1
|
Kuznetsov A, Sheshil A, Smolin E, Grudtsov V, Ryazantsev D, Shustinskiy M, Tikhonova T, Kitiashvili I, Vechorko V, Komarova N. Detection of α-Galactosidase A Reaction in Samples Extracted from Dried Blood Spots Using Ion-Sensitive Field Effect Transistors. SENSORS (BASEL, SWITZERLAND) 2024; 24:3681. [PMID: 38894470 PMCID: PMC11175248 DOI: 10.3390/s24113681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024]
Abstract
Fabry disease is a lysosomal storage disorder caused by a significant decrease in the activity or absence of the enzyme α-galactosidase A. The diagnostics of Fabry disease during newborn screening are reasonable, due to the availability of enzyme replacement therapy. This paper presents an electrochemical method using complementary metal-oxide semiconductor (CMOS)-compatible ion-sensitive field effect transistors (ISFETs) with hafnium oxide-sensitive surfaces for the detection of α-galactosidase A activity in dried blood spot extracts. The capability of ISFETs to detect the reaction catalyzed by α-galactosidase A was demonstrated. The buffer composition was optimized to provide suitable conditions for both enzyme and ISFET performance. The use of ISFET structures as sensor elements allowed for the label-free detection of enzymatic reactions with melibiose, a natural substrate of α-galactosidase A, instead of a synthetic fluorogenic one. ISFET chips were packaged with printed circuit boards and microfluidic reaction chambers to enable long-term signal measurement using a custom device. The packaged sensors were demonstrated to discriminate between normal and inhibited GLA activity in dried blood spots extracts. The described method offers a promising solution for increasing the widespread distribution of newborn screening of Fabry disease.
Collapse
Affiliation(s)
- Alexander Kuznetsov
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| | - Andrey Sheshil
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| | - Eugene Smolin
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| | - Vitaliy Grudtsov
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| | - Dmitriy Ryazantsev
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| | - Mark Shustinskiy
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| | - Tatiana Tikhonova
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| | - Irakli Kitiashvili
- Municipal Clinical Hospital No. 15 named after O.M. Filatov, 23 Veshnyakovskaya St., Moscow 111539, Russia
| | - Valerii Vechorko
- Municipal Clinical Hospital No. 15 named after O.M. Filatov, 23 Veshnyakovskaya St., Moscow 111539, Russia
| | - Natalia Komarova
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 32A Leninsky Prospekt, Moscow 119334, Russia
| |
Collapse
|
2
|
Deng M, Zhou H, He S, Qiu H, Wang Y, Zhao AY, Mu Y, Li F, Zhao AZ. Systematic gene therapy derived from an investigative study of AAV2/8 vector gene therapy for Fabry disease. Orphanet J Rare Dis 2023; 18:275. [PMID: 37670350 PMCID: PMC10481556 DOI: 10.1186/s13023-023-02894-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Fabry disease (FD) is a progressive multisystemic disease characterized by a lysosomal enzyme deficiency. A lack of α-galactosidase A (α-Gal A) activity results in the progressive systemic accumulation of its substrates, including globotriaosylceramide (Gb3) and globotriaosylsphingosine (Lyso-Gb3), which results in renal, cardiac, and/or cerebrovascular disease and early death. Enzyme replacement therapy (ERT) is the current standard of care for FD; however, it has important limitations, including a low half-life, limited distribution, and requirement of lifelong biweekly infusions of recombinant enzymes. METHODS Herein, we evaluated a gene therapy approach using an episomal adeno-associated viral 2/8 (AAV2/8) vector that encodes the human GLA cDNA driven by a liver-specific expression cassette in a mouse model of FD that lacks α-Gal A activity and progressively accumulates Gb3 and Lyso-Gb3 in plasma and tissues. RESULTS A pharmacology and toxicology study showed that administration of AAV2/8-hGLA vectors (AAV2/8-hGLA) in FD mice without immunosuppression resulted in significantly increased plasma and tissue α-Gal A activity and substantially normalized Gb3 and Lyso-Gb3 content. CONCLUSIONS Moreover, the plasma enzymatic activity of α-Gal A continued to be stably expressed for up to 38 weeks and sometimes even longer, indicating that AAV2/8-hGLA is effective in treating FD mice, and that α-Gal A is continuously and highly expressed in the liver, secreted into plasma, and absorbed by various tissues. These findings provide a basis for the clinical development of AAV2/8-hGLA.
Collapse
Affiliation(s)
- Mulan Deng
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China
| | - Hongyu Zhou
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China
| | - Shaomei He
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China
| | - Haoheng Qiu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China
| | - Yanping Wang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China
| | - April Yuanyi Zhao
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China
| | - Yunping Mu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China.
| | - Fanghong Li
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China.
| | - Allan Zijian Zhao
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, Guangdong Province, People's Republic of China.
| |
Collapse
|
3
|
Sun S, Li X, Mariappan M. Signal sequences encode information for protein folding in the endoplasmic reticulum. J Cell Biol 2023; 222:213733. [PMID: 36459117 PMCID: PMC9723807 DOI: 10.1083/jcb.202203070] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/22/2022] [Accepted: 10/26/2022] [Indexed: 12/05/2022] Open
Abstract
One-third of newly synthesized proteins in mammals are translocated into the endoplasmic reticulum (ER) through the Sec61 translocon. How protein translocation coordinates with chaperone availability in the ER to promote protein folding remains unclear. We find that marginally hydrophobic signal sequences and transmembrane domains cause transient retention at the Sec61 translocon and require the luminal BiP chaperone for efficient protein translocation. Using a substrate-trapping proteomic approach, we identify that nascent proteins bearing marginally hydrophobic signal sequences accumulate on the cytosolic side of the Sec61 translocon. Sec63 is co-translationally recruited to the translocation site and mediates BiP binding to incoming polypeptides. BiP binding not only releases translocationally paused nascent chains but also ensures protein folding in the ER. Increasing hydrophobicity of signal sequences bypasses Sec63/BiP-dependent translocation, but translocated proteins are prone to misfold and aggregate in the ER under limited BiP availability. Thus, the signal sequence-guided protein folding may explain why signal sequences are diverse and use multiple protein translocation pathways.
Collapse
Affiliation(s)
- Sha Sun
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT
| | - Xia Li
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT
| | - Malaiyalam Mariappan
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT
| |
Collapse
|
4
|
Hwang S, Lee BH, Kim WS, Kim DS, Cheon CK, Lee CH, Choi Y, Choi JH, Kim JH, Yoo HW. A phase II, multicenter, open-label trial to evaluate the safety and efficacy of ISU303 (Agalsidase beta) in patients with Fabry disease. Medicine (Baltimore) 2022; 101:e30345. [PMID: 36123934 PMCID: PMC9478233 DOI: 10.1097/md.0000000000030345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Fabry disease (FD) is caused by a deficiency in the activity of the lysosomal enzyme, α-galactosidase A (α-Gal A), which leads to globotriaosylceramide (Gb3) deposition in multiple tissues. The current management of FD is enzyme replacement therapy (ERT). We report on the efficacy and safety of a new agalsidase beta, ISU303, in FD. METHODS Ten patients (7 males, 3 females) were enrolled and administered a 1 mg/kg dose of ISU303, every other week for 6 months. The primary endpoint was the normalization of plasma Gb3 level. The secondary endpoints were the changes from baseline in urine Gb3 and the plasma and urine lyso-globotriaosylsphingosine (lyso-Gb3) level. Echocardiography, renal function test, and pain-related quality of life were also assessed before and after administration. Safety evaluation was performed including vital signs, laboratory tests, electrocardiograms, antibody screening tests, and adverse events at each visit. RESULTS At 22 weeks of treatment, plasma and urine Gb3 level decreased by a mean of 4.01 ± 1.29 μg/mL (range 2.50-5.70) (P = .005) and 1.12 ± 1.98 μg/mg Cr. (range 0.04-5.65) (P = .017), respectively. However, no significant difference was observed in plasma and urine lyso-Gb3 levels. Echocardiography also was not changed. Renal function and pain-related quality of life showed improvements, but there was no clinical significance. No severe adverse events were observed. Only 1 patient developed an anti-drug antibody without neutralizing activity during the trial. CONCLUSION This study showed the efficacy and safety of ISU303. Treatment with ISU303 significantly resulted in plasma and urine Gb3 decrease in patients with FD. These results suggest that ISU303 is safe and effective and can alternative ERT for FD.
Collapse
Affiliation(s)
- Soojin Hwang
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
- Medical Genetics Center, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Woo-Shik Kim
- Department of Internal Medicine, Kyung Hee University College of Medicine, Seoul, Korea
| | - Dae-Seong Kim
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Chong Kun Cheon
- Department of Pediatrics, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Chang Hwa Lee
- Department of Internal Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Yunha Choi
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin-Ho Choi
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Ja Hye Kim
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
- Medical Genetics Center, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Korea
- *Correspondence: Han-Wook Yoo, Department of Pediatrics, Asan Medical Center Children’s Hospital, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea (e-mail: )
| |
Collapse
|
5
|
Wang J, Li J, Li M, Ma K, Wang D, Su L, Zhang X, Tang BZ. Nanolab in a Cell: Crystallization-Induced In Situ Self-Assembly for Cancer Theranostic Amplification. J Am Chem Soc 2022; 144:14388-14395. [PMID: 35900284 DOI: 10.1021/jacs.2c06111] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Conducting crystallization-assisted self-assembly in living biosystems to obtain large-size nanoparticles and achieve a specific physiological purpose remains an appealing yet significantly challenging task. In this study, we designed Au(I)-disulfide nanosheets containing an aggregation-induced emission photosensitizer, namely, NSs@TTVP, which exhibited pH-responsive crystallization-driven self-assembly capability in lysosomes of cancer cells and tumor tissues of mice. The crystallization process endowed NSs@TTVP with a microscale morphology, stronger fluorescence output, and highly enhanced reactive oxygen species production efficiency. The in vivo results demonstrated that NSs@TTVP shows both long-term retention in tumors and extensive destruction to cancer cells, making it supremely powerful for fluorescence imaging-guided tumor tracking and inhibition.
Collapse
Affiliation(s)
- Jianxing Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China.,School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jie Li
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Meng Li
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ke Ma
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dong Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Lei Su
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Xueji Zhang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Ben Zhong Tang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China.,School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
6
|
Puhl AC, Ekins S. Advancing the Research and Development of Enzyme Replacement Therapies for Lysosomal Storage Diseases. GEN BIOTECHNOLOGY 2022; 1:156-162. [PMID: 35706761 PMCID: PMC9192161 DOI: 10.1089/genbio.2021.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
With the increasing interest in developing gene therapies for rare diseases, it is easy to overlook that there are numerous rare lysosomal storage diseases (LSD) with treatments that have been approved by regulatory agencies in the United States and Europe. These primarily consist of enzyme replacement therapies (ERT), which are recombinant human proteins that are delivered for the life of the patient via different routes and may have distinct safety and distribution advantages over gene therapies. The research and development of ERT is a lengthy and expensive process, which is usually performed in academic laboratories before transfer to pharmaceutical companies and is hence a process ripe for disruption. There may still be considerable scientific and investment potential for ERT, however we need to develop a pipeline of proteins analogous to what has been created in some open science efforts as well as apply technologies to decrease manufacturing costs. In this Perspective, we illustrate the opportunity to fill the rare LSD treatment gap with ERTs while gene therapies are in development for these life-shortening diseases.
Collapse
Affiliation(s)
- Ana C. Puhl
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina, USA
- Address correspondence to: Ana C. Puhl, Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, USA.
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina, USA
- Address correspondence to: Sean Ekins, Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, USA.
| |
Collapse
|
7
|
Mendoza R, Ferrer-Miralles N, Corchero JL. Eukaryotic Aggresomes: Protocols and Tips for Their Production, Purification , and Handling. Methods Mol Biol 2022; 2406:417-435. [PMID: 35089572 DOI: 10.1007/978-1-0716-1859-2_25] [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] [Indexed: 06/14/2023]
Abstract
Aggresomes are insoluble protein aggregates found in eukaryotic cells when the intracellular machinery is overtitered by, for example, the overexpression of a recombinant protein. These protein nanoparticles have become excellent models in studies devoted to elucidate protein aggregation processes in eukaryotic cells, like those involved in "conformational disorders" linked to neurodegenerative diseases. Since the presence of such protein aggregates is a hallmark of these conditions, they constitute an excellent target for new therapeutic approaches for such devastating pathologies. Moreover, and following the pathway opened a few years ago by bacterial inclusion bodies, eukaryotic aggresomes have been proposed as a new type of carrier-free, self-immobilized biocatalysts for use in biotechnology and biomedicine. Altogether, unraveling the characteristics and putative applications of naturally occurring protein aggregates has received an increasing interest during the last years. For that, availability of protocols allowing the production and purification of aggresomes constitute a valuable tool to boost research in the abovementioned fields. In this chapter, we describe both upstream and downstream protocols to obtain aggresomes produced in human cells, using as a model the recombinant human enzyme alpha-galactosidase A (GLA), together with technical tips and advices when working and analyzing eukaryotic aggresomes.
Collapse
Affiliation(s)
- Rosa Mendoza
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Neus Ferrer-Miralles
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - José Luis Corchero
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain.
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| |
Collapse
|
8
|
Abstract
Lysosomes are the main degradative organelles of almost all eukaryotic cells. They fulfil a crucial function in cellular homeostasis, and impairments in lysosomal function are connected to a continuously increasing number of pathological conditions. In recent years, lysosomes are furthermore emerging as control centers of cellular metabolism, and major regulators of cellular signaling were shown to be activated at the lysosomal surface. To date, >300 proteins were demonstrated to be located in/at the lysosome, and the lysosomal proteome and interactome is constantly growing. For the identification of these proteins, and their involvement in cellular mechanisms or disease progression, mass spectrometry (MS)-based proteomics has proven its worth in a large number of studies. In this review, we are recapitulating the application of MS-based approaches for the investigation of the lysosomal proteome, and their application to a diverse set of research questions. Numerous strategies were applied for the enrichment of lysosomes or lysosomal proteins and their identification by MS-based methods. This allowed for the characterization of the lysosomal proteome, the investigation of lysosome-related disorders, the utilization of lysosomal proteins as biomarkers for diseases, and the characterization of lysosome-related cellular mechanisms. While these >60 studies provide a comprehensive picture of the lysosomal proteome across several model organisms and pathological conditions, various proteomics approaches have not been applied to lysosomes yet, and a large number of questions are still left unanswered.
Collapse
Affiliation(s)
- Pathma Muthukottiappan
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Nussallee 11, 53115 Bonn, Germany.
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Nussallee 11, 53115 Bonn, Germany.
| |
Collapse
|
9
|
Strittmatter T, Egli S, Bertschi A, Plieninger R, Bojar D, Xie M, Fussenegger M. Gene switch for l-glucose-induced biopharmaceutical production in mammalian cells. Biotechnol Bioeng 2021; 118:2220-2233. [PMID: 33629358 DOI: 10.1002/bit.27730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/23/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022]
Abstract
In this study, we designed and built a gene switch that employs metabolically inert l-glucose to regulate transgene expression in mammalian cells via d-idonate-mediated control of the bacterial regulator LgnR. To this end, we engineered a metabolic cascade in mammalian cells to produce the inducer molecule d-idonate from its precursor l-glucose by ectopically expressing the Paracoccus species 43P-derived catabolic enzymes LgdA, LgnH, and LgnI. To obtain ON- and OFF-switches, we fused LgnR to the human transcriptional silencer domain Krüppel associated box (KRAB) and the viral trans-activator domain VP16, respectively. Thus, these artificial transcription factors KRAB-LgnR or VP16-LgnR modulated cognate promoters containing LgnR-specific binding sites in a d-idonate-dependent manner as a direct result of l-glucose metabolism. In a proof-of-concept experiment, we show that the switches can control production of the model biopharmaceutical rituximab in both transiently and stably transfected HEK-293T cells, as well as CHO-K1 cells. Rituximab production reached 5.9 µg/ml in stably transfected HEK-293T cells and 3.3 µg/ml in stably transfected CHO-K1 cells.
Collapse
Affiliation(s)
- Tobias Strittmatter
- Department of Biosystems, Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sabina Egli
- Department of Biosystems, Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Adrian Bertschi
- Department of Biosystems, Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Richard Plieninger
- Department of Biosystems, Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Daniel Bojar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts, USA
| | - Mingqi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Martin Fussenegger
- Department of Biosystems, Science and Engineering, ETH Zurich, Basel, Switzerland.,Faculty of Science, University of Basel, Mattenstrasse, Basel, Switzerland
| |
Collapse
|
10
|
Yasuda M, Huston MW, Pagant S, Gan L, St Martin S, Sproul S, Richards D, Ballaron S, Hettini K, Ledeboer A, Falese L, Cao L, Lu Y, Holmes MC, Meyer K, Desnick RJ, Wechsler T. AAV2/6 Gene Therapy in a Murine Model of Fabry Disease Results in Supraphysiological Enzyme Activity and Effective Substrate Reduction. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:607-619. [PMID: 32775495 PMCID: PMC7396970 DOI: 10.1016/j.omtm.2020.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/02/2020] [Indexed: 01/14/2023]
Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the alpha-galactosidase A (GLA) gene, which encodes the exogalactosyl hydrolase, alpha-galactosidase A (α-Gal A). Deficient α-Gal A activity results in the progressive, systemic accumulation of its substrates, globotriaosylceramide (Gb3) and globotriaosylsphingosine (Lyso-Gb3), leading to renal, cardiac, and/or cerebrovascular disease and early demise. The current standard treatment for Fabry disease is enzyme replacement therapy, which necessitates lifelong biweekly infusions of recombinant enzyme. A more long-lasting treatment would benefit Fabry patients. Here, a gene therapy approach using an episomal adeno-associated viral 2/6 (AAV2/6) vector that encodes the human GLA cDNA driven by a liver-specific expression cassette was evaluated in a Fabry mouse model that lacks α-Gal A activity and progressively accumulates Gb3 and Lyso-Gb3 in plasma and tissues. A detailed 3-month pharmacology and toxicology study showed that administration of a clinical-scale-manufactured AAV2/6 vector resulted in markedly increased plasma and tissue α-Gal A activities, and essentially normalized Gb3 and Lyso-Gb3 at key sites of pathology. Further optimization of vector design identified the clinical lead vector, ST-920, which produced several-fold higher plasma and tissue α-Gal A activity levels with a good safety profile. Together, these studies provide the basis for the clinical development of ST-920.
Collapse
Affiliation(s)
- Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Silvere Pagant
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lin Gan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Scott Sproul
- Sangamo Therapeutics, Inc., Brisbane, CA 94005, USA
| | | | | | | | | | | | - Liching Cao
- Sangamo Therapeutics, Inc., Brisbane, CA 94005, USA
| | - Yanmei Lu
- Sangamo Therapeutics, Inc., Brisbane, CA 94005, USA
| | | | | | - Robert J Desnick
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
11
|
Levites EV, Kirikovich SS, Dolgova EV, Proskurina AS, Ritter GS, Ostanin АA, Chernykh ER, Bogachev SS. <i>In vitro</i> assay of biological activity of a national preparation of macrophage activating factor (GcMAF-RF). Vavilovskii Zhurnal Genet Selektsii 2020; 24:284-291. [PMID: 33659810 PMCID: PMC7905294 DOI: 10.18699/vj20.621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
В статье сообщается о разработанном оригинальном способе получения витамин D3-связывающего
белка (DBP) и его конвертации в макрофаг-активирующий фактор GcMAF-RF. Согласно разработанному
регламенту, DBP получали из плазмы крови человека, применяя аффинную колоночную хроматографию, очи-
щали и модифицировали до GcMAF-RF с использованием цитоиммобилизованных гликозидаз (бета-галакто-
зидаза и нейраминидаза). Принадлежность полученного полипептида к Gc-группе глобулинов плазмы крови
подтверждали вестерн-блотом с использованием специфических антител. Полученный полипептид по своим
молекулярным свойствам соответствует описанному в литературе белку GсMAF, находящемуся на стадии кли-
нических испытаний в США, Британии, Израиле и Японии (Saisei Mirai, Reno Integrative Medical Center, Immuno
Biotech Ltd, Efranat, Catalytic Longevity). Биологическую активность препарата GcMAF-RF определяли по индук-
ции у перитонеальных макрофагов мыши фагоцитарной активности и способности продуцировать моноок-
сид азота (NO) in vitro. Фагоцитарную активность макрофагов оценивали по эффективности захвата магнитных
шариков. Степень активации макрофагов рассчитывали по отношению числа захваченных шариков к общему
числу макрофагов. Уровень продукции NO оценивали по накоплению монооксида азота в культуральных су-
пернатантах перитонеальных макрофагов колориметрическим методом с использованием реактива Грисса.
Показано, что GcMAF-RF кратно увеличивает фагоцитарную активность макрофагов и достоверно увеличивает
продукцию ими монооксида азота. Выделенный оригинальным способом активатор макрофагов GcMAF-RF по
своим характеристикам (согласно материалам, опубликованным в печати) соответствует препаратам GcMAF,
представляемым на рынке зарубежными компаниями, и может рассматриваться как новый отечественный био-
логически активный препарат с широким спектром действия. Наибольший интерес вызывает его способность
через активацию макрофагов усиливать адаптивный иммунитет организма. В этой связи предполагаются два
направления терапевтического применения препарата GcMAF-RF. Препарат может быть востребован в области
лечения онкологических заболеваний и, кроме того, может быть использован при лечении ряда нейродегене-
ративных патологий и иммунодефицитных состояний.
Collapse
Affiliation(s)
- E. V. Levites
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences
| | - S. S. Kirikovich
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences
| | - E. V. Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences
| | - A. S. Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences
| | - G. S. Ritter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences; Novosibirsk State University
| | | | | | - S. S. Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences
| |
Collapse
|
12
|
Ostanin AA, Kirikovich SS, Dolgova EV, Proskurina АS, Chernykh ER, Bogachev SS. A thorny pathway of macrophage activating factor (GcMAF): from bench to bedside. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Vitamin D3 Binding Protein (DBP) is a multifunctional glycoprotein whose main role is to transport vitamin D3 and its metabolites, but it also is the precursor of the macrophage activating factor (GcMAF). DBP is converted to GcMAF as a result of site-specific selective deglycosylation under the action of β-galactosidase and sialidase, localized on activated B and T cells, respectively. GcMAF exerts its biological activity primarily as the capability of activating macrophages by enhancing their phagocytic function and producing ROS. Activation results in elevated expression of the specific macrophageal surface receptors involved in the recognition of tumor-associated antigens, as well as in the implementation of direct anticancer activity by inducing the apoptosis or necrosis of tumor cells. Increased interest in GcMAF is associated with its potential to be used in the clinic as a new antitumor drug. Besides its anti-tumor activity, GcMAF exerts a potential against a number of viral and neurodegenerative diseases associated with increased activity of N-acetylgalactosaminidase (nagalase) in the blood serum of patients. Nagalase is an enzyme that completely (rather than selectively) deglycosylates DBP so it cannot be converted to GcMAF, leading to immunodeficiency. Circulating DBP is composed of unmodified and O-glycosylated molecules with the glycosylation degree being dependent on the allelic variants of the gene encoding DBP. The role of DBP in the resistance of organism against a number of diseases is supported by the increased risk of a variety of severe illnesses (amyotrophic lateral sclerosis, colorectal cancer etc.) in patients deficient for GcMAF due to homozygosity for defective DBP alleles. In this review, we also will examine in detail the current data i) on the structure and functions of DBP, as the main precursor of GcMAF, ii) on the main mechanisms of GcMAF anticancer effect, iii) on the tumor strategy for neutralizing GcMAF activity, iv) on the results of GcMAF clinical trials in various cancers; and will discuss the available controversies regarding the positioning of GcMAF as an effective antitumor drug.
Collapse
|
13
|
Zhu X, Yin L, Theisen M, Zhuo J, Siddiqui S, Levy B, Presnyak V, Frassetto A, Milton J, Salerno T, Benenato KE, Milano J, Lynn A, Sabnis S, Burke K, Besin G, Lukacs CM, Guey LT, Finn PF, Martini PG. Systemic mRNA Therapy for the Treatment of Fabry Disease: Preclinical Studies in Wild-Type Mice, Fabry Mouse Model, and Wild-Type Non-human Primates. Am J Hum Genet 2019; 104:625-637. [PMID: 30879639 DOI: 10.1016/j.ajhg.2019.02.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/01/2019] [Indexed: 11/27/2022] Open
Abstract
Fabry disease is an X-linked lysosomal storage disease caused by loss of alpha galactosidase A (α-Gal A) activity and is characterized by progressive accumulation of globotriaosylceramide and its analogs in all cells and tissues. Although enzyme replacement therapy (ERT) is considered standard of care, the long-term effects of ERT on renal and cardiac manifestations remain uncertain and thus novel therapies are desirable. We herein report preclinical studies evaluating systemic messenger RNA (mRNA) encoding human α-Gal A in wild-type (WT) mice, α-Gal A-deficient mice, and WT non-human primates (NHPs). The pharmacokinetics and distribution of h-α-Gal A mRNA encoded protein in WT mice demonstrated prolonged half-lives of α-Gal A in tissues and plasma. Single intravenous administration of h-α-Gal A mRNA to Gla-deficient mice showed dose-dependent protein activity and substrate reduction. Moreover, long duration (up to 6 weeks) of substrate reductions in tissues and plasma were observed after a single injection. Furthermore, repeat i.v. administration of h-α-Gal A mRNA showed a sustained pharmacodynamic response and efficacy in Fabry mice model. Lastly, multiple administrations to non-human primates confirmed safety and translatability. Taken together, these studies across species demonstrate preclinical proof-of-concept of systemic mRNA therapy for the treatment of Fabry disease and this approach may be useful for other lysosomal storage disorders.
Collapse
|
14
|
Rodríguez MC, Ceaglio N, Antuña S, Tardivo MB, Etcheverrigaray M, Prieto C. Production of Therapeutic Enzymes by Lentivirus Transgenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:25-54. [PMID: 31482493 DOI: 10.1007/978-981-13-7709-9_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Since ERT for several LSDs treatment has emerged at the beginning of the 1980s with Orphan Drug approval, patients' expectancy and life quality have been improved. Most LSDs treatment are based on the replaced of mutated or deficient protein with the natural or recombinant protein.One of the main ERT drawback is the high drug prices. Therefore, different strategies trying to optimize the global ERT biotherapeutic production have been proposed. LVs, a gene delivery tool, can be proposed as an alternative method to generate stable cell lines in manufacturing of recombinant proteins. Since LVs have been used in human gene therapy, clinical trials, safety testing assays and procedures have been developed. Moreover, one of the main advantages of LVs strategy to obtain manufacturing cell line is the short period required as well as the high protein levels achieved.In this chapter, we will focus on LVs as a recombinant protein production platform and we will present a case study that employs LVs to express in a manufacturing cell line, alpha-Galactosidase A (rhαGAL), which is used as ERT for Fabry disease treatment.
Collapse
Affiliation(s)
| | - Natalia Ceaglio
- Cell Culture Laboratory, UNL, CONICET, FBCB, Santa Fe, Argentina
| | | | | | | | - Claudio Prieto
- Cell Culture Laboratory, UNL, FBCB, Santa Fe, Argentina.
| |
Collapse
|
15
|
Rodríguez MC, Ceaglio N, Antuña S, Tardivo MB, Etcheverrigaray M, Prieto C. High yield process for the production of active human α-galactosidase a in CHO-K1 cells through lentivirus transgenesis. Biotechnol Prog 2017; 33:1334-1345. [PMID: 28840666 DOI: 10.1002/btpr.2538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/22/2017] [Indexed: 12/15/2022]
Abstract
Fabry disease is an X-linked recessive disorder caused by a deficiency in lysosomal α-Galactosidase A. Currently, two enzyme replacement therapies (ERT) are available. However, access to orphan drugs continues to be limited by their high price. Selection of adequate high-expression systems still constitutes a challenge for alleviating the cost of treatments. Several strategies have been implemented, with varying success, trying to optimize the production process of recombinant human α-Galactosidase A (rhαGAL) in Chinese hamster ovary (CHO-K1) cells. Herein, we describe for the first time the application of a strategy based on third-generation lentiviral particles (LP) transduction of suspension CHO-K1 cells to obtain high-producing rhαGAL clones (3.5 to 59.4 pg cell-1 d-1 ). After two purification steps, the active enzyme was recovered (2.4 × 106 U mg-1 ) with 98% purity and 60% overall yield. Michaelis-Menten analysis demonstrated that rhαGAL was capable of hydrolyzing the synthetic substrate 4MU-α-Gal at a comparable rate to Fabrazyme®, the current CHO-derived ERT available for Fabry disease. In addition, rhαGAL presented the same mannose-6-phosphate (M6P) content, about 40% higher acid sialic amount and 33% reduced content of the immunogenic type of sialic acid (Neu5Gc) than the corresponding ones for Fabrazyme®. In comparison with other rhαGAL production processes reported to date, our approach achieves the highest rhαGAL productivity preserving adequate activity and glycosylation pattern. Even more, considering the improved glycosylation characteristics of rhαGAL, which might provide advantages regarding pharmacokinetics, our enzyme could be postulated as a promising alternative for therapeutic use in Fabry disease. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:1334-1345, 2017.
Collapse
Affiliation(s)
- María Celeste Rodríguez
- Universidad Nacional del Litoral, CONICET, School of Biochemistry and Biological Sciences, Cell Culture Laboratory, Ciudad Universitaria, Paraje El Pozo, C.C. 242, Santa Fe, S3000ZAA, Argentina
| | - Natalia Ceaglio
- Universidad Nacional del Litoral, CONICET, School of Biochemistry and Biological Sciences, Cell Culture Laboratory, Ciudad Universitaria, Paraje El Pozo, C.C. 242, Santa Fe, S3000ZAA, Argentina
| | | | | | - Marina Etcheverrigaray
- Universidad Nacional del Litoral, CONICET, School of Biochemistry and Biological Sciences, Cell Culture Laboratory, Ciudad Universitaria, Paraje El Pozo, C.C. 242, Santa Fe, S3000ZAA, Argentina
| | - Claudio Prieto
- Universidad Nacional del Litoral, School of Biochemistry and Biological Sciences, Cell Culture Laboratory, Ciudad Universitaria, Paraje El Pozo, C.C. 242, Santa Fe, S3000ZAA, Argentina
| |
Collapse
|
16
|
Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev 2017; 118:109-134. [PMID: 28502768 PMCID: PMC5828774 DOI: 10.1016/j.addr.2017.05.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/06/2023]
Abstract
Lysosomes and lysosomal enzymes play a central role in numerous cellular processes, including cellular nutrition, recycling, signaling, defense, and cell death. Genetic deficiencies of lysosomal components, most commonly enzymes, are known as "lysosomal storage disorders" or "lysosomal diseases" (LDs) and lead to lysosomal dysfunction. LDs broadly affect peripheral organs and the central nervous system (CNS), debilitating patients and frequently causing fatality. Among other approaches, enzyme replacement therapy (ERT) has advanced to the clinic and represents a beneficial strategy for 8 out of the 50-60 known LDs. However, despite its value, current ERT suffers from several shortcomings, including various side effects, development of "resistance", and suboptimal delivery throughout the body, particularly to the CNS, lowering the therapeutic outcome and precluding the use of this strategy for a majority of LDs. This review offers an overview of the biomedical causes of LDs, their socio-medical relevance, treatment modalities and caveats, experimental alternatives, and future treatment perspectives.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University Maryland, College Park, MD 20742, USA.
| |
Collapse
|
17
|
Krauss U, Jäger VD, Diener M, Pohl M, Jaeger KE. Catalytically-active inclusion bodies-Carrier-free protein immobilizates for application in biotechnology and biomedicine. J Biotechnol 2017; 258:136-147. [PMID: 28465211 DOI: 10.1016/j.jbiotec.2017.04.033] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 02/08/2023]
Abstract
Bacterial inclusion bodies (IBs) consist of unfolded protein aggregates and represent inactive waste products often accumulating during heterologous overexpression of recombinant genes in Escherichia coli. This general misconception has been challenged in recent years by the discovery that IBs, apart from misfolded polypeptides, can also contain substantial amounts of active and thus correctly or native-like folded protein. The corresponding catalytically-active inclusion bodies (CatIBs) can be regarded as a biologically-active sub-micrometer sized biomaterial or naturally-produced carrier-free protein immobilizate. Fusion of polypeptide (protein) tags can induce CatIB formation paving the way towards the wider application of CatIBs in synthetic chemistry, biocatalysis and biomedicine. In the present review we summarize the history of CatIBs, present the molecular-biological tools that are available to induce CatIB formation, and highlight potential lines of application. In the second part findings regarding the formation, architecture, and structure of (Cat)IBs are summarized. Finally, an overview is presented about the available bioinformatic tools that potentially allow for the prediction of aggregation and thus (Cat)IB formation. This review aims at demonstrating the potential of CatIBs for biotechnology and hopefully contributes to a wider acceptance of this promising, yet not widely utilized, protein preparation.
Collapse
Affiliation(s)
- Ulrich Krauss
- Institut für Molekulare Enzymtechnologie, Heinrich-Heine Universität Düsseldorf, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany.
| | - Vera D Jäger
- Institut für Molekulare Enzymtechnologie, Heinrich-Heine Universität Düsseldorf, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany
| | - Martin Diener
- Institut für Molekulare Enzymtechnologie, Heinrich-Heine Universität Düsseldorf, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany
| | - Martina Pohl
- IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany
| | - Karl-Erich Jaeger
- Institut für Molekulare Enzymtechnologie, Heinrich-Heine Universität Düsseldorf, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany; IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany
| |
Collapse
|
18
|
Stütz AE, Wrodnigg TM. Carbohydrate-Processing Enzymes of the Lysosome: Diseases Caused by Misfolded Mutants and Sugar Mimetics as Correcting Pharmacological Chaperones. Adv Carbohydr Chem Biochem 2016; 73:225-302. [PMID: 27816107 DOI: 10.1016/bs.accb.2016.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lysosomal storage diseases are hereditary disorders caused by mutations on genes encoding for one of the more than fifty lysosomal enzymes involved in the highly ordered degradation cascades of glycans, glycoconjugates, and other complex biomolecules in the lysosome. Several of these metabolic disorders are associated with the absence or the lack of activity of carbohydrate-processing enzymes in this cell compartment. In a recently introduced therapy concept, for susceptible mutants, small substrate-related molecules (so-called pharmacological chaperones), such as reversible inhibitors of these enzymes, may serve as templates for the correct folding and transport of the respective protein mutant, thus improving its concentration and, consequently, its enzymatic activity in the lysosome. Carbohydrate-processing enzymes in the lysosome, related lysosomal diseases, and the scope and limitations of reported reversible inhibitors as pharmacological chaperones are discussed with a view to possibly extending and improving research efforts in this area of orphan diseases.
Collapse
Affiliation(s)
- Arnold E Stütz
- Glycogroup, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Tanja M Wrodnigg
- Glycogroup, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| |
Collapse
|
19
|
Abstract
Research and drug developments fostered under orphan drug product development programs have greatly assisted the introduction of efficient and safe enzyme-based therapies for a range of rare disorders. The introduction and regulatory approval of 20 different recombinant enzymes has enabled, often for the first time, effective enzyme-replacement therapy for some lysosomal storage disorders, including Gaucher (imiglucerase, taliglucerase, and velaglucerase), Fabry (agalsidase alfa and beta), and Pompe (alglucosidase alfa) diseases and mucopolysaccharidoses I (laronidase), II (idursulfase), IVA (elosulfase), and VI (galsulfase). Approved recombinant enzymes are also now used as therapy for myocardial infarction (alteplase, reteplase, and tenecteplase), cystic fibrosis (dornase alfa), chronic gout (pegloticase), tumor lysis syndrome (rasburicase), leukemia (L-asparaginase), some collagen-based disorders such as Dupuytren's contracture (collagenase), severe combined immunodeficiency disease (pegademase bovine), detoxification of methotrexate (glucarpidase), and vitreomacular adhesion (ocriplasmin). The development of these efficacious and safe enzyme-based therapies has occurred hand in hand with some remarkable advances in the preparation of the often specifically designed recombinant enzymes; the manufacturing expertise necessary for commercial production; our understanding of underlying mechanisms operative in the different diseases; and the mechanisms of action of the relevant recombinant enzymes. Together with information on these mechanisms, safety findings recorded so far on the various adverse events and problems of immunogenicity of the recombinant enzymes used for therapy are presented.
Collapse
|
20
|
Eukaryotic aggresomes: from a model of conformational diseases to an emerging type of immobilized biocatalyzers. Appl Microbiol Biotechnol 2015; 100:559-69. [DOI: 10.1007/s00253-015-7107-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/14/2015] [Accepted: 10/15/2015] [Indexed: 12/28/2022]
|
21
|
Rodríguez-Carmona E, Mendoza R, Ruiz-Cánovas E, Ferrer-Miralles N, Abasolo I, Schwartz S, Villaverde A, Corchero JL. A novel bio-functional material based on mammalian cell aggresomes. Appl Microbiol Biotechnol 2015; 99:7079-88. [PMID: 26003454 DOI: 10.1007/s00253-015-6684-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/30/2015] [Accepted: 05/07/2015] [Indexed: 11/27/2022]
Abstract
Aggresomes are protein aggregates found in mammalian cells when the intracellular protein degradation machinery is over-titered. Despite that they abound in cells producing recombinant proteins of biomedical and biotechnological interest, the physiological roles of these protein clusters and the functional status of the embedded proteins remain basically unexplored. In this work, we have determined for the first time that, like in bacterial inclusion bodies, deposition of recombinant proteins into aggresomes does not imply functional inactivation. As a model, human α-galactosidase A (GLA) has been expressed in mammalian cells as enzymatically active, mechanically stable aggresomes showing higher thermal stability than the soluble GLA version. Since aggresomes are easily produced and purified, we propose these particles as novel functional biomaterials with potential as carrier-free, self-immobilized catalyzers in biotechnology and biomedicine.
Collapse
Affiliation(s)
- Escarlata Rodríguez-Carmona
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Meghdari M, Gao N, Abdullahi A, Stokes E, Calhoun DH. Carboxyl-terminal truncations alter the activity of the human α-galactosidase A. PLoS One 2015; 10:e0118341. [PMID: 25719393 PMCID: PMC4342250 DOI: 10.1371/journal.pone.0118341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 01/13/2015] [Indexed: 12/17/2022] Open
Abstract
Fabry disease is an X-linked inborn error of glycolipid metabolism caused by deficiency of the human lysosomal enzyme, α-galactosidase A (αGal), leading to strokes, myocardial infarctions, and terminal renal failure, often leading to death in the fourth or fifth decade of life. The enzyme is responsible for the hydrolysis of terminal α-galactoside linkages in various glycolipids. Enzyme replacement therapy (ERT) has been approved for the treatment of Fabry disease, but adverse reactions, including immune reactions, make it desirable to generate improved methods for ERT. One approach to circumvent these adverse reactions is the development of derivatives of the enzyme with more activity per mg. It was previously reported that carboxyl-terminal deletions of 2 to 10 amino acids led to increased activity of about 2 to 6-fold. However, this data was qualitative or semi-quantitative and relied on comparison of the amounts of mRNA present in Northern blots with αGal enzyme activity using a transient expression system in COS-1 cells. Here we follow up on this report by constructing and purifying mutant enzymes with deletions of 2, 4, 6, 8, and 10 C-terminal amino acids (Δ2, Δ4, Δ6, Δ8, Δ10) for unambiguous quantitative enzyme assays. The results reported here show that the kcat/Km approximately doubles with deletions of 2, 4, 6 and 10 amino acids (0.8 to 1.7-fold effect) while a deletion of 8 amino acids decreases the kcat/Km (7.2-fold effect). These results indicate that the mutated enzymes with increased activity constructed here would be expected to have a greater therapeutic effect on a per mg basis, and could therefore reduce the likelihood of adverse infusion related reactions in Fabry patients receiving ERT treatment. These results also illustrate the principle that in vitro mutagenesis can be used to generate αGal derivatives with improved enzyme activity.
Collapse
Affiliation(s)
- Mariam Meghdari
- Chemistry Dept., City College of New York, New York, NY, USA
| | - Nicholas Gao
- Chemistry Dept., City College of New York, New York, NY, USA
| | - Abass Abdullahi
- Biology & Medical Lab Technology, Bronx Community College, Bronx, NY, USA
| | - Erin Stokes
- Chemistry Dept., City College of New York, New York, NY, USA
| | - David H. Calhoun
- Chemistry Dept., City College of New York, New York, NY, USA
- * E-mail:
| |
Collapse
|
23
|
Golebiowski D, Bradbury AM, Kwon CS, van der Bom IMJ, Stoica L, Johnson AK, Wilson DU, Gray-Edwards HL, Hudson JA, Johnson JA, Randle AN, Whitlock BK, Sartin JL, Kühn AL, Gounis M, Asaad W, Martin DR, Sena-Esteves M. AAV Gene Therapy Strategies for Lysosomal Storage Disorders with Central Nervous System Involvement. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/978-1-4939-2306-9_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
24
|
Smith W, Jäntti J, Oja M, Saloheimo M. Comparison of intracellular and secretion-based strategies for production of human α-galactosidase A in the filamentous fungus Trichoderma reesei. BMC Biotechnol 2014; 14:91. [PMID: 25344685 PMCID: PMC4219008 DOI: 10.1186/s12896-014-0091-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/09/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Trichoderma reesei is known as a good producer of industrial proteins but has hitherto been less successful in the production of therapeutic proteins. In order to elucidate the bottlenecks of heterologous protein production, human α-galactosidase A (GLA) was chosen as a model therapeutic protein. Fusion partners were designed to compare the effects of secretion using a cellobiohydrolase I (CBHI) carrier and intracellular production using a gamma zein peptide from maize (ZERA) which accumulates inside the endoplasmic reticulum (ER). The two strategies were compared on the basis of expression levels, purification performance, enzymatic activity, bioreactor cultivations, and transcriptional profiling. RESULTS Constructs were cloned into the cbh1 locus of the T. reesei strain Rut-C30. The secretion and intracellular strains produced 20 mg/l and 636 mg/l of GLA respectively. Purifications of secreted product were accomplished using Step-Tactin affinity columns and for intracellular product, a method was developed for gravity-based density separation and protein body solubilisation. The secreted protein had similar specific activity to that of the commercially available mammalian form. The intracellular version had 5-10-fold lower activity due to the enzymes incompatibility with alkaline pH. The secretion strain achieved 10% lower total biomass than either the parental or the intracellular strain. The patterns of gene induction for intracellular and parental strains were similar, whereas the secretion strain had a broader spectrum of gene expression level changes. Identification of the genes involved indicated strong secretion stress in the secretion strain and to a lesser extent also in intracellular production. Genes involved in the unfolded protein response (UPR) and ER-associated degradation were induced by GLA production, including; hac1, pdi1, prp1, cnx1, der1, and bap31. CONCLUSIONS Active human α-galactosidase could most effectively be produced intracellularly in Trichoderma reesei at >0.5 g/l by avoidance of the extracellular environment, although purification was challenging due to specific activity losses. Strain analysis revealed that in addition to the issues with secreted proteases, the processes of secretion stress including UPR and ER degradation remain as bottlenecks for heterologous protein production. Genetic engineering to eliminate these bottlenecks is the logical path towards establishing a strain capable of producing sensitive heterologous proteins.
Collapse
|
25
|
Hoogendoorn S, van Puijvelde GHM, Kuiper J, van der Marel GA, Overkleeft HS. A multivalent ligand for the mannose-6-phosphate receptor for endolysosomal targeting of an activity-based probe. Angew Chem Int Ed Engl 2014; 53:10975-8. [PMID: 25163608 DOI: 10.1002/anie.201406842] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Indexed: 12/26/2022]
Abstract
The ubiquitously expressed mannose-6-phosphate receptors (MPRs) are a promising class of receptors for targeted compound delivery into the endolysosomal compartments of a variety of cell types. The development of a synthetic, multivalent, mannose-6-phosphate (M6P) glycopeptide-based MPR ligand is described. The conjugation of this ligand to fluorescent DCG-04, an activity-based probe for cysteine cathepsins, enabled fluorescent readout of its receptor-targeting properties. The resulting M6P-cluster-BODIPY-DCG-04 probe was shown to efficiently label cathepsins in cell lysates as well as in live cells. Furthermore, the introduction of the 6-O-phosphates leads to a completely altered uptake profile in COS and dendritic cells compared to a mannose-containing ligand. Competition with mannose-6-phosphate abolished all uptake of the probe in COS cells, and we conclude that the mannose-6-phosphate cluster targets the MPR and ensures the targeted delivery of cargo bound to the cluster into the endolysosomal pathway.
Collapse
Affiliation(s)
- Sascha Hoogendoorn
- Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2300 RA, Leiden (The Netherlands)
| | | | | | | | | |
Collapse
|
26
|
Hoogendoorn S, van Puijvelde GHM, Kuiper J, van der Marel GA, Overkleeft HS. A Multivalent Ligand for the Mannose-6-Phosphate Receptor for Endolysosomal Targeting of an Activity-Based Probe. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201406842] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
27
|
Kim COK, Oh ES, Park MS. First-in-human study with new recombinant agalsidase beta (ISU303) in healthy subjects. J Clin Pharmacol 2014; 54:675-81. [PMID: 24408305 DOI: 10.1002/jcph.262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 01/06/2014] [Indexed: 11/05/2022]
Abstract
ISU303 is a new recombinant agalsidase beta (Agal) enzyme replacement therapy under investigation for Fabry disease, caused by a deficiency in α-galactosidase A activity that leads to fatty deposits in tissues. We evaluated the pharmacokinetic (PK) parameters, safety and tolerability of ISU303 in healthy adult volunteers. The study was a dose block-randomized, double-blinded, placebo-controlled, single-dosing, and dose escalation phase 1 clinical trial. A total of 18 healthy subjects were enrolled (0.3 mg/kg, n = 6; 1.0 mg/kg, n = 6; placebo, n = 6). Blood samples for PK analysis were collected according to planned time. The PK parameters in each 0.3 and 1.0 mg/kg Agal group were as follows: Cmax (mU/mL) 43.19 ± 5.9 and 195.86 ± 32.3; AUClast (h·mU/mL) 207.91 ± 25.1 and 939.96 ± 158.3; t1/2 (hours) 1.13 ± 0.3 and 1.46 ± 0.2; Cl (mL/min/kg) 1.79 ± 0.2 and 1.34 ± 0.2, respectively. There were seven adverse events (AE) overall. All AEs were resolved without any complications. None were related to the study drug. There were no immunogenicity or any significant infusion-related reactions. The new Agal product exhibited a dose-dependent PK and was well tolerated with no significant AEs in healthy adult volunteers.
Collapse
Affiliation(s)
- Choon O K Kim
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University Health System, Seoul, Korea
| | | | | |
Collapse
|
28
|
Huang L, Pike D, Sleat DE, Nanda V, Lobel P. Potential pitfalls and solutions for use of fluorescent fusion proteins to study the lysosome. PLoS One 2014; 9:e88893. [PMID: 24586430 PMCID: PMC3931630 DOI: 10.1371/journal.pone.0088893] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/13/2014] [Indexed: 01/06/2023] Open
Abstract
Use of fusion protein tags to investigate lysosomal proteins can be complicated by the acidic, protease-rich environment of the lysosome. Potential artifacts include degradation or release of the tag and acid quenching of fluorescence. Tagging can also affect protein folding, glycosylation and/or trafficking. To specifically investigate the use of fluorescent tags to reveal lysosomal localization, we tested mCherry derivatives as C-terminal tags for Niemann-Pick disease type C protein 2 (NPC2), a luminal lysosomal protein. Full-length mCherry was released from the NPC2 chimera while deletion of the 11 N-terminal residues of mCherry generated a cleavage-resistant (cr) fluorescent variant. Insertion of proline linkers between NPC2 and crmCherry had little effect while Gly-Ser linkers promoted cleavage. The NPC2-crmCherry fusion was targeted to the lysosome and restored function in NPC2-deficient cells. Fusion of crmCherry to known and candidate lysosomal proteins revealed that the linkers had different effects on lysosomal localization. Direct fusion of crmCherry impaired mannose 6-phosphorylation and lysosomal targeting of the lysosomal protease tripeptidyl peptidase I (TPP1), while insertion of linkers corrected the defects. Molecular modeling suggested structural bases for the effects of different linkers on NPC2 and TPP1 fusion proteins. While mCherry fusion proteins can be useful tools for studying the lysosome and related organelles, our findings underscore the potential artifacts associated with such applications.
Collapse
Affiliation(s)
- Ling Huang
- . Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
- Department of Pharmacology Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States of America
| | - Douglas Pike
- . Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States of America
| | - David E. Sleat
- . Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States of America
| | - Vikas Nanda
- . Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States of America
| | - Peter Lobel
- . Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States of America
| |
Collapse
|
29
|
Gregoire S, Zhang S, Costanzo J, Wilson K, Fernandez EJ, Kwon I. Cis-suppression to arrest protein aggregation in mammalian cells. Biotechnol Bioeng 2013; 111:462-74. [PMID: 24114411 DOI: 10.1002/bit.25119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/18/2013] [Accepted: 09/09/2013] [Indexed: 12/20/2022]
Abstract
Protein misfolding and aggregation are implicated in numerous human diseases and significantly lower production yield of proteins expressed in mammalian cells. Despite the importance of understanding and suppressing protein aggregation in mammalian cells, a protein design and selection strategy to modulate protein misfolding/aggregation in mammalian cells has not yet been reported. In this work, we address the particular challenge presented by mutation-induced protein aggregation in mammalian cells. We hypothesize that an additional mutation(s) can be introduced in an aggregation-prone protein variant, spatially near the original mutation, to suppress misfolding and aggregation (cis-suppression). As a model protein, we chose human copper, zinc superoxide dismutase mutant (SOD1(A4V) ) containing an alanine to valine mutation at residue 4, associated with the familial form of amyotrophic lateral sclerosis. We used the program RosettaDesign to identify Phe20 in SOD1(A4V) as a key residue responsible for SOD1(A4V) conformational destabilization. This information was used to rationally develop a pool of candidate mutations at the Phe20 site. After two rounds of mammalian-cell based screening of the variants, three novel SOD1(A4V) variants with a significantly reduced aggregation propensity inside cells were selected. The enhanced stability and reduced aggregation propensity of the three novel SOD1(A4V) variants were verified using cell fractionation and in vitro stability assays.
Collapse
Affiliation(s)
- Simpson Gregoire
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, 22904-4741
| | | | | | | | | | | |
Collapse
|
30
|
Bastin G, Heximer SP. Rab family proteins regulate the endosomal trafficking and function of RGS4. J Biol Chem 2013; 288:21836-49. [PMID: 23733193 DOI: 10.1074/jbc.m113.466888] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RGS4, a heterotrimeric G-protein inhibitor, localizes to plasma membrane (PM) and endosomal compartments. Here, we examined Rab-mediated control of RGS4 internalization and recycling. Wild type and constitutively active Rab5 decreased RGS4 PM levels while increasing its endosomal targeting. Rab5, however, did not appreciably affect the PM localization or function of the M1 muscarinic receptor (M1R)/Gq signaling cascade. RGS4-containing endosomes co-localized with subsets of Rab5-, transferrin receptor-, and Lamp1/Lysotracker-marked compartments suggesting RGS4 traffics through PM recycling or acidified endosome pathways. Rab7 activity promoted TGN association, whereas Rab7(dominant negative) trapped RGS4 in late endosomes. Furthermore, RGS4 was found to co-localize with an endosomal pool marked by Rab11, the protein that mediates recycling/sorting of proteins to the PM. The Cys-12 residue in RGS4 appeared important for its Rab11-mediated trafficking to the PM. Rab11(dominant negative) decreased RGS4 PM levels and increased the number of RGS4-containing endosomes. Inhibition of Rab11 activity decreased RGS4 function as an inhibitor of M1R activity without affecting localization and function of the M1R/Gq signaling complex. Thus, both Rab5 activation and Rab11 inhibition decreased RGS4 function in a manner that is independent from their effects on the localization and function of the M1R/Gq signaling complex. This is the first study to implicate Rab GTPases in the intracellular trafficking of an RGS protein. Thus, Rab GTPases may be novel molecular targets for the selective regulation of M1R-mediated signaling via their specific effects on RGS4 trafficking and function.
Collapse
Affiliation(s)
- Guillaume Bastin
- Department of Physiology, Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | | |
Collapse
|
31
|
Desnick RJ, Schuchman EH. Enzyme replacement therapy for lysosomal diseases: lessons from 20 years of experience and remaining challenges. Annu Rev Genomics Hum Genet 2013; 13:307-35. [PMID: 22970722 DOI: 10.1146/annurev-genom-090711-163739] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In 1964, Christian de Duve first suggested that enzyme replacement might prove therapeutic for lysosomal storage diseases (LSDs). Early efforts identified the major obstacles, including the inability to produce large quantities of the normal enzymes, the lack of animal models for proof-of-concept studies, and the potentially harmful immune responses to the "foreign" normal enzymes. Subsequently, the identification of receptor-mediated targeting of lysosomal enzymes, the cloning and overexpression of human lysosomal genes, and the generation of murine models markedly facilitated the development of enzyme replacement therapy (ERT). However, ERT did not become a reality until the early 1990s, when its safety and effectiveness were demonstrated for the treatment of type 1 Gaucher disease. Today, ERT is approved for six LSDs, and clinical trials with recombinant human enzymes are ongoing in several others. Here, we review the lessons learned from 20 years of experience, with an emphasis on the general principles for effective ERT and the remaining challenges.
Collapse
Affiliation(s)
- R J Desnick
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
32
|
Gregoire S, Kwon I. A revisited folding reporter for quantitative assay of protein misfolding and aggregation in mammalian cells. Biotechnol J 2012; 7:1297-307. [PMID: 22623352 DOI: 10.1002/biot.201200103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/15/2012] [Accepted: 05/21/2012] [Indexed: 01/12/2023]
Abstract
Protein misfolding and aggregation play important roles in many physiological processes. These include pathological protein aggregation in neurodegenerative diseases and biopharmaceutical protein aggregation during production in mammalian cells. To develop a simple non-invasive assay for protein misfolding and aggregation in mammalian cells, the folding reporter green fluorescent protein (GFP) system, originally developed for bacterial cells, was evaluated. As a folding reporter, GFP was fused to the C-terminus of a panel of human copper/zinc superoxide dismutase (SOD1) mutants with varying misfolding/aggregation propensities. Flow cytometric analysis of transfected HEK293T and NSC-34 cells revealed that the mean fluorescence intensities of the cells expressing GFP fusion of SOD1 variants exhibited an inverse correlation with the misfolding/aggregation propensities of the four SOD1 variants. Our results support the hypothesis that the extent of misfolding/aggregation of a target protein in mammalian cells can be quantitatively estimated by measuring the mean fluorescence intensity of the cells expressing GFP fusion. The assay method developed herein will facilitate the understanding of aggregation process of SOD1 variants and the identification of aggregation inhibitors. The method also has great promise for misfolding/aggregation studies of other proteins in mammalian cells.
Collapse
Affiliation(s)
- Simpson Gregoire
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22904, USA
| | | |
Collapse
|
33
|
Gregoire S, Irwin J, Kwon I. Techniques for Monitoring Protein Misfolding and Aggregation in Vitro and in Living Cells. KOREAN J CHEM ENG 2012; 29:693-702. [PMID: 23565019 PMCID: PMC3615250 DOI: 10.1007/s11814-012-0060-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protein misfolding and aggregation have been considered important in understanding many neurodegenerative diseases and recombinant biopharmaceutical production. Therefore, various traditional and modern techniques have been utilized to monitor protein aggregation in vitro and in living cells. Fibril formation, morphology and secondary structure content of amyloidogenic proteins in vitro have been monitored by molecular probes, TEM/AFM, and CD/FTIR analyses, respectively. Protein aggregation in living cells has been qualitatively or quantitatively monitored by numerous molecular folding reporters based on either fluorescent protein or enzyme. Aggregation of a target protein is directly correlated to the changes in fluorescence or enzyme activity of the folding reporter fused to the target protein, which allows non-invasive monitoring aggregation of the target protein in living cells. Advances in the techniques used to monitor protein aggregation in vitro and in living cells have greatly facilitated the understanding of the molecular mechanism of amyloidogenic protein aggregation associated with neurodegenerative diseases, optimizing culture conditions to reduce aggregation of biopharmaceuticals expressed in living cells, and screening of small molecule libraries in the search for protein aggregation inhibitors.
Collapse
Affiliation(s)
- Simpson Gregoire
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia22904
| | - Jacob Irwin
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia22904
| | - Inchan Kwon
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia22904
- Institutes on Aging, University of Virginia, Charlottesville, Virginia22904
| |
Collapse
|
34
|
Corchero JL, Mendoza R, Lorenzo J, Rodríguez-Sureda V, Domínguez C, Vázquez E, Ferrer-Miralles N, Villaverde A. Integrated approach to produce a recombinant, his-tagged human α-galactosidase a in mammalian cells. Biotechnol Prog 2011; 27:1206-17. [DOI: 10.1002/btpr.637] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 02/16/2011] [Indexed: 11/06/2022]
|
35
|
Treleaven CM, Tamsett T, Fidler JA, Taksir TV, Cheng SH, Shihabuddin LS, Dodge JC. Comparative analysis of acid sphingomyelinase distribution in the CNS of rats and mice following intracerebroventricular delivery. PLoS One 2011; 6:e16313. [PMID: 21283548 PMCID: PMC3026829 DOI: 10.1371/journal.pone.0016313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/09/2010] [Indexed: 12/04/2022] Open
Abstract
Niemann-Pick A (NPA) disease is a lysosomal storage disorder (LSD) caused by a deficiency in acid sphingomyelinase (ASM) activity. Previously, we reported that biochemical and functional abnormalities observed in ASM knockout (ASMKO) mice could be partially alleviated by intracerebroventricular (ICV) infusion of hASM. We now show that this route of delivery also results in widespread enzyme distribution throughout the rat brain and spinal cord. However, enzyme diffusion into CNS parenchyma did not occur in a linear dose-dependent fashion. Moreover, although the levels of hASM detected in the rat CNS were determined to be within the range shown to be therapeutic in ASMKO mice, the absolute amounts represented less than 1% of the total dose administered. Finally, our results also showed that similar levels of enzyme distribution are achieved across rodent species when the dose is normalized to CNS weight as opposed to whole body weight. Collectively, these data suggest that the efficacy observed following ICV delivery of hASM in ASMKO mice could be scaled to CNS of the rat.
Collapse
Affiliation(s)
| | - Thomas Tamsett
- Genzyme Corporation, Framingham, Massachusetts, United States of America
| | - Jonathan A. Fidler
- Genzyme Corporation, Framingham, Massachusetts, United States of America
| | - Tatyana V. Taksir
- Genzyme Corporation, Framingham, Massachusetts, United States of America
| | - Seng H. Cheng
- Genzyme Corporation, Framingham, Massachusetts, United States of America
| | | | - James C. Dodge
- Genzyme Corporation, Framingham, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
36
|
Pohl S, Marschner K, Storch S, Braulke T. Glycosylation- and phosphorylation-dependent intracellular transport of lysosomal hydrolases. Biol Chem 2009; 390:521-7. [PMID: 19426136 DOI: 10.1515/bc.2009.076] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lysosomes contain more than 50 soluble hydrolases that are targeted to lysosomes in a mannose 6-phosphate (Man6P)-dependent manner. The phosphorylation of man- nose residues on high mannose-type oligosaccharides of newly synthesized lysosomal enzymes is catalyzed by two multimeric enzymes, GlcNAc-1-phosphotransferase and GlcNAc-1-phosphodiester-alpha-N-acetylglucosaminidase, allowing the binding to two distinct Man6P receptors in the Golgi apparatus. Inherited defects in the GlcNAc-1-phosphotransferase complex result in missorting and cellular loss of lysosomal enzymes, and the subsequent lysosomal dysfunction causes the lysosomal storage disorders mucolipidosis types II and III. Biosynthetic studies and the availability of Man6P receptor-deficient mouse models have provided new insights into the structural requirements for preferential binding of subsets of lysosomal enzymes to Man6P receptors as well as the identification of alternative targeting pathways.
Collapse
Affiliation(s)
- Sandra Pohl
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | | | | | | |
Collapse
|
37
|
Benjamin ER, Flanagan JJ, Schilling A, Chang HH, Agarwal L, Katz E, Wu X, Pine C, Wustman B, Desnick RJ, Lockhart DJ, Valenzano KJ. The pharmacological chaperone 1-deoxygalactonojirimycin increases alpha-galactosidase A levels in Fabry patient cell lines. J Inherit Metab Dis 2009; 32:424-40. [PMID: 19387866 DOI: 10.1007/s10545-009-1077-0] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 01/30/2009] [Accepted: 03/03/2009] [Indexed: 01/06/2023]
Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the gene encoding alpha-galactosidase A (alpha-Gal A), with consequent accumulation of its major glycosphingolipid substrate, globotriaosylceramide (GL-3). Over 500 Fabry mutations have been reported; approximately 60% are missense. The iminosugar 1-deoxygalactonojirimycin (DGJ, migalastat hydrochloride, AT1001) is a pharmacological chaperone that selectively binds alpha-Gal A, increasing physical stability, lysosomal trafficking, and cellular activity. To identify DGJ-responsive mutant forms of alpha-Gal A, the effect of DGJ incubation on alpha-Gal A levels was assessed in cultured lymphoblasts from males with Fabry disease representing 75 different missense mutations, one insertion, and one splice-site mutation. Baseline alpha-Gal A levels ranged from 0 to 52% of normal. Increases in alpha-Gal A levels (1.5- to 28-fold) after continuous DGJ incubation for 5 days were seen for 49 different missense mutant forms with varying EC(50) values (820 nmol/L to >1 mmol/L). Amino acid substitutions in responsive forms were located throughout both structural domains of the enzyme. Half of the missense mutant forms associated with classic (early-onset) Fabry disease and a majority (90%) associated with later-onset Fabry disease were responsive. In cultured fibroblasts from males with Fabry disease, the responses to DGJ were comparable to those of lymphoblasts with the same mutation. Importantly, elevated GL-3 levels in responsive Fabry fibroblasts were reduced after DGJ incubation, indicating that increased mutant alpha-Gal A levels can reduce accumulated substrate. These data indicate that DGJ merits further evaluation as a treatment for patients with Fabry disease with various missense mutations.
Collapse
Affiliation(s)
- E R Benjamin
- Amicus Therapeutics, 6 Cedar Brook Drive, Cranbury, NJ 08512, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Oliveira JP, Ferreira S, Barceló J, Gaspar P, Carvalho F, Sá Miranda MC, Månsson JE. Effect of single-nucleotide polymorphisms of the 5' untranslated region of the human α-galactosidase gene on enzyme activity, and their frequencies in Portuguese caucasians. J Inherit Metab Dis 2008; 31 Suppl 2:S247-53. [PMID: 18979223 DOI: 10.1007/s10545-008-0818-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2007] [Revised: 07/28/2008] [Accepted: 07/29/2008] [Indexed: 11/29/2022]
Abstract
BACKGROUND The α-galactosidase gene (GLA) has three single-nucleotide polymorphisms in the 5' untranslated region of exon 1, respectively g.1150G>A, g.1168G>A, g.1170C>T. The g.1150A allele is associated with increased plasma α-galactosidase (α-Gal) activity in hemizygotes, while the others are regarded as biologically neutral. The primary goal of this investigation was to test the hypothesis, raised by a clinical observation and results of a family study, that the g.1170T allele polymorphism is associated with lower α-Gal expression. SUBJECTS AND METHODS Plasma and leukocyte α-Gal activities were assayed in unrelated healthy young adults of both sexes, who had been genotyped for GLA exon 1, and enzyme activity values in carriers of any of the polymorphisms were compared to those of individuals with the standard genotype; GLA exon 1 was genotyped in males who had α-Gal activity in dried blood spots lower than 2 SD below the cohort average. RESULTS AND CONCLUSIONS Mean α-Gal leukocyte activity was ∼ 25% higher in subjects with the g.1170C or CC genotype than in those with the alternative genotypes (p < 0.05). The frequency of the g.1170T allele in subjects with low α-Gal activity in dried blood spots was 4-fold higher (p < 0.05) than in the general population. As in hemizygotes, the g.1150A heterozygote identified in this study had plasma α-Gal activity more than 2-fold above the normal mean. The g.1168A allele did not affect enzyme activity. Surprisingly, females with the standard GLA exon 1 genotype had significantly higher plasma α-Gal activity than genetically comparable males.
Collapse
Affiliation(s)
- J P Oliveira
- Serviço de Genética, Faculdade de Medicina, Universidade do Porto, Alameda Hernâni Monteiro, 4200-319, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
39
|
Lübke T, Lobel P, Sleat DE. Proteomics of the lysosome. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:625-35. [PMID: 18977398 DOI: 10.1016/j.bbamcr.2008.09.018] [Citation(s) in RCA: 201] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 09/24/2008] [Accepted: 09/30/2008] [Indexed: 01/05/2023]
Abstract
Defects in lysosomal function have been associated with numerous monogenic human diseases typically classified as lysosomal storage diseases. However, there is increasing evidence that lysosomal proteins are also involved in more widespread human diseases including cancer and Alzheimer disease. Thus, there is a continuing interest in understanding the cellular functions of the lysosome and an emerging approach to this is the identification of its constituent proteins by proteomic analyses. To date, the mammalian lysosome has been shown to contain approximately 60 soluble luminal proteins and approximately 25 transmembrane proteins. However, recent proteomic studies based upon affinity purification of soluble components or subcellular fractionation to obtain both soluble and membrane components suggest that there may be many more of both classes of protein resident within this organelle than previously appreciated. Discovery of such proteins has important implications for understanding the function and the dynamics of the lysosome but can also lead the way towards the discovery of the genetic basis for human diseases of hitherto unknown etiology. Here, we describe current approaches to lysosomal proteomics and data interpretation and review the new lysosomal proteins that have recently emerged from such studies.
Collapse
Affiliation(s)
- Torben Lübke
- Zentrum Biochemie und Molekulare Zellbiologie, Abteilung Biochemie II, Georg-August Universität Göttingen, 37073 Göttingen, Germany
| | | | | |
Collapse
|
40
|
Liu J, Hong Z, Ding J, Liu J, Zhang J, Chen S. Predominant release of lysosomal enzymes by newborn rat microglia after LPS treatment revealed by proteomic studies. J Proteome Res 2008; 7:2033-49. [PMID: 18380473 DOI: 10.1021/pr7007779] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Growing evidence suggest that microglia may play an important role in the pathogenesis of neurodegenerative disease including Parkinson's disease, Alzheimer's disease, and so forth. The activation of microglia may cause neuronal damage through the release of reactive oxygen species and proinflammatory cytokines. However, the early response of microglial cells remains unclear before cells can secrete the proinflammatory cytokines. Here, a time course analysis showed the earliest expression of inducible nitric oxide synthase and cyclooxygenase-2 at 3 and 24 h following lipopolysaccharide (LPS) treatment. To further define initial response proteins of microglia after LPS treatment, we utilized a novel mass spectrometry-based quantitative proteomic technique termed SILAC (for stable isotope labeling by amino acids in cell culture) to compare the protein profiles of the cell culture-conditioned media of 1 h LPS-treated microglia as compared with controls. The proteomic analysis identified 77 secreted proteins using SignalP; of these, 28 proteins were associated with lysosome of cells and 13 lysosome-related proteins displayed significant changes in the relative abundance after 1 h LPS treatment. Four proteins were further evaluated with Western blot, demonstrating good agreement with quantitative proteomic data. These results suggested that microglia first released some lysosomal enzymes which may be involved in neuronal damage process. Furthermore, ammonium chloride, which inhibits microglia lysosomal enzyme activity, could prevent microglia from causing neuronal injury. Hence, in addition to the numerous novel proteins that are potentially important in microglial activation-mediated neurodegeneration revealed by the search, the study has indicated that the early release of lysosomal enzymes in microglial cells would contribute to LPS-activated inflammatory response.
Collapse
Affiliation(s)
- Jun Liu
- Department of Neurology & Institute of Neurology, Ruijin Hospital, Shanghai Jiatong University School of Medicine, Shanghai, China
| | | | | | | | | | | |
Collapse
|
41
|
Sleat DE, Jadot M, Lobel P. Lysosomal proteomics and disease. Proteomics Clin Appl 2007; 1:1134-46. [PMID: 21136763 DOI: 10.1002/prca.200700250] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Indexed: 11/07/2022]
Abstract
A recent trend in proteomic studies has been to analyze macromolecular complexes such as subcellular organelles instead of complete cells or tissues. This "divide and conquer" approach circumvents some of the formidable problems associated with whole proteome analyses and allows focus on a subset of proteins that may be involved in a particular process or disease of interest. One organelle that has been the focus of considerable attention in proteomic studies is the lysosome, an acidic, membrane-delimited compartment that plays an essential role in the degradation and recycling of biological macromolecules. Lysosomal proteomics have been driven in part by the well-established involvement of this organelle in numerous human diseases, but also by the availability of approaches to selectively visualize and/or isolate subsets of lysosomal proteins. In terms of clinical application, proteomic studies of the lysosome have led to the identification of gene defects in three human hereditary diseases. This review summarizes past progress, current limitations and future directions in the field of lysosomal proteomics.
Collapse
Affiliation(s)
- David E Sleat
- Center for Advanced Biotechnology and Medicine, and Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | | | | |
Collapse
|
42
|
Chen F, Gordon R, Ioannou Y. NPC1 late endosomes contain elevated levels of non-esterified ('free') fatty acids and an abnormally glycosylated form of the NPC2 protein. Biochem J 2006; 390:549-61. [PMID: 15896196 PMCID: PMC1198935 DOI: 10.1042/bj20050236] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
NPC (Niemann-Pick type C) disease is a rare lipidosis characterized by the accumulation of LDL (low-density lipoprotein)-derived non-esterified cholesterol in the E/L (endosomal/lysosomal) system. The gene products that are responsible for the two NPC complementation groups are distinct and dissimilar, yet their cellular and disease phenotypes are virtually indistinguishable. To investigate the relationship between NPC1 and NPC2 and their potential role in NPC disease pathogenesis, we have developed a method for the rapid and efficient isolation of late endocytic vesicles from mouse liver by magnetic chromatography. Late endosomes from Wt (wild-type) and NPC1 mice were found to differ not only in their cholesterol and sphingomyelin content, as expected, but also in their non-esterified ('free') fatty acid content, with NPC1 vesicles showing an approx. 7-fold increase in non-esterified fatty acid levels compared with Wt vesicles. Furthermore, we show that the NPC2 protein is in an incompletely deglycosylated form in NPC1 late endosomes by a mechanism that is specific to the NPC2 protein and not a global aberration of protein glycosylation/deglycosylation or trafficking, since NPC2 secreted from NPC1 cells is indistinguishable from that secreted from Wt cells. Also, a greater proportion of the normally soluble cellular NPC2 protein partitions with detergent-insoluble late endosomal internal membrane domains in NPC1 vesicles. In addition, we show that, although a small amount of the NPC2 protein associates with these membranes in Wt vesicles, this localization becomes much more pronounced in NPC1 vesicles. These results suggest that the function of the NPC2 protein may be compromised as well in NPC1 endosomes, which might explain the paradoxical phenotypic similarities of the two NPC disease complementation groups.
Collapse
Affiliation(s)
- Fannie W. Chen
- *Department of Human Genetics, The Mount Sinai School of Medicine, New York, NY 10029, U.S.A
| | - Ronald E. Gordon
- †Department of Pathology, The Mount Sinai School of Medicine, New York, NY 10029, U.S.A
| | - Yiannis A. Ioannou
- *Department of Human Genetics, The Mount Sinai School of Medicine, New York, NY 10029, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
43
|
Germain DP. Développement clinique de l’agalsidase β pour le traitement de la maladie de Fabry. Med Sci (Paris) 2005. [DOI: 10.1051/medsci/20052111s57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
44
|
Garzuly F, Maródi L, Erdös M, Grubits J, Varga Z, Gelpi E, Rohonyi B, Mázló M, Molnár A, Budka H. Megadolichobasilar anomaly with thrombosis in a family with Fabry's disease and a novel mutation in the α-galactosidase A gene. Brain 2005; 128:2078-83. [PMID: 15947062 DOI: 10.1093/brain/awh546] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Fabry's disease is an X-linked lysosomal storage disorder. alpha-Galactosidase deficiency leads to accumulation of globotriaosylceramide mainly in endothelial and smooth muscle cells. Cerebrovascular symptoms with predominant affection of the vertebrobasilar circulation are one of the major sources of morbidity in Fabry's disease. We present a Hungarian family with Fabry's disease caused by a new mutation in the alpha-galactosidase A gene (GLA), and describe a variant expression of the disease. Megadolichobasilar anomaly was diagnosed in two male patients in the family who died of thrombosis. In another female patient who had suffered from disturbance of the vertebrobasilar circulation, a strongly dilated basilar artery without thrombosis was found at autopsy. Another three family members had basilar strokes and large and elongated basilar arteries on MRI. Genetic analysis disclosed a c.47T-->C missense mutation resulting in L16P in the amino acid sequence of the alpha-galactosidase protein. This report suggests that megadolichobasilar anomaly is potentially life-threatening, and that L16P is a disease-causing mutation in patients with Fabry's disease. Early enzyme replacement therapy may prevent the development of these irreversible cerebrovascular complications.
Collapse
Affiliation(s)
- Ferenc Garzuly
- Department of Neurology, Markusovszky Hospital, Szombathely, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disease that is caused by a deficiency of arylsulfatase A (ASA). The deficiency results in the intralysosomal accumulation of the acidic sphingolipid 3-O-sulfogalactosyl-ceramide (sulfatide). Patients suffer from progressive demyelination and die from multiple neurological deficits. Curative treatment is not available. ASA bears mannose 6-phosphate residues which function as recognition markers in endosome/lysosome-specific targeting pathways. The endocytic targeting route can be exploited to deliver exogenous ASA to the lysosomes of ASA-deficient cells. ASA knockout mice, which develop a disorder related to MLD, have therefore been treated by ex vivo and in vivo gene therapy. Following transplantation of bone marrow cells overexpressing ASA from a retroviral vector, donor-type cells secrete ASA, which is endocytosed by recipient cells. The enzyme transfer results in the metabolic cross-correction of recipient cells and the improvement of biochemical, histological and clinical parameters. For the transfer of the ASA cDNA to non-dividing cells, adenovirus, adeno-associated virus and lentivirus vectors have been constructed. Such vectors might be particularly advantageous for direct ASA gene delivery to the brain, which is the main site of disease in MLD.
Collapse
Affiliation(s)
- Ulrich Matzner
- Rheinische Friedrich-Wilhelms-Universität, Institut für Physiologische Chemie, Nussallee 11, D-53115 Bonn, Germany.
| | | |
Collapse
|
46
|
Yasuda K, Chang HH, Wu HL, Ishii S, Fan JQ. Efficient and rapid purification of recombinant human alpha-galactosidase A by affinity column chromatography. Protein Expr Purif 2005; 37:499-506. [PMID: 15358377 DOI: 10.1016/j.pep.2004.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2004] [Revised: 07/04/2004] [Indexed: 10/26/2022]
Abstract
The lysosomal enzyme alpha-galactosidase A (alpha-Gal A) metabolizes neutral glycosphingolipids that possess alpha-galactoside residues at the non-reducing terminus, and inherited defects in the activity of alpha-Gal A lead to Fabry disease. We describe here an efficient and rapid purification procedure for recombinant alpha-Gal A by sequential Concanavalin A (Con A)-Sepharose and immobilized thio-alpha-galactoside (thio-Gal) agarose column chromatography. Optimal elution conditions for both columns were obtained using overexpressed human alpha-Gal A. We recommend the use of a mixture of 0.9 M methyl alpha-mannoside and 0.9 M methyl alpha-glucoside in 0.1 M acetate buffer (pH 6.0) with 0.1 M NaCl for the maximum recovery of glycoproteins with multiple high-mannose type sugar chains from Con A column chromatography, and that the Con A column should not be reused for the purification of glycoproteins that are used for structural studies. Binding of the enzyme to the thio-Gal column requires acidic condition at pH 4.8. A galactose-containing buffer (25 mM citrate-phosphate buffer, pH 5.5, with 0.1 M galactose, and 0.1 M NaCl) was used to elute alpha-Gal A. This procedure is especially useful for the purification of mutant forms of alpha-Gal A, which are not stable under conventional purification techniques. A protocol that purifies an intracellular mutant alpha-Gal A (M279I) expressed in COS-7 cells within 6h at 62% overall yield is presented.
Collapse
Affiliation(s)
- Kayo Yasuda
- Department of Human Genetics, Mount Sinai School of Medicine, Box 1498, Fifth Avenue at 100th Street, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
Although the first description of a lysosomal storage disorder was that of Tay-Sachs disease in 1881, the lysosome was not discovered until 1955, by Christian De Duve. The first demonstration by Hers in 1963 of a link between an enzyme deficiency and a storage disorder (Pompe's disease) paved the way for a series of seminal discoveries about the intracellular biology of these enzymes and their substrates, culminating in the successful treatment of Gaucher's disease with beta-glucosidase in the early 1990s. It is now recognized that these disorders are not simply a consequence of pure storage, but result from perturbation of complex cell signalling mechanisms. These in turn give rise to secondary structural and biochemical changes, which have important implications for therapy. Significant challenges remain, particularly the treatment of central nervous system disease. It is hoped that recent advances in our understanding of lysosomal biology will enable successful therapies to be developed.
Collapse
Affiliation(s)
- Ashok Vellodi
- Metabolic Unit, Great Ormond Street Hospital for Children NHS Trust, London, UK.
| |
Collapse
|
48
|
Zhou YB, Liu F, Zhu ZD, Zhu H, Zhang X, Wang ZQ, Liu JH, Han ZG. N-glycosylation is required for efficient secretion of a novel human secreted glycoprotein, hPAP21. FEBS Lett 2004; 576:401-7. [PMID: 15498570 DOI: 10.1016/j.febslet.2004.09.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2004] [Revised: 09/10/2004] [Accepted: 09/14/2004] [Indexed: 11/23/2022]
Abstract
The present study reported the isolation and characterization of a novel human secreted protein, named as hPAP21 (human protease-associated domain-containing protein, 21 kDa), encoded by the hypothetical gene chromosome 2 open reading frame 7 (C2orf7) that contains signal peptide in its N-terminus, without transmembrane domain, except for PA domain in its middle. Western blotting assay indicated that the c-Myc tagged hPAP21 could be secreted into culture medium in the transfected Chinese hamster ovary cells. However, the molecular weights, whatever intracellular (28 kDa) or extracellular (30 kDa) forms, are larger than that of the prediction. To define whether the glycosylation was important process for its secretion, endoglycosidase H (Endo H) and PNGase F (PNG F) were employed to evaluate the effect of glycosylation types on secretion of hPAP21. Interestingly, the extracellular forms were primarily sensitive to PNG F, not Endo H, implying that complex N-glycosylation could be required for the secretion of hPAP21. Furthermore, N-glycosylation of Asn171 was confirmed as potential crucial process for the secretory protein via site-directed mutagenesis assay. All data will be contributed to the understanding of molecular functions of hPAP21.
Collapse
Affiliation(s)
- Yu-Bo Zhou
- College of Life Science and Biotechnology, Shanghai Jiaotong University, 1954 Huashan Road, Shanghai 200030, China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Garman SC, Garboczi DN. The molecular defect leading to Fabry disease: structure of human alpha-galactosidase. J Mol Biol 2004; 337:319-35. [PMID: 15003450 DOI: 10.1016/j.jmb.2004.01.035] [Citation(s) in RCA: 284] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Revised: 01/16/2004] [Accepted: 01/21/2004] [Indexed: 01/01/2023]
Abstract
Fabry disease is an X-linked lysosomal storage disease afflicting 1 in 40,000 males with chronic pain, vascular degeneration, cardiac impairment, and other symptoms. Deficiency in the lysosomal enzyme alpha-galactosidase (alpha-GAL) causes an accumulation of its substrate, which ultimately leads to Fabry disease symptoms. Here, we present the structure of the human alpha-GAL glycoprotein determined by X-ray crystallography. The structure is a homodimer with each monomer containing a (beta/alpha)8 domain with the active site and an antiparallel beta domain. N-linked carbohydrate appears at six sites in the glycoprotein dimer, revealing the basis for lysosomal transport via the mannose-6-phosphate receptor. To understand how the enzyme cleaves galactose from glycoproteins and glycolipids, we also determined the structure of the complex of alpha-GAL with its catalytic product. The catalytic mechanism of the enzyme is revealed by the location of two aspartic acid residues (D170 and D231), which act as a nucleophile and an acid/base, respectively. As a point mutation in alpha-GAL can lead to Fabry disease, we have catalogued and plotted the locations of 245 missense and nonsense mutations in the three-dimensional structure. The structure of human alpha-GAL brings Fabry disease into the realm of molecular diseases, where insights into the structural basis of the disease phenotypes might help guide the clinical treatment of patients.
Collapse
Affiliation(s)
- Scott C Garman
- Structural Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Twinbrook II, 12441 Parklawn Drive, Rockville, MD 20852, USA.
| | | |
Collapse
|
50
|
Collette J, Bocock JP, Ahn K, Chapman RL, Godbold G, Yeyeodu S, Erickson AH. Biosynthesis and alternate targeting of the lysosomal cysteine protease cathepsin L. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 241:1-51. [PMID: 15548418 DOI: 10.1016/s0074-7696(04)41001-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Upregulation of cathepsin L expression, whether during development or cell transformation, or mediated by ectopic expression from a plasmid, alters the targeting of the protease and thus its physiological function. Upregulated procathepsin L is targeted to small dense core vesicles and to the dense cores of multivesicular bodies, as well as to lysosomes and to the plasma membrane for selective secretion. The multivesicular vesicles resemble secretory lysosomes characterized in specialized cell types in that they are endosomes that stably store an upregulated protein and they possess the tetraspanin CD63. Morphologically the multivesicular endosomes also resemble late endosomes, but they store procathepsin L, not the active protease, and they are not the major site for LAMP-1 accumulation. Distinction between the lysosomal proenzyme and active protease thus identifies two populations of multivesicular endosomes in fibroblasts, one a storage compartment and one an enzymatically active compartment. A distinctive targeting pathway using aggregation is utilized to enrich the storage endosomes with a particular lysosomal protease that can potentially activate and be secreted.
Collapse
Affiliation(s)
- John Collette
- University of Miami School of Medicine, Department of Molecular and Cellular Pharmacology, Miami, Florida 33101 USA
| | | | | | | | | | | | | |
Collapse
|