1
|
Mandolfo O, Parker H, Usman A, Learmonth YI, Holley RJ, MacDonald A, McKay T, Bigger B. Generation of a novel immunodeficient mouse model of Mucopolysaccharidosis type IIIA to test human stem cell-based therapies. Mol Genet Metab 2024; 143:108533. [PMID: 39059269 DOI: 10.1016/j.ymgme.2024.108533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Mucopolysaccharidosis Type IIIA (MPSIIIA) is a rare inherited lysosomal storage disease caused by mutations in the SGSH gene. This genetic variation results in the deficiency of the N-sulfoglucosamine sulfohydrolase enzyme, preventing the breakdown of heparan sulfate within lysosomes. The progressive accumulation of partially degraded substrate ultimately leads to brain pathology, for which there is currently no approved treatment. An established MPSIIIA mouse model has proved to be a vital asset to test several brain-targeting strategies. Nonetheless, the assessment of human stem cell-based products, an emerging research field, necessitates the use of an immunocompromised xenogeneic disease model. In the present study, we addressed this issue by generating a highly immunodeficient mouse model of MPSIIIA (NOD/SCID/GammaC chain null-MPSIIIA) through five generations of crossing an established MPSIIIA mouse model and a NOD/SCID/GammaC chain null (NSG) mouse. The immune system composition, behavioural phenotype and histopathological hallmarks of the NSG-MPSIIIA model were then evaluated. We demonstrated that NSG-MPSIIIA mice display compromised adaptive immunity, ultimately facilitating the successful engraftment of human iPSC-derived neural progenitor cells in the brain up to three months post-delivery. Furthermore, female NSG-MPSIIIA exhibit spatial working memory deficits and hyperactive behaviour, similar to MPSIIIA mice, which usually manifest around 5 months of age. NSG-MPSIIIA mice also developed primary disease-related neuropathological features in common with the MPSIIIA model, including lysosomal enlargement with storage of excess sulphated heparan sulphate and increased gliosis in several areas of the brain. In the future, the NSG-MPSIIIA mouse model holds the potential to serve as a valuable platform for evaluating human stem-cell based therapies for MPSIIIA patients.
Collapse
Affiliation(s)
- Oriana Mandolfo
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester M13 9PT, UK
| | - Helen Parker
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Asma'u Usman
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester M13 9PT, UK
| | - Yuko Ishikawa Learmonth
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester M13 9PT, UK
| | - Rebecca J Holley
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester M13 9PT, UK
| | - Andrew MacDonald
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Tristan McKay
- Centre for Bioscience, The Manchester Metropolitan University, E206 John Dalton Building, Manchester M1 5GD, UK
| | - Brian Bigger
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester M13 9PT, UK; Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, EH16 4UU, Edinburgh, UK.
| |
Collapse
|
2
|
Kim S, Przybilla MJ, Whitley CB, Ou L, Al-Kofahi M, Jarnes JR. Identification of a novel fusion Iduronidase with improved activity in the cardiovascular system. Mol Genet Metab Rep 2022; 33:100917. [PMID: 36159322 PMCID: PMC9489536 DOI: 10.1016/j.ymgmr.2022.100917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 12/04/2022] Open
Abstract
Background Lysosomal diseases are a group of over 70 rare genetic conditions in which a protein deficiency (most often an enzyme deficiency) leads to multi-system disease. Current therapies for lysosomal diseases are limited in their ability to treat certain tissues that are major contributors to morbidity and mortality, such as the central nervous system (CNS) and cardiac valves. For this study, the lysosomal disease mucopolysaccharidosis type I (MPS I) was selected as the disease model. In MPS I, mutations in the IDUA gene cause a deficiency of the α-L-iduronidase (IDUA) enzyme activity, leading to disease pathology in tissues throughout the body, including the CNS and cardiac valves. Current therapies have been unable to prevent neurodevelopmental deficits and cardiac valvular disease in patients with MPS I. This study aimed to evaluate the delivery of IDUA enzyme, via a novel gene therapy construct, to target tissues. Methods MPS I mice were hydrodynamically injected through the tail vein with plasmids containing either a codon-optimized cDNA encoding the wild-type IDUA protein or one of four modified IDUAs under the control of the liver-specific human α1-antitrypsin (hAAT) promoter. Two modified IDUAs contained a ligand for the CB1 receptor, which is a highly expressed receptor in the CNS. Iduronidase activity levels were measured in the tissues and plasma using an enzyme activity assay. Results The modified IDUAs did not appear to have improved activity levels in the brain compared with the unmodified IDUA. However, one modified IDUA exhibited higher activity levels than the unmodified IDUA in the heart (p = 0.0211). This modified iduronidase (LT-IDUA) contained a sequence for a six amino acid peptide termed LT. LT-IDUA was further characterized using a noncompartmental pharmacokinetic approach that directly analyzed enzyme activity levels after gene delivery. LT-IDUA had a 2-fold higher area under the curve (AUC) than the unmodified IDUA (p = 0.0034) when AUC was estimated using enzyme activity levels in the plasma. Conclusion The addition of a six amino acid peptide improved iduronidase's activity levels in the heart and plasma. The short length of this LT peptide facilitates its use as fusion enzymes encoded as gene therapy or administered as enzyme replacement therapy. More broadly, the LT peptide may aid in developing therapies for numerous lysosomal diseases.
Collapse
Affiliation(s)
- Sarah Kim
- Gene Therapy and Diagnostic Laboratory, Department of Pediatrics, University of Minnesota, Medical School, 516 Delaware St SE, 13th Floor, Rm 13-118 Minneapolis, MN 55455, USA.,Department of Experimental and Clinical Pharmacology, University of Minnesota, College of Pharmacy, 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN 55455, USA
| | - Michael J Przybilla
- Gene Therapy and Diagnostic Laboratory, Department of Pediatrics, University of Minnesota, Medical School, 516 Delaware St SE, 13th Floor, Rm 13-118 Minneapolis, MN 55455, USA
| | - Chester B Whitley
- Gene Therapy and Diagnostic Laboratory, Department of Pediatrics, University of Minnesota, Medical School, 516 Delaware St SE, 13th Floor, Rm 13-118 Minneapolis, MN 55455, USA.,Department of Experimental and Clinical Pharmacology, University of Minnesota, College of Pharmacy, 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN 55455, USA
| | - Li Ou
- Gene Therapy and Diagnostic Laboratory, Department of Pediatrics, University of Minnesota, Medical School, 516 Delaware St SE, 13th Floor, Rm 13-118 Minneapolis, MN 55455, USA
| | - Mahmoud Al-Kofahi
- Department of Experimental and Clinical Pharmacology, University of Minnesota, College of Pharmacy, 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN 55455, USA
| | - Jeanine R Jarnes
- Gene Therapy and Diagnostic Laboratory, Department of Pediatrics, University of Minnesota, Medical School, 516 Delaware St SE, 13th Floor, Rm 13-118 Minneapolis, MN 55455, USA.,Department of Experimental and Clinical Pharmacology, University of Minnesota, College of Pharmacy, 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN 55455, USA
| |
Collapse
|
3
|
Katrekar D, Yen J, Xiang Y, Saha A, Meluzzi D, Savva Y, Mali P. Efficient in vitro and in vivo RNA editing via recruitment of endogenous ADARs using circular guide RNAs. Nat Biotechnol 2022; 40:938-945. [PMID: 35145312 PMCID: PMC9232839 DOI: 10.1038/s41587-021-01171-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Recruiting endogenous adenosine deaminases using exogenous guide RNAs to edit cellular RNAs is a promising therapeutic strategy, but editing efficiency and durability remain low using current guide RNA designs. In this study, we engineered circular ADAR-recruiting guide RNAs (cadRNAs) to enable more efficient programmable adenosine-to-inosine RNA editing without requiring co-delivery of any exogenous proteins. Using these cadRNAs, we observed robust and durable RNA editing across multiple sites and cell lines, in both untranslated and coding regions of RNAs, and high transcriptome-wide specificity. Additionally, we increased transcript-level specificity for the target adenosine by incorporating interspersed loops in the antisense domains, reducing bystander editing. In vivo delivery of cadRNAs via adeno-associated viruses enabled 53% RNA editing of the mPCSK9 transcript in C57BL/6J mice livers and 12% UAG-to-UGG RNA correction of the amber nonsense mutation in the IDUA-W392X mouse model of mucopolysaccharidosis type I-Hurler syndrome. cadRNAs enable efficient programmable RNA editing in vivo with diverse protein modulation and gene therapeutic applications.
Collapse
Affiliation(s)
- Dhruva Katrekar
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - James Yen
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Yichen Xiang
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Anushka Saha
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Dario Meluzzi
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | | | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
4
|
Braunlin E, Abrahante JE, McElmurry R, Evans M, Smith M, Seelig D, O'Sullivan MG, Tolar J, Whitley CB, McIvor RS. Contribution of the innate and adaptive immune systems to aortic dilation in murine mucopolysaccharidosis type I. Mol Genet Metab 2022; 135:193-205. [PMID: 35165009 PMCID: PMC9109621 DOI: 10.1016/j.ymgme.2022.01.104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/23/2021] [Accepted: 01/31/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Adult immunocompetent male C57Bl/6 mucopolysaccharidosis, type I (MPSI) mice develop aortic insufficiency (AI), dilated ascending aortas and decreased cardiac function, findings not observed in immune incompetent adult male NSG MPSI mice. We sought to determine why. METHODS Cardiac ultrasound measurements of ascending aorta and left ventricular dimensions and Doppler interrogation for AI were performed in 6-month-old male B6 MPSI (N = 12), WT (N = 6), NSG MPSI (N = 8), NSG (N = 6) mice. Urinary glycosaminoglycans, RNA sequencing with quantitative PCR were performed and aortic pathology assessed by routine and immunohistochemical staining on subsets of murine aortas. RESULTS Ascending aortic diameters were significantly greater, left ventricular function significantly decreased, and AI significantly more frequent in B6 MPSI mice compared to NSG MPSI mice (p < 0.0001, p = 0.008 and p = 0.02, respectively); NSG and B6 WT mice showed no changes. Urinary glycosaminoglycans were significantly greater in B6 and NSG MPSI mice and both were significantly elevated compared to WT controls (p = 0.003 and p < 0.0001, respectively). By RNA sequencing, all 11 components of the inflammasome pathway were upregulated in B6 MUT, but only Aim2 and Ctsb in NSG MUT mice and none in WT controls. Both B6 and NSG MUT mice demonstrated variably-severe intramural inflammation, vacuolated cells, elastin fragmentation and disarray, and intense glycosaminoglycans on histological staining. B6 MPSI mice demonstrated numerous medial MAC2+ macrophages and adventitial CD3+ T-cells while MAC2+ macrophages were sparse and CD3+ T-cells absent in NSG MPSI mice. CONCLUSIONS Aortic dilation, AI and decreased cardiac function occur in immunocompetent B6 MPSI male mice but not in immune incompetent NSG MPSI mice, unrelated to GAG excretion, upregulation of Ctsb, or routine histologic appearance. Upregulation of all components of the inflammasome pathway in B6 MUT, but not NSG MUT mice, and abundant medial MAC2 and adventitial CD3 infiltrates in B6, but not NSG, MPSI aortas differentiated the two strains. These results suggest that the innate and adaptive immune systems play a role in these cardiac findings which may be relevant to human MPSI.
Collapse
Affiliation(s)
- Elizabeth Braunlin
- Department of Pediatrics University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Juan E Abrahante
- University of Minnesota Informatics Institute University of Minnesota, Minneapolis, MN, USA.
| | - Ron McElmurry
- Department of Pediatrics University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Michael Evans
- Biostatistical Design and Analysis Center Clinical and Translational Science Institute University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Miles Smith
- Department of Genetics, Cell Biology and Development University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Davis Seelig
- Comparative Pathology Shared Resource, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, St. Paul, MN, USA.
| | - M Gerard O'Sullivan
- Comparative Pathology Shared Resource, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, St. Paul, MN, USA.
| | - Jakub Tolar
- Department of Blood and Marrow Transplant University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Chester B Whitley
- Gene Therapy Center Department of Pediatrics University of Minnesota Medical School Minneapolis, MN, USA.
| | - R Scott McIvor
- Department of Genetics, Cell Biology and Development University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Belur LR, Romero M, Lee J, Podetz-Pedersen KM, Nan Z, Riedl MS, Vulchanova L, Kitto KF, Fairbanks CA, Kozarsky KF, Orchard PJ, Frey WH, Low WC, McIvor RS. Comparative Effectiveness of Intracerebroventricular, Intrathecal, and Intranasal Routes of AAV9 Vector Administration for Genetic Therapy of Neurologic Disease in Murine Mucopolysaccharidosis Type I. Front Mol Neurosci 2021; 14:618360. [PMID: 34040503 PMCID: PMC8141728 DOI: 10.3389/fnmol.2021.618360] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/30/2021] [Indexed: 12/02/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is an inherited metabolic disorder caused by deficiency of the lysosomal enzyme alpha-L-iduronidase (IDUA). The two current treatments [hematopoietic stem cell transplantation (HSCT) and enzyme replacement therapy (ERT)], are insufficiently effective in addressing neurologic disease, in part due to the inability of lysosomal enzyme to cross the blood brain barrier. With a goal to more effectively treat neurologic disease, we have investigated the effectiveness of AAV-mediated IDUA gene delivery to the brain using several different routes of administration. Animals were treated by either direct intracerebroventricular (ICV) injection, by intrathecal (IT) infusion into the cerebrospinal fluid, or by intranasal (IN) instillation of AAV9-IDUA vector. AAV9-IDUA was administered to IDUA-deficient mice that were either immunosuppressed with cyclophosphamide (CP), or immunotolerized at birth by weekly injections of human iduronidase. In animals treated by ICV or IT administration, levels of IDUA enzyme ranged from 3- to 1000-fold that of wild type levels in all parts of the microdissected brain. In animals administered vector intranasally, enzyme levels were 100-fold that of wild type in the olfactory bulb, but enzyme expression was close to wild type levels in other parts of the brain. Glycosaminoglycan levels were reduced to normal in ICV and IT treated mice, and in IN treated mice they were normalized in the olfactory bulb, or reduced in other parts of the brain. Immunohistochemical analysis showed extensive IDUA expression in all parts of the brain of ICV treated mice, while IT treated animals showed transduction that was primarily restricted to the hind brain with some sporadic labeling seen in the mid- and fore brain. At 6 months of age, animals were tested for spatial navigation, memory, and neurocognitive function in the Barnes maze; all treated animals were indistinguishable from normal heterozygous control animals, while untreated IDUA deficient animals exhibited significant learning and spatial navigation deficits. We conclude that IT and IN routes are acceptable and alternate routes of administration, respectively, of AAV vector delivery to the brain with effective IDUA expression, while all three routes of administration prevent the emergence of neurocognitive deficiency in a mouse MPS I model.
Collapse
Affiliation(s)
- Lalitha R. Belur
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Megan Romero
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Junggu Lee
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Kelly M. Podetz-Pedersen
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Zhenhong Nan
- Department of Neurosurgery and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Maureen S. Riedl
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Kelley F. Kitto
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, United States
| | - Carolyn A. Fairbanks
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, United States
| | | | - Paul J. Orchard
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - William H. Frey
- HealthPartners Neurosciences, Regions Hospital, St. Paul, MN, United States
| | - Walter C. Low
- Department of Neurosurgery and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - R. Scott McIvor
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Derrick-Roberts A, Kaidonis X, Jackson MR, Liaw WC, Ding X, Ong C, Ranieri E, Sharp P, Fletcher J, Byers S. Comparative analysis of brain pathology in heparan sulphate storing mucopolysaccharidoses. Mol Genet Metab 2020; 131:197-205. [PMID: 32739280 DOI: 10.1016/j.ymgme.2020.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/22/2020] [Accepted: 07/19/2020] [Indexed: 10/23/2022]
Abstract
The cause of neurodegeneration in MPS mouse models is the focus of much debate and what the underlying cause of disease pathology in MPS mice is. The timing of development of pathology and when this can be reversed or impacted is the key to developing suitable therapies in MPS. This study is the first of its kind to correlate the biochemical changes with the functional outcome as assessed using non-invasive behaviour testing across multiple mucopolysaccharidosis (MPS) mouse models. In the MPS brain, the primary lysosomal enzyme dysfunction leads to accumulation of primary glycosaminoglycans (GAGs) with gangliosides (GM2 and GM3) being the major secondary storage products. With a focus on the neuropathology, a time course experiment was conducted in MPS I, MPS IIIA, MPS VII (severe and attenuated models) in order to understand the relative timing and level of GAG and ganglioside accumulation and how this correlates to behaviour deficits. Time course analysis from 1 to 6 months of age was conducted on brain samples to assess primary GAG (uronic acid), β-hexosaminidase enzyme activity and levels of GM2 and GM3 gangliosides. This was compared to a battery of non-invasive behaviour tests including open field, inverted grid, rotarod and water cross maze were assessed to determine effects on motor function, activity and learning ability. The results show that the GAG and ganglioside accumulation begins prior to the onset of detectable changes in learning ability and behaviour. Interestingly, the highest levels of GAG and ganglioside accumulation was observed in the MPS IIIA mouse despite having 3% residual enzyme activity. Deficits in motor function were clearly observed in the severe Gusmps/mps, which were significantly delayed in the attenuated Gustm(L175F)Sly model despite their minimal increase in detectable enzyme activity. This suggests that genotype and residual enzyme activity are not indicative of severity of disease pathology in MPS disease and there exists a window when there are considerable storage products without detectable functional deficits which may allow an alteration to occur with therapy.
Collapse
Affiliation(s)
- Ainslie Derrick-Roberts
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia; Discipline of Paediatrics, The University of Adelaide, Adelaide, Australia.
| | - Xenia Kaidonis
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia; Discipline of Genetics and Evolution, University of Adelaide, Australia
| | - Matilda R Jackson
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia; Discipline of Genetics and Evolution, University of Adelaide, Australia
| | - Wan Chin Liaw
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia
| | - XiaoDan Ding
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia
| | - Chun Ong
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia
| | - Enzo Ranieri
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia
| | - Peter Sharp
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia
| | - Janice Fletcher
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia
| | - Sharon Byers
- Genetics and Molecular Pathology, SA Pathology (WCH site), Adelaide, Australia; Discipline of Paediatrics, The University of Adelaide, Adelaide, Australia; Discipline of Genetics and Evolution, University of Adelaide, Australia
| |
Collapse
|
7
|
Hampe CS, Eisengart JB, Lund TC, Orchard PJ, Swietlicka M, Wesley J, McIvor RS. Mucopolysaccharidosis Type I: A Review of the Natural History and Molecular Pathology. Cells 2020; 9:cells9081838. [PMID: 32764324 PMCID: PMC7463646 DOI: 10.3390/cells9081838] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/14/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is a rare autosomal recessive inherited disease, caused by deficiency of the enzyme α-L-iduronidase, resulting in accumulation of the glycosaminoglycans (GAGs) dermatan and heparan sulfate in organs and tissues. If untreated, patients with the severe phenotype die within the first decade of life. Early diagnosis is crucial to prevent the development of fatal disease manifestations, prominently cardiac and respiratory disease, as well as cognitive impairment. However, the initial symptoms are nonspecific and impede early diagnosis. This review discusses common phenotypic manifestations in the order in which they develop. Similarities and differences in the three animal models for MPS I are highlighted. Earliest symptoms, which present during the first 6 months of life, include hernias, coarse facial features, recurrent rhinitis and/or upper airway obstructions in the absence of infection, and thoracolumbar kyphosis. During the next 6 months, loss of hearing, corneal clouding, and further musculoskeletal dysplasias develop. Finally, late manifestations including lower airway obstructions and cognitive decline emerge. Cardiac symptoms are common in MPS I and can develop in infancy. The underlying pathogenesis is in the intra- and extracellular accumulation of partially degraded GAGs and infiltration of cells with enlarged lysosomes causing tissue expansion and bone deformities. These interfere with the proper arrangement of collagen fibrils, disrupt nerve fibers, and cause devastating secondary pathophysiological cascades including inflammation, oxidative stress, and other disruptions to intracellular and extracellular homeostasis. A greater understanding of the natural history of MPS I will allow early diagnosis and timely management of the disease facilitating better treatment outcomes.
Collapse
Affiliation(s)
- Christiane S. Hampe
- Immusoft Corp, Seattle, WA 98103, USA; (M.S.); (J.W.)
- Correspondence: ; Tel.: +1-206-554-9181
| | - Julie B. Eisengart
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; (J.B.E.); (T.C.L.); (P.J.O.)
| | - Troy C. Lund
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; (J.B.E.); (T.C.L.); (P.J.O.)
| | - Paul J. Orchard
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; (J.B.E.); (T.C.L.); (P.J.O.)
| | | | - Jacob Wesley
- Immusoft Corp, Seattle, WA 98103, USA; (M.S.); (J.W.)
| | - R. Scott McIvor
- Immusoft Corp, Minneapolis, MN 55413, USA; or
- Department of Genetics, Cell Biology and Development and Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55413, USA
| |
Collapse
|
8
|
Induced Pluripotent Stem Cell Derivation and Ex Vivo Gene Correction Using a Mucopolysaccharidosis Type 1 Disease Mouse Model. Stem Cells Int 2019; 2019:6978303. [PMID: 31065277 PMCID: PMC6466856 DOI: 10.1155/2019/6978303] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/18/2018] [Accepted: 01/06/2019] [Indexed: 12/17/2022] Open
Abstract
Mucopolysaccharidosis type 1 (MPS-1), also known as Hurler's disease, is a congenital metabolic disorder caused by a mutation in the alpha-L-iduronidase (IDUA) gene, which results in the loss of lysosomal enzyme function for the degradation of glycosaminoglycans. Here, we demonstrate the proof of concept of ex vivo gene editing therapy using induced pluripotent stem cell (iPSC) and CRISPR/Cas9 technologies with MPS-1 model mouse cell. Disease-affected iPSCs were generated from Idua knockout mouse embryonic fibroblasts, which carry a disrupting neomycin-resistant gene cassette (Neor) in exon VI of the Idua gene. Double guide RNAs were used to remove the Neor sequence, and various lengths of donor templates were used to reconstruct the exon VI sequence. A quantitative PCR-based screening method was used to identify Neor removal. The sequence restoration without any indel mutation was further confirmed by Sanger sequencing. After induced fibroblast differentiation, the gene-corrected iPSC-derived fibroblasts demonstrated Idua function equivalent to the wild-type iPSC-derived fibroblasts. The Idua-deficient cells were competent to be reprogrammed to iPSCs, and pluripotency was maintained through CRISPR/CAS9-mediated gene correction. These results support the concept of ex vivo gene editing therapy using iPSC and CRISPR/Cas9 technologies for MPS-1 patients.
Collapse
|
9
|
Azario I, Pievani A, Del Priore F, Antolini L, Santi L, Corsi A, Cardinale L, Sawamoto K, Kubaski F, Gentner B, Bernardo ME, Valsecchi MG, Riminucci M, Tomatsu S, Aiuti A, Biondi A, Serafini M. Neonatal umbilical cord blood transplantation halts skeletal disease progression in the murine model of MPS-I. Sci Rep 2017; 7:9473. [PMID: 28842642 PMCID: PMC5573317 DOI: 10.1038/s41598-017-09958-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/01/2017] [Indexed: 12/23/2022] Open
Abstract
Umbilical cord blood (UCB) is a promising source of stem cells to use in early haematopoietic stem cell transplantation (HSCT) approaches for several genetic diseases that can be diagnosed at birth. Mucopolysaccharidosis type I (MPS-I) is a progressive multi-system disorder caused by deficiency of lysosomal enzyme α-L-iduronidase, and patients treated with allogeneic HSCT at the onset have improved outcome, suggesting to administer such therapy as early as possible. Given that the best characterized MPS-I murine model is an immunocompetent mouse, we here developed a transplantation system based on murine UCB. With the final aim of testing the therapeutic efficacy of UCB in MPS-I mice transplanted at birth, we first defined the features of murine UCB cells and demonstrated that they are capable of multi-lineage haematopoietic repopulation of myeloablated adult mice similarly to bone marrow cells. We then assessed the effectiveness of murine UCB cells transplantation in busulfan-conditioned newborn MPS-I mice. Twenty weeks after treatment, iduronidase activity was increased in visceral organs of MPS-I animals, glycosaminoglycans storage was reduced, and skeletal phenotype was ameliorated. This study explores a potential therapy for MPS-I at a very early stage in life and represents a novel model to test UCB-based transplantation approaches for various diseases.
Collapse
Affiliation(s)
- Isabella Azario
- Dulbecco Telethon Institute, Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, 20900, Italy
| | - Alice Pievani
- Dulbecco Telethon Institute, Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, 20900, Italy
| | - Federica Del Priore
- Dulbecco Telethon Institute, Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, 20900, Italy
| | - Laura Antolini
- Centro di Biostatistica per l'epidemiologia clinica, Department of Health Sciences, University of Milano-Bicocca, Monza, 20900, Italy
| | - Ludovica Santi
- Dulbecco Telethon Institute, Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, 20900, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University, Rome, 00161, Italy
| | - Lucia Cardinale
- Dulbecco Telethon Institute, Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, 20900, Italy
| | - Kazuki Sawamoto
- Department of Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE, 19803, USA
| | - Francyne Kubaski
- Department of Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE, 19803, USA.,Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Maria Grazia Valsecchi
- Centro di Biostatistica per l'epidemiologia clinica, Department of Health Sciences, University of Milano-Bicocca, Monza, 20900, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University, Rome, 00161, Italy
| | - Shunji Tomatsu
- Department of Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE, 19803, USA
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), San Raffaele Scientific Institute, Milan, 20132, Italy.,Vita Salute San Raffaele University, Milan, 20132, Italy
| | - Andrea Biondi
- Department of Pediatrics, University of Milano-Bicocca, Monza, 20900, Italy
| | - Marta Serafini
- Dulbecco Telethon Institute, Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, 20900, Italy.
| |
Collapse
|
10
|
Rodriguez NS, Yanuaria L, Parducho KMR, Garcia IM, Varghese BA, Grubbs BH, Miki T. Liver-Directed Human Amniotic Epithelial Cell Transplantation Improves Systemic Disease Phenotype in Hurler Syndrome Mouse Model. Stem Cells Transl Med 2017; 6:1583-1594. [PMID: 28585336 PMCID: PMC5689764 DOI: 10.1002/sctm.16-0449] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/13/2017] [Accepted: 04/04/2017] [Indexed: 12/18/2022] Open
Abstract
Mucopolysaccharidosis type 1 (MPS1) is an inherited lysosomal storage disorder caused by a deficiency in the glycosaminoglycan (GAG)‐degrading enzyme α‐l‐iduronidase (IDUA). In affected patients, the systemic accumulation of GAGs results in skeletal dysplasia, neurological degeneration, multiple organ dysfunction, and early death. Current therapies, including enzyme replacement and bone marrow transplant, improve life expectancy but the benefits to skeletal and neurological phenotypes are limited. In this study, we tested the therapeutic efficacy of liver‐directed transplantation of a placental stem cell, which possesses multilineage differentiation potential, low immunogenicity, and high lysosomal enzyme activity. Unfractionated human amniotic epithelial cells (hAECs) were transplanted directly into the liver of immunodeficient Idua knockout mouse neonates. The hAECs engraftment was immunohistochemically confirmed with anti‐human mitochondria staining. Enzyme activity assays indicated that hAECs transplantation restored IDUA function in the liver and significantly decreased urinary GAG excretion. Histochemical and micro‐computed tomography analyses revealed reduced GAG deposition in the phalanges joints and composition/morphology improvement of cranial and facial bones. Neurological assessment in the hAEC treated mice showed significant improvement of sensorimotor coordination in the hAEC treated mice compared to untreated mice. Results confirm that partial liver cell replacement with placental stem cells can provide long‐term (>20 weeks) and systemic restoration of enzyme function, and lead to significant phenotypic improvement in the MPS1 mouse model. This preclinical data indicate that liver‐directed placental stem cell transplantation may improve skeletal and neurological phenotypes of MPS1 patients. Stem Cells Translational Medicine2017;6:1583–1594
Collapse
Affiliation(s)
| | - Lisa Yanuaria
- Department of SurgeryBiochemistry & Molecular Biology
| | | | | | | | - Brendan H. Grubbs
- Department of Obstetrics and GynecologyKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Toshio Miki
- Department of SurgeryBiochemistry & Molecular Biology
| |
Collapse
|
11
|
Substrate Deprivation Therapy to Reduce Glycosaminoglycan Synthesis Improves Aspects of Neurological and Skeletal Pathology in MPS I Mice. Diseases 2017; 5:diseases5010005. [PMID: 28933358 PMCID: PMC5456338 DOI: 10.3390/diseases5010005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 12/17/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is the most common form of the MPS group of genetic diseases. MPS I results from a deficiency in the lysosomal enzyme α-l-iduronidase, leading to accumulation of undegraded heparan and dermatan sulphate glycosaminoglycan (GAG) chains in patient cells. MPS children suffer from multiple organ failure and die in their teens to early twenties. In particular, MPS I children also suffer from profound mental retardation and skeletal disease that restricts growth and movement. Neither brain nor skeletal disease is adequately treated by current therapy approaches. To overcome these barriers to effective therapy we have developed and tested a treatment called substrate deprivation therapy (SDT). MPS I knockout mice were treated with weekly intravenous injections of 1 mg/kg rhodamine B for six months to assess the efficacy of SDT. Mice were assessed using biochemistry, micro-CT and a battery of behaviour tests to determine the outcome of treatment. A reduction in female bodyweight gain was observed with the treatment as well as a decrease in lung GAG. Behavioural studies showed slight improvements in inverted grid and significant improvements in learning ability for female MPS I mice treated with rhodamine B. Skeletal disease also improved with a reduction in bone mineral volume observed. Overall, rhodamine B is safe to administer to MPS I knockout mice where it had an effect on improving aspects of neurological and skeletal disease symptoms and may therefore provide a potential therapy or adjunct therapy for MPS I patients.
Collapse
|
12
|
Vance M, Llanga T, Bennett W, Woodard K, Murlidharan G, Chungfat N, Asokan A, Gilger B, Kurtzberg J, Samulski RJ, Hirsch ML. AAV Gene Therapy for MPS1-associated Corneal Blindness. Sci Rep 2016; 6:22131. [PMID: 26899286 PMCID: PMC4761992 DOI: 10.1038/srep22131] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/01/2016] [Indexed: 11/25/2022] Open
Abstract
Although cord blood transplantation has significantly extended the lifespan of mucopolysaccharidosis type 1 (MPS1) patients, over 95% manifest cornea clouding with about 50% progressing to blindness. As corneal transplants are met with high rejection rates in MPS1 children, there remains no treatment to prevent blindness or restore vision in MPS1 children. Since MPS1 is caused by mutations in idua, which encodes alpha-L-iduronidase, a gene addition strategy to prevent, and potentially reverse, MPS1-associated corneal blindness was investigated. Initially, a codon optimized idua cDNA expression cassette (opt-IDUA) was validated for IDUA production and function following adeno-associated virus (AAV) vector transduction of MPS1 patient fibroblasts. Then, an AAV serotype evaluation in human cornea explants identified an AAV8 and 9 chimeric capsid (8G9) as most efficient for transduction. AAV8G9-opt-IDUA administered to human corneas via intrastromal injection demonstrated widespread transduction, which included cells that naturally produce IDUA, and resulted in a >10-fold supraphysiological increase in IDUA activity. No significant apoptosis related to AAV vectors or IDUA was observed under any conditions in both human corneas and MPS1 patient fibroblasts. The collective preclinical data demonstrate safe and efficient IDUA delivery to human corneas, which may prevent and potentially reverse MPS1-associated cornea blindness.
Collapse
Affiliation(s)
- Melisa Vance
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Telmo Llanga
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA.,Department of Ophthalmology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Will Bennett
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA.,Department of Ophthalmology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kenton Woodard
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Giridhar Murlidharan
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Neil Chungfat
- Department of Ophthalmology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Aravind Asokan
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Brian Gilger
- College of Veterinary Medicine, NCSU-CVM, Clinical Sciences, Raleigh, NC, USA
| | - Joanne Kurtzberg
- Department of Pediatrics, Duke University, Durham, NC, 27710, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA.,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Matthew L Hirsch
- Gene Therapy Center, University of North Carolina at Chapel Hill, NC, 27599, USA.,Department of Ophthalmology, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
13
|
Kim C, Kwak MJ, Cho SY, Ko AR, Rheey J, Kwon JY, Chung Y, Jin DK. Decreased performance in IDUA knockout mouse mimic limitations of joint function and locomotion in patients with Hurler syndrome. Orphanet J Rare Dis 2015; 10:121. [PMID: 26407983 PMCID: PMC4582722 DOI: 10.1186/s13023-015-0337-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/06/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mucopolysaccharidosis type I (MPS I) is caused by the deficiency of alpha-L-iduronidase (IDUA), which is involved in the degradation of glycosaminoglycans (GAGs), such as heparan sulfate and dermatan sulfate in the lysosome. It has been reported that joint symptoms are almost universal in MPS I patients, and even in the case of attenuated disease, they are the first symptom that brings a child to medical attention. However, functional tests and biological markers have not been published for the evaluation of the limitations in joint and locomotion in animal model-mimicking MPS. METHODS We generated IDUA knockout (KO) mice to observe whether they present impairment of joint function. KO mice were characterized phenotypically and tested dual-energy X-ray absorptiometry analysis (DEXA), open-field, rotarod, and grip strength. RESULTS The IDUA KO mice, generated by disruption between exon 6 and exon 9, exhibited clinical and laboratory findings, such as high urinary GAGs excretion, GAGs accumulation in various tissues, and significantly increased bone mineral density (BMD) in both female and male mice in the DEXA of the femur and whole bone. Remarkably, we observed a decrease in grasp function, decreased performance in the rotarod test, and hypo-activity in the open-field test, which mimic the limitations of joint mobility and decreased motor performance in the 6-min walk test in patients with MPS I. CONCLUSIONS We generated a new IDUA KO mouse, tested open field, rotarod and grip strength and demonstrated decrease in grip strength, decreased performance and hypo-activity, which may be useful for investigating therapeutic approaches, and studying the pathogenesis of joint and locomotion symptoms in MPS I.
Collapse
Affiliation(s)
- Chihwa Kim
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea.,Present Address: MOGAM Biotechnology Institute, Yongin, Republic of Korea
| | - Min Jung Kwak
- Department of Pediatrics, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Sung Yoon Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Ah-Ra Ko
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Jinguen Rheey
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology, Seoul, Republic of Korea
| | - Jeong-Yi Kwon
- Department of Physical and Rehabilitation Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yokyung Chung
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Dong-Kyu Jin
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea.
| |
Collapse
|
14
|
Satzer D, DiBartolomeo C, Ritchie MM, Storino C, Liimatainen T, Hakkarainen H, Idiyatullin D, Mangia S, Michaeli S, Parr AM, Low WC. Assessment of dysmyelination with RAFFn MRI: application to murine MPS I. PLoS One 2015; 10:e0116788. [PMID: 25680196 PMCID: PMC4334512 DOI: 10.1371/journal.pone.0116788] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/11/2014] [Indexed: 12/17/2022] Open
Abstract
Type I mucopolysaccharidosis (MPS I) is an autosomal recessive lysosomal storage disorder with neurological features. Humans and laboratory animals with MPS I exhibit various white matter abnormalities involving the corpus callosum and other regions. In this study, we first validated a novel MRI technique, entitled Relaxation Along a Fictitious Field in the rotating frame of rank n (RAFFn), as a measure of myelination and dysmyelination in mice. We then examined differences between MPS I mice and heterozygotes using RAFF5 and histology. RAFF5 (i.e., RAFFn with n = 5) relaxation time constants were highly correlated with histological myelin density (R2 = 0.68, P<0.001), and RAFF5 clearly distinguished between the hypomyelinated and dysmyelinated shiverer mouse and the wild-type mouse. Bloch-McConnell theoretical analysis revealed slower exchange correlation times and smaller exchange-induced relaxation rate constants for RAFF4 and RAFF5 compared to RAFF1-3, T1ρ, and T2ρ. These data suggest that RAFF5 may assess methylene protons in myelin lipids and proteins, though other mechanisms (e.g. detection of myelin-bound water) may also explain the sensitivity of RAFF5 to myelin. In MPS I mice, mean RAFF5 relaxation time constants were significantly larger for the striatum (P = 0.004) and internal capsule (P = 0.039), and marginally larger for the fornix (P = 0.15). Histological assessment revealed no differences between MPS I mice and heterozygotes in myelin density or corpus callosum thickness. Taken together, these findings support subtle dysmyelination in the brains of mice with MPS I. Dysmyelination may result from myelin lipid abnormalities caused by the absence of α-L-iduronidase. Our findings may help to explain locomotor and cognitive deficits seen in mice with MPS I.
Collapse
Affiliation(s)
- David Satzer
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Christina DiBartolomeo
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Michael M. Ritchie
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Christine Storino
- Center of Magnetic Resonance Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Timo Liimatainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hanne Hakkarainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Djaudat Idiyatullin
- Center of Magnetic Resonance Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Silvia Mangia
- Center of Magnetic Resonance Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Shalom Michaeli
- Center of Magnetic Resonance Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ann M. Parr
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
15
|
Mendez DC, Stover AE, Rangel AD, Brick DJ, Nethercott HE, Torres MA, Khalid O, Wong AM, Cooper JD, Jester JV, Monuki ES, McGuire C, Le SQ, Kan SH, Dickson PI, Schwartz PH. A novel, long-lived, and highly engraftable immunodeficient mouse model of mucopolysaccharidosis type I. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:14068. [PMID: 26052536 PMCID: PMC4449030 DOI: 10.1038/mtm.2014.68] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/04/2014] [Accepted: 12/14/2014] [Indexed: 02/06/2023]
Abstract
Mucopolysaccharidosis type I (MPS I) is an inherited α-L-iduronidase (IDUA, I) deficiency in which glycosaminoglycan (GAG) accumulation causes progressive multisystem organ dysfunction, neurological impairment, and death. Current MPS I mouse models, based on a NOD/SCID (NS) background, are short-lived, providing a very narrow window to assess the long-term efficacy of therapeutic interventions. They also develop thymic lymphomas, making the assessment of potential tumorigenicity of human stem cell transplantation problematic. We therefore developed a new MPS I model based on a NOD/SCID/Il2rγ (NSG) background. This model lives longer than 1 year and is tumor-free during that time. NSG MPS I (NSGI) mice exhibit the typical phenotypic features of MPS I including coarsened fur and facial features, reduced/abnormal gait, kyphosis, and corneal clouding. IDUA is undetectable in all tissues examined while GAG levels are dramatically higher in most tissues. NSGI brain shows a significant inflammatory response and prominent gliosis. Neurological MPS I manifestations are evidenced by impaired performance in behavioral tests. Human neural and hematopoietic stem cells were found to readily engraft, with human cells detectable for at least 1 year posttransplantation. This new MPS I model is thus suitable for preclinical testing of novel pluripotent stem cell-based therapy approaches.
Collapse
Affiliation(s)
- Daniel C Mendez
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| | - Alexander E Stover
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| | - Anthony D Rangel
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| | - David J Brick
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| | - Hubert E Nethercott
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| | - Marissa A Torres
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| | - Omar Khalid
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| | - Andrew Ms Wong
- King's College, London, Institute of Psychiatry, Psychology & Neuroscience , London, UK
| | - Jonathan D Cooper
- King's College, London, Institute of Psychiatry, Psychology & Neuroscience , London, UK
| | - James V Jester
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California, USA ; Department of Biomedical Engineering, Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California, USA
| | - Edwin S Monuki
- Department of Pathology and Laboratory Medicine, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, California, USA ; Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine , Irvine, California, USA
| | - Cian McGuire
- Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center , Torrance, California, USA
| | - Steven Q Le
- Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center , Torrance, California, USA
| | - Shih-Hsin Kan
- Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center , Torrance, California, USA
| | - Patricia I Dickson
- Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center , Torrance, California, USA
| | - Philip H Schwartz
- National Human Neural Stem Cell Resource, Centers for Neuroscience and Translational Research, CHOC Children's Research Institute , Orange, California, USA
| |
Collapse
|
16
|
Aronovich EL, Hackett PB. Lysosomal storage disease: gene therapy on both sides of the blood-brain barrier. Mol Genet Metab 2015; 114:83-93. [PMID: 25410058 PMCID: PMC4312729 DOI: 10.1016/j.ymgme.2014.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 12/17/2022]
Abstract
Most lysosomal storage disorders affect the nervous system as well as other tissues and organs of the body. Previously, the complexities of these diseases, particularly in treating neurologic abnormalities, were too great to surmount. However, based on recent developments there are realistic expectations that effective therapies are coming soon. Gene therapy offers the possibility of affordable, comprehensive treatment associated with these diseases currently not provided by standards of care. With a focus on correction of neurologic disease by systemic gene therapy of mucopolysaccharidoses types I and IIIA, we review some of the major recent advances in viral and non-viral vectors, methods of their delivery and strategies leading to correction of both the nervous and somatic tissues as well as evaluation of functional correction of neurologic manifestations in animal models. We discuss two questions: what systemic gene therapy strategies work best for correction of both somatic and neurologic abnormalities in a lysosomal storage disorder and is there evidence that targeting peripheral tissues (e.g., in the liver) has a future for ameliorating neurologic disease in patients?
Collapse
Affiliation(s)
- Elena L Aronovich
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, United States.
| | - Perry B Hackett
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, United States
| |
Collapse
|
17
|
Janson CG, Romanova LG, Leone P, Nan Z, Belur L, McIvor RS, Low WC. Comparison of Endovascular and Intraventricular Gene Therapy With Adeno-Associated Virus-α-L-Iduronidase for Hurler Disease. Neurosurgery 2014; 74:99-111. [PMID: 24077583 DOI: 10.1227/neu.0000000000000157] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Hurler disease (mucopolysaccharidosis type I [MPS-I]) is an inherited metabolic disorder characterized by deficiency of the lysosomal enzyme α-L-iduronidase (IDUA). Currently, the only therapies for MPS-I, enzyme replacement and hematopoietic stem cell transplantation, are generally ineffective for central nervous system manifestations. OBJECTIVE To test whether brain-targeted gene therapy with recombinant adeno-associated virus (rAAV5)-IDUA vectors in an MPS-I transgenic mouse model would reverse the pathological hallmarks. METHODS Gene therapy approaches were compared using intraventricular or endovascular delivery with a marker (rAAV5-green fluorescent protein) or therapeutic (rAAV5-IDUA) vector. To improve the efficiency of brain delivery, we tested different applications of hyperosmolar mannitol to disrupt the blood-brain barrier or ependymal-brain interface. RESULTS Intraventricular delivery of 1 × 10 viral particles of rAAV5-IDUA with systemic 5 g/kg mannitol co-administration resulted in IDUA expression throughout the brain, with global enzyme activity >200% of the baseline level in age-matched, wild-type mice. Endovascular delivery of 1 × 10 viral particles of rAAV5-IDUA to the carotid artery with 29.1% mannitol blood-brain barrier disruption resulted in mainly ipsilateral brain IDUA expression and ipsilateral brain enzyme activity 42% of that in wild-type mice. Quantitative assays for glycosaminoglycans showed a significant decrease in both hemispheres after intraventricular delivery and in the ipsilateral hemisphere after endovascular delivery compared with untreated MPS-I mice. Immunohistochemistry for ganglioside GM3, another disease marker, showed reversal of neuronal inclusions in areas with IDUA co-expression in both delivery methods. CONCLUSION Physiologically relevant biochemical correction is possible with neurosurgical or endovascular gene therapy approaches for MPS-I. Intraventricular or endovascular delivery of rAAV5-IDUA was effective in reversing brain pathology, but in the latter method, effects were limited to the ipsilateral hemisphere.
Collapse
Affiliation(s)
- Christopher G Janson
- *Department of Neurosurgery, ‡Department of Neurology, ¶Department of Medicine, and ‖Genetics and Cell Biology, University of Minnesota, School of Medicine §Cell & Gene Therapy Center, University of Medicine and Dentistry of New Jersey School of Medicine
| | | | | | | | | | | | | |
Collapse
|
18
|
Ou L, Herzog TL, Wilmot CM, Whitley CB. Standardization of α-L-iduronidase enzyme assay with Michaelis-Menten kinetics. Mol Genet Metab 2014; 111:113-5. [PMID: 24332804 PMCID: PMC4014300 DOI: 10.1016/j.ymgme.2013.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 11/11/2013] [Accepted: 11/11/2013] [Indexed: 11/30/2022]
Abstract
The lack of methodological uniformity in enzyme assays has been a long-standing difficulty, a problem for bench researchers, for the interpretation of clinical diagnostic tests, and an issue for investigational drug review. Illustrative of the problem, α-L-iduronidase enzyme catalytic activity is frequently measured with the substrate 4-methylumbelliferyl-α-L-iduronide (4MU-iduronide); however, final substrate concentrations used in different assays vary greatly, ranging from 25 μM to 1425 μM (Km ≈ 180 μM) making it difficult to compare results between laboratories. In this study, α-L-iduronidase was assayed with 15 different substrate concentrations. The resulting activity levels from the same specimens varied greatly with different substrate concentrations but, as a group, obeyed the expectations of Michaelis-Menten kinetics. Therefore, for the sake of improved comparability, it is proposed that α-L-iduronidase enzyme assays should be conducted either (1) under substrate saturating conditions; or (2) when concentrations are significantly below substrate saturation, with results standardized by arithmetic adjustment that considers Michaelis-Menten kinetics. The approach can be generalized to many other enzyme assays.
Collapse
Affiliation(s)
- Li Ou
- Department of Genetics, Cell Biology and Development, PWB 13-146, 516 Delaware Str SE, Minneapolis, MN 55455, USA; Molecular, Cellular, Developmental Biology & Genetics Graduate Program, PWB 13-146, 516 Delaware Str SE, Minneapolis, MN 55455, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA
| | - Tyler L Herzog
- Department of Biochemistry, Molecular Biology and Biophysics, 5-120 NHH, 312 Church Str SE, Minneapolis, MN 55455, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA
| | - Carrie M Wilmot
- Department of Biochemistry, Molecular Biology and Biophysics, 5-120 NHH, 312 Church Str SE, Minneapolis, MN 55455, USA
| | - Chester B Whitley
- Molecular, Cellular, Developmental Biology & Genetics Graduate Program, PWB 13-146, 516 Delaware Str SE, Minneapolis, MN 55455, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA.
| |
Collapse
|
19
|
Ou L, Herzog T, Koniar BL, Gunther R, Whitley CB. High-dose enzyme replacement therapy in murine Hurler syndrome. Mol Genet Metab 2014; 111:116-22. [PMID: 24100243 PMCID: PMC4014311 DOI: 10.1016/j.ymgme.2013.09.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 11/21/2022]
Abstract
Mucopolysaccharidosis type I (MPS I) is an autosomal recessive disease that is systemic, including progressive neurodegeneration, mental retardation and death before the age of 10 years. MPS I results from deficiency of α-L-iduronidase (IDUA) in lysosomes and subsequent accumulation of glycosaminoglycans (GAG). Clinical enzyme replacement therapy (ERT) with intravenous laronidase reverses some aspects of MPS I disease (e.g., hepatomegaly, splenomegaly, glycosaminoglycanuria) and ameliorates others (e.g., pulmonary function, cardiac disease, arthropathy, exercise tolerance). However, neurologic benefits are thought to be negligible because the blood-brain barrier (BBB) blocks enzyme from reaching the central nervous system (CNS). We considered the possibility that a very high dose of intravenous laronidase might be able to traverse the BBB in small quantities, and provide some metabolic correction in the brain. To address this question, high-dose laronidase was administered (11.6 mg/kg, once per week, 4 weeks) to adult MPS I mice. IDUA enzyme activity in the cortex of treated mice increased to 97% of that in wild type mice (p<0.01). GAG levels in cortex were reduced by 63% of that from untreated MPS I mice (p<0.05). Further, immunohistochemical analysis showed that treatment reduced secondary GM3-ganglioside accumulation in treated MPS I mice. Water T-maze tests showed that the learning abnormality in MPS I mice was reduced (p<0.0001). In summary, repeated, high-dose ERT facilitated laronidase transit across the BBB, reduced GAG accumulation within the CNS, and rescued cognitive impairment.
Collapse
Affiliation(s)
- Li Ou
- Department of Genetics, Cell Biology and Development, University of Minnesota, USA; Molecular, Cellular, Developmental Biology & Genetics Graduate Program, University of Minnesota, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA
| | - Tyler Herzog
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA
| | - Brenda L Koniar
- Research Animal Resources, University of Minnesota, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA
| | - Roland Gunther
- Research Animal Resources, University of Minnesota, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA
| | - Chester B Whitley
- Molecular, Cellular, Developmental Biology & Genetics Graduate Program, University of Minnesota, USA; Gene Therapy Center, Department of Pediatrics, University of Minnesota, USA.
| |
Collapse
|
20
|
Aronovich EL, Hall BC, Bell JB, McIvor RS, Hackett PB. Quantitative analysis of α-L-iduronidase expression in immunocompetent mice treated with the Sleeping Beauty transposon system. PLoS One 2013; 8:e78161. [PMID: 24205141 PMCID: PMC3804460 DOI: 10.1371/journal.pone.0078161] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/16/2013] [Indexed: 12/23/2022] Open
Abstract
The Sleeping Beauty transposon system, a non-viral, integrating vector that can deliver the alpha-L-iduronidase-encoding gene, is efficient in correcting mucopolysaccharidosis type I in NOD/SCID mice. However, in previous studies we failed to attain reliable long-term alpha-L-iduronidase expression in immunocompetent mice. Here, we focused on achieving sustained high-level expression in immunocompetent C57BL/6 mice. In our standard liver-directed treatment we hydrodynamically infuse mice with plasmids containing a SB transposon-encoding human alpha-L-iduronidase, along with a source of SB transposase. We sought to 1) minimize expression of the therapeutic enzyme in antigen-presenting cells, while avoiding promoter shutdown and gender bias, 2) increase transposition efficiency and 3) improve immunosuppression. By using a liver-specific promoter to drive IDUA expression, the SB100X hyperactive transposase and transient cyclophosphamide immunosuppression we achieved therapeutic-level (>100 wild-type) stabilized expression for 1 year in 50% of C57BL/6 mice. To gain insights into the causes of variability in transgene expression, we quantified the rates of alpha-L-iduronidase activity decay vis-a-vis transposition and transgene maintenance using the data obtained in this and previous studies. Our analyses showed that immune responses are the most important variable to control in order to prevent loss of transgene expression. Cumulatively, our results allow transition to pre-clinical studies of SB-mediated alpha-L-iduronidase expression and correction of mucopolysaccharidosis type I in animal models.
Collapse
Affiliation(s)
- Elena L. Aronovich
- Department of Genetics, Cell Biology and Development and the Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| | - Bryan C. Hall
- Department of Genetics, Cell Biology and Development and the Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jason B. Bell
- Department of Genetics, Cell Biology and Development and the Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - R. Scott McIvor
- Department of Genetics, Cell Biology and Development and the Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Perry B. Hackett
- Department of Genetics, Cell Biology and Development and the Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
21
|
Nan Z, Shekels L, Ryabinin O, Evavold C, Nelson MS, Khan SA, Deans RJ, Mays RW, Low WC, Gupta P. Intracerebroventricular transplantation of human bone marrow-derived multipotent progenitor cells in an immunodeficient mouse model of mucopolysaccharidosis type I (MPS-I). Cell Transplant 2013; 21:1577-93. [PMID: 22472595 DOI: 10.3727/096368912x636894] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS-I; Hurler syndrome) is an inborn error of metabolism caused by lack of the functional lysosomal glycosaminoglycan (GAG)-degrading enzyme α-L-iduronidase (IDUA). Without treatment, the resulting GAG accumulation causes multisystem dysfunction and death within the first decade. Current treatments include allogeneic hematopoietic stem cell transplantation (HSCT) and enzyme replacement therapy. HSCT ameliorates clinical features and extends life but is not available to all patients, and inadequately corrects the most devastating features of the disease including mental retardation and skeletal deformities. Recent developments suggest that stem cells can be used to deliver needed enzymes to the central nervous system. To test this concept, we transplanted bone marrow-derived normal adult human MultiStem® cells into the cerebral lateral ventricles of immunodeficient MPS-I neonatal mice. Transplanted cells and human-specific DNA were detected in the hippocampal formation, striatum, and other areas of the central nervous system. Brain tissue assays revealed significant long-term decrease in GAG levels in the hippocampus and striatum. Sensorimotor testing 6 months after transplantation demonstrated significantly improved rotarod performance of transplanted mice in comparison to nontransplanted and sham-transplanted control animals. These results suggest that a single injection of MultiStem cells into the cerebral ventricles of neonatal MPS-I mice induces sustained reduction in GAG accumulation within the brain, and modest long-term improvement in sensorimotor function.
Collapse
Affiliation(s)
- Zhenhong Nan
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Vaquer G, Rivière F, Mavris M, Bignami F, Llinares-Garcia J, Westermark K, Sepodes B. Animal models for metabolic, neuromuscular and ophthalmological rare diseases. Nat Rev Drug Discov 2013; 12:287-305. [PMID: 23493083 DOI: 10.1038/nrd3831] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Animal models are important tools in the discovery and development of treatments for rare diseases, particularly given the small populations of patients in which to evaluate therapeutic candidates. Here, we provide a compilation of mammalian animal models for metabolic, neuromuscular and ophthalmological orphan-designated conditions based on information gathered by the European Medicines Agency's Committee for Orphan Medicinal Products (COMP) since its establishment in 2000, as well as from a review of the literature. We discuss the predictive value of the models and their advantages and limitations with the aim of highlighting those that are appropriate for the preclinical evaluation of novel therapies, thereby facilitating further drug development for rare diseases.
Collapse
Affiliation(s)
- Guillaume Vaquer
- Human Medicines Special Areas, Human Medicines Development and Evaluation, European Medicines Agency, London E14 4HB, UK
| | | | | | | | | | | | | |
Collapse
|
23
|
Baldo G, Mayer FQ, Martinelli B, Dilda A, Meyer F, Ponder KP, Giugliani R, Matte U. Evidence of a progressive motor dysfunction in Mucopolysaccharidosis type I mice. Behav Brain Res 2012; 233:169-75. [PMID: 22580166 DOI: 10.1016/j.bbr.2012.04.051] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Revised: 04/24/2012] [Accepted: 04/28/2012] [Indexed: 10/28/2022]
Abstract
Mucopolysaccharidosis (MPS) type I (Hurler syndrome) is a lysosomal storage disorder characterized by deficiency of alpha-L-iduronidase (IDUA), intracellular storage of glycosaminoglycans (GAGs) and progressive neurological pathology. The MPS I mouse model provides an opportunity to study the pathophysiology of this disorder and to determine the efficacy of novel therapies. Previous work has demonstrated a series of abnormalities in MPS I mice behavior, but so far some important brain functions have not been addressed. Therefore, in the present study we aimed to determine if MPS I mice have motor abnormalities, and at what age they become detectable. MPS I and normal male mice from 2 to 8 months of age were tested in open-field for locomotor activity, hindlimb gait analysis and hang wire performance. We were able to detect a progressive reduction in the crossings and rearings in the open field test and in the hang wire test in MPS I mice from 4 months, as well as a reduction in the gait length at 8 months. Histological examination of 8-month old mice cortex and cerebellum revealed storage of GAGs in Purkinje cells and neuroinflammation, evidenced by GFAP immunostaining. However TUNEL staining was negative, suggesting that death does not occur. Our findings suggest that MPS I mice have a progressive motor dysfunction, which is not caused by loss of neuron cells but might be related to a neuroinflammatory process.
Collapse
Affiliation(s)
- Guilherme Baldo
- Gene Therapy Center - Research Center - Hospital de Clinicas de Porto Alegre, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Wolf DA, Hanson LR, Aronovich EL, Nan Z, Low WC, Frey WH, McIvor RS. Lysosomal enzyme can bypass the blood-brain barrier and reach the CNS following intranasal administration. Mol Genet Metab 2012; 106:131-4. [PMID: 22420937 DOI: 10.1016/j.ymgme.2012.02.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 02/05/2012] [Indexed: 11/28/2022]
Abstract
Here we provide the first evidence that therapeutic levels of a lysosomal enzyme can bypass the blood-brain barrier following intranasal administration. α-L-iduronidase (IDUA) activity was detected throughout the brains of IDUA-deficient mice following a single intranasal treatment with concentrated Aldurazyme® (laronidase) and was also detected after intranasal treatment with an adeno-associated virus (AAV) vector expressing human IDUA. These results suggest that intranasal routes of delivery may be efficacious in the treatment of lysosomal storage disorders.
Collapse
Affiliation(s)
- Daniel A Wolf
- Gene Therapy Program, Institute of Human Genetics, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Wolf DA, Lenander AW, Nan Z, Braunlin EA, Podetz-Pedersen KM, Whitley CB, Gupta P, Low WC, McIvor RS. Increased longevity and metabolic correction following syngeneic BMT in a murine model of mucopolysaccharidosis type I. Bone Marrow Transplant 2011; 47:1235-40. [PMID: 22179554 PMCID: PMC4465813 DOI: 10.1038/bmt.2011.239] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mucopolysaccharidosis type I (MPS I) is an autosomal recessive inherited disease caused by deficiency of the glycosidase α-L-iduronidase (IDUA). Deficiency of IDUA leads to lysosomal accumulation of the glycosaminoglycans (GAG) heparan and dermatan sulfate and associated multi-systemic disease, the most severe form known as Hurler syndrome. Since 1981, the treatment of Hurler patients has often included allogeneic bone marrow transplantation (BMT) from a matched donor. However, mouse models of the disease were not developed until 1997. To further characterize the MPS I mouse model and to study the effectiveness of BMT in these animals, we engrafted a cohort (n=33) of 4–8 week-old Idua−/− animals with high levels (88.4 ± 10.3%) of wild-type donor marrow. Engrafted animals displayed an increased lifespan, preserved cardiac function, partially restored IDUA activity in peripheral organs, and decreased GAG accumulation in both peripheral organs and in the brain. However, levels of GAG and GM3 ganglioside in the brain remained elevated in comparison to unaffected animals. Since these results are similar to those observed in Hurler patients following BMT, this murine transplantation model can be used to evaluate the effects of novel, more effective methods of delivering IDUA to the brain as an adjunct to BMT.
Collapse
Affiliation(s)
- D A Wolf
- Gene Therapy Program, Institute of Human Genetics, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wolf DA, Lenander AW, Nan Z, Belur LR, Whitley CB, Gupta P, Low WC, McIvor RS. Direct gene transfer to the CNS prevents emergence of neurologic disease in a murine model of mucopolysaccharidosis type I. Neurobiol Dis 2011; 43:123-33. [PMID: 21397026 DOI: 10.1016/j.nbd.2011.02.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 02/23/2011] [Accepted: 02/27/2011] [Indexed: 12/31/2022] Open
Abstract
The mucopolysaccharidoses (MPSs) are a group of 11 storage diseases caused by disruptions in glycosaminoglycan (GAG) catabolism, leading to their accumulation in lysosomes. Resultant multisystemic disease is manifested by growth delay, hepatosplenomegaly, skeletal dysplasias, cardiopulmonary obstruction, and, in severe MPS I, II, III, and VII, progressive neurocognitive decline. Some MPSs are treated by allogeneic hematopoietic stem cell transplantation (HSCT) and/or recombinant enzyme replacement therapy (ERT), but effectiveness is limited by central nervous system (CNS) access across the blood-brain barrier. To provide a high level of gene product to the CNS, we tested neonatal intracerebroventricular (ICV) infusion of an adeno-associated virus (AAV) serotype 8 vector transducing the human α-L-iduronidase gene in MPS I mice. Supranormal levels of iduronidase activity in the brain (including 40× normal levels in the hippocampus) were associated with transduction of neurons in motor and limbic areas identifiable by immunofluorescence staining. The treatment prevented accumulation of GAG and GM3 ganglioside storage materials and emergence of neurocognitive dysfunction in a modified Morris water maze test. The results suggest the potential of improved outcome for MPSs and other neurological diseases when a high level of gene expression can be achieved by direct, early administration of vector to the CNS.
Collapse
Affiliation(s)
- Daniel A Wolf
- Department of Genetics, Institute of Human Genetics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Heinecke KA, Peacock BN, Blazar BR, Tolar J, Seyfried TN. Lipid composition of whole brain and cerebellum in Hurler syndrome (MPS IH) mice. Neurochem Res 2011; 36:1669-76. [PMID: 21253856 DOI: 10.1007/s11064-011-0400-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2011] [Indexed: 11/29/2022]
Abstract
Hurler syndrome (MPS IH) is caused by a mutation in the gene encoding alpha-L-iduronidase (IDUA) and leads to the accumulation of partially degraded glycosaminoglycans (GAGs). Ganglioside content is known to increase secondary to GAG accumulation. Most studies in organisms with MPS IH have focused on changes in gangliosides GM3 and GM2, without the study of other lipids. We evaluated the total lipid distribution in the whole brain and cerebellum of MPS IH (Idua⁻/⁻) and control (Idua(+/?)) mice at 6 months and at 12 months of age. The content of total sialic acid and levels of gangliosides GM3, GM2, and GD3 were greater in the whole brains of Idua⁻/⁻ mice then in Idua (+/?) mice at 12 months of age. No other significant lipid differences were found in either whole brain or in cerebellum at either age. The accumulation of ganglioside GD3 suggests that neurodegeneration occurs in the Idua⁻/⁻) mouse brain, but not to the extent seen in human MPS IH brain.
Collapse
Affiliation(s)
- Karie A Heinecke
- Department of Biology, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA 02467, USA
| | | | | | | | | |
Collapse
|
28
|
Cotugno G, Tessitore A, Capalbo A, Annunziata P, Strisciuglio C, Faella A, Aurilio M, Di Tommaso M, Russo F, Mancini A, De Leonibus E, Aloj L, Auricchio A. Different serum enzyme levels are required to rescue the various systemic features of the mucopolysaccharidoses. Hum Gene Ther 2010; 21:555-69. [PMID: 20021231 DOI: 10.1089/hum.2009.189] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mucopolysaccharidoses (MPSs) are lysosomal storage disorders characterized by progressive accumulation of glycosaminoglycans (GAGs) in various tissues. Enzyme replacement therapy (ERT) for several MPSs is available to date. However, the efficacy of ERT is limited, in particular in compartments such as bone, cartilage, the brain, and the eyes. We selected a rodent model of an MPS, with no central nervous system storage, to study the impact, on systemic features of the disease, of various stable levels of exogenous enzymes produced by adeno-associated viral vector (AAV)-mediated liver gene transfer. Low levels (6% of normal) of circulating enzyme were enough to reduce storage and inflammation in the visceral organs and to ameliorate skull abnormalities; intermediate levels (11% of normal) were required to reduce urinary GAG excretion; and high levels (>or=50% of normal) rescued abnormalities of the long bones and motor activity. These data will be instrumental to design appropriate clinical protocols based on either enzyme or gene replacement therapy for MPS and to predict their impact on the pathological features of MPS.
Collapse
Affiliation(s)
- Gabriella Cotugno
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wang D, Shukla C, Liu X, Schoeb TR, Clarke LA, Bedwell DM, Keeling KM. Characterization of an MPS I-H knock-in mouse that carries a nonsense mutation analogous to the human IDUA-W402X mutation. Mol Genet Metab 2010; 99:62-71. [PMID: 19751987 PMCID: PMC2795040 DOI: 10.1016/j.ymgme.2009.08.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 08/17/2009] [Accepted: 08/17/2009] [Indexed: 02/01/2023]
Abstract
Here we report the characterization of a knock-in mouse model for the autosomal recessive disorder mucopolysaccharidosis type I-Hurler (MPS I-H), also known as Hurler syndrome. MPS I-H is the most severe form of alpha-l-iduronidase deficiency. alpha-l-iduronidase (encoded by the IDUA gene) is a lysosomal enzyme that participates in the degradation of dermatan sulfate and heparan sulfate. Using gene replacement methodology, a nucleotide change was introduced into the mouse Idua locus that resulted in a nonsense mutation at codon W392. The Idua-W392X mutation is analogous to the human IDUA-W402X mutation commonly found in MPS I-H patients. We found that the phenotype in homozygous Idua-W392X mice closely correlated with the human MPS I-H disease. Homozygous W392X mice showed no detectable alpha-l-iduronidase activity. We observed a defect in GAG degradation as evidenced by an increase in sulfated GAGs excreted in the urine and stored in multiple tissues. Histology and electron microscopy also revealed evidence of GAG storage in all tissues examined. Additional assessment revealed bone abnormalities and altered metabolism within the Idua-W392X mouse. This new mouse will provide an important tool to investigate therapeutic approaches for MPS I-H that cannot be addressed using current MPS I-H animal models.
Collapse
Affiliation(s)
- Dan Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Charu Shukla
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xiaoli Liu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Trenton R. Schoeb
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lorne A. Clarke
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, CA
| | - David M. Bedwell
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kim M. Keeling
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
30
|
Aronovich EL, Bell JB, Khan SA, Belur LR, Gunther R, Koniar B, Schachern PA, Parker JB, Carlson CS, Whitley CB, McIvor RS, Gupta P, Hackett PB. Systemic correction of storage disease in MPS I NOD/SCID mice using the sleeping beauty transposon system. Mol Ther 2009; 17:1136-44. [PMID: 19384290 DOI: 10.1038/mt.2009.87] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Sleeping Beauty (SB) transposon system is a nonviral vector that directs transgene integration into vertebrate genomes. We hydrodynamically delivered SB transposon plasmids encoding human alpha-L-iduronidase (hIDUA) at two DNA doses, with and without an SB transposase gene, to NOD.129(B6)-Prkdc(scid) IDUA(tm1Clk)/J mice. In transposon-treated, nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice with mucopolysaccharidosis type I (MPS I), plasma IDUA persisted for 18 weeks at levels up to several hundred-fold wild-type (WT) activity, depending on DNA dose and gender. IDUA activity was present in all examined somatic organs, as well as in the brain, and correlated with both glycosaminoglycan (GAG) reduction in these organs and level of expression in the liver, the target of transposon delivery. IDUA activity was higher in the treated males than in females. In females, omission of transposase source resulted in significantly lower IDUA levels and incomplete GAG reduction in some organs, confirming the positive effect of transposition on long-term IDUA expression and correction of the disease. The SB transposon system proved efficacious in correcting several clinical manifestations of MPS I in mice, including thickening of the zygomatic arch, hepatomegaly, and accumulation of foamy macrophages in bone marrow and synovium, implying potential effectiveness of this approach in treatment of human MPS I.
Collapse
Affiliation(s)
- Elena L Aronovich
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, 55455, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|