1
|
Hintze J, Fraumeni R, de Haan N, Miller RL, Saraswat M, Yang Z, Clausen H, Marth JD. Compositional and topological determinants of a physiological Ashwell-Morell receptor ligand. Proc Natl Acad Sci U S A 2025; 122:e2427129122. [PMID: 40208943 PMCID: PMC12012520 DOI: 10.1073/pnas.2427129122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025] Open
Abstract
The hepatocyte Ashwell-Morell receptor (AMR) is the prototypical mammalian lectin and the first cell receptor isolated. This recycling endocytic receptor of the plasma membrane determines the concentrations of hundreds of circulating glycoproteins in the blood and plays important roles in host responses to and outcomes of infection. The compositional and topological determinants of a physiological AMR ligand have remained unclear with contradictory findings reported. Previous studies established that the AMR binds multivalent galactose on desialylated triantennary or higher-branched N-glycans with little to no binding to galactose on biantennary forms. However, the vast majority of circulating blood glycoproteins are modified by biantennary N-glycans, rendering them unlikely to be ligands bound and eliminated by the AMR. Separately, other studies reported that AMR ligands include sialylated N-glycans, and specifically α2-6, but not α2-3, sialic acid linkages. Herein, we investigated the composition and topology of AMR ligands using a known physiological AMR ligand, intestinal alkaline phosphatase (IAP). Recombinant active IAP was produced in glycoengineered cells with either biantennary or higher valency triantennary and tetra-antennary N-glycan structures, and further with and without either α2-6 or α2-3 sialic acid linkages. These closely homogenous IAP monomer glycoforms assemble as dimers with similar enzymatic activity and were compared in AMR binding and clearance assays. Our results indicate that the AMR does not significantly bind IAP when its N-glycans are predominantly sialylated with either α2-6 or α2-3 sialic acid linkages. Multivalent desialylated AMR ligands may, however, appear when IAP monomers dimerize, resulting in the close proximation of biantennary N-glycans.
Collapse
Affiliation(s)
- John Hintze
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Robert Fraumeni
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Noortje de Haan
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden2300, The Netherlands
| | - Rebecca L. Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Mayank Saraswat
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, CopenhagenN 2200, Denmark
| | - Jamey D. Marth
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA92037
| |
Collapse
|
2
|
Padhy A, Gupta M, Das A, Farook I, Dutta T, Datta S, Datta R, Gupta SS. Lysosome-Specific Delivery of β-Glucosidase Enzyme Using Protein-Glycopolypeptide Conjugate via Protein Engineering and Bioconjugation. Bioconjug Chem 2025; 36:383-394. [PMID: 39988831 DOI: 10.1021/acs.bioconjchem.4c00430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Lysosomal enzyme replacement therapy (ERT) holds potential for treating lysosomal storage disorders, but achieving targeted delivery of deficient therapeutic enzymes remains a significant challenge. This study presents a novel approach for the lysosome-specific delivery of the β-glucosidase (B8CYA8) enzyme by covalently conjugating lysosome-targeting mannose-6-phosphate functionalized glycopolypeptides (M6P-GP). We used a protein-glycopolypeptide conjugate developed through advanced protein engineering and bioconjugation techniques. By conjugating β-glucosidase to M6P-GP that has a high affinity for the cation-independent mannose-6-phosphate receptors (CI-MPR) and lysosomal receptors, we enhance the enzyme's selective intracellular uptake and lysosome-specific localization. To attain maximum activity of the near-native enzyme after delivery, we have designed and synthesized an acetal linkage containing the pH-responsive linker maleimide-acetal-azide (MAA), which will cleave in the lysosomal acidic pH to detach the glycopolypeptide from the protein backbone. We demonstrated the efficient cellular uptake of the protein-glycopolypeptide conjugate and showed targeted lysosome delivery, leading to increased enzymatic activity compared to untreated cells. Our results proved that the approach mainly improves the specificity and efficiency of enzyme delivery, particularly into lysosomes, which may enable new methods for ERT. These findings suggest that protein-glycopolypeptide conjugates could represent a class of bioconjugates to design targeted enzyme therapies, offering a pathway to the effective treatment of Gaucher disease (GD) and potentially other related lysosomal storage disorders.
Collapse
Affiliation(s)
- Abinash Padhy
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| | - Mani Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| | - Apurba Das
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| | - Isha Farook
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| | - Tahiti Dutta
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| | - Supratim Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
- Center for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| | - Rupak Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| | - Sayam Sen Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, Nadia, West Bengal 741246, India
| |
Collapse
|
3
|
He KJ, Gong G. Prognostic prediction and immune infiltration analysis based on lysosome and senescence state identifies MMP12 as a novel therapy target in gastric cancer. Int Immunopharmacol 2024; 143:113344. [PMID: 39401475 DOI: 10.1016/j.intimp.2024.113344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/21/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND AND AIMS As humans undergo the aging process, they become more vulnerable to various types of cancers, including gastric cancer (GC), which is frequently associated with aging. The senescent phenotype is closely linked to lysosomes, but research on the combined impact of senescence and lysosomes on GC prognosis is scarce. METHODS To construct and validate a prognostic model for gastric cancer (GC), we obtained gene expression and clinical data of GC patients from Cancer Genome Atlas (TCGA) databases. We employed Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression for model construction and ConsensusClusterPlus R package for generating cluster heatmaps. The model's predictive ability was evaluated through Kaplan-Meier survival analysis and ROC curve analysis. Our analysis included an assessment of the senescence and lysosome state using expression profiles and immune infiltration analysis through CIBERSORT methods. Finally, we validated potential gene targets through cellular experiments. RESULTS "In this research, we discovered two subtypes of gastric cancer (GC), Cluster 1 and Cluster 2. These subtypes are characterized by the presence of lysosomes and senescence, and we have identified distinct molecular features unique to each subtype. We observed that Cluster 2 had a lower survival prognosis compared to Cluster 1. Additionally, we have developed a risk prediction model that takes into consideration the presence of lysosomes and senescence. Patients in the high-risk group, as predicted by our model, experienced shorter survival times. Further analysis included immune infiltration, immune checkpoint, and chemotherapy evaluation of GC patients. We have displayed the frequency of mutations and copy number variations (CNVs) in visual formats. Our cellular experiments demonstrated that the MMP12 gene serves as a protective factor in GC cells." CONCLUSIONS In conclusion, we have clarified the extensive relationship between lysosomes and senescence in GC and developed a risk signature to forecast the prognosis of GC patients. MMP12 could be a promising protective factor for GC patients and might present a novel concept for anticipating the efficacy of targeted therapies and immunotherapies in GC patients.
Collapse
Affiliation(s)
- Ke-Jie He
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou City, Zhejiang Province, China.
| | - Guoyu Gong
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
4
|
Tomsen-Melero J, Moltó-Abad M, Merlo-Mas J, Díaz-Riascos ZV, Cristóbal-Lecina E, Soldevila A, Altendorfer-Kroath T, Danino D, Ionita I, Pedersen JS, Snelling L, Clay H, Carreño A, Corchero JL, Pulido D, Casas J, Veciana J, Schwartz S, Sala S, Font A, Birngruber T, Royo M, Córdoba A, Ventosa N, Abasolo I, González-Mira E. Targeted nanoliposomes to improve enzyme replacement therapy of Fabry disease. SCIENCE ADVANCES 2024; 10:eadq4738. [PMID: 39671483 PMCID: PMC11801267 DOI: 10.1126/sciadv.adq4738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024]
Abstract
The central nervous system represents a major target tissue for therapeutic approach of numerous lysosomal storage disorders. Fabry disease arises from the lack or dysfunction of the lysosomal alpha-galactosidase A (GLA) enzyme, resulting in substrate accumulation and multisystemic clinical manifestations. Current enzyme replacement therapies (ERTs) face limited effectiveness due to poor enzyme biodistribution in target tissues and inability to reach the brain. We present an innovative drug delivery strategy centered on a peptide-targeted nanoliposomal formulation, designated as nanoGLA, engineered to selectively deliver a recombinant human GLA (rhGLA) to target tissues. In a Fabry mouse model, nanoGLA demonstrated improved efficacy, inducing a notable reduction in Gb3 deposits in contrast to non-nanoformulated GLA, even in the brain, highlighting the potential of the nanoGLA to address both systemic and cerebrovascular manifestations of Fabry disease. The EMA has granted the Orphan Drug Designation to this product, underscoring the potential clinical superiority of nanoGLA over authorized ERTs and encouraging to advance it toward clinical translation.
Collapse
Affiliation(s)
- Judit Tomsen-Melero
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Marc Moltó-Abad
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Josep Merlo-Mas
- Nanomol Technologies SL, Campus de la UAB, 08193 Bellaterra, Spain
| | - Zamira V. Díaz-Riascos
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Functional Validaton & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Edgar Cristóbal-Lecina
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | | | - Thomas Altendorfer-Kroath
- JOANNEUM RESEARCH–Institute for Biomedical Research and Technologies (HEALTH), Neue Stiftingtalstraße 2, 8010 Graz, Austria
| | - Dganit Danino
- Cryo-EM Laboratory of Soft Matter, Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, 32000 Haifa, Israel
- Cryo-EM and Self-Assembly Laboratory, Guangdong-Technion Israel Institute of Technology, Shantou, China
| | - Inbal Ionita
- Cryo-EM Laboratory of Soft Matter, Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Jan Skov Pedersen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark
| | | | - Hazel Clay
- Labcorp Drug Development, Harrogate HG3 IPY, UK
| | - Aida Carreño
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - José L. Corchero
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i de Microbiologia, Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Daniel Pulido
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Josefina Casas
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
| | - Jaume Veciana
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Simó Schwartz
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Servei de Bioquímica, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Santi Sala
- Nanomol Technologies SL, Campus de la UAB, 08193 Bellaterra, Spain
| | | | - Thomas Birngruber
- JOANNEUM RESEARCH–Institute for Biomedical Research and Technologies (HEALTH), Neue Stiftingtalstraße 2, 8010 Graz, Austria
| | - Miriam Royo
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Alba Córdoba
- Nanomol Technologies SL, Campus de la UAB, 08193 Bellaterra, Spain
| | - Nora Ventosa
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Ibane Abasolo
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Functional Validaton & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Servei de Bioquímica, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Elisabet González-Mira
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| |
Collapse
|
5
|
Tian W, Zagami C, Chen J, Blomberg AL, Guiu LS, Skovbakke SL, Goletz S. Cell-based glycoengineering of extracellular vesicles through precise genome editing. N Biotechnol 2024; 83:101-109. [PMID: 39079597 DOI: 10.1016/j.nbt.2024.07.004] [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: 05/07/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/03/2024]
Abstract
Engineering of extracellular vesicles (EVs) towards more efficient targeting and uptake to specific cells has large potentials for their application as therapeutics. Carbohydrates play key roles in various biological interactions and are essential for EV biology. The extent to which glycan modification of EVs can be achieved through genetic glycoengineering of their parental cells has not been explored yet. Here we introduce targeted glycan modification of EVs through cell-based glycoengineering via modification of various enzymes in the glycosylation machinery. In a "simple cell" strategy, we modified major glycosylation pathways by knocking-out (KO) essential genes for N-glycosylation (MGAT1), O-GalNAc glycosylation (C1GALT1C1), glycosphingolipids (B4GALT5/6), glycosaminoglycans (B4GALT7) and sialylation (GNE) involved in the elongation or biosynthesis of the glycans in HEK293F cells. The gene editing led to corresponding glycan changes on the cells as demonstrated by differential lectin staining. Small EVs (sEVs) isolated from the cells showed overall corresponding glycan changes, but also some unexpected differences to their parental cell including enrichment preference for certain glycan structures and absence of other glycan types. The genetic glycoengineering did not significantly impact sEVs production, size distribution, or syntenin-1 biomarker expression, while a clonal influence on sEVs production yields was observed. Our findings demonstrate the successful implementation of sEVs glycoengineering via genetic modification of the parental cell and a stable source for generation of glycoengineered sEVs. The utilization of glycoengineered sEVs offers a promising opportunity to study the role of glycosylation in EV biology, as well as to facilitate the optimization of sEVs for therapeutic purposes.
Collapse
Affiliation(s)
- Weihua Tian
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Chiara Zagami
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jiasi Chen
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anne Louise Blomberg
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Laura Salse Guiu
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Sarah Line Skovbakke
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
6
|
Jaroentomeechai T, Karlsson R, Goerdeler F, Teoh FKY, Grønset MN, de Wit D, Chen YH, Furukawa S, Psomiadou V, Hurtado-Guerrero R, Vidal-Calvo EE, Salanti A, Boltje TJ, van den Bos LJ, Wunder C, Johannes L, Schjoldager KT, Joshi HJ, Miller RL, Clausen H, Vakhrushev SY, Narimatsu Y. Mammalian cell-based production of glycans, glycopeptides and glycomodules. Nat Commun 2024; 15:9668. [PMID: 39516489 PMCID: PMC11549445 DOI: 10.1038/s41467-024-53738-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Access to defined glycans and glycoconjugates is pivotal for discovery, dissection, and harnessing of a range of biological functions orchestrated by cellular glycosylation processes and the glycome. We previously employed genetic glycoengineering by nuclease-based gene editing to develop sustainable production of designer glycoprotein therapeutics and cell-based glycan arrays that display glycans in their natural context at the cell surface. However, access to human glycans in formats and quantities that allow structural studies of molecular interactions and use of glycans in biomedical applications currently rely on chemical and chemoenzymatic syntheses associated with considerable labor, waste, and costs. Here, we develop a sustainable and scalable method for production of glycans in glycoengineered mammalian cells by employing secreted Glycocarriers with repeat glycosylation acceptor sequence motifs for different glycans. The Glycocarrier technology provides a flexible production platform for glycans in different formats, including oligosaccharides, glycopeptides, and multimeric glycomodules, and offers wide opportunities for use in bioassays and biomedical applications.
Collapse
Affiliation(s)
- Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Richard Karlsson
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Felix Goerdeler
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fallen Kai Yik Teoh
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Magnus Nørregaard Grønset
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dylan de Wit
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Sanae Furukawa
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Venetia Psomiadou
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Ramon Hurtado-Guerrero
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Biocomputation and Physics of Complex Systems, University of Zaragoza, Zaragoza, Spain
- Fundación ARAID, Zaragoza, Spain
| | - Elena Ethel Vidal-Calvo
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas J Boltje
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Nijmegen, The Netherlands
| | | | - Christian Wunder
- Institut Curie, Cellular and Chemical Biology Unit, PSL Research University, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Ludger Johannes
- Institut Curie, Cellular and Chemical Biology Unit, PSL Research University, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Katrine T Schjoldager
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hiren J Joshi
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca L Miller
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yoshiki Narimatsu
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
- GlycoDisplay ApS, Copenhagen, Denmark.
| |
Collapse
|
7
|
Voss M. Proteolytic cleavage of Golgi glycosyltransferases by SPPL3 and other proteases and its implications for cellular glycosylation. Biochim Biophys Acta Gen Subj 2024; 1868:130668. [PMID: 38992482 DOI: 10.1016/j.bbagen.2024.130668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
Glycosylation of proteins and lipids is of fundamental importance in multicellular eukaryotes. The vast diversity of glycan structures observed is generated in the Golgi apparatus by the concerted activity of >100 distinct enzymes, which include glycosyltransferases and other glycan-modifying enzymes. Well-known for decades, the majority of these enzymes is released from the Golgi apparatus and subsequently secreted into the extracellular space following endoproteolytic cleavage, but the underlying molecular mechanisms and the physiological implications have remained unexplored. This review will summarize our current knowledge of Golgi enzyme proteolysis and secretion and will discuss its conceptual implications for the regulation of cellular glycosylation and the organization of the Golgi apparatus. A particular focus will lie on the intramembrane protease SPPL3, which recently emerged as key protease facilitating Golgi enzyme release and has since been shown to affect a multitude of glycosylation-dependent physiological processes.
Collapse
Affiliation(s)
- Matthias Voss
- Institute of Biochemistry, Kiel University, Kiel, Germany.
| |
Collapse
|
8
|
Tian W, Blomberg AL, Steinberg KE, Henriksen BL, Jørgensen JS, Skovgaard K, Skovbakke SL, Goletz S. Novel genetically glycoengineered human dendritic cell model reveals regulatory roles of α2,6-linked sialic acids in DC activation of CD4+ T cells and response to TNFα. Glycobiology 2024; 34:cwae042. [PMID: 38873803 DOI: 10.1093/glycob/cwae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
Dendritic cells (DCs) are central for the initiation and regulation of appropriate immune responses. While several studies suggest important regulatory roles of sialoglycans in DC biology, our understanding is still inadequate primarily due to a lack of appropriate models. Previous approaches based on enzymatic- or metabolic-glycoengineering and primary cell isolation from genetically modified mice have limitations related to specificity, stability, and species differences. This study addresses these challenges by introducing a workflow to genetically glycoengineer the human DC precursor cell line MUTZ-3, described to differentiate and maturate into fully functional dendritic cells, using CRISPR-Cas9, thereby providing and validating the first isogenic cell model for investigating glycan alteration on human DC differentiation, maturation, and activity. By knocking out (KO) the ST6GAL1 gene, we generated isogenic cells devoid of ST6GAL1-mediated α(2,6)-linked sialylation, allowing for a comprehensive investigation into its impact on DC function. Glycan profiling using lectin binding assay and functional studies revealed that ST6GAL1 KO increased the expression of important antigen presenting and co-stimulatory surface receptors and a specifically increased activation of allogenic human CD4 + T cells. Additionally, ST6GAL1 KO induces significant changes in surface marker expression and cytokine response to TNFα-induced maturation, and it affects migration and the endocytic capacity. These results indicate that genetic glycoengineering of the isogenic MUTZ-3 cellular model offers a valuable tool to study how specific glycan structures influence human DC biology, contributing to our understanding of glycoimmunology.
Collapse
Affiliation(s)
- Weihua Tian
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Anne Louise Blomberg
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Kaylin Elisabeth Steinberg
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Betina Lyngfeldt Henriksen
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Josefine Søborg Jørgensen
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Kerstin Skovgaard
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Sarah Line Skovbakke
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| | - Steffen Goletz
- Biotherapeutic Glycoengineering and Immunology, Section for Medical Biotechnology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs Lyngby 2800, Denmark
| |
Collapse
|
9
|
Germain DP, Linhart A. Pegunigalsidase alfa: a novel, pegylated recombinant alpha-galactosidase enzyme for the treatment of Fabry disease. Front Genet 2024; 15:1395287. [PMID: 38680424 PMCID: PMC11045972 DOI: 10.3389/fgene.2024.1395287] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
Fabry disease, a rare X-linked genetic disorder, results from pathogenic variants in GLA, leading to deficient lysosomal α-galactosidase A enzyme activity and multi-organ manifestations. Since 2001, enzyme replacement therapy (ERT), using agalsidase alfa or agalsidase beta, has been the mainstay treatment, albeit with limitations such as rapid clearance and immunogenicity. Pegunigalsidase alfa, a novel PEGylated recombinant alpha-galactosidase, offers promise as an alternative. Produced in plant cells, pegunigalsidase alfa exhibits enhanced stability, prolonged half-life, and reduced immunogenicity due to pegylation. A phase 1/2 clinical trial demonstrated Gb3 clearance from renal capillary endothelial cells and its 48-month extension study revealed notable outcomes in renal function preservation. Three phase 3 clinical trials (BRIDGE, BRIGHT, and BALANCE) have shown favorable efficacy and safety profile, although caution is warranted in interpreting the results of BRIDGE and BRIGHT which lacked control groups. In BALANCE, the pivotal phase 3 trial comparing pegunigalsidase alfa with agalsidase beta, an intention-to-treat analysis of the eGFR decline over 2 years showed that the intergroup difference [95%confidence interval] in the median slope was -0.36 mL/min/1.73 m2/year [-2.44; 1.73]. The confidence interval had a lower limit above the prespecified value of -3 mL/min/1.73 m2/year and included zero. Despite challenges such as occasional hypersensitivity reactions and immune-complex-mediated glomerulonephritis, pegunigalsidase alfa approval by the European Medicines Agency and the Food and Drug Administration represents a significant addition to Fabry disease therapeutic landscape providing an option for patients in whom enzyme replacement therapy with current formulations is poorly tolerated or poorly effective.
Collapse
Affiliation(s)
- Dominique P. Germain
- Division of Medical Genetics, University of Versailles–St Quentin en Yvelines (UVSQ), Paris–Saclay University, Montigny, France
- Second Department of Medicine, Charles University, General University Hospital, Prague, Czechia
| | - Ales Linhart
- Second Department of Medicine, Charles University, General University Hospital, Prague, Czechia
| |
Collapse
|
10
|
Nilsson J, Eriksson P, Naguib MM, Jax E, Sihlbom C, Olsson BM, Lundkvist Å, Olsen B, Järhult JD, Larson G, Ellström P. Expression of influenza A virus glycan receptor candidates in mallard, chicken, and tufted duck. Glycobiology 2024; 34:cwad098. [PMID: 38127648 PMCID: PMC10987293 DOI: 10.1093/glycob/cwad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Influenza A virus (IAV) pandemics result from interspecies transmission events within the avian reservoir and further into mammals including humans. Receptor incompatibility due to differently expressed glycan structures between species has been suggested to limit zoonotic IAV transmission from the wild bird reservoir as well as between different bird species. Using glycoproteomics, we have studied the repertoires of expressed glycan structures with focus on putative sialic acid-containing glycan receptors for IAV in mallard, chicken and tufted duck; three bird species with different roles in the zoonotic ecology of IAV. The methodology used pinpoints specific glycan structures to specific glycosylation sites of identified glycoproteins and was also used to successfully discriminate α2-3- from α2-6-linked terminal sialic acids by careful analysis of oxonium ions released from glycopeptides in tandem MS/MS (MS2), and MS/MS/MS (MS3). Our analysis clearly demonstrated that all three bird species can produce complex N-glycans including α2-3-linked sialyl Lewis structures, as well as both N- and O- glycans terminated with both α2-3- and α2-6-linked Neu5Ac. We also found the recently identified putative IAV receptor structures, Man-6P N-glycopeptides, in all tissues of the three bird species. Furthermore, we found many similarities in the repertoires of expressed receptors both between the bird species investigated and to previously published data from pigs and humans. Our findings of sialylated glycan structures, previously anticipated to be mammalian specific, in all three bird species may have major implications for our understanding of the role of receptor incompatibility in interspecies transmission of IAV.
Collapse
Affiliation(s)
- Jonas Nilsson
- Department of Laboratory Medicine, University of Gothenburg, Sahlgrenska University Hospital, Vita Stråket 12, Gothenburg SE-413 45, Sweden
- Laboratory of Clinical Chemistry, Sahlgrenska University Hospital, Bruna Stråket 16, Gothenburg SE-413 45, Sweden
- Proteomics Core Facility, University of Gothenburg, Sahlgrenska Academy, Medicinaregatan 9E, Gothenburg SE-405 30, Sweden
| | - Per Eriksson
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| | - Mahmoud M Naguib
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Husargatan 3, Uppsala University, Uppsala, SE-75237, Sweden
| | - Elinor Jax
- Department of Migration, Max Planck Institute of Animal Behavior, Am Obstberg 1, Radolfzell, Baden-Württemberg DE-78315, Germany
| | - Carina Sihlbom
- Proteomics Core Facility, University of Gothenburg, Sahlgrenska Academy, Medicinaregatan 9E, Gothenburg SE-405 30, Sweden
| | - Britt-Marie Olsson
- Proteomics Core Facility, University of Gothenburg, Sahlgrenska Academy, Medicinaregatan 9E, Gothenburg SE-405 30, Sweden
| | - Åke Lundkvist
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Husargatan 3, Uppsala University, Uppsala, SE-75237, Sweden
| | - Björn Olsen
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| | - Josef D Järhult
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, University of Gothenburg, Sahlgrenska University Hospital, Vita Stråket 12, Gothenburg SE-413 45, Sweden
- Laboratory of Clinical Chemistry, Sahlgrenska University Hospital, Bruna Stråket 16, Gothenburg SE-413 45, Sweden
| | - Patrik Ellström
- Zoonosis Science Center, Department of Medical Sciences, Husargatan 3, Uppsala University, Uppsala, SE-75185, Sweden
| |
Collapse
|
11
|
Thalén NB, Barzadd MM, Lundqvist M, Rodhe J, Andersson M, Bidkhori G, Possner D, Su C, Nilsson J, Eisenhut P, Malm M, Karlsson A, Vestin J, Forsberg J, Nordling E, Mardinoglu A, Volk AL, Sandegren A, Rockberg J. Tuning of CHO secretional machinery improve activity of secreted therapeutic sulfatase 150-fold. Metab Eng 2024; 81:157-166. [PMID: 38081506 DOI: 10.1016/j.ymben.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 10/12/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023]
Abstract
Rare diseases are, despite their name, collectively common and millions of people are affected daily of conditions where treatment often is unavailable. Sulfatases are a large family of activating enzymes related to several of these diseases. Heritable genetic variations in sulfatases may lead to impaired activity and a reduced macromolecular breakdown within the lysosome, with several severe and lethal conditions as a consequence. While therapeutic options are scarce, treatment for some sulfatase deficiencies by recombinant enzyme replacement are available. The recombinant production of such sulfatases suffers greatly from both low product activity and yield, further limiting accessibility for patient groups. To mitigate the low product activity, we have investigated cellular properties through computational evaluation of cultures with varying media conditions and comparison of two CHO clones with different levels of one active sulfatase variant. Transcriptome analysis identified 18 genes in secretory pathways correlating with increased sulfatase production. Experimental validation by upregulation of a set of three key genes improved the specific enzymatic activity at varying degree up to 150-fold in another sulfatase variant, broadcasting general production benefits. We also identified a correlation between product mRNA levels and sulfatase activity that generated an increase in sulfatase activity when expressed with a weaker promoter. Furthermore, we suggest that our proposed workflow for resolving bottlenecks in cellular machineries, to be useful for improvements of cell factories for other biologics as well.
Collapse
Affiliation(s)
- Niklas Berndt Thalén
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Mona Moradi Barzadd
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Magnus Lundqvist
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | | | | | - Gholamreza Bidkhori
- Science for Life Laboratory, KTH - Royal Institute of Technology, Solna, 171 65, Sweden; AIVIVO Ltd. Unit 25, Bio-innovation centre, Cambridge Science park, Cambridge, UK
| | | | - Chao Su
- SOBI AB, Tomtebodavägen 23A, Stockholm, Sweden
| | | | - Peter Eisenhut
- ACIB - Austrian Centre of Industrial Biotechnology, Krenngasse 37, 8010 Graz, Austria; BOKU - University of Natural Resources and Life Sciences, Department of Biotechnology, Vienna, 1190, Austria
| | - Magdalena Malm
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | - Alice Karlsson
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | | | | | | | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Solna, 171 65, Sweden
| | - Anna-Luisa Volk
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden
| | | | - Johan Rockberg
- Dept. of Protein science, KTH - Royal Institute of Technology, Stockholm, SE-106 91, Sweden.
| |
Collapse
|
12
|
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: 19] [Impact Index Per Article: 9.5] [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
|
13
|
Glinšek K, Bozovičar K, Bratkovič T. CRISPR Technologies in Chinese Hamster Ovary Cell Line Engineering. Int J Mol Sci 2023; 24:ijms24098144. [PMID: 37175850 PMCID: PMC10179654 DOI: 10.3390/ijms24098144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
The Chinese hamster ovary (CHO) cell line is a well-established platform for the production of biopharmaceuticals due to its ability to express complex therapeutic proteins with human-like glycopatterns in high amounts. The advent of CRISPR technology has opened up new avenues for the engineering of CHO cell lines for improved protein production and enhanced product quality. This review summarizes recent advances in the application of CRISPR technology for CHO cell line engineering with a particular focus on glycosylation modulation, productivity enhancement, tackling adventitious agents, elimination of problematic host cell proteins, development of antibiotic-free selection systems, site-specific transgene integration, and CRISPR-mediated gene activation and repression. The review highlights the potential of CRISPR technology in CHO cell line genome editing and epigenetic engineering for the more efficient and cost-effective development of biopharmaceuticals while ensuring the safety and quality of the final product.
Collapse
Affiliation(s)
- Katja Glinšek
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Krištof Bozovičar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tomaž Bratkovič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
14
|
Kalkan AK, Palaz F, Sofija S, Elmousa N, Ledezma Y, Cachat E, Rios-Solis L. Improving recombinant protein production in CHO cells using the CRISPR-Cas system. Biotechnol Adv 2023; 64:108115. [PMID: 36758652 DOI: 10.1016/j.biotechadv.2023.108115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/28/2022] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Chinese hamster ovary (CHO) cells are among the most widely used mammalian cell lines in the biopharmaceutical industry. Therefore, it is not surprising that significant efforts have been made around the engineering of CHO cells using genetic engineering methods such as the CRISPR-Cas system. In this review, we summarize key recent studies that have used different CRISPR-Cas systems such as Cas9, Cas13 or dCas9 fused with effector domains to improve recombinant protein (r-protein) production in CHO cells. Here, every relevant stage of production was considered, underscoring the advantages and limitations of these systems, as well as discussing their bottlenecks and probable solutions. A special emphasis was given on how these systems could disrupt and/or regulate genes related to glycan composition, which has relevant effects over r-protein properties and in vivo activity. Furthermore, the related promising future applications of CRISPR to achieve a tunable, reversible, or highly stable editing of CHO cells are discussed. Overall, the studies covered in this review show that despite the complexity of mammalian cells, the synthetic biology community has developed many mature strategies to improve r-protein production using CHO cells. In this regard, CRISPR-Cas technology clearly provides efficient and flexible genetic manipulation and allows for the generation of more productive CHO cell lines, leading to more cost-efficient production of biopharmaceuticals, however, there is still a need for many emerging techniques in CRISPR to be reported in CHO cells; therefore, more research in these cells is needed to realize the full potential of this technology.
Collapse
Affiliation(s)
- Ali Kerem Kalkan
- Department of Bioengineering and Imperial College Centre for Synthetic Biology, Imperial College London, London, UK; Environmental Engineering Department, Gebze Technical University, Turkey
| | - Fahreddin Palaz
- Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Semeniuk Sofija
- Centre for Engineering Biology, University of Edinburgh, Edinburgh EH9 3BF, UK; Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Nada Elmousa
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh EH9 3DW, UK
| | - Yuri Ledezma
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh EH9 3DW, UK; Biology Department, Faculty of Pure and Natural Sciences, Universidad Mayor de San Andrés, Bolivia
| | - Elise Cachat
- Centre for Engineering Biology, University of Edinburgh, Edinburgh EH9 3BF, UK; Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences University of Edinburgh, Edinburgh EH9 3BF, UK; UK Centre for Mammalian Synthetic Biology, University of Edinburgh, Edinburgh EH8 9YL, UK
| | - Leonardo Rios-Solis
- Centre for Engineering Biology, University of Edinburgh, Edinburgh EH9 3BF, UK; Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh EH9 3DW, UK; School of Natural and Environmental Sciences, Molecular Biology and Biotechnology Division, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK.
| |
Collapse
|
15
|
van Kuilenburg ABP, Hollak CEM, Travella A, Jacobs M, Gentilini LD, Leen R, der Vlugt KMMGV, Stet FSB, Goorden SMI, van der Veen S, Criscuolo M, Papouchado M. Development of a Biosimilar of Agalsidase Beta for the Treatment of Fabry Disease: Preclinical Evaluation. Drugs R D 2023:10.1007/s40268-023-00421-x. [PMID: 37083901 DOI: 10.1007/s40268-023-00421-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Fabry disease (FD) is a rare lysosomal storage disorder caused by a deficiency of the enzyme α-galactosidase A (aGal A). Since 2001, two different enzyme replacement therapies have been authorized, with agalsidase beta being used in most parts of the Western world. Currently, biosimilars of several expensive enzyme therapies are under development to improve their accessibility for patients. We present the preclinical results of the development of a biosimilar to agalsidase beta. METHODS Produced in a Chinese hamster ovary (CHO)-cell system, the biosimilar aGal A Biosidus (AGABIO), was compared with agalsidase beta with respect to amino acid sequence, glycosylation, specific α-galactosidase activity, stability in plasma, and effects on cultured human Fabry fibroblasts and Fabry mice. RESULTS AGABIO had the same amino acid composition and similar glycosylation, enzymatic activity, and stability as compared with agalsidase beta. After uptake in fibroblasts, α-galactosidase A activity increased in a dose-dependent manner, with maximum uptake observed after 24 h, which remained stable until at least 48 h. Both enzymes were localized to lysosomes. Reduction of accumulated globotriaosylceramide (Gb3) and lysoGb3 in cultured Fabry fibroblasts by AGABIO and agalsidase beta showed comparable dose-response curves. In Fabry knockout mice, after a single injection, both enzymes were rapidly cleared from the plasma and showed equal reductions in tissue and plasma sphingolipids. Repeated dose studies in rats did not raise any safety concerns. Anti-drug antibodies from patients with FD treated with agalsidase beta showed equal neutralization activity toward AGABIO. CONCLUSION These findings support the biosimilarity of AGABIO in comparison with agalsidase beta. The clinical study phase is currently under development.
Collapse
Affiliation(s)
- André B P van Kuilenburg
- Amsterdam UMC location University of Amsterdam, Laboratory Genetic Metabolic Diseases F0-220, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.
| | - Carla E M Hollak
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
- Medicine for Society, Platform at Amsterdam, UMC-University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - René Leen
- Amsterdam UMC location University of Amsterdam, Laboratory Genetic Metabolic Diseases F0-220, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Karen M M Ghauharali-van der Vlugt
- Amsterdam UMC location University of Amsterdam, Laboratory Genetic Metabolic Diseases F0-220, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Femke S Beers Stet
- Amsterdam UMC location University of Amsterdam, Laboratory Genetic Metabolic Diseases F0-220, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Susan M I Goorden
- Amsterdam UMC location University of Amsterdam, Laboratory Genetic Metabolic Diseases F0-220, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Sanne van der Veen
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
- Medicine for Society, Platform at Amsterdam, UMC-University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
16
|
Amiri S, Adibzadeh S, Ghanbari S, Rahmani B, Kheirandish MH, Farokhi-Fard A, Dastjerdeh MS, Davami F. CRISPR-interceded CHO cell line development approaches. Biotechnol Bioeng 2023; 120:865-902. [PMID: 36597180 DOI: 10.1002/bit.28329] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/28/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
For industrial production of recombinant protein biopharmaceuticals, Chinese hamster ovary (CHO) cells represent the most widely adopted host cell system, owing to their capacity to produce high-quality biologics with human-like posttranslational modifications. As opposed to random integration, targeted genome editing in genomic safe harbor sites has offered CHO cell line engineering a new perspective, ensuring production consistency in long-term culture and high biotherapeutic expression levels. Corresponding the remarkable advancements in knowledge of CRISPR-Cas systems, the use of CRISPR-Cas technology along with the donor design strategies has been pushed into increasing novel scenarios in cell line engineering, allowing scientists to modify mammalian genomes such as CHO cell line quickly, readily, and efficiently. Depending on the strategies and production requirements, the gene of interest can also be incorporated at single or multiple loci. This review will give a gist of all the most fundamental recent advancements in CHO cell line development, such as different cell line engineering approaches along with donor design strategies for targeted integration of the desired construct into genomic hot spots, which could ultimately lead to the fast-track product development process with consistent, improved product yield and quality.
Collapse
Affiliation(s)
- Shahin Amiri
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Setare Adibzadeh
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Samaneh Ghanbari
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Behnaz Rahmani
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad H Kheirandish
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - Aref Farokhi-Fard
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mansoureh S Dastjerdeh
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Davami
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
17
|
Hallows WC, Skvorak K, Agard N, Kruse N, Zhang X, Zhu Y, Botham RC, Chng C, Shukla C, Lao J, Miller M, Sero A, Viduya J, Ismaili MHA, McCluskie K, Schiffmann R, Silverman AP, Shen JS, Huisman GW. Optimizing human α-galactosidase for treatment of Fabry disease. Sci Rep 2023; 13:4748. [PMID: 36959353 PMCID: PMC10036536 DOI: 10.1038/s41598-023-31777-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 03/17/2023] [Indexed: 03/25/2023] Open
Abstract
Fabry disease is caused by a deficiency of α-galactosidase A (GLA) leading to the lysosomal accumulation of globotriaosylceramide (Gb3) and other glycosphingolipids. Fabry patients experience significant damage to the heart, kidney, and blood vessels that can be fatal. Here we apply directed evolution to generate more stable GLA variants as potential next generation treatments for Fabry disease. GLAv05 and GLAv09 were identified after screening more than 12,000 GLA variants through 8 rounds of directed evolution. Both GLAv05 and GLAv09 exhibit increased stability at both lysosomal and blood pH, stability to serum, and elevated enzyme activity in treated Fabry fibroblasts (19-fold) and GLA-/- podocytes (10-fold). GLAv05 and GLAv09 show improved pharmacokinetics in mouse and non-human primates. In a Fabry mouse model, the optimized variants showed prolonged half-lives in serum and relevant tissues, and a decrease of accumulated Gb3 in heart and kidney. To explore the possibility of diminishing the immunogenic potential of rhGLA, amino acid residues in sequences predicted to bind MHC II were targeted in late rounds of GLAv09 directed evolution. An MHC II-associated peptide proteomics assay confirmed a reduction in displayed peptides for GLAv09. Collectively, our findings highlight the promise of using directed evolution to generate enzyme variants for more effective treatment of lysosomal storage diseases.
Collapse
Affiliation(s)
| | - Kristen Skvorak
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Nick Agard
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
- Genentech, South San Francisco, CA, 94080, USA
| | - Nikki Kruse
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Xiyun Zhang
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
- Fornia BioSolutions Inc US, Hayward, CA, 94545, USA
| | - Yu Zhu
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Rachel C Botham
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Chinping Chng
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Charu Shukla
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Jessica Lao
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
- Octant, Emeryville, CA, 94608, USA
| | - Mathew Miller
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Antoinette Sero
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Judy Viduya
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Moulay Hicham Alaoui Ismaili
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
- Glycomine, San Mateo, CA, 94070, USA
| | - Kerryn McCluskie
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
- Glycomine, San Mateo, CA, 94070, USA
| | - Raphael Schiffmann
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX, 75246, USA
- 4D Molecular Therapeutics, Emeryville, CA, 94608, USA
| | - Adam P Silverman
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| | - Jin-Song Shen
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX, 75246, USA
- 4D Molecular Therapeutics, Emeryville, CA, 94608, USA
| | - Gjalt W Huisman
- Codexis Inc.,, 200 Penobscot Drive, Redwood City, CA, 94063, USA
| |
Collapse
|
18
|
Sørensen DM, Büll C, Madsen TD, Lira-Navarrete E, Clausen TM, Clark AE, Garretson AF, Karlsson R, Pijnenborg JFA, Yin X, Miller RL, Chanda SK, Boltje TJ, Schjoldager KT, Vakhrushev SY, Halim A, Esko JD, Carlin AF, Hurtado-Guerrero R, Weigert R, Clausen H, Narimatsu Y. Identification of global inhibitors of cellular glycosylation. Nat Commun 2023; 14:948. [PMID: 36804936 PMCID: PMC9941569 DOI: 10.1038/s41467-023-36598-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Small molecule inhibitors of glycosylation enzymes are valuable tools for dissecting glycan functions and potential drug candidates. Screening for inhibitors of glycosyltransferases are mainly performed by in vitro enzyme assays with difficulties moving candidates to cells and animals. Here, we circumvent this by employing a cell-based screening assay using glycoengineered cells expressing tailored reporter glycoproteins. We focused on GalNAc-type O-glycosylation and selected the GalNAc-T11 isoenzyme that selectively glycosylates endocytic low-density lipoprotein receptor (LDLR)-related proteins as targets. Our screen of a limited small molecule compound library did not identify selective inhibitors of GalNAc-T11, however, we identify two compounds that broadly inhibited Golgi-localized glycosylation processes. These compounds mediate the reversible fragmentation of the Golgi system without affecting secretion. We demonstrate how these inhibitors can be used to manipulate glycosylation in cells to induce expression of truncated O-glycans and augment binding of cancer-specific Tn-glycoprotein antibodies and to inhibit expression of heparan sulfate and binding and infection of SARS-CoV-2.
Collapse
Affiliation(s)
- Daniel Madriz Sørensen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Christian Büll
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University, 6525 AJ, Nijmegen, The Netherlands
| | - Thomas D Madsen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erandi Lira-Navarrete
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
- The Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquillor s/n, Campus Rio Ebro, 50018, Zaragoza, Spain
- Fundación ARAID, 50018, Zaragoza, Spain
| | - Thomas Mandel Clausen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Alex E Clark
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Aaron F Garretson
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Richard Karlsson
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Johan F A Pijnenborg
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Xin Yin
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Rebecca L Miller
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Sumit K Chanda
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Thomas J Boltje
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Katrine T Schjoldager
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Adnan Halim
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Aaron F Carlin
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ramon Hurtado-Guerrero
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
- The Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquillor s/n, Campus Rio Ebro, 50018, Zaragoza, Spain
- Fundación ARAID, 50018, Zaragoza, Spain
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark.
| | - Yoshiki Narimatsu
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark.
- GlycoDisplay ApS, Copenhagen, Denmark.
| |
Collapse
|
19
|
Chen YH, Tian W, Yasuda M, Ye Z, Song M, Mandel U, Kristensen C, Povolo L, Marques ARA, Čaval T, Heck AJR, Sampaio JL, Johannes L, Tsukimura T, Desnick R, Vakhrushev SY, Yang Z, Clausen H. A universal GlycoDesign for lysosomal replacement enzymes to improve circulation time and biodistribution. Front Bioeng Biotechnol 2023; 11:1128371. [PMID: 36911201 PMCID: PMC9999025 DOI: 10.3389/fbioe.2023.1128371] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/06/2023] [Indexed: 03/14/2023] Open
Abstract
Currently available enzyme replacement therapies for lysosomal storage diseases are limited in their effectiveness due in part to short circulation times and suboptimal biodistribution of the therapeutic enzymes. We previously engineered Chinese hamster ovary (CHO) cells to produce α-galactosidase A (GLA) with various N-glycan structures and demonstrated that elimination of mannose-6-phosphate (M6P) and conversion to homogeneous sialylated N-glycans prolonged circulation time and improved biodistribution of the enzyme following a single-dose infusion into Fabry mice. Here, we confirmed these findings using repeated infusions of the glycoengineered GLA into Fabry mice and further tested whether this glycoengineering approach, Long-Acting-GlycoDesign (LAGD), could be implemented on other lysosomal enzymes. LAGD-engineered CHO cells stably expressing a panel of lysosomal enzymes [aspartylglucosamine (AGA), beta-glucuronidase (GUSB), cathepsin D (CTSD), tripeptidyl peptidase (TPP1), alpha-glucosidase (GAA) or iduronate 2-sulfatase (IDS)] successfully converted all M6P-containing N-glycans to complex sialylated N-glycans. The resulting homogenous glycodesigns enabled glycoprotein profiling by native mass spectrometry. Notably, LAGD extended the plasma half-life of all three enzymes tested (GLA, GUSB, AGA) in wildtype mice. LAGD may be widely applicable to lysosomal replacement enzymes to improve their circulatory stability and therapeutic efficacy.
Collapse
Affiliation(s)
- Yen-Hsi Chen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,GlycoDisplay ApS, Copenhagen, Denmark
| | - Weihua Tian
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zilu Ye
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ming Song
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulla Mandel
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Lorenzo Povolo
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Tomislav Čaval
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science4Life, Utrecht University and Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science4Life, Utrecht University and Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Julio Lopes Sampaio
- Institut Curie, PSL Research University, Cellular and Chemical Biology, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, Cellular and Chemical Biology, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Takahiro Tsukimura
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Functional Bioanalysis, Meiji Pharmaceutical University, Tokyo, Japan
| | - Robert Desnick
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk AS, Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Truberg J, Hobohm L, Jochimsen A, Desel C, Schweizer M, Voss M. Endogenous tagging reveals a mid-Golgi localization of the glycosyltransferase-cleaving intramembrane protease SPPL3. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119345. [PMID: 36007678 DOI: 10.1016/j.bbamcr.2022.119345] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Numerous Golgi-resident enzymes implicated in glycosylation are regulated by the conserved intramembrane protease SPPL3. SPPL3-catalyzed endoproteolysis separates Golgi enzymes from their membrane anchors, enabling subsequent release from the Golgi and secretion. Experimentally altered SPPL3 expression changes glycosylation patterns, yet the regulation of SPPL3-mediated Golgi enzyme cleavage is not understood and conflicting results regarding the subcellular localization of SPPL3 have been reported. Here, we used precise genome editing to generate isogenic cell lines expressing N- or C-terminally tagged SPPL3 from its endogenous locus. Using these cells, we conducted co-localization analyses of tagged endogenous SPPL3 and Golgi markers under steady-state conditions and upon treatment with drugs disrupting Golgi organization. Our data demonstrate that endogenous SPPL3 is Golgi-resident and found predominantly in the mid-Golgi. We find that endogenous SPPL3 co-localizes with its substrates but similarly with non-substrate type II proteins, demonstrating that in addition to co-localization in the Golgi other substrate-intrinsic properties govern SPPL3-mediated intramembrane proteolysis. Given the prevalence of SPPL3-mediated cleavage among Golgi-resident proteins our results have important implications for the regulation of SPPL3 and its role in the organization of the Golgi glycosylation machinery.
Collapse
Affiliation(s)
- Jule Truberg
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, D-24118 Kiel, Germany
| | - Laura Hobohm
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, D-24118 Kiel, Germany
| | - Alexander Jochimsen
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, D-24118 Kiel, Germany
| | - Christine Desel
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, D-24118 Kiel, Germany
| | - Michaela Schweizer
- Morphology and Electron Microscopy, University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), 20251 Hamburg, Germany
| | - Matthias Voss
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, D-24118 Kiel, Germany.
| |
Collapse
|
21
|
Van Coillie J, Schulz MA, Bentlage AEH, de Haan N, Ye Z, Geerdes DM, van Esch WJE, Hafkenscheid L, Miller RL, Narimatsu Y, Vakhrushev SY, Yang Z, Vidarsson G, Clausen H. Role of N-Glycosylation in FcγRIIIa interaction with IgG. Front Immunol 2022; 13:987151. [PMID: 36189205 PMCID: PMC9524020 DOI: 10.3389/fimmu.2022.987151] [Citation(s) in RCA: 19] [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: 07/05/2022] [Accepted: 08/16/2022] [Indexed: 01/09/2023] Open
Abstract
Immunoglobulins G (IgG) and their Fc gamma receptors (FcγRs) play important roles in our immune system. The conserved N-glycan in the Fc region of IgG1 impacts interaction of IgG with FcγRs and the resulting effector functions, which has led to the design of antibody therapeutics with greatly improved antibody-dependent cell cytotoxicity (ADCC) activities. Studies have suggested that also N-glycosylation of the FcγRIII affects receptor interactions with IgG, but detailed studies of the interaction of IgG1 and FcγRIIIa with distinct N-glycans have been hindered by the natural heterogeneity in N-glycosylation. In this study, we employed comprehensive genetic engineering of the N-glycosylation capacities in mammalian cell lines to express IgG1 and FcγRIIIa with different N-glycan structures to more generally explore the role of N-glycosylation in IgG1:FcγRIIIa binding interactions. We included FcγRIIIa variants of both the 158F and 158V allotypes and investigated the key N-glycan features that affected binding affinity. Our study confirms that afucosylated IgG1 has the highest binding affinity to oligomannose FcγRIIIa, a glycan structure commonly found on Asn162 on FcγRIIIa expressed by NK cells but not monocytes or recombinantly expressed FcγRIIIa.
Collapse
Affiliation(s)
- Julie Van Coillie
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Morten A. Schulz
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arthur E. H. Bentlage
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Noortje de Haan
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Zilu Ye
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Lise Hafkenscheid
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca L. Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yoshiki Narimatsu
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- GlycoDisplay ApS, Copenhagen, Denmark
| | - Sergey Y. Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- GlycoDisplay ApS, Copenhagen, Denmark
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Biel TG, Faison T, Matthews AM, Zou G, Ortega-Rodriguez U, Pegues MA, Azer N, Gomez F, Johnson S, Rogstad S, Chen K, Xie H, Agarabi C, Rao VA, Ju T. An etanercept O-glycovariant with enhanced potency. Mol Ther Methods Clin Dev 2022; 25:124-135. [PMID: 35402630 PMCID: PMC8957051 DOI: 10.1016/j.omtm.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/01/2022] [Indexed: 11/30/2022]
Abstract
Most therapeutic proteins are glycosylated with N-glycans and/or O-glycans. N-glycans on therapeutic proteins have been extensively studied for their control strategy and impact on drug product quality. However, knowledge of O-glycosylation in therapeutic protein production and its impact on product quality remains elusive. To address this gap, we generated an O-glycoengineered Chinese Hamster Ovary (CHO) cell line platform to modulate O-glycosylation of therapeutic proteins and investigated the impact of O-glycans on the physicochemical and biological properties of etanercept. Our results demonstrate that this CHO cell line platform produces controlled O-glycosylation profiles containing either truncated O-glycans (sialylTn and/or Tn), or sialylCore 3 alone, or sialylCore 1 with sialylTn or sialylCore 3 O-glycans on endogenous and recombinant proteins. Moreover, the platform demonstrated exclusive modulation of O-glycosylation without affecting N-glycosylation. Importantly, certain O-glycans on etanercept enhanced tumor necrosis factor-α binding affinity and consequent potency. This is the first report that describes the systematic establishment of an O-glycoengineered CHO cell line platform with direct evidence that supports the applicability of the platform in the production of engineered proteins with desired O-glycans. This platform is valuable for identifying O-glycosylation as a critical quality attribute of biotherapeutics using the quality by design principle.
Collapse
Affiliation(s)
- Thomas G Biel
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Talia Faison
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alicia M Matthews
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Guozhang Zou
- Division of Hematology and Oncology Products, Office of New Drugs, Vaccine Production Program, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Uriel Ortega-Rodriguez
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Melissa A Pegues
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Nicole Azer
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Fabiola Gomez
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Sarah Johnson
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Sarah Rogstad
- Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kang Chen
- Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hang Xie
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Cyrus Agarabi
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - V Ashutosh Rao
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Tongzhong Ju
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
23
|
Hobohm L, Koudelka T, Bahr FH, Truberg J, Kapell S, Schacht SS, Meisinger D, Mengel M, Jochimsen A, Hofmann A, Heintz L, Tholey A, Voss M. N-terminome analyses underscore the prevalence of SPPL3-mediated intramembrane proteolysis among Golgi-resident enzymes and its role in Golgi enzyme secretion. Cell Mol Life Sci 2022; 79:185. [PMID: 35279766 PMCID: PMC8918473 DOI: 10.1007/s00018-022-04163-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/07/2022] [Accepted: 01/22/2022] [Indexed: 12/17/2022]
Abstract
Golgi membrane proteins such as glycosyltransferases and other glycan-modifying enzymes are key to glycosylation of proteins and lipids. Secretion of soluble Golgi enzymes that are released from their membrane anchor by endoprotease activity is a wide-spread yet largely unexplored phenomenon. The intramembrane protease SPPL3 can specifically cleave select Golgi enzymes, enabling their secretion and concomitantly altering global cellular glycosylation, yet the entire range of Golgi enzymes cleaved by SPPL3 under physiological conditions remains to be defined. Here, we established isogenic SPPL3-deficient HEK293 and HeLa cell lines and applied N-terminomics to identify substrates cleaved by SPPL3 and released into cell culture supernatants. With high confidence, our study identifies more than 20 substrates of SPPL3, including entirely novel substrates. Notably, our N-terminome analyses provide a comprehensive list of SPPL3 cleavage sites demonstrating that SPPL3-mediated shedding of Golgi enzymes occurs through intramembrane proteolysis. Through the use of chimeric glycosyltransferase constructs we show that transmembrane domains can determine cleavage by SPPL3. Using our cleavage site data, we surveyed public proteome data and found that SPPL3 cleavage products are present in human blood. We also generated HEK293 knock-in cells expressing the active site mutant D271A from the endogenous SPPL3 locus. Immunoblot analyses revealed that secretion of select novel substrates such as the key mucin-type O-glycosylation enzyme GALNT2 is dependent on endogenous SPPL3 protease activity. In sum, our study expands the spectrum of known physiological substrates of SPPL3 corroborating its significant role in Golgi enzyme turnover and secretion as well as in the regulation of global glycosylation pathways.
Collapse
Affiliation(s)
- Laura Hobohm
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
| | - Tomas Koudelka
- Systematic Proteome Research and Bioanalytics, Institute for Experimental Medicine, Kiel University, 24105, Kiel, Germany
| | - Fenja H Bahr
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
| | - Jule Truberg
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
| | - Sebastian Kapell
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Sarah-Sophie Schacht
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
- Institute of Immunology, University Medical Center Schleswig-Holstein, 24105, Kiel, Germany
| | - Daniel Meisinger
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
| | - Marion Mengel
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
| | - Alexander Jochimsen
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
| | - Anna Hofmann
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
| | - Lukas Heintz
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany
- Institute for Cellular and Integrative Physiology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Andreas Tholey
- Systematic Proteome Research and Bioanalytics, Institute for Experimental Medicine, Kiel University, 24105, Kiel, Germany
| | - Matthias Voss
- Institute of Biochemistry, Kiel University, Rudolf-Höber-Str. 1, 24118, Kiel, Germany.
| |
Collapse
|
24
|
Zhong X, D’Antona AM, Scarcelli JJ, Rouse JC. New Opportunities in Glycan Engineering for Therapeutic Proteins. Antibodies (Basel) 2022; 11:5. [PMID: 35076453 PMCID: PMC8788452 DOI: 10.3390/antib11010005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/22/2021] [Accepted: 12/31/2021] [Indexed: 11/17/2022] Open
Abstract
Glycans as sugar polymers are important metabolic, structural, and physiological regulators for cellular and biological functions. They are often classified as critical quality attributes to antibodies and recombinant fusion proteins, given their impacts on the efficacy and safety of biologics drugs. Recent reports on the conjugates of N-acetyl-galactosamine and mannose-6-phosphate for lysosomal degradation, Fab glycans for antibody diversification, as well as sialylation therapeutic modulations and O-linked applications, have been fueling the continued interest in glycoengineering. The current advancements of the human glycome and the development of a comprehensive network in glycosylation pathways have presented new opportunities in designing next-generation therapeutic proteins.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, 610 Main Street, Cambridge, MA 02139, USA;
| | - Aaron M. D’Antona
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, 610 Main Street, Cambridge, MA 02139, USA;
| | - John J. Scarcelli
- BioProcess R&D, Biotherapeutics Pharmaceutical Sciences, Medicinal Sciences, Pfizer Worldwide R&D, 1 Burtt Road, Andover, MA 01810, USA;
| | - Jason C. Rouse
- Analytical R&D, Biotherapeutics Pharmaceutical Sciences, Medicinal Sciences, Pfizer Worldwide R&D, 1 Burtt Road, Andover, MA 01810, USA;
| |
Collapse
|
25
|
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: 35] [Impact Index Per Article: 8.8] [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
|
26
|
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: 1.3] [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
|
27
|
Gao G, Li C, Fan W, Zhang M, Li X, Chen W, Li W, Liang R, Li Z, Zhu X. Brilliant glycans and glycosylation: Seq and ye shall find. Int J Biol Macromol 2021; 189:279-291. [PMID: 34389387 DOI: 10.1016/j.ijbiomac.2021.08.054] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 01/30/2023]
Abstract
Proteoglycosylation is the addition of monosaccharides or glycans to the protein peptide chain. This is a common post-translational modification of proteins with a variety of biological functions. At present, more than half of all biopharmaceuticals in clinic are modified by glycosylation. Most glycoproteins are potential drug targets and biomarkers for disease diagnosis. Therefore, in-depth study of glycan structure of glycoproteins will ultimately improve the sensitivity and specificity of glycoproteins for clinical disease detection. With the deepening of research, the function and application value of glycans and glycosylation has gradually emerged. This review systematically introduces the latest research progress of glycans and glycosylation. It encompasses six cancers, four viruses, and their latest discoveries in Alzheimer's disease, allergic diseases, congenital diseases, gastrointestinal diseases, inflammation, and aging.
Collapse
Affiliation(s)
- Guanwen Gao
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy, Free University of Berlin, Berlin 14195, Germany
| | - Wenguo Fan
- Department of Anesthesiology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Mingtao Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xinming Li
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Wenqing Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Weiquan Li
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Runzhang Liang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Zesong Li
- Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| | - Xiao Zhu
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| |
Collapse
|
28
|
García-García A, Serna S, Yang Z, Delso I, Taleb V, Hicks T, Artschwager R, Vakhrushev SY, Clausen H, Angulo J, Corzana F, Reichardt NC, Hurtado-Guerrero R. FUT8-Directed Core Fucosylation of N-glycans Is Regulated by the Glycan Structure and Protein Environment. ACS Catal 2021; 11:9052-9065. [PMID: 35662980 PMCID: PMC9161449 DOI: 10.1021/acscatal.1c01698] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/24/2021] [Indexed: 12/17/2022]
Abstract
FUT8 is an essential α-1,6-fucosyltransferase that fucosylates the innermost GlcNAc of N-glycans, a process called core fucosylation. In vitro, FUT8 exhibits substrate preference for the biantennary complex N-glycan oligosaccharide (G0), but the role of the underlying protein/peptide to which N-glycans are attached remains unclear. Here, we explored the FUT8 enzyme with a series of N-glycan oligosaccharides, N-glycopeptides, and an Asn-linked oligosaccharide. We found that the underlying peptide plays a role in fucosylation of paucimannose (low mannose) and high-mannose N-glycans but not for complex-type N-glycans. Using saturation transfer difference (STD) NMR spectroscopy, we demonstrate that FUT8 recognizes all sugar units of the G0 N-glycan and most of the amino acid residues (Asn-X-Thr) that serve as a recognition sequon for the oligosaccharyltransferase (OST). The largest STD signals were observed in the presence of GDP, suggesting that prior FUT8 binding to GDP-β-l-fucose (GDP-Fuc) is required for an optimal recognition of N-glycans. We applied genetic engineering of glycosylation capacities in CHO cells to evaluate FUT8 core fucosylation of high-mannose and complex-type N-glycans in cells with a panel of well-characterized therapeutic N-glycoproteins. This confirmed that core fucosylation mainly occurs on complex-type N-glycans, although clearly only at selected glycosites. Eliminating the capacity for complex-type glycosylation in cells (KO mgat1) revealed that glycosites with complex-type N-glycans when converted to high mannose lost the core Fuc. Interestingly, however, for erythropoietin that is uncommon among the tested glycoproteins in efficiently acquiring tetra-antennary N-glycans, two out of three N-glycosites obtained Fuc on the high-mannose N-glycans. An examination of the N-glycosylation sites of several protein crystal structures indicates that core fucosylation is mostly affected by the accessibility and nature of the N-glycan and not by the nature of the underlying peptide sequence. These data have further elucidated the different FUT8 acceptor substrate specificities both in vitro and in vivo in cells, revealing different mechanisms for promoting core fucosylation.
Collapse
Affiliation(s)
- Ana García-García
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, Zaragoza 50018, Spain
| | - Sonia Serna
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Donostia San Sebastián 20014, Spain
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Ignacio Delso
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Víctor Taleb
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, Zaragoza 50018, Spain
| | - Thomas Hicks
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Raik Artschwager
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Donostia San Sebastián 20014, Spain
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Jesús Angulo
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.,Departamento de Química Orgánica, Universidad de Sevilla, Sevilla 41012, Spain.,Instituto de Investigaciones Químicas (CSIC-US), Avda. Américo Vespucio, 49, Seville 41092, Spain
| | - Francisco Corzana
- Departamento de Química, Universidad de La Rioja, Centro de Investigación en Síntesis Química, Logroño E-26006, Spain
| | - Niels C Reichardt
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Donostia San Sebastián 20014, Spain.,CIBER-BBN, Paseo Miramón 182, San Sebastian 20014, Spain
| | - Ramon Hurtado-Guerrero
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, Zaragoza 50018, Spain.,Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark.,Fundación ARAID, Zaragoza 50018, Spain
| |
Collapse
|
29
|
Anderluh M, Berti F, Bzducha‐Wróbel A, Chiodo F, Colombo C, Compostella F, Durlik K, Ferhati X, Holmdahl R, Jovanovic D, Kaca W, Lay L, Marinovic‐Cincovic M, Marradi M, Ozil M, Polito L, Reina‐Martin JJ, Reis CA, Sackstein R, Silipo A, Švajger U, Vaněk O, Yamamoto F, Richichi B, van Vliet SJ. Emerging glyco-based strategies to steer immune responses. FEBS J 2021; 288:4746-4772. [PMID: 33752265 PMCID: PMC8453523 DOI: 10.1111/febs.15830] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/12/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Glycan structures are common posttranslational modifications of proteins, which serve multiple important structural roles (for instance in protein folding), but also are crucial participants in cell-cell communications and in the regulation of immune responses. Through the interaction with glycan-binding receptors, glycans are able to affect the activation status of antigen-presenting cells, leading either to induction of pro-inflammatory responses or to suppression of immunity and instigation of immune tolerance. This unique feature of glycans has attracted the interest and spurred collaborations of glyco-chemists and glyco-immunologists to develop glycan-based tools as potential therapeutic approaches in the fight against diseases such as cancer and autoimmune conditions. In this review, we highlight emerging advances in this field, and in particular, we discuss on how glycan-modified conjugates or glycoengineered cells can be employed as targeting devices to direct tumor antigens to lectin receptors on antigen-presenting cells, like dendritic cells. In addition, we address how glycan-based nanoparticles can act as delivery platforms to enhance immune responses. Finally, we discuss some of the latest developments in glycan-based therapies, including chimeric antigen receptor (CAR)-T cells to achieve targeting of tumor-associated glycan-specific epitopes, as well as the use of glycan moieties to suppress ongoing immune responses, especially in the context of autoimmunity.
Collapse
Affiliation(s)
- Marko Anderluh
- Chair of Pharmaceutical ChemistryFaculty of PharmacyUniversity of LjubljanaSlovenia
| | | | - Anna Bzducha‐Wróbel
- Department of Biotechnology and Food MicrobiologyWarsaw University of Life Sciences‐SGGWPoland
| | - Fabrizio Chiodo
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| | - Cinzia Colombo
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Federica Compostella
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanItaly
| | - Katarzyna Durlik
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Xhenti Ferhati
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Rikard Holmdahl
- Division of Medical Inflammation ResearchDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Dragana Jovanovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Wieslaw Kaca
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Luigi Lay
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Milena Marinovic‐Cincovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Marco Marradi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Musa Ozil
- Department of ChemistryFaculty of Arts and SciencesRecep Tayyip Erdogan University RizeTurkey
| | | | | | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortugal
- IPATIMUP‐Institute of Molecular Pathology and ImmunologyInstituto de Ciências Biomédicas Abel SalazarUniversity of PortoPortugal
| | - Robert Sackstein
- Department of Translational Medicinethe Translational Glycobiology InstituteHerbert Wertheim College of MedicineFlorida International UniversityMiamiFLUSA
| | - Alba Silipo
- Department of Chemical SciencesUniversity of Naples Federico IIComplesso Universitario Monte Sant’AngeloNapoliItaly
| | - Urban Švajger
- Blood Transfusion Center of SloveniaLjubljanaSlovenia
| | - Ondřej Vaněk
- Department of BiochemistryFaculty of ScienceCharles UniversityPragueCzech Republic
| | - Fumiichiro Yamamoto
- Immunohematology & Glycobiology LaboratoryJosep Carreras Leukaemia Research InstituteBadalonaSpain
| | - Barbara Richichi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| |
Collapse
|
30
|
A molecular genetics view on Mucopolysaccharidosis Type II. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 788:108392. [PMID: 34893157 DOI: 10.1016/j.mrrev.2021.108392] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Abstract
Mucopolysaccharidosis Type II (MPS II) is an X-linked recessive genetic disorder that primarily affects male patients. With an incidence of 1 in 100,000 male live births, the disease is one of the orphan diseases. MPS II symptoms are caused by mutations in the lysosomal iduronate-2-sulfatase (IDS) gene. The mutations cause a loss of enzymatic performance and result in the accumulation of glycosaminoglycans (GAGs), heparan sulfate and dermatan sulfate, which are no longer degradable. This inadvertent accumulation causes damage in multiple organs and leads either to a severe neurological course or to an attenuated course of the disease, although the exact relationship between mutation, extent of GAG accumulation and disease progression is not yet fully understood. This review is intended to present current diagnostic procedures and therapeutic interventions. In times when the genetic profile of patients plays an increasingly important role in the assessment of therapeutic success and future drug design, we chose to further elucidate the impact of genetic diversity within the IDS gene on disease phenotype and potential implications in current diagnosis, prognosis and therapy. We report recent advances in the structural biological elucidation of I2S enzyme that that promises to improve our future understanding of the molecular damage of the hundreds of IDS gene variants and will aid damage prediction of novel mutations in the future.
Collapse
|
31
|
Kurhade SE, Weiner JD, Gao FP, Farrell MP. Functionalized High Mannose-Specific Lectins for the Discovery of Type I Mannosidase Inhibitors. Angew Chem Int Ed Engl 2021; 60:12313-12318. [PMID: 33728787 PMCID: PMC8131250 DOI: 10.1002/anie.202101249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/27/2021] [Indexed: 01/01/2023]
Abstract
An engineered cyanovirin-N homologue that exhibits specificity for high mannose N-glycans has been constructed to aid type I α 1,2-mannosidase inhibitor discovery and development. Engineering the lectins C-terminus permitted facile functionalization with fluorophores via a sortase and click strategy. The resulting lectin constructs exhibit specificity for cells presenting high mannose N-glycans. Importantly, these lectin constructs can also be applied to specifically assess changes in cell surface glycosylation induced by type I mannosidase inhibitors. Testing the utility of these lectin constructs led to the discovery of type I mannosidase inhibitors with nanomolar potency. Cumulatively, these findings reveal the specificity and utility of the functionalized cyanovirin-N homologue constructs, and highlight their potential in analytical contexts that require high mannose-specific lectins.
Collapse
Affiliation(s)
- Suresh E Kurhade
- Department of Medicinal Chemistry, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| | - Jack D Weiner
- Department of Medicinal Chemistry, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| | - Fei Philip Gao
- Protein Production Group, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| | - Mark P Farrell
- Department of Medicinal Chemistry, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| |
Collapse
|
32
|
Kurhade SE, Weiner JD, Gao FP, Farrell MP. Functionalized High Mannose‐Specific Lectins for the Discovery of Type I Mannosidase Inhibitors. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Suresh E. Kurhade
- Department of Medicinal Chemistry The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| | - Jack D. Weiner
- Department of Medicinal Chemistry The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| | - Fei Philip Gao
- Protein Production Group The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| | - Mark P. Farrell
- Department of Medicinal Chemistry The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| |
Collapse
|
33
|
Donini R, Haslam SM, Kontoravdi C. Glycoengineering Chinese hamster ovary cells: a short history. Biochem Soc Trans 2021; 49:915-931. [PMID: 33704400 PMCID: PMC8106501 DOI: 10.1042/bst20200840] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/26/2021] [Accepted: 02/08/2021] [Indexed: 12/25/2022]
Abstract
Biotherapeutic glycoproteins have revolutionised the field of pharmaceuticals, with new discoveries and continuous improvements underpinning the rapid growth of this industry. N-glycosylation is a critical quality attribute of biotherapeutic glycoproteins that influences the efficacy, half-life and immunogenicity of these drugs. This review will focus on the advances and future directions of remodelling N-glycosylation in Chinese hamster ovary (CHO) cells, which are the workhorse of recombinant biotherapeutic production, with particular emphasis on antibody products, using strategies such as cell line and protein backbone engineering.
Collapse
Affiliation(s)
- Roberto Donini
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Stuart M. Haslam
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
| | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| |
Collapse
|
34
|
Dellas N, Liu J, Botham RC, Huisman GW. Adapting protein sequences for optimized therapeutic efficacy. Curr Opin Chem Biol 2021; 64:38-47. [PMID: 33933937 DOI: 10.1016/j.cbpa.2021.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022]
Abstract
Therapeutic proteins alleviate disease pathology by supplementing missing or defective native proteins, sequestering superfluous proteins, or by acting through designed non-natural mechanisms. Although therapeutic proteins often have the same amino acid sequence as their native counterpart, their maturation paths from expression to the site of physiological activity are inherently different, and optimizing protein sequences for properties that 100s of millions of years of evolution did not need to address presents an opportunity to develop better biological treatments. Because therapeutic proteins are inherently non-natural entities, optimization for their desired function should be considered analogous to that of small molecule drug candidates, which are optimized through expansive combinatorial variation by the medicinal chemist. Here, we review recent successes and challenges of protein engineering for optimized therapeutic efficacy.
Collapse
Affiliation(s)
- Nikki Dellas
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA.
| | - Joyce Liu
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA
| | - Rachel C Botham
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA
| | - Gjalt W Huisman
- Codexis Inc., 200 Penobscot Dr, Redwood City, CA, 94063, USA
| |
Collapse
|
35
|
Wu YS, Khanna R, Schmith V, Lun Y, Shen JS, Garcia A, Dungan L, Perry A, Martin L, Tsai PC, Hamler R, Das AM, Schiffmann R, Johnson FK. Migalastat Tissue Distribution: Extrapolation From Mice to Humans Using Pharmacokinetic Modeling and Comparison With Agalsidase Beta Tissue Distribution in Mice. Clin Pharmacol Drug Dev 2021; 10:1075-1088. [PMID: 33876577 PMCID: PMC8453552 DOI: 10.1002/cpdd.941] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/01/2021] [Indexed: 11/09/2022]
Abstract
Approved therapies for Fabry disease (FD) include migalastat, an oral pharmacological chaperone, and agalsidase beta and agalsidase alfa, 2 forms of enzyme replacement therapy. Broad tissue distribution may be beneficial for clinical efficacy in FD, which has severe manifestations in multiple organs. Here, migalastat and agalsidase beta biodistribution were assessed in mice and modeled using physiologically based pharmacokinetic (PBPK) analysis, and migalastat biodistribution was subsequently extrapolated to humans. In mice, migalastat concentration was highest in kidneys and the small intestine, 2 FD-relevant organs. Agalsidase beta was predominantly sequestered in the liver and spleen (organs unaffected in FD). PBPK modeling predicted that migalastat 123 mg every other day resulted in concentrations exceeding the in vitro half-maximal effective concentration in kidneys, small intestine, skin, heart, and liver in human subjects. However, extrapolation of mouse agalsidase beta concentrations to humans was unsuccessful. In conclusion, migalastat may distribute to tissues that are inaccessible to intravenous agalsidase beta in mice, and extrapolation of mouse migalastat concentrations to humans showed adequate tissue penetration, particularly in FD-relevant organs.
Collapse
Affiliation(s)
- Yi Shuan Wu
- Nuventra Pharma Sciences, Durham, North Carolina, USA
| | - Richie Khanna
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | | | - Yi Lun
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | - Jin-Song Shen
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | | | - Leo Dungan
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | - Anthony Perry
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | - Lukas Martin
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | - Pai-Chi Tsai
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | - Rick Hamler
- Amicus Therapeutics, Inc., Cranbury, New Jersey, USA
| | - Anibh M Das
- Clinic for Paediatric Nephrology, Hepatology and Metabolic Disorders, Hannover Medical School, Hannover, Germany
| | | | | |
Collapse
|
36
|
Xia Q, Huang X, Huang J, Zheng Y, March ME, Li J, Wei Y. The Role of Autophagy in Skeletal Muscle Diseases. Front Physiol 2021; 12:638983. [PMID: 33841177 PMCID: PMC8027491 DOI: 10.3389/fphys.2021.638983] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle is the most abundant type of tissue in human body, being involved in diverse activities and maintaining a finely tuned metabolic balance. Autophagy, characterized by the autophagosome–lysosome system with the involvement of evolutionarily conserved autophagy-related genes, is an important catabolic process and plays an essential role in energy generation and consumption, as well as substance turnover processes in skeletal muscles. Autophagy in skeletal muscles is finely tuned under the tight regulation of diverse signaling pathways, and the autophagy pathway has cross-talk with other pathways to form feedback loops under physiological conditions and metabolic stress. Altered autophagy activity characterized by either increased formation of autophagosomes or inhibition of lysosome-autophagosome fusion can lead to pathological cascades, and mutations in autophagy genes and deregulation of autophagy pathways have been identified as one of the major causes for a variety of skeleton muscle disorders. The advancement of multi-omics techniques enables further understanding of the molecular and biochemical mechanisms underlying the role of autophagy in skeletal muscle disorders, which may yield novel therapeutic targets for these disorders.
Collapse
Affiliation(s)
- Qianghua Xia
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Xubo Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jieru Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yongfeng Zheng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Michael E March
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jin Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yongjie Wei
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
37
|
Cadaoas J, Hu H, Boyle G, Gomero E, Mosca R, Jayashankar K, Machado M, Cullen S, Guzman B, van de Vlekkert D, Annunziata I, Vellard M, Kakkis E, Koppaka V, d’Azzo A. Galactosialidosis: preclinical enzyme replacement therapy in a mouse model of the disease, a proof of concept. Mol Ther Methods Clin Dev 2021; 20:191-203. [PMID: 33426146 PMCID: PMC7782203 DOI: 10.1016/j.omtm.2020.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Galactosialidosis is a rare lysosomal storage disease caused by a congenital defect of protective protein/cathepsin A (PPCA) and secondary deficiency of neuraminidase-1 and β-galactosidase. PPCA is a lysosomal serine carboxypeptidase that functions as a chaperone for neuraminidase-1 and β-galactosidase within a lysosomal multi-protein complex. Combined deficiency of the three enzymes leads to accumulation of sialylated glycoproteins and oligosaccharides in tissues and body fluids and manifests in a systemic disease pathology with severity mostly correlating with the type of mutation(s) and age of onset of the symptoms. Here, we describe a proof-of-concept, preclinical study toward the development of enzyme replacement therapy for galactosialidosis, using a recombinant human PPCA. We show that the recombinant enzyme, taken up by patient-derived fibroblasts, restored cathepsin A, neuraminidase-1, and β-galactosidase activities. Long-term, bi-weekly injection of the recombinant enzyme in a cohort of mice with null mutation at the PPCA (CTSA) locus (PPCA -/- ), a faithful model of the disease, demonstrated a dose-dependent, systemic internalization of the enzyme by cells of various organs, including the brain. This resulted in restoration/normalization of the three enzyme activities, resolution of histopathology, and reduction of sialyloligosacchariduria. These positive results underscore the benefits of a PPCA-mediated enzyme replacement therapy for the treatment of galactosialidosis.
Collapse
Affiliation(s)
| | - Huimin Hu
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | - Elida Gomero
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Rosario Mosca
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | - Mike Machado
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Sean Cullen
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Belle Guzman
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Diantha van de Vlekkert
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Ida Annunziata
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | - Emil Kakkis
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Vish Koppaka
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Alessandra d’Azzo
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
38
|
Kok K, Zwiers KC, Boot RG, Overkleeft HS, Aerts JMFG, Artola M. Fabry Disease: Molecular Basis, Pathophysiology, Diagnostics and Potential Therapeutic Directions. Biomolecules 2021; 11:271. [PMID: 33673160 PMCID: PMC7918333 DOI: 10.3390/biom11020271] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023] Open
Abstract
Fabry disease (FD) is a lysosomal storage disorder (LSD) characterized by the deficiency of α-galactosidase A (α-GalA) and the consequent accumulation of toxic metabolites such as globotriaosylceramide (Gb3) and globotriaosylsphingosine (lysoGb3). Early diagnosis and appropriate timely treatment of FD patients are crucial to prevent tissue damage and organ failure which no treatment can reverse. LSDs might profit from four main therapeutic strategies, but hitherto there is no cure. Among the therapeutic possibilities are intravenous administered enzyme replacement therapy (ERT), oral pharmacological chaperone therapy (PCT) or enzyme stabilizers, substrate reduction therapy (SRT) and the more recent gene/RNA therapy. Unfortunately, FD patients can only benefit from ERT and, since 2016, PCT, both always combined with supportive adjunctive and preventive therapies to clinically manage FD-related chronic renal, cardiac and neurological complications. Gene therapy for FD is currently studied and further strategies such as substrate reduction therapy (SRT) and novel PCTs are under investigation. In this review, we discuss the molecular basis of FD, the pathophysiology and diagnostic procedures, together with the current treatments and potential therapeutic avenues that FD patients could benefit from in the future.
Collapse
Affiliation(s)
- Ken Kok
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Kimberley C Zwiers
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Rolf G Boot
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Hermen S Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Johannes M F G Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Marta Artola
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
39
|
Narimatsu Y, Büll C, Chen YH, Wandall HH, Yang Z, Clausen H. Genetic glycoengineering in mammalian cells. J Biol Chem 2021; 296:100448. [PMID: 33617880 PMCID: PMC8042171 DOI: 10.1016/j.jbc.2021.100448] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Advances in nuclease-based gene-editing technologies have enabled precise, stable, and systematic genetic engineering of glycosylation capacities in mammalian cells, opening up a plethora of opportunities for studying the glycome and exploiting glycans in biomedicine. Glycoengineering using chemical, enzymatic, and genetic approaches has a long history, and precise gene editing provides a nearly unlimited playground for stable engineering of glycosylation in mammalian cells to explore and dissect the glycome and its many biological functions. Genetic engineering of glycosylation in cells also brings studies of the glycome to the single cell level and opens up wider use and integration of data in traditional omics workflows in cell biology. The last few years have seen new applications of glycoengineering in mammalian cells with perspectives for wider use in basic and applied glycosciences, and these have already led to discoveries of functions of glycans and improved designs of glycoprotein therapeutics. Here, we review the current state of the art of genetic glycoengineering in mammalian cells and highlight emerging opportunities.
Collapse
Affiliation(s)
- Yoshiki Narimatsu
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark; GlycoDisplay ApS, Copenhagen, Denmark.
| | - Christian Büll
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark.
| | | | - Hans H Wandall
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Zhang Yang
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark; GlycoDisplay ApS, Copenhagen, Denmark
| | - Henrik Clausen
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
40
|
Weissmann C, Albanese AA, Contreras NE, Gobetto MN, Castellanos LCS, Uchitel OD. Ion channels and pain in Fabry disease. Mol Pain 2021; 17:17448069211033172. [PMID: 34284652 PMCID: PMC8299890 DOI: 10.1177/17448069211033172] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/29/2022] Open
Abstract
Fabry disease (FD) is a progressive, X-linked inherited disorder of glycosphingolipid metabolism due to deficient or absent lysosomal α-galactosidase A (α-Gal A) activity which results in progressive accumulation of globotriaosylceramide (Gb3) and related metabolites. One prominent feature of Fabry disease is neuropathic pain. Accumulation of Gb3 has been documented in dorsal root ganglia (DRG) as well as other neurons, and has lately been associated with the mechanism of pain though the pathophysiology is still unclear. Small fiber (SF) neuropathy in FD differs from other entities in several aspects related to the perception of pain, alteration of fibers as well as drug therapies used in the practice with patients, with therapies far from satisfying. In order to develop better treatments, more information on the underlying mechanisms of pain is needed. Research in neuropathy has gained momentum from the development of preclinical models where different aspects of pain can be modelled and further analyzed. This review aims at describing the different in vitro and FD animal models that have been used so far, as well as some of the insights gained from their use. We focus especially in recent findings associated with ion channel alterations -that apart from the vascular alterations-, could provide targets for improved therapies in pain.
Collapse
Affiliation(s)
- Carina Weissmann
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Adriana A Albanese
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Natalia E Contreras
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - María N Gobetto
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Libia C Salinas Castellanos
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Osvaldo D Uchitel
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| |
Collapse
|
41
|
Wang F, Moen DR, Sauni C, Kan SH, Li S, Le SQ, Lomenick B, Zhang X, Ekins S, Singamsetty S, Wood J, Dickson PI, Chou TF. Enzyme Replacement Therapy for Mucopolysaccharidosis IIID using Recombinant Human α- N-Acetylglucosamine-6-Sulfatase in Neonatal Mice. Mol Pharm 2020; 18:214-227. [PMID: 33320673 DOI: 10.1021/acs.molpharmaceut.0c00831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There is currently no cure or effective treatment available for mucopolysaccharidosis type IIID (MPS IIID, Sanfilippo syndrome type D), a lysosomal storage disorder (LSD) caused by the deficiency of α-N-acetylglucosamine-6-sulfatase (GNS). The clinical symptoms of MPS IIID, like other subtypes of Sanfilippo syndrome, are largely localized to the central nervous system (CNS), and any treatments aiming to ameliorate or reverse the catastrophic and fatal neurologic decline caused by this disease need to be delivered across the blood-brain barrier. Here, we report a proof-of-concept enzyme replacement therapy (ERT) for MPS IIID using recombinant human α-N-acetylglucosamine-6-sulfatase (rhGNS) via intracerebroventricular (ICV) delivery in a neonatal MPS IIID mouse model. We overexpressed and purified rhGNS from CHO cells with a specific activity of 3.9 × 104 units/mg protein and a maximal enzymatic activity at lysosomal pH (pH 5.6), which was stable for over one month at 4 °C in artificial cerebrospinal fluid (CSF). We demonstrated that rhGNS was taken up by MPS IIID patient fibroblasts via the mannose 6-phosphate (M6P) receptor and reduced intracellular glycosaminoglycans to normal levels. The delivery of 5 μg of rhGNS into the lateral cerebral ventricle of neonatal MPS IIID mice resulted in normalization of the enzymatic activity in brain tissues; rhGNS was found to be enriched in lysosomes in MPS IIID-treated mice relative to the control. Furthermore, a single dose of rhGNS was able to reduce the accumulated heparan sulfate and β-hexosaminidase. Our results demonstrate that rhGNS delivered into CSF is a potential therapeutic option for MPS IIID that is worthy of further development.
Collapse
Affiliation(s)
- Feng Wang
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Derek R Moen
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | - Chelsee Sauni
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | - Shih-Hsin Kan
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Research Administration, CHOC Children's Hospital, Orange, California 92868, United States
| | - Shan Li
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Steven Q Le
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Brett Lomenick
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, California 91125, United States
| | - Xiaoyi Zhang
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States
| | - Sean Ekins
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | | | - Jill Wood
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | - Patricia I Dickson
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Tsui-Fen Chou
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States.,Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
42
|
Abasolo I, Seras-Franzoso J, Moltó-Abad M, Díaz-Riascos V, Corchero JL, Pintos-Morell G, Schwartz S. Nanotechnology-based approaches for treating lysosomal storage disorders, a focus on Fabry disease. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1684. [PMID: 33314628 DOI: 10.1002/wnan.1684] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/08/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022]
Abstract
Lysosomal storage disorders (LSDs) are a group of rare diseases in which the defect of a lysosomal protein results in a pathogenic accumulation of nonmetabolized products within the cells. The main treatment for LSDs is enzyme replacement therapy (ERT), consisting in the exogenous administration a recombinant protein to replace the defective one. Although several diseases such as Gaucher, Fabry, and Pompe are treated following this approach, ERT is limited to LSDs without severe neuronal affectation because recombinant enzymes do not cross the blood-brain barrier. Moreover, ERT shows additional drawbacks, including enzyme low half-life, poor bioavailability, and immunogenic responses. In this scenario, nanotechnology-based drug delivery systems (DDS) have been proposed as solution to overcome these limitations and improve the efficacy of ERT. The present review summarizes distinct approaches followed by our group and collaborators on the use of DDS for restoring lysosomal enzymes in disease-affected cells. During the last decade, we have been exploring different synthetic nanoparticles, from electrolytic complexes, to liposomes and aggresomes, for the delivery of α-galactosidase A (GLA) enzyme. Studies were mainly conducted on Fabry disease models, but results can be also extrapolated to other LSDs, as well as to other diseases treated with alternative therapeutic proteins. The advantages and disadvantages of different DDS, the difficulties from working with very labile and highly glycosylated enzymes and the relevance of using appropriate targeting moieties is thoroughly discussed. Finally, the use of natural DDS, namely extracellular vesicles (EVs) is also introduced. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Ibane Abasolo
- Functional Validation & Preclinical Research, Drug Delivery & Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut of Research (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Joaquin Seras-Franzoso
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.,Drug Delivery & Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut of Research (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Marc Moltó-Abad
- Functional Validation & Preclinical Research, Drug Delivery & Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut of Research (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.,Division of Rare Diseases, Reference Center for Hereditary Metabolic Disorders (CSUR, XUEC, MetabERN, and CIBER-ER), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Vanessa Díaz-Riascos
- Functional Validation & Preclinical Research, Drug Delivery & Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut of Research (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - José Luis Corchero
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.,Institut de Biotecnologia i de Biomedicina (IBB) and Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Guillem Pintos-Morell
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.,Drug Delivery & Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut of Research (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Division of Rare Diseases, Reference Center for Hereditary Metabolic Disorders (CSUR, XUEC, MetabERN, and CIBER-ER), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Simó Schwartz
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.,Drug Delivery & Targeting Group, CIBBIM-Nanomedicine, Vall d'Hebron Institut of Research (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
43
|
Glycomic analysis of host response reveals high mannose as a key mediator of influenza severity. Proc Natl Acad Sci U S A 2020; 117:26926-26935. [PMID: 33046650 PMCID: PMC7604487 DOI: 10.1073/pnas.2008203117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Influenza virus infection causes a range of outcomes from mild illness to death. The molecular mechanisms leading to these differential host responses are currently unknown. Herein, we identify the induction of high mannose, a glycan epitope, as a key mediator of severe disease outcome. We propose a mechanism in which activation of the unfolded protein response (UPR) upon influenza virus infection induces cell surface high mannose, which is then recognized by the innate immune lectin MBL2, activating the complement cascade and leading to subsequent inflammation. This work is the first to systematically study host glycomic changes in response to influenza virus infection, identifying high mannose as a key feature of differential host response. Influenza virus infections cause a wide variety of outcomes, from mild disease to 3 to 5 million cases of severe illness and ∼290,000 to 645,000 deaths annually worldwide. The molecular mechanisms underlying these disparate outcomes are currently unknown. Glycosylation within the human host plays a critical role in influenza virus biology. However, the impact these modifications have on the severity of influenza disease has not been examined. Herein, we profile the glycomic host responses to influenza virus infection as a function of disease severity using a ferret model and our lectin microarray technology. We identify the glycan epitope high mannose as a marker of influenza virus-induced pathogenesis and severity of disease outcome. Induction of high mannose is dependent upon the unfolded protein response (UPR) pathway, a pathway previously shown to associate with lung damage and severity of influenza virus infection. Also, the mannan-binding lectin (MBL2), an innate immune lectin that negatively impacts influenza outcomes, recognizes influenza virus-infected cells in a high mannose-dependent manner. Together, our data argue that the high mannose motif is an infection-associated molecular pattern on host cells that may guide immune responses leading to the concomitant damage associated with severity.
Collapse
|
44
|
Zhang M, Zhao J, Lv Y, Wang W, Feng C, Zou W, Su L, Jiao J. Histone Variants and Histone Modifications in Neurogenesis. Trends Cell Biol 2020; 30:869-880. [PMID: 33011018 DOI: 10.1016/j.tcb.2020.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022]
Abstract
During embryonic brain development, neurogenesis requires the orchestration of gene expression to regulate neural stem cell (NSC) fate specification. Epigenetic regulation with specific emphasis on the modes of histone variants and histone post-translational modifications are involved in interactive gene regulation of central nervous system (CNS) development. Here, we provide a broad overview of the regulatory system of histone variants and histone modifications that have been linked to neurogenesis and diseases. We also review the crosstalk between different histone modifications and discuss how the 3D genome affects cell fate dynamics during brain development. Understanding the mechanisms of epigenetic regulation in neurogenesis has shifted the paradigm from single gene regulation to synergistic interactions to ensure healthy embryonic neurogenesis.
Collapse
Affiliation(s)
- Mengtian Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinyue Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuqing Lv
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenwen Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, University of Science and Technology of China, Hefei 230000, China
| | - Chao Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenzheng Zou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Libo Su
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
45
|
Kightlinger W, Warfel KF, DeLisa MP, Jewett MC. Synthetic Glycobiology: Parts, Systems, and Applications. ACS Synth Biol 2020; 9:1534-1562. [PMID: 32526139 PMCID: PMC7372563 DOI: 10.1021/acssynbio.0c00210] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Indexed: 12/11/2022]
Abstract
Protein glycosylation, the attachment of sugars to amino acid side chains, can endow proteins with a wide variety of properties of great interest to the engineering biology community. However, natural glycosylation systems are limited in the diversity of glycoproteins they can synthesize, the scale at which they can be harnessed for biotechnology, and the homogeneity of glycoprotein structures they can produce. Here we provide an overview of the emerging field of synthetic glycobiology, the application of synthetic biology tools and design principles to better understand and engineer glycosylation. Specifically, we focus on how the biosynthetic and analytical tools of synthetic biology have been used to redesign glycosylation systems to obtain defined glycosylation structures on proteins for diverse applications in medicine, materials, and diagnostics. We review the key biological parts available to synthetic biologists interested in engineering glycoproteins to solve compelling problems in glycoscience, describe recent efforts to construct synthetic glycoprotein synthesis systems, and outline exemplary applications as well as new opportunities in this emerging space.
Collapse
Affiliation(s)
- Weston Kightlinger
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Tech E136, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Tech B486, Evanston, Illinois 60208, United States
| | - Katherine F. Warfel
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Tech E136, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Tech B486, Evanston, Illinois 60208, United States
| | - Matthew P. DeLisa
- Department
of Microbiology, Cornell University, 123 Wing Drive, Ithaca, New York 14853, United States
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New York 14853, United States
- Nancy
E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, New York 14853, United States
| | - Michael C. Jewett
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Tech E136, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Tech B486, Evanston, Illinois 60208, United States
| |
Collapse
|
46
|
Abstract
Following the success of and the high demand for recombinant protein-based therapeutics during the last 25 years, the pharmaceutical industry has invested significantly in the development of novel treatments based on biologics. Mammalian cells are the major production systems for these complex biopharmaceuticals, with Chinese hamster ovary (CHO) cell lines as the most important players. Over the years, various engineering strategies and modeling approaches have been used to improve microbial production platforms, such as bacteria and yeasts, as well as to create pre-optimized chassis host strains. However, the complexity of mammalian cells curtailed the optimization of these host cells by metabolic engineering. Most of the improvements of titer and productivity were achieved by media optimization and large-scale screening of producer clones. The advances made in recent years now open the door to again consider the potential application of systems biology approaches and metabolic engineering also to CHO. The availability of a reference genome sequence, genome-scale metabolic models and the growing number of various “omics” datasets can help overcome the complexity of CHO cells and support design strategies to boost their production performance. Modular design approaches applied to engineer industrially relevant cell lines have evolved to reduce the time and effort needed for the generation of new producer cells and to allow the achievement of desired product titers and quality. Nevertheless, important steps to enable the design of a chassis platform similar to those in use in the microbial world are still missing. In this review, we highlight the importance of mammalian cellular platforms for the production of biopharmaceuticals and compare them to microbial platforms, with an emphasis on describing novel approaches and discussing still open questions that need to be resolved to reach the objective of designing enhanced modular chassis CHO cell lines.
Collapse
|
47
|
Multiplex secretome engineering enhances recombinant protein production and purity. Nat Commun 2020; 11:1908. [PMID: 32313013 PMCID: PMC7170862 DOI: 10.1038/s41467-020-15866-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 03/31/2020] [Indexed: 01/20/2023] Open
Abstract
Host cell proteins (HCPs) are process-related impurities generated during biotherapeutic protein production. HCPs can be problematic if they pose a significant metabolic demand, degrade product quality, or contaminate the final product. Here, we present an effort to create a "clean" Chinese hamster ovary (CHO) cell by disrupting multiple genes to eliminate HCPs. Using a model of CHO cell protein secretion, we predict that the elimination of unnecessary HCPs could have a non-negligible impact on protein production. We analyze the HCP content of 6-protein, 11-protein, and 14-protein knockout clones. These cell lines exhibit a substantial reduction in total HCP content (40%-70%). We also observe higher productivity and improved growth characteristics in specific clones. The reduced HCP content facilitates purification of a monoclonal antibody. Thus, substantial improvements can be made in protein titer and purity through large-scale HCP deletion, providing an avenue to increased quality and affordability of high-value biopharmaceuticals.
Collapse
|
48
|
Kytidou K, Artola M, Overkleeft HS, Aerts JMFG. Plant Glycosides and Glycosidases: A Treasure-Trove for Therapeutics. FRONTIERS IN PLANT SCIENCE 2020; 11:357. [PMID: 32318081 PMCID: PMC7154165 DOI: 10.3389/fpls.2020.00357] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/11/2020] [Indexed: 05/10/2023]
Abstract
Plants contain numerous glycoconjugates that are metabolized by specific glucosyltransferases and hydrolyzed by specific glycosidases, some also catalyzing synthetic transglycosylation reactions. The documented value of plant-derived glycoconjugates to beneficially modulate metabolism is first addressed. Next, focus is given to glycosidases, the central theme of the review. The therapeutic value of plant glycosidases is discussed as well as the present production in plant platforms of therapeutic human glycosidases used in enzyme replacement therapies. The increasing knowledge on glycosidases, including structure and catalytic mechanism, is described. The novel insights have allowed the design of functionalized highly specific suicide inhibitors of glycosidases. These so-called activity-based probes allow unprecedented visualization of glycosidases cross-species. Here, special attention is paid on the use of such probes in plant science that promote the discovery of novel enzymes and the identification of potential therapeutic inhibitors and chaperones.
Collapse
Affiliation(s)
- Kassiani Kytidou
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Marta Artola
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Herman S. Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Johannes M. F. G. Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| |
Collapse
|
49
|
Copoiu L, Malhotra S. The current structural glycome landscape and emerging technologies. Curr Opin Struct Biol 2020; 62:132-139. [PMID: 32006784 DOI: 10.1016/j.sbi.2019.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 11/19/2022]
Abstract
Carbohydrates represent one of the building blocks of life, along with nucleic acids, proteins and lipids. Although glycans are involved in a wide range of processes from embryogenesis to protein trafficking and pathogen infection, we are still a long way from deciphering the glycocode. In this review, we aim to present a few of the challenges that researchers working in the area of glycobiology can encounter and what strategies can be utilised to overcome them. Our goal is to paint a comprehensive picture of the current saccharide landscape available in the Protein Data Bank (PDB). We also review recently updated repositories relevant to the topic proposed, the impact of software development on strategies to structurally solve carbohydrate moieties, and state-of-the-art molecular and cellular biology methods that can shed some light on the function and structure of glycans.
Collapse
Affiliation(s)
- Liviu Copoiu
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Sony Malhotra
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, Malet Street, London WC1E 7HX, United Kingdom.
| |
Collapse
|
50
|
Marqvorsen MHS, Araman C, van Kasteren SI. Going Native: Synthesis of Glycoproteins and Glycopeptides via Native Linkages To Study Glycan-Specific Roles in the Immune System. Bioconjug Chem 2019; 30:2715-2726. [PMID: 31580646 PMCID: PMC6873266 DOI: 10.1021/acs.bioconjchem.9b00588] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/30/2019] [Indexed: 12/16/2022]
Abstract
Glycosylation plays a myriad of roles in the immune system: Certain glycans can interact with specific immune receptors to kickstart a pro-inflammatory response, whereas other glycans can do precisely the opposite and ameliorate the immune response. Specific glycans and glycoforms can themselves become the targets of the adaptive immune system, leading to potent antiglycan responses that can lead to the killing of altered self- or pathogenic species. This hydra-like set of roles glycans play is of particular importance in cancer immunity, where it influences the anticancer immune response, likely playing pivotal roles in tumor survival or clearance. The complexity of carbohydrate biology requires synthetic access to glycoproteins and glycopeptides that harbor homogeneous glycans allowing the probing of these systems with high precision. One particular complicating factor in this is that these synthetic structures are required to be as close to the native structures as possible, as non-native linkages can themselves elicit immune responses. In this Review, we discuss examples and current strategies for the synthesis of natively linked single glycoforms of peptides and proteins that have enabled researchers to gain new insights into glycoimmunology, with a particular focus on the application of these reagents in cancer immunology.
Collapse
Affiliation(s)
- Mikkel H. S. Marqvorsen
- Leiden
Institute of Chemistry, Institute for Chemical Immunology Gorlaeus
Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Can Araman
- Leiden
Institute of Chemistry, Institute for Chemical Immunology Gorlaeus
Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Sander I. van Kasteren
- Leiden
Institute of Chemistry, Institute for Chemical Immunology Gorlaeus
Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|