1
|
Del Grosso A, Parlanti G, Mezzena R, Cecchini M. Current treatment options and novel nanotechnology-driven enzyme replacement strategies for lysosomal storage disorders. Adv Drug Deliv Rev 2022; 188:114464. [PMID: 35878795 DOI: 10.1016/j.addr.2022.114464] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/26/2022] [Accepted: 07/19/2022] [Indexed: 11/01/2022]
Abstract
Lysosomal storage disorders (LSDs) are a vast group of more than 50 clinically identified metabolic diseases. They are singly rare, but they affect collectively 1 on 5,000 live births. They result in most of the cases from an enzymatic defect within lysosomes, which causes the subsequent augmentation of unwanted substrates. This accumulation process leads to plenty of clinical signs, determined by the specific substrate and accumulation area. The majority of LSDs present a broad organ and tissue engagement. Brain, connective tissues, viscera and bones are usually afflicted. Among them, brain disease is markedly frequent (two-thirds of LSDs). The most clinically employed approach to treat LSDs is enzyme replacement therapy (ERT), which is practiced by administering systemically the missed or defective enzyme. It represents a healthful strategy for 11 LSDs at the moment, but it solves the pathology only in the case of Gaucher disease. This approach, in fact, is not efficacious in the case of LSDs that have an effect on the central nervous system (CNS) due to the existence of the blood-brain barrier (BBB). Additionally, ERT suffers from several other weak points, such as low penetration of the exogenously administered enzyme to poorly vascularized areas, the development of immunogenicity and infusion-associated reactions (IARs), and, last but not least, the very high cost and lifelong needed. To ameliorate these weaknesses lot of efforts have been recently spent around the development of innovative nanotechnology-driven ERT strategies. They may boost the power of ERT and minimize adverse reactions by loading enzymes into biodegradable nanomaterials. Enzyme encapsulation into biocompatible liposomes, micelles, and polymeric nanoparticles, for example, can protect enzymatic activity, eliminating immunologic reactions and premature enzyme degradation. It can also permit a controlled release of the payload, ameliorating pharmacokinetics and pharmacodynamics of the drug. Additionally, the potential to functionalize the surface of the nanocarrier with targeting agents (antibodies or peptides), could promote the passage through biological barriers. In this review we examined the clinically applied ERTs, highlighting limitations that do not allow to completely cure the specific LSD. Later, we critically consider the nanotechnology-based ERT strategies that have beenin-vitroand/orin-vivotested to improve ERT efficacy.
Collapse
Affiliation(s)
- Ambra Del Grosso
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Gabriele Parlanti
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Roberta Mezzena
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
2
|
Lehmann RJ, Jolly LA, Johnson BV, Lord MS, Kim HN, Saville JT, Fuller M, Byers S, Derrick-Roberts AL. Impaired neural differentiation of MPS IIIA patient induced pluripotent stem cell-derived neural progenitor cells. Mol Genet Metab Rep 2021; 29:100811. [PMID: 34712574 PMCID: PMC8531667 DOI: 10.1016/j.ymgmr.2021.100811] [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: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 11/23/2022] Open
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is characterised by a progressive neurological decline leading to early death. It is caused by bi-allelic loss-of-function mutations in SGSH encoding sulphamidase, a lysosomal enzyme required for heparan sulphate glycosaminoglycan (HS GAG) degradation, that results in the progressive build-up of HS GAGs in multiple tissues most notably the central nervous system (CNS). Skin fibroblasts from two MPS IIIA patients who presented with an intermediate and a severe clinical phenotype, respectively, were reprogrammed into induced pluripotent stem cells (iPSCs). The intermediate MPS IIIA iPSCs were then differentiated into neural progenitor cells (NPCs) and subsequently neurons. The patient derived fibroblasts, iPSCs, NPCs and neurons all displayed hallmark biochemical characteristics of MPS IIIA including reduced sulphamidase activity and increased accumulation of an MPS IIIA HS GAG biomarker. Proliferation of MPS IIIA iPSC-derived NPCs was reduced compared to control, but could be partially rescued by reintroducing functional sulphamidase enzyme, or by doubling the concentration of the mitogen fibroblast growth factor 2 (FGF2). Whilst both control heparin, and MPS IIIA HS GAGs had a similar binding affinity for FGF2, only the latter inhibited FGF signalling, suggesting accumulated MPS IIIA HS GAGs disrupt the FGF2:FGF2 receptor:HS signalling complex. Neuronal differentiation of MPS IIIA iPSC-derived NPCs was associated with a reduction in the expression of neuronal cell marker genes βIII-TUBULIN, NF-H and NSE, revealing reduced neurogenesis compared to control. A similar result was achieved by adding MPS IIIA HS GAGs to the culture medium during neuronal differentiation of control iPSC-derived NPCs. This study demonstrates the generation of MPS IIIA iPSCs, and NPCs, the latter of which display reduced proliferation and neurogenic capacity. Reduced NPC proliferation can be explained by a model in which soluble MPS IIIA HS GAGs compete with cell surface HS for FGF2 binding. The mechanism driving reduced neurogenesis remains to be determined but appears downstream of MPS IIIA HS GAG accumulation.
Collapse
Affiliation(s)
- Rebecca J. Lehmann
- Genetics and Molecular Pathology, SA Pathology (at the Women's and Children's Hospital), 72 King William Rd, North Adelaide, SA 5006, Australia
- Department of Molecular and Cellular Biology, University of Adelaide, Adelaide, SA 5005, Australia
| | - Lachlan A. Jolly
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Brett V. Johnson
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Megan S. Lord
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| | - Ha Na Kim
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| | - Jennifer T. Saville
- Genetics and Molecular Pathology, SA Pathology (at the Women's and Children's Hospital), 72 King William Rd, North Adelaide, SA 5006, Australia
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology (at the Women's and Children's Hospital), 72 King William Rd, North Adelaide, SA 5006, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Sharon Byers
- Genetics and Molecular Pathology, SA Pathology (at the Women's and Children's Hospital), 72 King William Rd, North Adelaide, SA 5006, Australia
- Department of Molecular and Cellular Biology, University of Adelaide, Adelaide, SA 5005, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Ainslie L.K. Derrick-Roberts
- Genetics and Molecular Pathology, SA Pathology (at the Women's and Children's Hospital), 72 King William Rd, North Adelaide, SA 5006, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
3
|
Douglass ML, Beard H, Shoubridge A, Nazri N, King B, Trim PJ, Duplock SK, Snel MF, Hopwood JJ, Hemsley KM. Is SGSH heterozygosity a risk factor for early-onset neurodegenerative disease? J Inherit Metab Dis 2021; 44:763-776. [PMID: 33423317 DOI: 10.1002/jimd.12359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 02/05/2023]
Abstract
Lysosomal dysfunction may be an important factor in the pathogenesis of neurodegenerative disorders such as Parkinson's disease (PD). Heterozygous mutations in the gene encoding the lysosomal enzyme glucocerebrosidase (GBA1) have been found in PD patients, and some but not all mutations in other lysosomal enzyme genes, for example, NPC1 and MCOLN1 have been associated with PD. We have examined the behaviour and brain structure of mice carrying a D31N mutation in the sulphamidase (Sgsh) gene which encodes a lysosomal sulphatase. Female heterozygotes and wildtype mice aged 12-, 15-, 18- and 21-months of age underwent motor phenotyping and the brain was comprehensively evaluated for disease-associated lesions. Heterozygous mice exhibited impaired performance in the negative geotaxis test when compared with wildtype mice. Whilst the brain of Sgsh heterozygotes aged up to 21-months did not exhibit any of the gross features of PD, Alzheimer's disease or the neurodegenerative lysosomal storage disorders, for example, loss of striatal dopamine, reduced GBA activity, α-synuclein-positive inclusions, perturbation of lipid synthesis, or cerebellar Purkinje cell drop-out, we noted discrete structural aberrations in the dendritic tree of cortical pyramidal neurons in 21-month old animals. The overt disease lesions and resultant phenotypic changes previously described in individuals with heterozygous mutations in lysosomal enzyme genes such as glucocerebrosidase may be enzyme dependent. By better understanding why deficiency in, or mutant forms of some but not all lysosomal proteins leads to heightened risk or earlier onset of classical neurodegenerative disorders, novel disease-causing mechanisms may be identified.
Collapse
Affiliation(s)
- Meghan L Douglass
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Helen Beard
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Andrew Shoubridge
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Nazzmer Nazri
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Barbara King
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Paul J Trim
- Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Mass Spectrometry Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, SAHMRI, Adelaide, South Australia, Australia
| | - Stephen K Duplock
- Mass Spectrometry Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, SAHMRI, Adelaide, South Australia, Australia
| | - Marten F Snel
- Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Mass Spectrometry Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, SAHMRI, Adelaide, South Australia, Australia
| | - John J Hopwood
- Hopwood Centre for Neurobiology, Lifelong Health Theme, SAHMRI, Adelaide, South Australia, Australia
| | - Kim M Hemsley
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
4
|
Harm TA, Hostetter SJ, Nenninger AS, Valentine BN, Ellinwood NM, Smith JD. Temporospatial Development of Neuropathologic Findings in a Canine Model of Mucopolysaccharidosis IIIB. Vet Pathol 2020; 58:205-222. [PMID: 33205707 DOI: 10.1177/0300985820960128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mucopolysaccharidosis (MPS) IIIB is a neuropathic lysosomal storage disease characterized by the deficient activity of a lysosomal enzyme obligate for the degradation of the glycosaminoglycan (GAG) heparan sulfate (HS). The pathogenesis of neurodegeneration in MPS IIIB is incompletely understood. Large animal models are attractive for pathogenesis and therapeutic studies due to their larger size, outbred genetics, longer lifespan, and naturally occurring MPS IIIB disease. However, the temporospatial development of neuropathologic changes has not been reported for canine MPS IIIB. Here we describe lesions in 8 brain regions, cervical spinal cord, and dorsal root ganglion (DRG) in a canine model of MPS IIIB that includes dogs aged from 2 to 26 months of age. Pathological changes in the brain included early microscopic vacuolation of glial cells initially observed at 2 months, and vacuolation of neurons initially observed at 10 months. Inclusions within affected cells variably stained positively with PAS and LFB stains. Quantitative immunohistochemistry demonstrated increased glial expression of GFAP and Iba1 in dogs with MPS IIIB compared to age-matched controls at all time points, suggesting neuroinflammation occurs early in disease. Loss of Purkinje cells was initially observed at 10 months and was pronounced in 18- and 26-month-old dogs with MPS IIIB. Our results support the dog as a replicative model of MPS IIIB neurologic lesions and detail the pathologic and neuroinflammatory changes in the spinal cord and DRG of MPS IIIB-affected dogs.
Collapse
Affiliation(s)
| | - Shannon J Hostetter
- 70724Iowa State University, Ames, IA, USA.,Current address: Department of Veterinary Pathology, University of Georgia, Athens, GA, USA
| | | | | | | | | |
Collapse
|
5
|
Story BD, Miller ME, Bradbury AM, Million ED, Duan D, Taghian T, Faissler D, Fernau D, Beecy SJ, Gray-Edwards HL. Canine Models of Inherited Musculoskeletal and Neurodegenerative Diseases. Front Vet Sci 2020; 7:80. [PMID: 32219101 PMCID: PMC7078110 DOI: 10.3389/fvets.2020.00080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Mouse models of human disease remain the bread and butter of modern biology and therapeutic discovery. Nonetheless, more often than not mouse models do not reproduce the pathophysiology of the human conditions they are designed to mimic. Naturally occurring large animal models have predominantly been found in companion animals or livestock because of their emotional or economic value to modern society and, unlike mice, often recapitulate the human disease state. In particular, numerous models have been discovered in dogs and have a fundamental role in bridging proof of concept studies in mice to human clinical trials. The present article is a review that highlights current canine models of human diseases, including Alzheimer's disease, degenerative myelopathy, neuronal ceroid lipofuscinosis, globoid cell leukodystrophy, Duchenne muscular dystrophy, mucopolysaccharidosis, and fucosidosis. The goal of the review is to discuss canine and human neurodegenerative pathophysiologic similarities, introduce the animal models, and shed light on the ability of canine models to facilitate current and future treatment trials.
Collapse
Affiliation(s)
- Brett D. Story
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
- University of Florida College of Veterinary Medicine, Gainesville, FL, United States
| | - Matthew E. Miller
- Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Allison M. Bradbury
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily D. Million
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Dominik Faissler
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Sidney J. Beecy
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, United States
| | - Heather L. Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
6
|
Bigger BW, Begley DJ, Virgintino D, Pshezhetsky AV. Anatomical changes and pathophysiology of the brain in mucopolysaccharidosis disorders. Mol Genet Metab 2018; 125:322-331. [PMID: 30145178 DOI: 10.1016/j.ymgme.2018.08.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/09/2018] [Accepted: 08/09/2018] [Indexed: 11/28/2022]
Abstract
Mucopolysaccharidosis (MPS) disorders are caused by deficiencies in lysosomal enzymes, leading to impaired glycosaminoglycan (GAG) degradation. The resulting GAG accumulation in cells and connective tissues ultimately results in widespread tissue and organ dysfunction. The seven MPS types currently described are heterogeneous and progressive disorders, with somatic and neurological manifestations depending on the type of accumulating GAG. Heparan sulfate (HS) is one of the GAGs stored in patients with MPS I, II, and VII and the main GAG stored in patients with MPS III. These disorders are associated with significant central nervous system (CNS) abnormalities that can manifest as impaired cognition, hyperactive and/or aggressive behavior, epilepsy, hydrocephalus, and sleeping problems. This review discusses the anatomical and pathophysiological CNS changes accompanying HS accumulation as well as the mechanisms believed to cause CNS abnormalities in MPS patients. The content of this review is based on presentations and discussions on these topics during a meeting on the brain in MPS attended by an international group of MPS experts.
Collapse
Affiliation(s)
- Brian W Bigger
- Stem Cell & Neurotherapies Laboratory, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK.
| | - David J Begley
- Drug Delivery Group, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy and Histology Unit, Bari University School of Medicine, Bari, Italy
| | - Alexey V Pshezhetsky
- Departments of Pediatrics and Biochemistry, CHU Sainte-Justine, Research Center, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
7
|
Winner LK, Marshall NR, Jolly RD, Trim PJ, Duplock SK, Snel MF, Hemsley KM. Evaluation of Disease Lesions in the Developing Canine MPS IIIA Brain. JIMD Rep 2018; 43:91-101. [PMID: 29923090 PMCID: PMC6323028 DOI: 10.1007/8904_2018_110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 04/04/2018] [Accepted: 04/26/2018] [Indexed: 01/26/2023] Open
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA) is an inherited neurodegenerative disease of childhood that results in early death. Post-mortem studies have been carried out on human MPS IIIA brain, but little is known about early disease development. Here, we utilised the Huntaway dog model of MPS IIIA to evaluate disease lesion development from 2 to 24 weeks of age. A significant elevation in primarily stored heparan sulphate was observed in all brain regions assessed in MPS IIIA pups ≤9.5 weeks of age. There was a significant elevation in secondarily stored ganglioside (GM3 36:1) in ≤9.5-week-old MPS IIIA pup cerebellum, and other brain regions also exhibited accumulation of this lipid with time. The number of neural stem cells and neuronal precursor cells was essentially unchanged in MPS IIIA dog brain (c.f. unaffected) over the time course assessed, a finding corroborated by neuron cell counts. We observed early neuroinflammatory changes in young MPS IIIA pup brain, with significantly increased numbers of activated microglia recorded in all but one brain region in MPS IIIA pups ≤9.5 weeks of age (c.f. age-matched unaffected pups). In conclusion, infant-paediatric-stage MPS IIIA canine brain exhibits substantial and progressive primary and secondary substrate accumulation, coupled with early and robust microgliosis. Whilst early initiation of treatment is likely to be required to maintain optimal neurological function, the brain's neurodevelopmental potential appears largely unaffected by the disease process; further investigations confirming this are warranted.
Collapse
Affiliation(s)
- Leanne K. Winner
- grid.430453.5Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Neil R. Marshall
- grid.148374.d0000 0001 0696 9806Institute of Veterinary, Animal and Biomedical Science, Massey University, Palmerston North, New Zealand
| | - Robert D. Jolly
- grid.148374.d0000 0001 0696 9806Institute of Veterinary, Animal and Biomedical Science, Massey University, Palmerston North, New Zealand
| | - Paul J. Trim
- grid.430453.5Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Stephen K. Duplock
- grid.430453.5Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Marten F. Snel
- grid.430453.5Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Kim M. Hemsley
- grid.430453.5Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, SA Australia
| |
Collapse
|
8
|
Salazar DA, Rodríguez-López A, Herreño A, Barbosa H, Herrera J, Ardila A, Barreto GE, González J, Alméciga-Díaz CJ. Systems biology study of mucopolysaccharidosis using a human metabolic reconstruction network. Mol Genet Metab 2016; 117:129-39. [PMID: 26276570 DOI: 10.1016/j.ymgme.2015.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 07/30/2015] [Accepted: 08/01/2015] [Indexed: 12/11/2022]
Abstract
Mucopolysaccharidosis (MPS) is a group of lysosomal storage diseases (LSD), characterized by the deficiency of a lysosomal enzyme responsible for the degradation of glycosaminoglycans (GAG). This deficiency leads to the lysosomal accumulation of partially degraded GAG. Nevertheless, deficiency of a single lysosomal enzyme has been associated with impairment in other cell mechanism, such as apoptosis and redox balance. Although GAG analysis represents the main biomarker for MPS diagnosis, it has several limitations that can lead to a misdiagnosis, whereby the identification of new biomarkers represents an important issue for MPS. In this study, we used a system biology approach, through the use of a genome-scale human metabolic reconstruction to understand the effect of metabolism alterations in cell homeostasis and to identify potential new biomarkers in MPS. In-silico MPS models were generated by silencing of MPS-related enzymes, and were analyzed through a flux balance and variability analysis. We found that MPS models used approximately 2286 reactions to satisfy the objective function. Impaired reactions were mainly involved in cellular respiration, mitochondrial process, amino acid and lipid metabolism, and ion exchange. Metabolic changes were similar for MPS I and II, and MPS III A to C; while the remaining MPS showed unique metabolic profiles. Eight and thirteen potential high-confidence biomarkers were identified for MPS IVB and VII, respectively, which were associated with the secondary pathologic process of LSD. In vivo evaluation of predicted intermediate confidence biomarkers (β-hexosaminidase and β-glucoronidase) for MPS IVA and VI correlated with the in-silico prediction. These results show the potential of a computational human metabolic reconstruction to understand the molecular mechanisms this group of diseases, which can be used to identify new biomarkers for MPS.
Collapse
Affiliation(s)
- Diego A Salazar
- Grupo Bioquímica Computacional y Bioinformática, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Alexander Rodríguez-López
- Institute for the Study of Inborn Errors of Metabolism, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Chemistry Department, School of Science, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Angélica Herreño
- Institute for the Study of Inborn Errors of Metabolism, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Hector Barbosa
- Institute for the Study of Inborn Errors of Metabolism, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Juliana Herrera
- Institute for the Study of Inborn Errors of Metabolism, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Andrea Ardila
- Institute for the Study of Inborn Errors of Metabolism, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Hospital Universitario San Ignacio, Bogotá D.C., Colombia
| | - George E Barreto
- Grupo Bioquímica Computacional y Bioinformática, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Janneth González
- Grupo Bioquímica Computacional y Bioinformática, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.
| |
Collapse
|