- Glycogroup, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Tanja M Wrodnigg
- Glycogroup, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| |
Collapse
|
45
|
Yamaguchi T, Amin MN, Toonstra C, Wang LX. Chemoenzymatic Synthesis and Receptor Binding of Mannose-6-Phosphate (M6P)-Containing Glycoprotein Ligands Reveal Unusual Structural Requirements for M6P Receptor Recognition. J Am Chem Soc 2016; 138:12472-85. [PMID: 27500601 DOI: 10.1021/jacs.6b05762] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mannose-6-phosphate (M6P)-terminated oligosaccharides are important signals for M6P-receptor-mediated targeting of newly synthesized hydrolases from Golgi to lysosomes, but the precise structural requirement for the M6P ligand-receptor recognition has not been fully understood due to the difficulties in obtaining homogeneous M6P-containing glycoproteins. We describe here a chemoenzymatic synthesis of homogeneous phosphoglycoproteins carrying natural M6P-containing N-glycans. The method includes the chemical synthesis of glycan oxazolines with varied number and location of the M6P moieties and their transfer to the GlcNAc-protein by an endoglycosynthase to provide homogeneous M6P-containing glycoproteins. Simultaneous attachment of two M6P-oligosaccahrides to a cyclic polypeptide was also accomplished to yield bivalent M6P-glycopeptides. Surface plasmon resonance binding studies reveal that a single M6P moiety located at the low α-1,3-branch of the oligomannose context is sufficient for a high-affinity binding to receptor CI-MPR, while the presence of a M6P moiety at the α-1,6-branch is dispensable. In addition, a binding study with the bivalent cyclic and linear polypeptides reveals that a close proximity of two M6P-oligosaccharide ligands is critical to achieve high affinity for the CI-MPR receptor. Taken together, the present study indicates that the location and valency of the M6P moieties and the right oligosaccharide context are all critical for high-affinity binding with the major M6P receptor. The chemoenzymatic method described here provides a new avenue for glycosylation remodeling of recombinant enzymes to enhance the uptake and delivery of enzymes to lysosomes in enzyme replacement therapy for the treatment of lysosomal storage diseases.
Collapse
Affiliation(s)
- Takahiro Yamaguchi
- Institute of Human Virology, University of Maryland School of Medicine , Baltimore, Maryland 21201, United States
| | - Mohammed N Amin
- Institute of Human Virology, University of Maryland School of Medicine , Baltimore, Maryland 21201, United States.,Department of Chemistry and Biochemistry, University of Maryland , College Park, Maryland 20742, United States
| | - Christian Toonstra
- Department of Chemistry and Biochemistry, University of Maryland , College Park, Maryland 20742, United States
| | - Lai-Xi Wang
- Institute of Human Virology, University of Maryland School of Medicine , Baltimore, Maryland 21201, United States.,Department of Chemistry and Biochemistry, University of Maryland , College Park, Maryland 20742, United States
| |
Collapse
|
46
|
Barkhuizen M, Anderson DG, Grobler AF. Advances in GBA-associated Parkinson's disease--Pathology, presentation and therapies. Neurochem Int 2015; 93:6-25. [PMID: 26743617 DOI: 10.1016/j.neuint.2015.12.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/29/2015] [Accepted: 12/04/2015] [Indexed: 12/27/2022]
Abstract
GBA mutations are to date the most common genetic risk factor for Parkinson's disease. The GBA gene encodes the lysomal hydrolase glucocerebrosidase. Whilst bi-allelic GBA mutations cause Gaucher disease, both mono- and bi-allelic mutations confer risk for Parkinson's disease. Clinically, Parkinson's disease patients with GBA mutations resemble idiopathic Parkinson's disease patients. However, these patients have a modest reduction in age-of-onset of disease and a greater incidence of cognitive decline. In some cases, GBA mutations are also responsible for familial Parkinson's disease. The accumulation of α-synuclein into Lewy bodies is the central neuropathological hallmark of Parkinson's disease. Pathologic GBA mutations reduce enzymatic function. A reduction in glucocerebrosidase function increases α-synuclein levels and propagation, which in turn inhibits glucocerebrosidase in a feed-forward cascade. This cascade is central to the neuropathology of GBA-associated Parkinson's disease. The lysosomal integral membrane protein type-2 is necessary for normal glucocerebrosidase function. Glucocerebrosidase dysfunction also increases in the accumulation of β-amyloid and amyloid-precursor protein, oxidative stress, neuronal susceptibility to metal ions, microglial and immune activation. These factors contribute to neuronal death. The Mendelian Parkinson's disease genes, Parkin and ATP13A2, intersect with glucocerebrosidase. These factors sketch a complex circuit of GBA-associated neuropathology. To clinically interfere with this circuit, central glucocerebrosidase function must be improved. Strategies based on reducing breakdown of mutant glucocerebrosidase and increasing the fraction that reaches the lysosome has shown promise. Breakdown can be reduced by interfering with the ability of heat-shock proteins to recognize mutant glucocerebrosidase. This underlies the therapeutic efficacy of certain pharmacological chaperones and histone deacetylase inhibitors. These therapies are promising for Parkinson's disease, regardless of mutation status. Recently, there has been a boom in studies investigating the role of glucocerebrosidase in the pathology of Parkinson's disease. This merits a comprehensive review of the current cell biological processes and pathological pictures involving Parkinson's disease associated with GBA mutations.
Collapse
Affiliation(s)
- Melinda Barkhuizen
- DST/NWU Preclinical Drug Development Platform, North-West University, Potchefstroom, 2520, South Africa; Department of Paediatrics, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6229, The Netherlands.
| | - David G Anderson
- Department of Neurology, Witwatersrand University Donald Gordon Medical Centre, Parktown, Johannesburg, 2193, South Africa
| | - Anne F Grobler
- DST/NWU Preclinical Drug Development Platform, North-West University, Potchefstroom, 2520, South Africa
| |
Collapse
|
47
|
Sánchez-Sánchez L, Tapia-Moreno A, Juarez-Moreno K, Patterson DP, Cadena-Nava RD, Douglas T, Vazquez-Duhalt R. Design of a VLP-nanovehicle for CYP450 enzymatic activity delivery. J Nanobiotechnology 2015; 13:66. [PMID: 26452461 PMCID: PMC4599659 DOI: 10.1186/s12951-015-0127-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/24/2015] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The intracellular delivery of enzymes for therapeutic use has a promising future for the treatment of several diseases such as genetic disorders and cancer. Virus-like particles offer an interesting platform for enzymatic delivery to targeted cells because of their great cargo capacity and the enhancement of the biocatalyst stability towards several factors important in the practical application of these nanoparticles. RESULTS We have designed a nano-bioreactor based on the encapsulation of a cytochrome P450 (CYP) inside the capsid derived from the bacteriophage P22. An enhanced peroxigenase, CYPBM3, was selected as a model enzyme because of its potential in enzyme prodrug therapy. A total of 109 enzymes per capsid were encapsulated with a 70 % retention of activity for cytochromes with the correct incorporation of the heme cofactor. Upon encapsulation, the stability of the enzyme towards protease degradation and acidic pH was increased. Cytochrome P450 activity was delivered into Human cervix carcinoma cells via transfecting P22-CYP nanoparticles with lipofectamine. CONCLUSION This work provides a clear demonstration of the potential of biocatalytic virus-like particles as medical relevant enzymatic delivery vehicles for clinical applications.
Collapse
Affiliation(s)
- Lorena Sánchez-Sánchez
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, 62250, Cuernavaca, Morelos, Mexico.
| | - Alejandro Tapia-Moreno
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
| | - Karla Juarez-Moreno
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
- Cátedras CONACyT affiliated to CNyN-UNAM, Ensenada, Mexico.
| | - Dustin P Patterson
- Department of Chemistry and Biochemistry, University of Texas at Tyler, Tyler, 75799, TX, USA.
| | - Ruben D Cadena-Nava
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
| | - Trevor Douglas
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA.
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
| |
Collapse
|
48
|
Large scale analysis of the mutational landscape in β-glucuronidase: A major player of mucopolysaccharidosis type VII. Gene 2015; 576:36-44. [PMID: 26415878 DOI: 10.1016/j.gene.2015.09.062] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/17/2015] [Accepted: 09/23/2015] [Indexed: 11/22/2022]
Abstract
The lysosomal storage disorders are a group of 50 unique inherited diseases characterized by unseemly lipid storage in lysosomes. These malfunctions arise due to genetic mutations that result in deficiency or reduced activities of the lysosomal enzymes, which are responsible for catabolism of biological macromolecules. Sly syndrome or mucopolysaccharidosis type VII is a lysosomal storage disorder associated with the deficiency of β-glucuronidase (EC 3.2.1.31) that catalyzes the hydrolysis of β-D-glucuronic acid residues from the non-reducing terminal of glycosaminoglycan. The effects of the disease causing mutations on the framework of the sequences and structure of β-glucuronidase (GUSBp) were analyzed utilizing a variety of bioinformatic tools. These analyses showed that 211 mutations may result in alteration of the biological activity of GUSBp, including previously experimentally validated mutations. Finally, we refined 90 disease causing mutations, which presumably cause a significant impact on the structure, function, and stability of GUSBp. Stability analyses showed that mutations p.Phe208Pro, p.Phe539Gly, p.Leu622Gly, p.Ile499Gly and p.Ile586Gly caused the highest impact on GUSBp stability and function because of destabilization of the protein structure. Furthermore, structures of wild type and mutant GUSBp were subjected to molecular dynamics simulation to examine the relative structural behaviors in the explicit conditions of water. In a broader view, the use of in silico approaches provided a useful understanding of the effect of single point mutations on the structure-function relationship of GUSBp.
Collapse
|
49
|
Damme M, Stroobants S, Lüdemann M, Rothaug M, Lüllmann-Rauch R, Beck HC, Ericsson A, Andersson C, Fogh J, D'Hooge R, Saftig P, Blanz J. Chronic enzyme replacement therapy ameliorates neuropathology in alpha-mannosidosis mice. Ann Clin Transl Neurol 2015; 2:987-1001. [PMID: 26817023 PMCID: PMC4693626 DOI: 10.1002/acn3.245] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/03/2015] [Accepted: 08/03/2015] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE The lysosomal storage disease alpha-mannosidosis is caused by the deficiency of the lysosomal acid hydrolase alpha-mannosidase (LAMAN) leading to lysosomal accumulation of neutral mannose-linked oligosaccharides throughout the body, including the brain. Clinical findings in alpha-mannosidosis include skeletal malformations, intellectual disabilities and hearing impairment. To date, no curative treatment is available. We previously developed a beneficial enzyme replacement therapy (ERT) regimen for alpha-mannosidase knockout mice, a valid mouse model for the human disease. However, humoral immune responses against the injected recombinant human alpha-mannosidase (rhLAMAN) precluded long-term studies and chronic treatment. METHODS Here, we describe the generation of an immune-tolerant alpha-mannosidosis mouse model that allowed chronic injection of rhLAMAN by transgenic expression of a catalytically inactive variant of human LAMAN in the knockout background. RESULTS Chronic ERT of rhLAMAN revealed pronounced effects on primary substrate storage throughout the brain, normalization of lysosomal enzyme activities and morphology as well as a decrease in microglia activation. The positive effect of long-term ERT on neuronal lysosomal function was reflected by an improvement of cognitive deficits and exploratory activity. in vivo and in vitro uptake measurements indicate rapid clearance of rhLAMAN from circulation and a broad uptake into different cell types of the nervous system. INTERPRETATION Our data contribute to the understanding of neurological disorders treatment by demonstrating that lysosomal enzymes such as rhLAMAN can penetrate into the brain and is able to ameliorate neuropathology.
Collapse
Affiliation(s)
- Markus Damme
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| | - Stijn Stroobants
- Laboratory of Biological Psychology University of Leuven B-3000 Leuven Belgium
| | - Meike Lüdemann
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| | | | | | - Hans Christian Beck
- Department of Biochemistry and Pharmacology Centre for Clinical Proteomics Odense University Hospital Sdr Boulevard 29 DK-5000 Odense C Denmark
| | | | | | - Jens Fogh
- Zymenex A/S Roskildevej 12C 3400 Hillerød Denmark
| | - Rudi D'Hooge
- Laboratory of Biological Psychology University of Leuven B-3000 Leuven Belgium
| | - Paul Saftig
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| | - Judith Blanz
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| |
Collapse
|
50
|
Abstract
Lysosomal storage diseases are a group of rare, inborn, metabolic errors characterized by deficiencies in normal lysosomal function and by intralysosomal accumulation of undegraded substrates. The past 25 years have been characterized by remarkable progress in the treatment of these diseases and by the development of multiple therapeutic approaches. These approaches include strategies aimed at increasing the residual activity of a missing enzyme (enzyme replacement therapy, hematopoietic stem cell transplantation, pharmacological chaperone therapy and gene therapy) and approaches based on reducing the flux of substrates to lysosomes. As knowledge has improved about the pathophysiology of lysosomal storage diseases, novel targets for therapy have been identified, and innovative treatment approaches are being developed.
Collapse
|