1
|
Giugliani R, de Siqueira ACM, Santos ES, Leão EKEA, Carvalho GDS, Santos MLSF, Raskin S, Martins AM. Heparan sulfate in cerebrospinal fluid as a biomarker to assess disease severity and for treatment monitoring in patients with Mucopolysaccharidosis Type II: a position statement. Orphanet J Rare Dis 2024; 19:436. [PMID: 39593190 PMCID: PMC11590453 DOI: 10.1186/s13023-024-03463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Patients with mucopolysaccharidosis type II (MPS II) can present with a severe neuronopathic phenotype or an attenuated non-neuronopathic phenotype. In the light of the recent development of drugs that cross the blood-brain barrier for treatment of neurologic MPS II symptoms, it is critical to define biomarkers that objectively differentiate phenotypes and monitor therapeutic outcomes of advanced treatments. In December 2023, a panel of Brazilian experts discussed the potential of quantifying heparan sulfate (HS) in the cerebrospinal fluid (CSF) as a biomarker for assessing neurological impairment in patients with MPS II, as well as the potential of the molecule as an objective parameter for therapeutic monitoring. Based on scientific evidence, the experts concluded that HS in CSF is predominantly derived from the brain and reflects neurological impairment in patients with MPS II. CSF HS levels may help differentiate between neuronopathic and non-neuronopathic forms of MPS II, with preliminary observations suggesting a potential threshold around 4,000 ng/mL when HS quantification is performed using the same method described in clinical studies of pabinafusp alfa. According to the authors, monitoring HS levels in CSF can serve as an objective parameter for assessing the effectiveness of treatment with drugs that cross the blood-brain barrier. The recommended timing of HS evaluations in CSF of patients with the severe phenotype is: (i) before treatment; (ii) six months after starting treatment; and (iii) two years after starting treatment. The same monitoring scheme is recommended for patients with the attenuated MPS II phenotype, however, after two years of treatment, the physician may elect to perform regular neurocognitive evaluations instead of measuring HS in CSF. Lastly, the authors reinforced the importance of evaluating adherence to treatment, including interruptions, to provide a more meaningful assessment of the treatment's real-world impact and to determine the ideal timing of CSF collection for therapeutic monitoring.
Collapse
Affiliation(s)
- Roberto Giugliani
- Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, Porto Alegre, 90035-903, Brasil.
- Dasa Genômica, Casa dos Raros, Porto Alegre, Brazil.
| | | | | | | | | | | | - Salmo Raskin
- Centro de Aconselhamento e Laboratório Genetika, Curitiba, Brazil
| | - Ana Maria Martins
- Centro de Referência em Erros Inatos do Metabolismo, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Elendu C, Babawale EA, Babarinde FO, Babatunde OD, Chukwu C, Chiegboka SF, Shode OP, Ngozi-ibeh JK, Njoku A, Ikokwu MN, Kaka GU, Hassan JI, Fatungase OO, Osifodunrin T, Udoeze CA, Ikeji VI. Neurological manifestations of lysosomal storage diseases. Ann Med Surg (Lond) 2024; 86:6619-6635. [PMID: 39525762 PMCID: PMC11543150 DOI: 10.1097/ms9.0000000000002611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/19/2024] [Indexed: 11/16/2024] Open
Abstract
Lysosomal storage diseases (LSDs) encompass a group of rare inherited metabolic disorders characterized by the accumulation of undegraded substrates within lysosomes, leading to multisystemic manifestations, including profound neurological involvement. This article provides a concise overview of the neurological manifestations of LSDs, with a focus on central nervous system (CNS) involvement and treatment strategies. While the paper intricacies of each LSD subtype and its associated CNS manifestations, it aims to provide a summary of the essential findings and implications. The neurological manifestations of LSDs encompass a spectrum of symptoms, including cognitive impairment, motor dysfunction, seizures, and sensory deficits, which significantly impact patients' quality of life and pose therapeutic challenges. Current treatment strategies primarily aim to alleviate symptoms and slow disease progression, with limited success in reversing established neurological damage. Enzyme replacement therapy, substrate reduction therapy, and emerging gene therapies hold promise for addressing CNS involvement in LSDs. However, challenges such as blood-brain barrier penetration and long-term efficacy remain. In addition to discussing treatment modalities, this article highlights the importance of early diagnosis, multidisciplinary care, and patient advocacy in optimizing outcomes for individuals affected by LSDs. Ethical considerations are also addressed, including equitable access to emerging treatments and integrating personalized medicine approaches. Overall, this article underscores the complex interplay between genetics, neuroscience, and clinical care in understanding and managing the neurological manifestations of LSDs while emphasizing the need for continued research and collaboration to advance therapeutic interventions and improve patient outcomes.
Collapse
|
3
|
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
|
4
|
Xu T, Heon-Roberts R, Moore T, Dubot P, Pan X, Guo T, Cairo CW, Holley R, Bigger B, Durcan TM, Levade T, Ausseil J, Amilhon B, Gorelik A, Nagar B, Sturiale L, Palmigiano A, Röckle I, Thiesler H, Hildebrandt H, Garozzo D, Pshezhetsky AV. Secondary deficiency of neuraminidase 1 contributes to CNS pathology in neurological mucopolysaccharidoses via hypersialylation of brain glycoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.587986. [PMID: 38712143 PMCID: PMC11071461 DOI: 10.1101/2024.04.26.587986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Mucopolysaccharidoses (MPS) are lysosomal storage diseases caused by defects in catabolism of glycosaminoglycans. MPS I, II, III and VII are associated with lysosomal accumulation of heparan sulphate and manifest with neurological deterioration. Most of these neurological MPS currently lack effective treatments. Here, we report that, compared to controls, neuraminidase 1 (NEU1) activity is drastically reduced in brain tissues of neurological MPS patients and in mouse models of MPS I, II, IIIA, IIIB and IIIC, but not of other neurological lysosomal disorders not presenting with heparan sulphate storage. We further show that accumulated heparan sulphate disrupts the lysosomal multienzyme complex of NEU1 with cathepsin A (CTSA), β-galactosidase (GLB1) and glucosamine-6-sulfate sulfatase (GALNS) necessary to maintain enzyme activity, and that NEU1 deficiency is linked to partial deficiencies of GLB1 and GALNS in cortical tissues and iPSC-derived cortical neurons of neurological MPS patients. Increased sialylation of N-linked glycans in brain samples of human MPS III patients and MPS IIIC mice implicated insufficient processing of brain N-linked sialylated glycans, except for polysialic acid, which was reduced in the brains of MPS IIIC mice. Correction of NEU1 activity in MPS IIIC mice by lentiviral gene transfer ameliorated previously identified hallmarks of the disease, including memory impairment, behavioural traits, and reduced levels of the excitatory synapse markers VGLUT1 and PSD95. Overexpression of NEU1 also restored levels of VGLUT1-/PSD95-positive puncta in cortical neurons derived from iPSC of an MPS IIIA patient. Together, our data demonstrate that heparan sulphate-induced secondary NEU1 deficiency and aberrant sialylation of glycoproteins implicated in synaptogenesis, memory, and behaviour constitute a novel pathological pathway in neurological MPS spectrum crucially contributing to CNS pathology. Graphical abstract
Collapse
|
5
|
Çopur O, Yazıcı H, Canbay E, Durmaz B, Canda E, Ucar SK, Coker M, Sozmen EY. Glycosaminoglycan-induced proinflammatory cytokine levels as disease marker in mucopolysaccharidosis. Cytokine 2024; 173:156410. [PMID: 37924740 DOI: 10.1016/j.cyto.2023.156410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/06/2023]
Abstract
Recently, it has been shown disturbances in oxidant/antioxidant system and increases in some inflammatory markers in animal studies and in some Mucopolysaccharidoses (MPSs) patients. In this study, we aimed to determine the oxidative stress/antioxidant parameters and pro-inflammatory cytokine levels in the serum of MPS patients, in order to evaluate the possible role of inflammation in these patient groups regarding to accumulated metabolites. MPS I (n = 3), MPS II (n = 8), MPS III (n = 4), MPS IVA (n = 3), MPS VI (n = 3), and VII (n = 1) patients and 20 age-matched healthy subjects were included into the study. There was no statistically significant change in activities of SOD, Catalase, GSH-Px and lipid peroxidation levels in erythrocytes between the MPS patients and healthy controls. While IL-1alpha (p = 0.054), IL-6 (p = 0.008) levels, and chitotriosidase activity (p = 0.003) elevated in MPS3 patients, IL1α (p = 0.006), IL-1β (p = 0.006), IL-6 (p = 0.006), IFNγ (p = 0.006), and NFκB (p = 0.006) levels increased in MPS-6 patients. Elevated levels of IL-6, IL1α and chitotriosidase activity demonstrated macrophage activation in MPSIII untreated with enzyme replacement. Our study showed for the first time that high levels of IL1α, IL-6, IL1β and NFκB were present in MPSVI patients, demonstrating the induction of inflammation by dermatan sulphate. The low level of paraoxonase in MPSVI patients may be a good marker for cardiac involvement. Overall, this study provides important insights into the relationship between lysosomal storage of glycosaminoglycan and inflammation in MPS patients. It highlights possible pathways for the increased release of inflammatory molecules and suggests new targets for the development of treatments.
Collapse
Affiliation(s)
- Oznur Çopur
- Ege University, Institute of Health Sciences, Department of Medical Biochemistry, Izmır, Turkiye; Ege University, Faculty of Medicine, Department of Medical Biochemistry, Izmır, Turkiye
| | - Havva Yazıcı
- Ege University, Faculty of Medicine, Department of Inherited Metabolic Disease, Izmır, Turkiye
| | - Erhan Canbay
- Ege University, Faculty of Medicine, Department of Medical Biochemistry, Izmır, Turkiye
| | - Burak Durmaz
- Ege University, Institute of Health Sciences, Department of Medical Biochemistry, Izmır, Turkiye; Ege University, Faculty of Medicine, Department of Medical Biochemistry, Izmır, Turkiye
| | - Ebru Canda
- Ege University, Faculty of Medicine, Department of Inherited Metabolic Disease, Izmır, Turkiye
| | - Sema Kalkan Ucar
- Ege University, Faculty of Medicine, Department of Inherited Metabolic Disease, Izmır, Turkiye
| | - Mahmut Coker
- Ege University, Faculty of Medicine, Department of Inherited Metabolic Disease, Izmır, Turkiye
| | - Eser Yıldırım Sozmen
- Ege University, Faculty of Medicine, Department of Medical Biochemistry, Izmır, Turkiye.
| |
Collapse
|
6
|
Usenko T, Bezrukova A, Basharova K, Baydakova G, Shagimardanova E, Blatt N, Rizvanov A, Limankin O, Novitskiy M, Shnayder N, Izyumchenko A, Nikolaev M, Zabotina A, Lavrinova A, Kulabukhova D, Nasyrova R, Palchikova E, Zalutskaya N, Miliukhina I, Barbitoff Y, Glotov O, Glotov A, Taraskina A, Neznanov N, Zakharova E, Pchelina S. Altered Sphingolipid Hydrolase Activities and Alpha-Synuclein Level in Late-Onset Schizophrenia. Metabolites 2023; 14:30. [PMID: 38248833 PMCID: PMC10819534 DOI: 10.3390/metabo14010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Recent data described that patients with lysosomal storage disorders (LSDs) may have clinical schizophrenia (SCZ) features. Disruption of lipid metabolism in SCZ pathogenesis was found. Clinical features of schizophrenia (SCZ) have been demonstrated in patients with several lysosomal storage disorders (LSDs). Taking into account the critical role of lysosomal function for neuronal cells' lysosomal dysfunction could be proposed in SCZ pathogenesis. The current study analyzed lysosomal enzyme activities and the alpha-synuclein level in the blood of patients with late-onset SCZ. In total, 52 SCZ patients with late-onset SCZ, 180 sporadic Parkinson's disease (sPD) patients, and 176 controls were recruited. The enzymatic activity of enzymes associated with mucopolysaccharidosis (alpha-L-Iduronidase (IDUA)), glycogenosis (acid alpha-glucosidase (GAA)) and sphingolipidosis (galactosylceramidase (GALC), glucocerebrosidase (GCase), alpha-galactosidase (GLA), acid sphingomyelinase (ASMase)) and concentration of lysosphingolipids (hexosylsphingosine (HexSph), globotriaosylsphingosine (LysoGb3), and lysosphingomyelin (LysoSM)) were measured using LC-MS/MS. The alpha-synuclein level was estimated in magnetically separated CD45+ blood cells using the enzyme-linked immunosorbent assay (ELISA). Additionally, NGS analysis of 11 LSDs genes was conducted in 21 early-onset SCZ patients and 23 controls using the gene panel PGRNseq-NDD. Decreased ASMase, increased GLA activities, and increased HexSpn, LysoGb3, and LysoSM concentrations along with an accumulation of the alpha-synuclein level were observed in late-onset SCZ patients in comparison to the controls (p < 0.05). Four rare deleterious variants among LSDs genes causing mucopolysaccharidosis type I (IDUA (rs532731688, rs74385837) and type III (HGSNAT (rs766835582)) and sphingolipidosis (metachromatic leukodystrophy (ARSA (rs201251634)) were identified in five patients from the group of early-onset SCZ patients but not in the controls. Our findings supported the role of sphingolipid metabolism in SCZ pathogenesis. Aberrant enzyme activities and compounds of sphingolipids associated with ceramide metabolism may lead to accumulation of alpha-synuclein and may be critical in SCZ pathogenesis.
Collapse
Affiliation(s)
- Tatiana Usenko
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Anastasia Bezrukova
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Katerina Basharova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Galina Baydakova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
- Research Center for Medical Genetics, 115478 Moscow, Russia
| | - Elena Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.S.); (N.B.); (A.R.)
| | - Nataliya Blatt
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.S.); (N.B.); (A.R.)
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.S.); (N.B.); (A.R.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| | - Oleg Limankin
- Psychiatric Hospital No. 1 Named after P. P. Kashchenko, 195009 Saint Petersburg, Russia;
- North-Western Medical University Named after P. I.I. Mechnikov of the Ministry of Health of the Russian Federation, 191015 Saint Petersburg, Russia
| | - Maxim Novitskiy
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
| | - Natalia Shnayder
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
| | - Artem Izyumchenko
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Mikhail Nikolaev
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Anna Zabotina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Anna Lavrinova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Darya Kulabukhova
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Regina Nasyrova
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
| | - Ekaterina Palchikova
- V.M. Bekhterev National Medical Research Center Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (E.P.); (N.Z.)
| | - Natalia Zalutskaya
- V.M. Bekhterev National Medical Research Center Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (E.P.); (N.Z.)
| | - Irina Miliukhina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
- Institute of the Human Brain of RAS, 197022 Saint Petersburg, Russia
| | - Yury Barbitoff
- D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproductology, 199034 Saint Petersburg, Russia; (Y.B.); (O.G.); (A.G.)
- Cerbalab Ltd., 197136 Saint Petersburg, Russia
- Bioinformatics Institute, 197342 Saint Petersburg, Russia
| | - Oleg Glotov
- D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproductology, 199034 Saint Petersburg, Russia; (Y.B.); (O.G.); (A.G.)
- Cerbalab Ltd., 197136 Saint Petersburg, Russia
- Pediatric Research and Clinical Center of Infectious Diseases, 197022 Saint Petersburg, Russia
| | - Andrey Glotov
- D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproductology, 199034 Saint Petersburg, Russia; (Y.B.); (O.G.); (A.G.)
- School of Medicine, St. Petersburg State University, 199034 Saint Petersburg, Russia
| | - Anastasia Taraskina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Nikolai Neznanov
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
- V.M. Bekhterev National Medical Research Center Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (E.P.); (N.Z.)
| | | | - Sofya Pchelina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| |
Collapse
|
7
|
Taherzadeh M, Zhang E, Londono I, De Leener B, Wang S, Cooper JD, Kennedy TE, Morales CR, Chen Z, Lodygensky GA, Pshezhetsky AV. Severe central nervous system demyelination in Sanfilippo disease. Front Mol Neurosci 2023; 16:1323449. [PMID: 38163061 PMCID: PMC10756675 DOI: 10.3389/fnmol.2023.1323449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Chronic progressive neuroinflammation is a hallmark of neurological lysosomal storage diseases, including mucopolysaccharidosis III (MPS III or Sanfilippo disease). Since neuroinflammation is linked to white matter tract pathology, we analyzed axonal myelination and white matter density in the mouse model of MPS IIIC HgsnatP304L and post-mortem brain samples of MPS III patients. Methods Brain and spinal cord tissues of human MPS III patients, 6-month-old HgsnatP304L mice and age- and sex-matching wild type mice were analyzed by immunofluorescence to assess levels of myelin-associated proteins, primary and secondary storage materials, and levels of microgliosis. Corpus callosum (CC) region was studied by transmission electron microscopy to analyze axon myelination and morphology of oligodendrocytes and microglia. Mouse brains were analyzed ex vivo by high-filed MRI using Diffusion Basis Spectrum Imaging in Python-Diffusion tensor imaging algorithms. Results Analyses of CC and spinal cord tissues by immunohistochemistry revealed substantially reduced levels of myelin-associated proteins including Myelin Basic Protein, Myelin Associated Glycoprotein, and Myelin Oligodendrocyte Glycoprotein. Furthermore, ultrastructural analyses revealed disruption of myelin sheath organization and reduced myelin thickness in the brains of MPS IIIC mice and human MPS IIIC patients compared to healthy controls. Oligodendrocytes (OLs) in the CC of MPS IIIC mice were scarce, while examination of the remaining cells revealed numerous enlarged lysosomes containing heparan sulfate, GM3 ganglioside or "zebra bodies" consistent with accumulation of lipids and myelin fragments. In addition, OLs contained swollen mitochondria with largely dissolved cristae, resembling those previously identified in the dysfunctional neurons of MPS IIIC mice. Ex vivo Diffusion Basis Spectrum Imaging revealed compelling signs of demyelination (26% increase in radial diffusivity) and tissue loss (76% increase in hindered diffusivity) in CC of MPS IIIC mice. Discussion Our findings demonstrate an important role for white matter injury in the pathophysiology of MPS III. This study also defines specific parameters and brain regions for MRI analysis and suggests that it may become a crucial non-invasive method to evaluate disease progression and therapeutic response.
Collapse
Affiliation(s)
- Mahsa Taherzadeh
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Erjun Zhang
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Irene Londono
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Benjamin De Leener
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
- NeuroPoly Lab, Institute of Biomedical Engineering, Department of Computer Engineering and Software Engineering, École Polytechnique de Montréal, Montreal, QC, Canada
| | - Sophie Wang
- Pediatric Storage Disorders Laboratory (PSDL), Departments of Pediatrics, Genetics and Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory (PSDL), Departments of Pediatrics, Genetics and Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Timothy E. Kennedy
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Carlos R. Morales
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Zesheng Chen
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Gregory A. Lodygensky
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Alexey V. Pshezhetsky
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
8
|
Ellison S, Parker H, Bigger B. Advances in therapies for neurological lysosomal storage disorders. J Inherit Metab Dis 2023; 46:874-905. [PMID: 37078180 DOI: 10.1002/jimd.12615] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Lysosomal Storage Disorders (LSDs) are a diverse group of inherited, monogenic diseases caused by functional defects in specific lysosomal proteins. The lysosome is a cellular organelle that plays a critical role in catabolism of waste products and recycling of macromolecules in the body. Disruption to the normal function of the lysosome can result in the toxic accumulation of storage products, often leading to irreparable cellular damage and organ dysfunction followed by premature death. The majority of LSDs have no curative treatment, with many clinical subtypes presenting in early infancy and childhood. Over two-thirds of LSDs present with progressive neurodegeneration, often in combination with other debilitating peripheral symptoms. Consequently, there is a pressing unmet clinical need to develop new therapeutic interventions to treat these conditions. The blood-brain barrier is a crucial hurdle that needs to be overcome in order to effectively treat the central nervous system (CNS), adding considerable complexity to therapeutic design and delivery. Enzyme replacement therapy (ERT) treatments aimed at either direct injection into the brain, or using blood-brain barrier constructs are discussed, alongside more conventional substrate reduction and other drug-related therapies. Other promising strategies developed in recent years, include gene therapy technologies specifically tailored for more effectively targeting treatment to the CNS. Here, we discuss the most recent advances in CNS-targeted treatments for neurological LSDs with a particular emphasis on gene therapy-based modalities, such as Adeno-Associated Virus and haematopoietic stem cell gene therapy approaches that encouragingly, at the time of writing are being evaluated in LSD clinical trials in increasing numbers. If safety, efficacy and improved quality of life can be demonstrated, these therapies have the potential to be the new standard of care treatments for LSD patients.
Collapse
Affiliation(s)
- S Ellison
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - H Parker
- Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - B Bigger
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Jacques CED, Guerreiro G, Lopes FF, de Souza CFM, Giugliani R, Vargas CR. Alterations of Plasmatic Biomarkers of Neurodegeneration in Mucopolysaccharidosis Type II Patients Under Enzyme Replacement Therapy. Cell Biochem Biophys 2023; 81:533-542. [PMID: 37470932 DOI: 10.1007/s12013-023-01149-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2023] [Indexed: 07/21/2023]
Abstract
Mucopolysaccharidosis type II (MPS II) is a disorder caused by a deficient activity of iduronate-2-sulfatase, a lysosomal enzyme responsible for degrading glycosaminoglycans (GAGs). The abnormal storage of GAGs within lysosomes disrupts cellular homeostasis and leads to a severe symptomatology. Patients present neuropsychiatric impairment characterized by mental retardation and impaired cognition. The aim of this study was to quantify four neurodegeneration biomarkers in plasma: brain-derived neurotrophic factor (BDNF), platelet-derived growth factor (PDGF-AA), neural cell adhesion molecule (NCAM) and cathepsin-D, as well as to identify possible correlations with urinary GAGs in seven patients undergoing treatment with ERT (Elaprase® 0.5 mg/kg of body weight). Patients with both severe and attenuated forms of MPS II showed signs of neurodegeneration in neuroimaging exams. Patients have a decrease in BDNF and PDGF-AA concentrations, and an increase in NCAM level compared to controls. No alterations in cathepsin-D concentration were seen. GAGs levels were higher in patients than in controls, but no significant correlations between GAGs and biomarkers were observed. These results evidence that patients have neurodegeneration and that monitoring these biomarkers might be useful for assessing this process. To this date, this is the first work to analyze these plasmatic markers of neurodegeneration in patients.
Collapse
Affiliation(s)
- Carlos Eduardo Diaz Jacques
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, R. Ramiro Barcelos, 2600, Porto Alegre, RS, CEP 90035-003, Brazil.
- Serviço de Genética Médica, HCPA, R. Ramiro Barcelos, 2350, Porto Alegre, RS, CEP 90050-903, Brazil.
| | - Gilian Guerreiro
- Serviço de Genética Médica, HCPA, R. Ramiro Barcelos, 2350, Porto Alegre, RS, CEP 90050-903, Brazil
- Faculdade de Farmácia, UFRGS, Av. Ipiranga, 2752, Porto Alegre, RS, CEP 90610-000, Brazil
| | - Franciele Fatima Lopes
- Serviço de Genética Médica, HCPA, R. Ramiro Barcelos, 2350, Porto Alegre, RS, CEP 90050-903, Brazil
| | | | - Roberto Giugliani
- Serviço de Genética Médica, HCPA, R. Ramiro Barcelos, 2350, Porto Alegre, RS, CEP 90050-903, Brazil
- Departamento de Genética, Instituto de Biociências, UFRGS, Av. Bento Gonçalves, 9500, Porto Alegre, RS, CEP 91501-970, Brazil
| | - Carmen Regla Vargas
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, R. Ramiro Barcelos, 2600, Porto Alegre, RS, CEP 90035-003, Brazil.
- Serviço de Genética Médica, HCPA, R. Ramiro Barcelos, 2350, Porto Alegre, RS, CEP 90050-903, Brazil.
- Faculdade de Farmácia, UFRGS, Av. Ipiranga, 2752, Porto Alegre, RS, CEP 90610-000, Brazil.
| |
Collapse
|
10
|
Spiewak J, Doykov I, Papandreou A, Hällqvist J, Mills P, Clayton PT, Gissen P, Mills K, Heywood WE. New Perspectives in Dried Blood Spot Biomarkers for Lysosomal Storage Diseases. Int J Mol Sci 2023; 24:10177. [PMID: 37373322 DOI: 10.3390/ijms241210177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Dried blood spots (DBSs) biomarkers are convenient for monitoring for specific lysosomal storage diseases (LSDs), but they could have relevance for other LSDs. To determine the specificity and utility of glycosphingolipidoses biomarkers against other LSDs, we applied a multiplexed lipid liquid chromatography tandem mass spectrometry assay to a DBS cohort of healthy controls (n = 10) and Gaucher (n = 4), Fabry (n = 10), Pompe (n = 2), mucopolysaccharidosis types I-VI (n = 52), and Niemann-Pick disease type C (NPC) (n = 5) patients. We observed no complete disease specificity for any of the markers tested. However, comparison among the different LSDs highlighted new applications and perspectives of the existing biomarkers. We observed elevations in glucosylceramide isoforms in the NPC and Gaucher patients relative to the controls. In NPC, there was a greater proportion of C24 isoforms, giving a specificity of 96-97% for NPC, higher than 92% for the NPC biomarker N-palmitoyl-O-phosphocholineserine ratio to lyso-sphingomyelin. We also observed significantly elevated levels of lyso-dihexosylceramide in Gaucher and Fabry disease as well as elevated lyso-globotriaosylceramide (Lyso-Gb3) in Gaucher disease and the neuronopathic forms of Mucopolysaccharidoses. In conclusion, DBS glucosylceramide isoform profiling has increased the specificity for the detection of NPC, thereby improving diagnostic accuracy. Low levels of lyso-lipids can be observed in other LSDs, which may have implications in their disease pathogenesis.
Collapse
Affiliation(s)
- Justyna Spiewak
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
| | - Ivan Doykov
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
| | - Apostolos Papandreou
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
- Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
- Department of Neurology, Great Ormond Street Hospital for Children, London WC1N 3JH, UK
| | - Jenny Hällqvist
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
| | - Philippa Mills
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
| | - Peter T Clayton
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
| | - Paul Gissen
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
- Department of Metabolic Medicine, Great Ormond Street Hospital for Children, London WC1N 3JH, UK
| | - Kevin Mills
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
| | - Wendy E Heywood
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1 1EH, UK
| |
Collapse
|
11
|
Lopes N, Maia ML, Pereira CS, Mondragão-Rodrigues I, Martins E, Ribeiro R, Gaspar A, Aguiar P, Garcia P, Cardoso MT, Rodrigues E, Leão-Teles E, Giugliani R, Coutinho MF, Alves S, Macedo MF. Leukocyte Imbalances in Mucopolysaccharidoses Patients. Biomedicines 2023; 11:1699. [PMID: 37371793 DOI: 10.3390/biomedicines11061699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Mucopolysaccharidoses (MPSs) are rare inherited lysosomal storage diseases (LSDs) caused by deficient activity in one of the enzymes responsible for glycosaminoglycans lysosomal degradation. MPS II is caused by pathogenic mutations in the IDS gene, leading to deficient activity of the enzyme iduronate-2-sulfatase, which causes dermatan and heparan sulfate storage in the lysosomes. In MPS VI, there is dermatan sulfate lysosomal accumulation due to pathogenic mutations in the ARSB gene, leading to arylsulfatase B deficiency. Alterations in the immune system of MPS mouse models have already been described, but data concerning MPSs patients is still scarce. Herein, we study different leukocyte populations in MPS II and VI disease patients. MPS VI, but not MPS II patients, have a decrease percentage of natural killer (NK) cells and monocytes when compared with controls. No alterations were identified in the percentage of T, invariant NKT, and B cells in both groups of MPS disease patients. However, we discovered alterations in the naïve versus memory status of both helper and cytotoxic T cells in MPS VI disease patients compared to control group. Indeed, MPS VI disease patients have a higher frequency of naïve T cells and, consequently, lower memory T cell frequency than control subjects. Altogether, these results reveal MPS VI disease-specific alterations in some leukocyte populations, suggesting that the type of substrate accumulated and/or enzyme deficiency in the lysosome may have a particular effect on the normal cellular composition of the immune system.
Collapse
Affiliation(s)
- Nuno Lopes
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria L Maia
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
| | - Cátia S Pereira
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
- Cell Activation & Gene Expression (CAGE), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Inês Mondragão-Rodrigues
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
- Cell Activation & Gene Expression (CAGE), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| | - Esmeralda Martins
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de Santo António, 4099-001 Porto, Portugal
| | - Rosa Ribeiro
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de Santo António, 4099-001 Porto, Portugal
| | - Ana Gaspar
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar e Universitário Lisboa Norte (CHULN), 1649-035 Lisbon, Portugal
| | - Patrício Aguiar
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar e Universitário Lisboa Norte (CHULN), 1649-035 Lisbon, Portugal
- Faculdade de Medicina da Universidade de Lisboa, Universidade de Lisboa, 1649-190 Lisbon, Portugal
| | - Paula Garcia
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar e Universitário de Coimbra, Centro de Desenvolvimento da Criança, 3000-075 Coimbra, Portugal
| | - Maria Teresa Cardoso
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
| | - Esmeralda Rodrigues
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
| | - Elisa Leão-Teles
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
| | - Roberto Giugliani
- Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, DASA e Casa dos Raros, Porto Alegre 90610-150, Brazil
| | - Maria F Coutinho
- Research and Development Unit, Department of Genetics, INSA, 4000-055 Porto, Portugal
| | - Sandra Alves
- Research and Development Unit, Department of Genetics, INSA, 4000-055 Porto, Portugal
| | - M Fátima Macedo
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
- Cell Activation & Gene Expression (CAGE), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| |
Collapse
|
12
|
Bugiani M, Abbink TEM, Edridge AWD, van der Hoek L, Hillen AEJ, van Til NP, Hu‐A‐Ng GV, Breur M, Aiach K, Drevot P, Hocquemiller M, Laufer R, Wijburg FA, van der Knaap MS. Focal lesions following intracerebral gene therapy for mucopolysaccharidosis IIIA. Ann Clin Transl Neurol 2023; 10:904-917. [PMID: 37165777 PMCID: PMC10270249 DOI: 10.1002/acn3.51772] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/11/2023] [Accepted: 03/19/2023] [Indexed: 05/12/2023] Open
Abstract
OBJECTIVE Mucopolysaccharidosis type IIIA (MPSIIIA) caused by recessive SGSH variants results in sulfamidase deficiency, leading to neurocognitive decline and death. No disease-modifying therapy is available. The AAVance gene therapy trial investigates AAVrh.10 overexpressing human sulfamidase (LYS-SAF302) delivered by intracerebral injection in children with MPSIIIA. Post-treatment MRI monitoring revealed lesions around injection sites. Investigations were initiated in one patient to determine the cause. METHODS Clinical and MRI details were reviewed. Stereotactic needle biopsies of a lesion were performed; blood and CSF were sampled. All samples were used for viral studies. Immunohistochemistry, electron microscopy, and transcriptome analysis were performed on brain tissue of the patient and various controls. RESULTS MRI revealed focal lesions around injection sites with onset from 3 months after therapy, progression until 7 months post therapy with subsequent stabilization and some regression. The patient had transient slight neurological signs and is following near-normal development. No evidence of viral or immunological/inflammatory cause was found. Immunohistochemistry showed immature oligodendrocytes and astrocytes, oligodendrocyte apoptosis, strong intracellular and extracellular sulfamidase expression and hardly detectable intracellular or extracellular heparan sulfate. No activation of the unfolded protein response was found. INTERPRETATION Results suggest that intracerebral gene therapy with local sulfamidase overexpression leads to dysfunction of transduced cells close to injection sites, with extracellular spilling of lysosomal enzymes. This alters extracellular matrix composition, depletes heparan sulfate, impairs astrocyte and oligodendrocyte function, and causes cystic white matter degeneration at the site of highest gene expression. The AAVance trial results will reveal the potential benefit-risk ratio of this therapy.
Collapse
Affiliation(s)
- Marianna Bugiani
- Department of PathologyAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Amsterdam Leukodystrophy CenterAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - Truus E. M. Abbink
- Amsterdam Leukodystrophy CenterAmsterdam University Medical CentersAmsterdamThe Netherlands
- Department of Child NeurologyEmma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Arthur W. D. Edridge
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection PreventionAmsterdam University Medical Centers, University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Centre for Global Child HealthAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - Lia van der Hoek
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection PreventionAmsterdam University Medical Centers, University of AmsterdamAmsterdamThe Netherlands
| | - Anne E. J. Hillen
- Amsterdam Leukodystrophy CenterAmsterdam University Medical CentersAmsterdamThe Netherlands
- Department of Child NeurologyEmma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Niek P. van Til
- Amsterdam Leukodystrophy CenterAmsterdam University Medical CentersAmsterdamThe Netherlands
- Department of Child NeurologyEmma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Gino V. Hu‐A‐Ng
- Amsterdam Leukodystrophy CenterAmsterdam University Medical CentersAmsterdamThe Netherlands
- Department of Child NeurologyEmma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Marjolein Breur
- Amsterdam Leukodystrophy CenterAmsterdam University Medical CentersAmsterdamThe Netherlands
- Department of Child NeurologyEmma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | | | | | | | | | - Frits A. Wijburg
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center “Sphinx”Amsterdam University Medical Centers, Academic Medical CenterAmsterdamThe Netherlands
| | - Marjo S. van der Knaap
- Amsterdam Leukodystrophy CenterAmsterdam University Medical CentersAmsterdamThe Netherlands
- Department of Child NeurologyEmma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdam1081 HVThe Netherlands
| |
Collapse
|
13
|
De Pasquale V, Esposito A, Scerra G, Scarcella M, Ciampa M, Luongo A, D’Alonzo D, Guaragna A, D’Agostino M, Pavone LM. N-Substituted l-Iminosugars for the Treatment of Sanfilippo Type B Syndrome. J Med Chem 2023; 66:1790-1808. [PMID: 36696678 PMCID: PMC9923752 DOI: 10.1021/acs.jmedchem.2c01617] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Sanfilippo syndrome comprises a group of four genetic diseases due to the lack or decreased activity of enzymes involved in heparan sulfate (HS) catabolism. HS accumulation in lysosomes and other cellular compartments results in tissue and organ dysfunctions, leading to a wide range of clinical symptoms including severe neurodegeneration. To date, no approved treatments for Sanfilippo disease exist. Here, we report the ability of N-substituted l-iminosugars to significantly reduce substrate storage and lysosomal dysfunctions in Sanfilippo fibroblasts and in a neuronal cellular model of Sanfilippo B subtype. Particularly, we found that they increase the levels of defective α-N-acetylglucosaminidase and correct its proper sorting toward the lysosomal compartment. Furthermore, l-iminosugars reduce HS accumulation by downregulating protein levels of exostosin glycosyltransferases. These results highlight an interesting pharmacological potential of these glycomimetics in Sanfilippo syndrome, paving the way for the development of novel therapeutic approaches for the treatment of such incurable disease.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department
of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| | - Anna Esposito
- Department
of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Gianluca Scerra
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Melania Scarcella
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Mariangela Ciampa
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Antonietta Luongo
- AORN
Sant’Anna e San Sebastiano, Via F. Palasciano, 81100 Caserta, Italy
| | - Daniele D’Alonzo
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cintia, 80126 Napoli, Italy
| | - Annalisa Guaragna
- Department
of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy,
| | - Massimo D’Agostino
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy,
| | - Luigi Michele Pavone
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy,
| |
Collapse
|
14
|
Sandhoff R, Sandhoff K. Neuronal Ganglioside and Glycosphingolipid (GSL) Metabolism and Disease : Cascades of Secondary Metabolic Errors Can Generate Complex Pathologies (in LSDs). ADVANCES IN NEUROBIOLOGY 2023; 29:333-390. [PMID: 36255681 DOI: 10.1007/978-3-031-12390-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Glycosphingolipids (GSLs) are a diverse group of membrane components occurring mainly on the surfaces of mammalian cells. They and their metabolites have a role in intercellular communication, serving as versatile biochemical signals (Kaltner et al, Biochem J 476(18):2623-2655, 2019) and in many cellular pathways. Anionic GSLs, the sialic acid containing gangliosides (GGs), are essential constituents of neuronal cell surfaces, whereas anionic sulfatides are key components of myelin and myelin forming oligodendrocytes. The stepwise biosynthetic pathways of GSLs occur at and lead along the membranes of organellar surfaces of the secretory pathway. After formation of the hydrophobic ceramide membrane anchor of GSLs at the ER, membrane-spanning glycosyltransferases (GTs) of the Golgi and Trans-Golgi network generate cell type-specific GSL patterns for cellular surfaces. GSLs of the cellular plasma membrane can reach intra-lysosomal, i.e. luminal, vesicles (ILVs) by endocytic pathways for degradation. Soluble glycoproteins, the glycosidases, lipid binding and transfer proteins and acid ceramidase are needed for the lysosomal catabolism of GSLs at ILV-membrane surfaces. Inherited mutations triggering a functional loss of glycosylated lysosomal hydrolases and lipid binding proteins involved in GSL degradation cause a primary lysosomal accumulation of their non-degradable GSL substrates in lysosomal storage diseases (LSDs). Lipid binding proteins, the SAPs, and the various lipids of the ILV-membranes regulate GSL catabolism, but also primary storage compounds such as sphingomyelin (SM), cholesterol (Chol.), or chondroitin sulfate can effectively inhibit catabolic lysosomal pathways of GSLs. This causes cascades of metabolic errors, accumulating secondary lysosomal GSL- and GG- storage that can trigger a complex pathology (Breiden and Sandhoff, Int J Mol Sci 21(7):2566, 2020).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Konrad Sandhoff
- LIMES, c/o Kekule-Institute for Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany.
| |
Collapse
|
15
|
MPSI Manifestations and Treatment Outcome: Skeletal Focus. Int J Mol Sci 2022; 23:ijms231911168. [PMID: 36232472 PMCID: PMC9569890 DOI: 10.3390/ijms231911168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/23/2022] Open
Abstract
Mucopolysaccharidosis type I (MPSI) (OMIM #252800) is an autosomal recessive disorder caused by pathogenic variants in the IDUA gene encoding for the lysosomal alpha-L-iduronidase enzyme. The deficiency of this enzyme causes systemic accumulation of glycosaminoglycans (GAGs). Although disease manifestations are typically not apparent at birth, they can present early in life, are progressive, and include a wide spectrum of phenotypic findings. Among these, the storage of GAGs within the lysosomes disrupts cell function and metabolism in the cartilage, thus impairing normal bone development and ossification. Skeletal manifestations of MPSI are often refractory to treatment and severely affect patients’ quality of life. This review discusses the pathological and molecular processes leading to impaired endochondral ossification in MPSI patients and the limitations of current therapeutic approaches. Understanding the underlying mechanisms responsible for the skeletal phenotype in MPSI patients is crucial, as it could lead to the development of new therapeutic strategies targeting the skeletal abnormalities of MPSI in the early stages of the disease.
Collapse
|
16
|
Secondary Mitochondrial Dysfunction as a Cause of Neurodegenerative Dysfunction in Lysosomal Storage Diseases and an Overview of Potential Therapies. Int J Mol Sci 2022; 23:ijms231810573. [PMID: 36142486 PMCID: PMC9503973 DOI: 10.3390/ijms231810573] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/05/2022] Open
Abstract
Mitochondrial dysfunction has been recognised a major contributory factor to the pathophysiology of a number of lysosomal storage disorders (LSDs). The cause of mitochondrial dysfunction in LSDs is as yet uncertain, but appears to be triggered by a number of different factors, although oxidative stress and impaired mitophagy appear to be common inhibitory mechanisms shared amongst this group of disorders, including Gaucher’s disease, Niemann–Pick disease, type C, and mucopolysaccharidosis. Many LSDs resulting from defects in lysosomal hydrolase activity show neurodegeneration, which remains challenging to treat. Currently available curative therapies are not sufficient to meet patients’ needs. In view of the documented evidence of mitochondrial dysfunction in the neurodegeneration of LSDs, along with the reciprocal interaction between the mitochondrion and the lysosome, novel therapeutic strategies that target the impairment in both of these organelles could be considered in the clinical management of the long-term neurodegenerative complications of these diseases. The purpose of this review is to outline the putative mechanisms that may be responsible for the reported mitochondrial dysfunction in LSDs and to discuss the new potential therapeutic developments.
Collapse
|
17
|
Pan X, Taherzadeh M, Bose P, Heon-Roberts R, Nguyen AL, Xu T, Pará C, Yamanaka Y, Priestman DA, Platt FM, Khan S, Fnu N, Tomatsu S, Morales CR, Pshezhetsky AV. Glucosamine amends CNS pathology in mucopolysaccharidosis IIIC mouse expressing misfolded HGSNAT. J Exp Med 2022; 219:e20211860. [PMID: 35704026 PMCID: PMC9204472 DOI: 10.1084/jem.20211860] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/26/2022] [Accepted: 05/02/2022] [Indexed: 02/03/2023] Open
Abstract
The majority of mucopolysaccharidosis IIIC (MPS IIIC) patients have missense variants causing misfolding of heparan sulfate acetyl-CoA:α-glucosaminide N-acetyltransferase (HGSNAT), which are potentially treatable with pharmacological chaperones. To test this approach, we generated a novel HgsnatP304L mouse model expressing misfolded HGSNAT Pro304Leu variant. HgsnatP304L mice present deficits in short-term and working/spatial memory 2-4 mo earlier than previously described constitutive knockout Hgsnat-Geo mice. HgsnatP304L mice also show augmented severity of neuroimmune response, synaptic deficits, and neuronal storage of misfolded proteins and gangliosides compared with Hgsnat-Geo mice. Expression of misfolded human Pro311Leu HGSNAT protein in cultured hippocampal Hgsnat-Geo neurons further reduced levels of synaptic proteins. Memory deficits and majority of brain pathology were rescued in mice receiving HGSNAT chaperone, glucosamine. Our data for the first time demonstrate dominant-negative effects of misfolded HGSNAT Pro304Leu variant and show that they are treatable by oral administration of glucosamine. This suggests that patients affected with mutations preventing normal folding of the enzyme can benefit from chaperone therapy.
Collapse
Affiliation(s)
- Xuefang Pan
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
| | - Mahsa Taherzadeh
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Poulomee Bose
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
| | - Rachel Heon-Roberts
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Annie L.A. Nguyen
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
| | - TianMeng Xu
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
| | - Camila Pará
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
| | - Yojiro Yamanaka
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | | | | | - Shaukat Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Nidhi Fnu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Carlos R. Morales
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Alexey V. Pshezhetsky
- Centre Hospitalier Universitaire Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Wiesinger AM, Bigger B, Giugliani R, Scarpa M, Moser T, Lampe C, Kampmann C, Lagler FB. The Inflammation in the Cytopathology of Patients With Mucopolysaccharidoses- Immunomodulatory Drugs as an Approach to Therapy. Front Pharmacol 2022; 13:863667. [PMID: 35645812 PMCID: PMC9136158 DOI: 10.3389/fphar.2022.863667] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/27/2022] [Indexed: 01/31/2023] Open
Abstract
Mucopolysaccharidoses (MPS) are a group of lysosomal storage diseases (LSDs), characterized by the accumulation of glycosaminoglycans (GAGs). GAG storage-induced inflammatory processes are a driver of cytopathology in MPS and pharmacological immunomodulation can bring improvements in brain, cartilage and bone pathology in rodent models. This manuscript reviews current knowledge with regard to inflammation in MPS patients and provides hypotheses for the therapeutic use of immunomodulators in MPS. Thus, we aim to set the foundation for a rational repurposing of the discussed molecules to minimize the clinical unmet needs still remaining despite enzyme replacement therapy (ERT) and hematopoietic stem cell transplantation (HSCT).
Collapse
Affiliation(s)
- Anna-Maria Wiesinger
- Institute of Congenital Metabolic Diseases, Paracelsus Medical University, Salzburg, Austria
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, Udine, Italy
- *Correspondence: Anna-Maria Wiesinger,
| | - Brian Bigger
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, Udine, Italy
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Roberto Giugliani
- Department of Genetics, Medical Genetics Service and Biodiscovery Laboratory, HCPA, UFRGS, Porto Alegre, Brazil
| | - Maurizio Scarpa
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, Udine, Italy
- Regional Coordinating Center for Rare Diseases, University Hospital Udine, Udine, Italy
| | - Tobias Moser
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Christina Lampe
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, Udine, Italy
- Department of Child and Adolescent Medicine, Center of Rare Diseases, University Hospitals Giessen/Marburg, Giessen, Germany
| | - Christoph Kampmann
- Department of Pediatric Cardiology, University Hospital Mainz, Mainz, Germany
| | - Florian B. Lagler
- Institute of Congenital Metabolic Diseases, Paracelsus Medical University, Salzburg, Austria
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, Udine, Italy
| |
Collapse
|
19
|
Li W, Cologna SM. Mass spectrometry-based proteomics in neurodegenerative lysosomal storage disorders. Mol Omics 2022; 18:256-278. [PMID: 35343995 PMCID: PMC9098683 DOI: 10.1039/d2mo00004k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The major function of the lysosome is to degrade unwanted materials such as lipids, proteins, and nucleic acids; therefore, deficits of the lysosomal system can result in improper degradation and trafficking of these biomolecules. Diseases associated with lysosomal failure can be lethal and are termed lysosomal storage disorders (LSDs), which affect 1 in 5000 live births collectively. LSDs are inherited metabolic diseases caused by mutations in single lysosomal and non-lysosomal proteins and resulting in the subsequent accumulation of macromolecules within. Most LSD patients present with neurodegenerative clinical symptoms, as well as damage in other organs. The discovery of new biomarkers is necessary to understand and monitor these diseases and to track therapeutic progress. Over the past ten years, mass spectrometry (MS)-based proteomics has flourished in the biomarker studies in many diseases, including neurodegenerative, and more specifically, LSDs. In this review, biomarkers of disease pathophysiology and monitoring of LSDs revealed by MS-based proteomics are discussed, including examples from Niemann-Pick disease type C, Fabry disease, neuronal ceroid-lipofuscinoses, mucopolysaccharidosis, Krabbe disease, mucolipidosis, and Gaucher disease.
Collapse
Affiliation(s)
- Wenping Li
- Department of Chemistry, University of Illinois at Chicago, USA.
| | | |
Collapse
|
20
|
Pardridge WM. Blood-brain barrier delivery for lysosomal storage disorders with IgG-lysosomal enzyme fusion proteins. Adv Drug Deliv Rev 2022; 184:114234. [PMID: 35307484 DOI: 10.1016/j.addr.2022.114234] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/25/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
The majority of lysosomal storage diseases affect the brain. Treatment of the brain with intravenous enzyme replacement therapy is not successful, because the recombinant lysosomal enzymes do not cross the blood-brain barrier (BBB). Biologic drugs, including lysosomal enzymes, can be re-engineered for BBB delivery as IgG-enzyme fusion proteins. The IgG domain of the fusion protein is a monoclonal antibody directed against an endogenous receptor-mediated transporter at the BBB, such as the insulin receptor or the transferrin receptor. This receptor transports the IgG across the BBB, in parallel with the endogenous receptor ligand, and the IgG acts as a molecular Trojan horse to ferry into brain the lysosomal enzyme genetically fused to the IgG. The IgG-enzyme fusion protein is bi-functional and retains both high affinity binding for the BBB receptor, and high lysosomal enzyme activity. IgG-lysosomal enzymes are presently in clinical trials for treatment of the brain in Mucopolysaccharidosis.
Collapse
|
21
|
Splicing Modulation as a Promising Therapeutic Strategy for Lysosomal Storage Disorders: The Mucopolysaccharidoses Example. Life (Basel) 2022; 12:life12050608. [PMID: 35629276 PMCID: PMC9146820 DOI: 10.3390/life12050608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/07/2022] [Accepted: 04/15/2022] [Indexed: 11/17/2022] Open
Abstract
Over recent decades, the many functions of RNA have become more evident. This molecule has been recognized not only as a carrier of genetic information, but also as a specific and essential regulator of gene expression. Different RNA species have been identified and novel and exciting roles have been unveiled. Quite remarkably, this explosion of novel RNA classes has increased the possibility for new therapeutic strategies that tap into RNA biology. Most of these drugs use nucleic acid analogues and take advantage of complementary base pairing to either mimic or antagonize the function of RNAs. Among the most successful RNA-based drugs are those that act at the pre-mRNA level to modulate or correct aberrant splicing patterns, which are caused by specific pathogenic variants. This approach is particularly tempting for monogenic disorders with associated splicing defects, especially when they are highly frequent among affected patients worldwide or within a specific population. With more than 600 mutations that cause disease affecting the pre-mRNA splicing process, we consider lysosomal storage diseases (LSDs) to be perfect candidates for this type of approach. Here, we introduce the overall rationale and general mechanisms of splicing modulation approaches and highlight the currently marketed formulations, which have been developed for non-lysosomal genetic disorders. We also extensively reviewed the existing preclinical studies on the potential of this sort of therapeutic strategy to recover aberrant splicing and increase enzyme activity in our diseases of interest: the LSDs. Special attention was paid to a particular subgroup of LSDs: the mucopolysaccharidoses (MPSs). By doing this, we hoped to unveil the unique therapeutic potential of the use of this sort of approach for LSDs as a whole.
Collapse
|
22
|
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
|
23
|
Takahashi K, Le SQ, Kan SH, Jansen MJ, Dickson PI, Cooper JD. Neuropathology of murine Sanfilippo D syndrome. Mol Genet Metab 2021; 134:323-329. [PMID: 34844863 DOI: 10.1016/j.ymgme.2021.11.010] [Citation(s) in RCA: 4] [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: 10/01/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022]
Abstract
Sanfilippo D syndrome (mucopolysaccharidosis type IIID) is a lysosomal storage disorder caused by the deficiency of N-acetylglucosamine-6-sulfatase (GNS). A mouse model was generated by constitutive knockout of the Gns gene. We studied affected mice and controls at 12, 24, 36, and 48 weeks of age for neuropathological markers of disease in the somatosensory cortex, primary motor cortex, ventral posterior nuclei of the thalamus, striatum, hippocampus, and lateral and medial entorhinal cortex. We found significantly increased immunostaining for glial fibrillary associated protein (GFAP), CD68 (a marker of activated microglia), and lysosomal-associated membrane protein-1 (LAMP-1) in Sanfilippo D mice compared to controls at 12 weeks of age in all brain regions. Intergroup differences were marked for GFAP and CD68 staining, with levels in Sanfilippo D mice consistently above controls at all age groups. Intergroup differences in LAMP-1 staining were more pronounced in 12- and 24-week age groups compared to 36- and 48-week groups, as control animals showed some LAMP-1 staining at later timepoints in some brain regions. We also evaluated the somatosensory cortex, medial entorhinal cortex, reticular nucleus of the thalamus, medial amygdala, and hippocampal hilus for subunit c of mitochondrial ATP synthase (SCMAS). We found a progressive accumulation of SCMAS in most brain regions of Sanfilippo D mice compared to controls by 24 weeks of age. Cataloging the regional neuropathology of Sanfilippo D mice may aid in understanding the disease pathogenesis and designing preclinical studies to test brain-directed treatments.
Collapse
Affiliation(s)
- Keigo Takahashi
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Steven Q Le
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Shih-Hsin Kan
- Children's Hospital Orange County Research Institute, Orange, CA 92868, USA
| | - Matthew J Jansen
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Patricia I Dickson
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Jonathan D Cooper
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
24
|
Chhabra M, Doherty GG, See NW, Gandhi NS, Ferro V. From Cancer to COVID-19: A Perspective on Targeting Heparan Sulfate-Protein Interactions. CHEM REC 2021; 21:3087-3101. [PMID: 34145723 PMCID: PMC8441866 DOI: 10.1002/tcr.202100125] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/01/2021] [Indexed: 12/16/2022]
Abstract
Heparan sulfate (HS) is a complex, polyanionic polysaccharide ubiquitously expressed on cell surfaces and in the extracellular matrix. HS interacts with numerous proteins to mediate a vast array of biological and pathological processes. Inhibition of HS-protein interactions is thus an attractive approach for new therapeutic development for cancer and infectious diseases, including COVID-19; however, synthesis of well-defined native HS oligosaccharides remains challenging. This has aroused significant interest in the development of HS mimetics which are more synthetically tractable and have fewer side effects, such as undesired anticoagulant activity. This account provides a perspective on the design and synthesis of different classes of HS mimetics with useful properties, and the development of various assays and molecular modelling tools to progress our understanding of their interactions with HS-binding proteins.
Collapse
Affiliation(s)
- Mohit Chhabra
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| | - Gareth G. Doherty
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| | - Nicholas W. See
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| | - Neha S. Gandhi
- School of Chemistry and PhysicsQueensland University of Technology4000BrisbaneQLDAustralia
| | - Vito Ferro
- School of Chemistry and Molecular BiosciencesThe University of Queensland4072BrisbaneQLDAustralia
| |
Collapse
|
25
|
Arguello A, Meisner R, Thomsen ER, Nguyen HN, Ravi R, Simms J, Lo I, Speckart J, Holtzman J, Gill TM, Chan D, Cheng Y, Chiu CL, Dugas JC, Fang M, Lopez IA, Solanoy H, Tsogtbaatar B, Zhu Y, Bhalla A, Henne KR, Henry AG, Delucchi A, Costanzo S, Harris JM, Diaz D, Scearce-Levie K, Sanchez PE. Iduronate-2-sulfatase transport vehicle rescues behavioral and skeletal phenotypes in a mouse model of Hunter syndrome. JCI Insight 2021; 6:145445. [PMID: 34622797 PMCID: PMC8525587 DOI: 10.1172/jci.insight.145445] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 09/01/2021] [Indexed: 02/05/2023] Open
Abstract
Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disorder caused by deficiency of the iduronate-2-sulfatase (IDS) enzyme, resulting in cellular accumulation of glycosaminoglycans (GAGs) throughout the body. Treatment of MPS II remains a considerable challenge as current enzyme replacement therapies do not adequately control many aspects of the disease, including skeletal and neurological manifestations. We developed an IDS transport vehicle (ETV:IDS) that is engineered to bind to the transferrin receptor; this design facilitates receptor-mediated transcytosis of IDS across the blood-brain barrier and improves its distribution into the brain while maintaining distribution to peripheral tissues. Here we show that chronic systemic administration of ETV:IDS in a mouse model of MPS II reduced levels of peripheral and central nervous system GAGs, microgliosis, and neurofilament light chain, a biomarker of neuronal injury. Additionally, ETV:IDS rescued auricular and skeletal abnormalities when introduced in adult MPS II mice. These effects were accompanied by improvements in several neurobehavioral domains, including motor skills, sensorimotor gating, and learning and memory. Together, these results highlight the therapeutic potential of ETV:IDS for treating peripheral and central abnormalities in MPS II. DNL310, an investigational ETV:IDS molecule, is currently in clinical trials as a potential treatment for patients with MPS II.
Collapse
Affiliation(s)
- Annie Arguello
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - René Meisner
- Denali Therapeutics Inc., South San Francisco, California, USA
| | | | - Hoang N Nguyen
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Ritesh Ravi
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Jeffrey Simms
- Behavioral Core, Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Iris Lo
- Behavioral Core, Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Jessica Speckart
- Behavioral Core, Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Julia Holtzman
- Behavioral Core, Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Thomas M Gill
- Behavioral Core, Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Darren Chan
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Yuhsiang Cheng
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Chi-Lu Chiu
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Jason C Dugas
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Meng Fang
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Isabel A Lopez
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Hilda Solanoy
- Denali Therapeutics Inc., South San Francisco, California, USA
| | | | - Yuda Zhu
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Akhil Bhalla
- Denali Therapeutics Inc., South San Francisco, California, USA
| | - Kirk R Henne
- Denali Therapeutics Inc., South San Francisco, California, USA
| | | | | | - Simona Costanzo
- Denali Therapeutics Inc., South San Francisco, California, USA
| | | | - Dolores Diaz
- Denali Therapeutics Inc., South San Francisco, California, USA
| | | | | |
Collapse
|
26
|
Differences in MPS I and MPS II Disease Manifestations. Int J Mol Sci 2021; 22:ijms22157888. [PMID: 34360653 PMCID: PMC8345985 DOI: 10.3390/ijms22157888] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Mucopolysaccharidosis (MPS) type I and II are two closely related lysosomal storage diseases associated with disrupted glycosaminoglycan catabolism. In MPS II, the first step of degradation of heparan sulfate (HS) and dermatan sulfate (DS) is blocked by a deficiency in the lysosomal enzyme iduronate 2-sulfatase (IDS), while, in MPS I, blockage of the second step is caused by a deficiency in iduronidase (IDUA). The subsequent accumulation of HS and DS causes lysosomal hypertrophy and an increase in the number of lysosomes in cells, and impacts cellular functions, like cell adhesion, endocytosis, intracellular trafficking of different molecules, intracellular ionic balance, and inflammation. Characteristic phenotypical manifestations of both MPS I and II include skeletal disease, reflected in short stature, inguinal and umbilical hernias, hydrocephalus, hearing loss, coarse facial features, protruded abdomen with hepatosplenomegaly, and neurological involvement with varying functional concerns. However, a few manifestations are disease-specific, including corneal clouding in MPS I, epidermal manifestations in MPS II, and differences in the severity and nature of behavioral concerns. These phenotypic differences appear to be related to different ratios between DS and HS, and their sulfation levels. MPS I is characterized by higher DS/HS levels and lower sulfation levels, while HS levels dominate over DS levels in MPS II and sulfation levels are higher. The high presence of DS in the cornea and its involvement in the arrangement of collagen fibrils potentially causes corneal clouding to be prevalent in MPS I, but not in MPS II. The differences in neurological involvement may be due to the increased HS levels in MPS II, because of the involvement of HS in neuronal development. Current treatment options for patients with MPS II are often restricted to enzyme replacement therapy (ERT). While ERT has beneficial effects on respiratory and cardiopulmonary function and extends the lifespan of the patients, it does not significantly affect CNS manifestations, probably because the enzyme cannot pass the blood-brain barrier at sufficient levels. Many experimental therapies, therefore, aim at delivery of IDS to the CNS in an attempt to prevent neurocognitive decline in the patients.
Collapse
|
27
|
Bose SK, White BM, Kashyap MV, Dave A, De Bie FR, Li H, Singh K, Menon P, Wang T, Teerdhala S, Swaminathan V, Hartman HA, Jayachandran S, Chandrasekaran P, Musunuru K, Jain R, Frank DB, Zoltick P, Peranteau WH. In utero adenine base editing corrects multi-organ pathology in a lethal lysosomal storage disease. Nat Commun 2021; 12:4291. [PMID: 34257302 PMCID: PMC8277817 DOI: 10.1038/s41467-021-24443-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 06/09/2021] [Indexed: 01/19/2023] Open
Abstract
In utero base editing has the potential to correct disease-causing mutations before the onset of pathology. Mucopolysaccharidosis type I (MPS-IH, Hurler syndrome) is a lysosomal storage disease (LSD) affecting multiple organs, often leading to early postnatal cardiopulmonary demise. We assessed in utero adeno-associated virus serotype 9 (AAV9) delivery of an adenine base editor (ABE) targeting the Idua G→A (W392X) mutation in the MPS-IH mouse, corresponding to the common IDUA G→A (W402X) mutation in MPS-IH patients. Here we show efficient long-term W392X correction in hepatocytes and cardiomyocytes and low-level editing in the brain. In utero editing was associated with improved survival and amelioration of metabolic, musculoskeletal, and cardiac disease. This proof-of-concept study demonstrates the possibility of efficiently performing therapeutic base editing in multiple organs before birth via a clinically relevant delivery mechanism, highlighting the potential of this approach for MPS-IH and other genetic diseases.
Collapse
Affiliation(s)
- Sourav K Bose
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brandon M White
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Meghana V Kashyap
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Apeksha Dave
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Felix R De Bie
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Haiying Li
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kshitiz Singh
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Pallavi Menon
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tiankun Wang
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shiva Teerdhala
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Vishal Swaminathan
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Heather A Hartman
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sowmya Jayachandran
- Division of Pediatric Cardiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Center for Pulmonary Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Prashant Chandrasekaran
- Division of Pediatric Cardiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Center for Pulmonary Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kiran Musunuru
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rajan Jain
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David B Frank
- Division of Pediatric Cardiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Center for Pulmonary Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Philip Zoltick
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - William H Peranteau
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Douek AM, Amiri Khabooshan M, Henry J, Stamatis SA, Kreuder F, Ramm G, Änkö ML, Wlodkowic D, Kaslin J. An Engineered sgsh Mutant Zebrafish Recapitulates Molecular and Behavioural Pathobiology of Sanfilippo Syndrome A/MPS IIIA. Int J Mol Sci 2021; 22:ijms22115948. [PMID: 34073041 PMCID: PMC8197930 DOI: 10.3390/ijms22115948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/29/2022] Open
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA, Sanfilippo syndrome type A), a paediatric neurological lysosomal storage disease, is caused by impaired function of the enzyme N-sulfoglucosamine sulfohydrolase (SGSH) resulting in impaired catabolism of heparan sulfate glycosaminoglycan (HS GAG) and its accumulation in tissues. MPS IIIA represents a significant proportion of childhood dementias. This condition generally leads to patient death in the teenage years, yet no effective therapy exists for MPS IIIA and a complete understanding of the mechanisms of MPS IIIA pathogenesis is lacking. Here, we employ targeted CRISPR/Cas9 mutagenesis to generate a model of MPS IIIA in the zebrafish, a model organism with strong genetic tractability and amenity for high-throughput screening. The sgshΔex5-6 zebrafish mutant exhibits a complete absence of Sgsh enzymatic activity, leading to progressive accumulation of HS degradation products with age. sgshΔex5-6 zebrafish faithfully recapitulate diverse CNS-specific features of MPS IIIA, including neuronal lysosomal overabundance, complex behavioural phenotypes, and profound, lifelong neuroinflammation. We further demonstrate that neuroinflammation in sgshΔex5-6 zebrafish is largely dependent on interleukin-1β and can be attenuated via the pharmacological inhibition of Caspase-1, which partially rescues behavioural abnormalities in sgshΔex5-6 mutant larvae in a context-dependent manner. We expect the sgshΔex5-6 zebrafish mutant to be a valuable resource in gaining a better understanding of MPS IIIA pathobiology towards the development of timely and effective therapeutic interventions.
Collapse
Affiliation(s)
- Alon M. Douek
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Mitra Amiri Khabooshan
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Jason Henry
- Neurotoxicology Lab, School of Science (Biosciences), RMIT University, Bundoora, VIC 3083, Australia; (J.H.); (D.W.)
| | - Sebastian-Alexander Stamatis
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Florian Kreuder
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
| | - Georg Ramm
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, VIC 3800, Australia;
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Minna-Liisa Änkö
- Centre for Reproductive Health and Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia;
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Donald Wlodkowic
- Neurotoxicology Lab, School of Science (Biosciences), RMIT University, Bundoora, VIC 3083, Australia; (J.H.); (D.W.)
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (A.M.D.); (M.A.K.); (S.-A.S.); (F.K.)
- Correspondence: ; Tel.: +61-3-9902-9613; Fax: +61-3-9902-9729
| |
Collapse
|
29
|
Paget TL, Parkinson-Lawrence EJ, Trim PJ, Autilio C, Panchal MH, Koster G, Echaide M, Snel MF, Postle AD, Morrison JL, Pérez-Gil J, Orgeig S. Increased Alveolar Heparan Sulphate and Reduced Pulmonary Surfactant Amount and Function in the Mucopolysaccharidosis IIIA Mouse. Cells 2021; 10:849. [PMID: 33918094 PMCID: PMC8070179 DOI: 10.3390/cells10040849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disease with significant neurological and skeletal pathologies. Respiratory dysfunction is a secondary pathology contributing to mortality in MPS IIIA patients. Pulmonary surfactant is crucial to optimal lung function and has not been investigated in MPS IIIA. We measured heparan sulphate (HS), lipids and surfactant proteins (SP) in pulmonary tissue and bronchoalveolar lavage fluid (BALF), and surfactant activity in healthy and diseased mice (20 weeks of age). Heparan sulphate, ganglioside GM3 and bis(monoacylglycero)phosphate (BMP) were increased in MPS IIIA lung tissue. There was an increase in HS and a decrease in BMP and cholesteryl esters (CE) in MPS IIIA BALF. Phospholipid composition remained unchanged, but BALF total phospholipids were reduced (49.70%) in MPS IIIA. There was a reduction in SP-A, -C and -D mRNA, SP-D protein in tissue and SP-A, -C and -D protein in BALF of MPS IIIA mice. Captive bubble surfactometry showed an increase in minimum and maximum surface tension and percent surface area compression, as well as a higher compressibility and hysteresis in MPS IIIA surfactant upon dynamic cycling. Collectively these biochemical and biophysical changes in alveolar surfactant are likely to be detrimental to lung function in MPS IIIA.
Collapse
Affiliation(s)
- Tamara L. Paget
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| | - Emma J. Parkinson-Lawrence
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| | - Paul J. Trim
- Proteomics, Metabolomics and MS-Imaging Core Facility, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (P.J.T.); (M.F.S.)
| | - Chiara Autilio
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Madhuriben H. Panchal
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Grielof Koster
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Mercedes Echaide
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Marten F. Snel
- Proteomics, Metabolomics and MS-Imaging Core Facility, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (P.J.T.); (M.F.S.)
| | - Anthony D. Postle
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Janna L. Morrison
- Early Origins Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Jésus Pérez-Gil
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Sandra Orgeig
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| |
Collapse
|
30
|
McBride KL, Flanigan KM. Update in the Mucopolysaccharidoses. Semin Pediatr Neurol 2021; 37:100874. [PMID: 33892850 DOI: 10.1016/j.spen.2021.100874] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022]
Abstract
The mucopolysaccharidoses (MPS) are a genetically heterogenous group of enzyme deficiencies marked by accumulation of glycosaminoglycans in lysosomes leading to multisystem disease. Although significant therapeutic advances have been made for the MPS disorders, including recombinant enzyme replacement approaches, the neuronopathic features of MPS lack adequate treatment. Gene therapies, including adeno-associated virus vectors targeting the central nervous system, hold significant promise for this group of disorders. Optimal outcomes of all therapies will require early disease identification and treatment, ideally by newborn screening.
Collapse
Affiliation(s)
- Kim L McBride
- The Center for Cardiovascular Research and the Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital; and the Ohio State University, Columbus, OH; Department of Pediatrics, the Ohio State University, Columbus, OH.
| | - Kevin M Flanigan
- Department of Pediatrics, the Ohio State University, Columbus, OH; Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital; and the Ohio State University, Columbus, OH.
| |
Collapse
|
31
|
Arylsulfatase K inactivation causes mucopolysaccharidosis due to deficient glucuronate desulfation of heparan and chondroitin sulfate. Biochem J 2021; 477:3433-3451. [PMID: 32856704 DOI: 10.1042/bcj20200546] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 11/17/2022]
Abstract
Mucopolysaccharidoses comprise a group of rare metabolic diseases, in which the lysosomal degradation of glycosaminoglycans (GAGs) is impaired due to genetically inherited defects of lysosomal enzymes involved in GAG catabolism. The resulting intralysosomal accumulation of GAG-derived metabolites consequently manifests in neurological symptoms and also peripheral abnormalities in various tissues like liver, kidney, spleen and bone. As each GAG consists of differently sulfated disaccharide units, it needs a specific, but also partly overlapping set of lysosomal enzymes to accomplish their complete degradation. Recently, we identified and characterized the lysosomal enzyme arylsulfatase K (Arsk) exhibiting glucuronate-2-sulfatase activity as needed for the degradation of heparan sulfate (HS), chondroitin sulfate (CS) and dermatan sulfate (DS). In the present study, we investigated the physiological relevance of Arsk by means of a constitutive Arsk knockout mouse model. A complete lack of glucuronate desulfation was demonstrated by a specific enzyme activity assay. Arsk-deficient mice show, in an organ-specific manner, a moderate accumulation of HS and CS metabolites characterized by 2-O-sulfated glucuronate moieties at their non-reducing ends. Pathophysiological studies reflect a rather mild phenotype including behavioral changes. Interestingly, no prominent lysosomal storage pathology like bone abnormalities were detected. Our results from the Arsk mouse model suggest a new although mild form of mucopolysacharidose (MPS), which we designate MPS type IIB.
Collapse
|
32
|
Parenti G, Medina DL, Ballabio A. The rapidly evolving view of lysosomal storage diseases. EMBO Mol Med 2021; 13:e12836. [PMID: 33459519 PMCID: PMC7863408 DOI: 10.15252/emmm.202012836] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Lysosomal storage diseases are a group of metabolic disorders caused by deficiencies of several components of lysosomal function. Most commonly affected are lysosomal hydrolases, which are involved in the breakdown and recycling of a variety of complex molecules and cellular structures. The understanding of lysosomal biology has progressively improved over time. Lysosomes are no longer viewed as organelles exclusively involved in catabolic pathways, but rather as highly dynamic elements of the autophagic-lysosomal pathway, involved in multiple cellular functions, including signaling, and able to adapt to environmental stimuli. This refined vision of lysosomes has substantially impacted on our understanding of the pathophysiology of lysosomal disorders. It is now clear that substrate accumulation triggers complex pathogenetic cascades that are responsible for disease pathology, such as aberrant vesicle trafficking, impairment of autophagy, dysregulation of signaling pathways, abnormalities of calcium homeostasis, and mitochondrial dysfunction. Novel technologies, in most cases based on high-throughput approaches, have significantly contributed to the characterization of lysosomal biology or lysosomal dysfunction and have the potential to facilitate diagnostic processes, and to enable the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Giancarlo Parenti
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,SSM School for Advanced Studies, Federico II University, Naples, Italy
| |
Collapse
|
33
|
Chazeirat T, Denamur S, Bojarski KK, Andrault PM, Sizaret D, Zhang F, Saidi A, Tardieu M, Linhardt RJ, Labarthe F, Brömme D, Samsonov SA, Lalmanach G, Lecaille F. The abnormal accumulation of heparan sulfate in patients with mucopolysaccharidosis prevents the elastolytic activity of cathepsin V. Carbohydr Polym 2020; 253:117261. [PMID: 33278943 DOI: 10.1016/j.carbpol.2020.117261] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/30/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023]
Abstract
Mucopolysaccharidosis (MPS) are rare inherited diseases characterized by accumulation of lysosomal glycosaminoglycans, including heparan sulfate (HS). Patients exhibit progressive multi-visceral dysfunction and shortened lifespan mainly due to a severe cardiac/respiratory decline. Cathepsin V (CatV) is a potent elastolytic protease implicated in extracellular matrix (ECM) remodeling. Whether CatV is inactivated by HS in lungs from MPS patients remained unknown. Herein, CatV colocalized with HS in MPS bronchial epithelial cells. HS level correlated positively with the severity of respiratory symptoms and negatively to the overall endopeptidase activity of cysteine cathepsins. HS bound tightly to CatV and impaired its activity. Withdrawal of HS by glycosidases preserved exogenous CatV activity, while addition of Surfen, a HS antagonist, restored elastolytic CatV-like activity in MPS samples. Our data suggest that the pathophysiological accumulation of HS may be deleterious for CatV-mediated ECM remodeling and for lung tissue homeostasis, thus contributing to respiratory disorders associated to MPS diseases.
Collapse
Affiliation(s)
- Thibault Chazeirat
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes Protéolytiques Dans l'Inflammation", Tours, France.
| | - Sophie Denamur
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes Protéolytiques Dans l'Inflammation", Tours, France; Pediatric Department, Reference Center for Inborn Errors of Metabolism ToTeM, CHRU Tours, France.
| | | | - Pierre-Marie Andrault
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Damien Sizaret
- Anatomical Pathology and Cytology Department, Bretonneau Hospital, CHRU Tours, France.
| | - Fuming Zhang
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA.
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes Protéolytiques Dans l'Inflammation", Tours, France.
| | - Marine Tardieu
- Pediatric Department, Reference Center for Inborn Errors of Metabolism ToTeM, CHRU Tours, France.
| | - Robert J Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA.
| | - François Labarthe
- Pediatric Department, Reference Center for Inborn Errors of Metabolism ToTeM, CHRU Tours, France; INSERM, UMR 1069, Nutrition, Croissance et Cancer (N2C), Tours, France.
| | - Dieter Brömme
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes Protéolytiques Dans l'Inflammation", Tours, France.
| | - Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes Protéolytiques Dans l'Inflammation", Tours, France.
| |
Collapse
|
34
|
Bhalla A, Ravi R, Fang M, Arguello A, Davis SS, Chiu CL, Blumenfeld JR, Nguyen HN, Earr TK, Wang J, Astarita G, Zhu Y, Fiore D, Scearce-Levie K, Diaz D, Cahan H, Troyer MD, Harris JM, Escolar ML. Characterization of Fluid Biomarkers Reveals Lysosome Dysfunction and Neurodegeneration in Neuronopathic MPS II Patients. Int J Mol Sci 2020; 21:ijms21155188. [PMID: 32707880 PMCID: PMC7432645 DOI: 10.3390/ijms21155188] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/19/2020] [Accepted: 07/19/2020] [Indexed: 12/13/2022] Open
Abstract
Mucopolysaccharidosis type II is a lysosomal storage disorder caused by a deficiency of iduronate-2-sulfatase (IDS) and characterized by the accumulation of the primary storage substrate, glycosaminoglycans (GAGs). Understanding central nervous system (CNS) pathophysiology in neuronopathic MPS II (nMPS II) has been hindered by the lack of CNS biomarkers. Characterization of fluid biomarkers has been largely focused on evaluating GAGs in cerebrospinal fluid (CSF) and the periphery; however, GAG levels alone do not accurately reflect the broad cellular dysfunction in the brains of MPS II patients. We utilized a preclinical mouse model of MPS II, treated with a brain penetrant form of IDS (ETV:IDS) to establish the relationship between markers of primary storage and downstream pathway biomarkers in the brain and CSF. We extended the characterization of pathway and neurodegeneration biomarkers to nMPS II patient samples. In addition to the accumulation of CSF GAGs, nMPS II patients show elevated levels of lysosomal lipids, neurofilament light chain, and other biomarkers of neuronal damage and degeneration. Furthermore, we find that these biomarkers of downstream pathology are tightly correlated with heparan sulfate. Exploration of the responsiveness of not only CSF GAGs but also pathway and disease-relevant biomarkers during drug development will be crucial for monitoring disease progression, and the development of effective therapies for nMPS II.
Collapse
Affiliation(s)
- Akhil Bhalla
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
- Correspondence: (A.B.); (M.L.E.)
| | - Ritesh Ravi
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Meng Fang
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Annie Arguello
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Sonnet S. Davis
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Chi-Lu Chiu
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Jessica R. Blumenfeld
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
- Department of Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hoang N. Nguyen
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Timothy K. Earr
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Junhua Wang
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Giuseppe Astarita
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Yuda Zhu
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Damian Fiore
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Kimberly Scearce-Levie
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Dolores Diaz
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Heather Cahan
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Matthew D. Troyer
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Jeffrey M. Harris
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA; (R.R.); (M.F.); (A.A.); (S.S.D.); (C.-L.C.); (J.R.B.); (H.N.N.); (T.K.E.); (J.W.); (G.A.); (Y.Z.); (D.F.); (K.S.-L.); (D.D.); (H.C.); (M.D.T.); (J.M.H.)
| | - Maria L. Escolar
- Department of Pediatrics, Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
- Correspondence: (A.B.); (M.L.E.)
| |
Collapse
|
35
|
Pathogenesis of Mucopolysaccharidoses, an Update. Int J Mol Sci 2020; 21:ijms21072515. [PMID: 32260444 PMCID: PMC7178160 DOI: 10.3390/ijms21072515] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 01/08/2023] Open
Abstract
The recent advancements in the knowledge of lysosomal biology and function have translated into an improved understanding of the pathophysiology of mucopolysaccharidoses (MPSs). The concept that MPS manifestations are direct consequences of lysosomal engorgement with undegraded glycosaminoglycans (GAGs) has been challenged by new information on the multiple biological roles of GAGs and by a new vision of the lysosome as a signaling hub involved in many critical cellular functions. MPS pathophysiology is now seen as the result of a complex cascade of secondary events that lead to dysfunction of several cellular processes and pathways, such as abnormal composition of membranes and its impact on vesicle fusion and trafficking; secondary storage of substrates; impairment of autophagy; impaired mitochondrial function and oxidative stress; dysregulation of signaling pathways. The characterization of this cascade of secondary cellular events is critical to better understand the pathophysiology of MPS clinical manifestations. In addition, some of these pathways may represent novel therapeutic targets and allow for the development of new therapies for these disorders.
Collapse
|
36
|
Elevated LysoGb3 Concentration in the Neuronopathic Forms of Mucopolysaccharidoses. Diagnostics (Basel) 2020; 10:diagnostics10030155. [PMID: 32183018 PMCID: PMC7151490 DOI: 10.3390/diagnostics10030155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/02/2020] [Accepted: 03/07/2020] [Indexed: 12/31/2022] Open
Abstract
Mucopolysaccharidoses (MPSs) are a group of lysosomal storage disorders associated with impaired glycosaminoglycans (GAGs) catabolism. In MPS I, II, III, and VII, heparan sulfate (HS) cannot be degraded because of the lack of sufficient activity of the respective enzymes, and its accumulation in the brain causes neurological symptoms. Globotriaosylsphingosine (LysoGb3), the deacylated form of globotriaosylceramide (Gb3), is described as a highly sensitive biomarker for another lysosomal storage disease—Fabry disease. The connection between MPSs and LysoGb3 has not yet been established. This study included 36—MPS I, 15—MPS II, 25—MPS III, 26—MPS IV, and 14—MPS VI patients who were diagnosed by biochemical and molecular methods and a control group of 250 males and 250 females. The concentration of lysosphingolipids (LysoSLs) was measured in dried blood spots by high pressure liquid chromatography—tandem mass spectrometry. We have demonstrated that LysoGb3 concentration was significantly elevated (p < 0.0001) in untreated MPS I (3.07 + 1.55 ng/mL), MPS II (5.24 + 2.13 ng/mL), and MPS III (6.82 + 3.69 ng/mL) patients, compared to the control group (0.87 + 0.55 ng/mL). LysoGb3 level was normal in MPS VI and MPS IVA (1.26 + 0.39 and 0.99 + 0.38 ng/mL, respectively). Activity of α-galactosidase A (α-Gal A), an enzyme deficient in Fabry disease, was not, however, inhibited by heparan sulfate in vitro, indicating that an increase of LysoGb3 level in MPS I, MPS II, and MPS III is an indirect effect of stored MPSs rather than a direct result of impairment of degradation of this compound by HS. Our findings indicate some association of elevated LysoGb3 concentration with the neuronopathic forms of MPSs. The pathological mechanism of which is still to be studied.
Collapse
|
37
|
Viana GM, Gonzalez EA, Alvarez MMP, Cavalheiro RP, do Nascimento CC, Baldo G, D’Almeida V, de Lima MA, Pshezhetsky AV, Nader HB. Cathepsin B-associated Activation of Amyloidogenic Pathway in Murine Mucopolysaccharidosis Type I Brain Cortex. Int J Mol Sci 2020; 21:ijms21041459. [PMID: 32093427 PMCID: PMC7073069 DOI: 10.3390/ijms21041459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 01/28/2023] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is caused by genetic deficiency of α-l-iduronidase and impairment of lysosomal catabolism of heparan sulfate and dermatan sulfate. In the brain, these substrates accumulate in the lysosomes of neurons and glial cells, leading to neuroinflammation and neurodegeneration. Their storage also affects lysosomal homeostasis-inducing activity of several lysosomal proteases including cathepsin B (CATB). In the central nervous system, increased CATB activity has been associated with the deposition of amyloid plaques due to an alternative pro-amyloidogenic processing of the amyloid precursor protein (APP), suggesting a potential role of this enzyme in the neuropathology of MPS I. In this study, we report elevated levels of protein expression and activity of CATB in cortex tissues of 6-month-old MPS I (Idua -/- mice. Besides, increased CATB leakage from lysosomes to the cytoplasm of Idua -/- cortical pyramidal neurons was indicative of damaged lysosomal membranes. The increased CATB activity coincided with an elevated level of the 16-kDa C-terminal APP fragment, which together with unchanged levels of β-secretase 1 was suggestive for the role of this enzyme in the amyloidogenic APP processing. Neuronal accumulation of Thioflavin-S-positive misfolded protein aggregates and drastically increased levels of neuroinflammatory glial fibrillary acidic protein (GFAP)-positive astrocytes and CD11b-positive activated microglia were observed in Idua -/- cortex by confocal fluorescent microscopy. Together, our results point to the existence of a novel CATB-associated alternative amyloidogenic pathway in MPS I brain induced by lysosomal storage and potentially leading to neurodegeneration.
Collapse
Affiliation(s)
- Gustavo Monteiro Viana
- Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04044-020, Brazil; (M.M.P.A.); (R.P.C.); (H.B.N.)
- Correspondence: (G.M.V); (A.V.P); Tel.: +55-11-55764438 (ext. 1188) (G.M.V.); Tel.: +1 (514)-345-4931 (ext. 2736) (A.V.P.)
| | - Esteban Alberto Gonzalez
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil; (E.A.G.); (G.B.)
| | - Marcela Maciel Palacio Alvarez
- Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04044-020, Brazil; (M.M.P.A.); (R.P.C.); (H.B.N.)
| | - Renan Pelluzzi Cavalheiro
- Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04044-020, Brazil; (M.M.P.A.); (R.P.C.); (H.B.N.)
| | - Cinthia Castro do Nascimento
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04024-002, Brazil; (C.C.d.N.); (V.D.)
| | - Guilherme Baldo
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil; (E.A.G.); (G.B.)
| | - Vânia D’Almeida
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04024-002, Brazil; (C.C.d.N.); (V.D.)
| | - Marcelo Andrade de Lima
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK;
| | - Alexey V. Pshezhetsky
- Division of Medical Genetics, CHU Ste-Justine Research Centre, Montreal, QC H3T 1C5, Canada
- Correspondence: (G.M.V); (A.V.P); Tel.: +55-11-55764438 (ext. 1188) (G.M.V.); Tel.: +1 (514)-345-4931 (ext. 2736) (A.V.P.)
| | - Helena Bonciani Nader
- Department of Biochemistry, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04044-020, Brazil; (M.M.P.A.); (R.P.C.); (H.B.N.)
| |
Collapse
|