1
|
Shammas H, Kloster Fog C, Klein P, Koustrup A, Pedersen MT, Bie AS, Mickle T, Petersen NHT, Kirkegaard Jensen T, Guenther S. Mechanistic insights into arimoclomol mediated effects on lysosomal function in Niemann-pick type C disease. Mol Genet Metab 2025; 145:109103. [PMID: 40215728 DOI: 10.1016/j.ymgme.2025.109103] [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: 10/25/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Niemann-Pick disease type C (NPC) is an ultra-rare, fatal neurodegenerative disease. It is characterized by lysosomal dysfunction with cytotoxic accumulation of unesterified cholesterol and glycosphingolipids in lysosomes, which causes neurodegeneration and peripheral organ dysfunction. Arimoclomol, an orally available small molecule, is the first FDA-approved treatment for NPC when used in combination with miglustat. Here, we present the results of a series of in vitro studies performed to explore the pathways by which arimoclomol targets the fundamentals of NPC etiology. While the precise cellular interactions of arimoclomol remain unclear, the increased translocation of the transcription factors EB and E3 (TFEB and TFE3) from the cytosol to the nucleus is a key initial step for triggering a cascade of downstream events that can rescue cellular functions. Activation of TFEB and TFE3 raises the expression rates of coordinated lysosomal expression and regulation (CLEAR) genes including NPC1 that are essential for the regulation of lysosomal function. The subsequent upregulation of CLEAR network proteins combined with increased unfolded protein response activation was shown to enlarge the pool of matured NPC1 capable of reaching the lysosome to reduce cholesterol accumulation. By also amplifying expression of CLEAR genes associated with autophagy, arimoclomol has the potential to act on different pathways and improve cell viability independent of NPC1 protein levels and functionality. In summary, the findings presented illustrate how arimoclomol improves lysosomal function and potentially autophagy flux to decrease lipid burden in NPC patient fibroblasts.
Collapse
|
2
|
Presa M, Bailey RM, Ray S, Bailey L, Tata S, Murphy T, Piec PA, Combs H, Gray SJ, Lutz C. Preclinical use of a clinically-relevant scAAV9/SUMF1 vector for the treatment of multiple sulfatase deficiency. COMMUNICATIONS MEDICINE 2025; 5:29. [PMID: 39870870 PMCID: PMC11772666 DOI: 10.1038/s43856-025-00734-9] [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: 06/27/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Multiple Sulfatase Deficiency (MSD) is a rare inherited lysosomal storage disorder characterized by loss of function mutations in the SUMF1 gene that manifests as a severe pediatric neurological disease. There are no available targeted therapies for MSD. METHODS We engineered a viral vector (AAV9/SUMF1) to deliver working copies of the SUMF1 gene and tested the vector in Sumf1 knock out mice that generally display a median lifespan of 10 days. Mice were injected as pre-symptomatic neonates via intracerebroventricular administration, or as post-symptomatic juveniles via intrathecal alone or combination intrathecal and intravenous delivery. Cohorts were assessed for survival, behavioral outcomes, and post-mortem for sulfatase activity. RESULTS We show that treatment of neonates extends survival up to 1-year post-injection. Importantly, delivery of SUMF1 through cerebral spinal fluid at 7 days of age alleviates MSD symptoms. The treated mice show wide distribution of the SUMF1 gene, no signs of toxicity or neuropathy, improved vision and cardiac function, and no behavioral deficits. One-year post treatment, tissues show increased sulfatase activity, indicating functional SUMF1. Further, a GLP toxicology study conducted in rats demonstrates favorable overall safety of this approach. CONCLUSIONS These preclinical studies highlight the potential of our AAV9/SUMF1 vector, the design of which is directly translatable for clinical use, as a gene replacement therapy for MSD patients.
Collapse
Affiliation(s)
- Maximiliano Presa
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Rachel M Bailey
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Somdatta Ray
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Lauren Bailey
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Saurabh Tata
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Tara Murphy
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | | | - Harold Combs
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Steven J Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Cathleen Lutz
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA.
| |
Collapse
|
3
|
Pham V, Tricoli L, Hong X, Wongkittichote P, Castruccio Castracani C, Guerra A, Schlotawa L, Adang LA, Kuhs A, Cassidy MM, Kane O, Tsai E, Presa M, Lutz C, Rivella SB, Ahrens-Nicklas RC. Hematopoietic stem cell gene therapy improves outcomes in a clinically relevant mouse model of multiple sulfatase deficiency. Mol Ther 2024; 32:3829-3846. [PMID: 39169621 PMCID: PMC11573602 DOI: 10.1016/j.ymthe.2024.08.015] [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: 03/14/2024] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
Multiple sulfatase deficiency (MSD) is a severe, lysosomal storage disorder caused by pathogenic variants in the gene SUMF1, encoding the sulfatase modifying factor formylglycine-generating enzyme. Patients with MSD exhibit functional deficiencies in all cellular sulfatases. The inability of sulfatases to break down their substrates leads to progressive and multi-systemic complications in patients, similar to those seen in single-sulfatase disorders such as metachromatic leukodystrophy and mucopolysaccharidoses IIIA. Here, we aimed to determine if hematopoietic stem cell transplantation with ex vivo SUMF1 lentiviral gene therapy could improve outcomes in a clinically relevant mouse model of MSD. We first tested our approach in MSD patient-derived cells and found that our SUMF1 lentiviral vector improved protein expression, sulfatase activities, and glycosaminoglycan accumulation. In vivo, we found that our gene therapy approach rescued biochemical deficits, including sulfatase activity and glycosaminoglycan accumulation, in affected organs of MSD mice treated post-symptom onset. In addition, treated mice demonstrated improved neuroinflammation and neurocognitive function. Together, these findings suggest that SUMF1 HSCT-GT can improve both biochemical and functional disease markers in the MSD mouse.
Collapse
Affiliation(s)
- Vi Pham
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lucas Tricoli
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Xinying Hong
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Parith Wongkittichote
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Carlo Castruccio Castracani
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Amaliris Guerra
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lars Schlotawa
- Department of Pediatrics and Adolescent Medicine, University Medical Center Goettingen, 37075 Goettingen, Germany; Translational Neuroinflammation and Automated Microscopy, Fraunhofer Institute for Translational Medicine and Pharmacology, 37075 Goettingen, Germany
| | - Laura A Adang
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Amanda Kuhs
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Margaret M Cassidy
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Owen Kane
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Emily Tsai
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maximiliano Presa
- The Jackson Laboratory, Rare Disease Translational Center, Bar Harbor, ME 04609, USA
| | - Cathleen Lutz
- The Jackson Laboratory, Rare Disease Translational Center, Bar Harbor, ME 04609, USA
| | - Stefano B Rivella
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; RNA Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca C Ahrens-Nicklas
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
4
|
Presa M, Pham V, Ray S, Piec PA, Ryan J, Billings T, Coombs H, Schlotawa L, Lund T, Ahrens-Nicklas RC, Lutz C. Bone marrow transplantation increases sulfatase activity in somatic tissues in a multiple sulfatase deficiency mouse model. COMMUNICATIONS MEDICINE 2024; 4:215. [PMID: 39448727 PMCID: PMC11502872 DOI: 10.1038/s43856-024-00648-y] [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: 10/28/2023] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Multiple Sulfatase Deficiency (MSD) is an ultra-rare autosomal recessive disorder characterized by deficient enzymatic activity of all known sulfatases. MSD patients frequently carry two loss of function mutations in the SUMF1 gene, encoding a formylglycine-generating enzyme (FGE) that activates 17 different sulfatases. MSD patients show common features of other lysosomal diseases like mucopolysaccharidosis and metachromatic leukodystrophy, including neurologic impairments, developmental delay, and visceromegaly. There are currently no approved therapies for MSD patients. Hematopoietic stem cell transplant (HSCT) has been applied with success in the treatment of certain lysosomal diseases. In HSCT, donor-derived myeloid cells are a continuous source of active sulfatase enzymes that can be taken up by sulfatase-deficient host cells. Thus, HSCT could be a potential approach for the treatment of MSD. METHODS To test this hypothesis, we used a clinically relevant mouse model for MSD, B6-Sumf1(S153P/S153P) mice, engrafted with bone marrow cells, Sumf1+/+, from B6-PtprcK302E mice (CD45.1 immunoreactive). RESULTS After 10 months post-transplant, flow cytometric analysis shows an average of 90% of circulating leukocytes of donor origin (Sumf1(+/+)). Enzymatic activity for ARSA, ARSB, and SGSH is significantly increased in spleen of B6-Sumf1(S153P/S153P) recipient mice. In non-lymphoid organs, only liver and heart show a significant correction of sulfatase activity and GAG accumulation. Frequency of inflammatory cells and lysosomal pathology is significantly reduced in liver and heart, while no significant improvement is detected in brain. CONCLUSIONS Our results indicate that HSCT could be a suitable approach to treat MSD-pathology affecting peripheral organs, however that benefit to CNS pathology might be limited.
Collapse
Affiliation(s)
- Maximiliano Presa
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Vi Pham
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Division of Human Genetics, Department of Pediatrics, Metabolic Disease Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Somdatta Ray
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | | | - Jennifer Ryan
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Timothy Billings
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Harold Coombs
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Lars Schlotawa
- Department of Pediatrics and Adolescent Medicine, University Medical Center Goettingen, Goettingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology - Tranlational Neuroinflammation and Automated Microscopy, Goettingen, Germany
| | - Troy Lund
- Division of Hematology-Oncology and Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Rebecca C Ahrens-Nicklas
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Division of Human Genetics, Department of Pediatrics, Metabolic Disease Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Cathleen Lutz
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME, USA.
| |
Collapse
|
5
|
Lin S, Robson AG, Thompson DA, Stepien KM, Lachmann R, Footitt E, Czyz O, Chandrasekhar S, Schiff E, Iosifidis C, Black GC, Michaelides M, Mahroo OA, Arno G, Webster AR. Non-syndromic retinal dystrophy associated with biallelic variation of SUMF1 and reduced leukocyte sulfatase activity. Clin Genet 2024; 106:505-511. [PMID: 38863195 PMCID: PMC7616411 DOI: 10.1111/cge.14573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Biallelic variants in SUMF1 are associated with multiple sulfatase deficiency (MSD), a rare lysosomal storage disorder typically diagnosed in early infancy or childhood, marked by severe neurodegeneration and early mortality. We present clinical and molecular characterisation of three unrelated patients aged 13 to 58 years with milder clinical manifestations due to SUMF1 disease variants, including two adult patients presenting with apparent non-syndromic retinal dystrophy. Whole genome sequencing identified biallelic SUMF1 variants in all three patients; Patient 1 homozygous for a complex allele c.[290G>T;293T>A]; p.[(Gly97Val);(Val98Glu)], Patient 2 homozygous for c.866A>G; p.(Tyr289Cys), and Patient 3 compound heterozygous for c.726-1G>C and p.(Tyr289Cys). Electroretinography indicated a rod-cone dystrophy with additional possible inner retinal dysfunction in all three patients. Biochemical studies confirmed reduced, but not absent, sulfatase enzyme activity in the absence of extra-ocular disease (Patient 1) or only mild systemic disease (Patients 2, 3). These cases are suggestive that non-null SUMF1 genotypes can cause an attenuated clinical phenotype, including retinal dystrophy without systemic complications, in adulthood.
Collapse
Affiliation(s)
- Siying Lin
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Anthony G Robson
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Dorothy A Thompson
- Tony Kriss Visual Electrophysiology Unit, Department of Clinical and Academic Department of Ophthalmology, Sight and Sound Centre, Great Ormond Street Hospital for Children, London, UK
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Karolina M Stepien
- Adult Inherited Metabolic Disorders, Salford Royal Organisation, Northern Care Alliance NHS Foundation Trust, London, UK
| | - Robin Lachmann
- Charles Dent Metabolic Unit, National Hospital for Neurology and Neurosurgery, London, UK
| | - Emma Footitt
- Department of Metabolic Paediatrics, Great Ormond Street Hospital, London, UK
| | - Ola Czyz
- Department of Metabolic Paediatrics, Great Ormond Street Hospital, London, UK
| | | | - Elena Schiff
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Christos Iosifidis
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Graeme C Black
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Michel Michaelides
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Omar A Mahroo
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
- Department of Ophthalmology, St Thomas' Hospital, London, UK
| | - Gavin Arno
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
- Division of Research, Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Andrew R Webster
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
6
|
Antonsdottir IM, Creese B, Klei L, DeMichele‐Sweet MAA, Weamer EA, Garcia‐Gonzalez P, Marquie M, Boada M, Alarcón‐Martín E, Valero S, Liu Y, Hooli B, Aarsland D, Selbaek G, Bergh S, Rongve A, Saltvedt I, Skjellegrind HK, Engdahl B, Andreassen OA, Borroni B, Mecocci P, Wedatilake Y, Mayeux R, Foroud T, Ruiz A, Lopez OL, Kamboh MI, Ballard C, Devlin B, Lyketsos C, Sweet RA. Genetic associations with psychosis and affective disturbance in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12472. [PMID: 38784964 PMCID: PMC11114588 DOI: 10.1002/trc2.12472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 11/01/2023] [Accepted: 01/30/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Individuals with Alzheimer's disease (AD) commonly experience neuropsychiatric symptoms of psychosis (AD+P) and/or affective disturbance (depression, anxiety, and/or irritability, AD+A). This study's goal was to identify the genetic architecture of AD+P and AD+A, as well as their genetically correlated phenotypes. METHODS Genome-wide association meta-analysis of 9988 AD participants from six source studies with participants characterized for AD+P AD+A, and a joint phenotype (AD+A+P). RESULTS AD+P and AD+A were genetically correlated. However, AD+P and AD+A diverged in their genetic correlations with psychiatric phenotypes in individuals without AD. AD+P was negatively genetically correlated with bipolar disorder and positively with depressive symptoms. AD+A was positively correlated with anxiety disorder and more strongly correlated than AD+P with depressive symptoms. AD+P and AD+A+P had significant estimated heritability, whereas AD+A did not. Examination of the loci most strongly associated with the three phenotypes revealed overlapping and unique associations. DISCUSSION AD+P, AD+A, and AD+A+P have both shared and divergent genetic associations pointing to the importance of incorporating genetic insights into future treatment development. Highlights It has long been known that psychotic and affective symptoms are often comorbid in individuals diagnosed with Alzheimer's disease. Here we examined for the first time the genetic architecture underlying this clinical observation, determining that psychotic and affective phenotypes in Alzheimer's disease are genetically correlated.Nevertheless, psychotic and affective phenotypes in Alzheimer's disease diverged in their genetic correlations with psychiatric phenotypes assessed in individuals without Alzheimer's disease. Psychosis in Alzheimer's disease was negatively genetically correlated with bipolar disorder and positively with depressive symptoms, whereas the affective phenotypes in Alzheimer's disease were positively correlated with anxiety disorder and more strongly correlated than psychosis with depressive symptoms.Psychosis in Alzheimer's disease, and the joint psychotic and affective phenotype, had significant estimated heritability, whereas the affective in AD did not.Examination of the loci most strongly associated with the psychotic, affective, or joint phenotypes revealed overlapping and unique associations.
Collapse
|
7
|
Pham V, Sertori Finoti L, Cassidy MM, Maguire JA, Gagne AL, Waxman EA, French DL, King K, Zhou Z, Gelb MH, Wongkittichote P, Hong X, Schlotawa L, Davidson BL, Ahrens-Nicklas RC. A novel iPSC model reveals selective vulnerability of neurons in multiple sulfatase deficiency. Mol Genet Metab 2024; 141:108116. [PMID: 38161139 PMCID: PMC10951942 DOI: 10.1016/j.ymgme.2023.108116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
Multiple sulfatase deficiency (MSD) is an ultra-rare, inherited lysosomal storage disease caused by mutations in the gene sulfatase modifying factor 1 (SUMF1). MSD is characterized by the functional deficiency of all sulfatase enzymes, leading to the storage of sulfated substrates including glycosaminoglycans (GAGs), sulfolipids, and steroid sulfates. Patients with MSD experience severe neurological impairment, hearing loss, organomegaly, corneal clouding, cardiac valve disease, dysostosis multiplex, contractures, and ichthyosis. Here, we generated a novel human model of MSD by reprogramming patient peripheral blood mononuclear cells to establish an MSD induced pluripotent stem cell (iPSC) line (SUMF1 p.A279V). We also generated an isogenic control iPSC line by correcting the pathogenic variant with CRISPR/Cas9 gene editing. We successfully differentiated these iPSC lines into neural progenitor cells (NPCs) and NGN2-induced neurons (NGN2-iN) to model the neuropathology of MSD. Mature neuronal cells exhibited decreased SUMF1 gene expression, increased lysosomal stress, impaired neurite outgrowth and maturation, reduced sulfatase activities, and GAG accumulation. Interestingly, MSD iPSCs and NPCs did not exhibit as severe of phenotypes, suggesting that as neurons differentiate and mature, they become more vulnerable to loss of SUMF1. In summary, we demonstrate that this human iPSC-derived neuronal model recapitulates the cellular and biochemical features of MSD. These cell models can be used as tools to further elucidate the mechanisms of MSD pathology and for the development of therapeutics.
Collapse
Affiliation(s)
- Vi Pham
- The Children's Hospital of Philadelphia, Division of Human Genetics and Metabolism, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA; University of Pennsylvania, Perelman School of Medicine, Department of Pediatrics, Philadelphia, PA 19104, USA.
| | - Livia Sertori Finoti
- The Children's Hospital of Philadelphia, Division of Human Genetics and Metabolism, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA.
| | - Margaret M Cassidy
- The Children's Hospital of Philadelphia, Division of Human Genetics and Metabolism, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA; University of Pennsylvania, Perelman School of Medicine, Department of Pediatrics, Philadelphia, PA 19104, USA.
| | - Jean Ann Maguire
- The Children's Hospital of Philadelphia, Center for Cellular and Molecular Therapeutics, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA.
| | - Alyssa L Gagne
- The Children's Hospital of Philadelphia, Center for Cellular and Molecular Therapeutics, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA.
| | - Elisa A Waxman
- The Children's Hospital of Philadelphia, Center for Cellular and Molecular Therapeutics, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA; Center for Epilepsy and NeuroDevelopmental Disorders (ENDD), The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Deborah L French
- The Children's Hospital of Philadelphia, Center for Cellular and Molecular Therapeutics, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA; Center for Epilepsy and NeuroDevelopmental Disorders (ENDD), The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Department of Pathology and Laboratory Medicine, Philadelphia, PA 19104, USA.
| | - Kaitlyn King
- The Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Zitao Zhou
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Michael H Gelb
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA.
| | - Parith Wongkittichote
- The Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Xinying Hong
- University of Pennsylvania, Perelman School of Medicine, Department of Pathology and Laboratory Medicine, Philadelphia, PA 19104, USA; The Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Lars Schlotawa
- University Medical Center Goettingen, Department of Pediatrics and Adolescent Medicine, Robert-Koch-Str. 40, 37075 Goettingen, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology - Translational Neuroinflammation and Automated Microscopy, Robert-Koch-Str. 40, 37075, Goettingen, Germany.
| | - Beverly L Davidson
- The Children's Hospital of Philadelphia, Center for Cellular and Molecular Therapeutics, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA; University of Pennsylvania, Perelman School of Medicine, Department of Pathology and Laboratory Medicine, Philadelphia, PA 19104, USA.
| | - Rebecca C Ahrens-Nicklas
- The Children's Hospital of Philadelphia, Division of Human Genetics and Metabolism, Colket Translational Research Building, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA; University of Pennsylvania, Perelman School of Medicine, Department of Pediatrics, Philadelphia, PA 19104, USA.
| |
Collapse
|
8
|
Liang S, Gao H, He T, Li L, Zhang X, Zhao L, Chen J, Xie Y, Bao J, Gao Y, Dai E, Wang Y. Association between SUMF1 polymorphisms and COVID-19 severity. BMC Genom Data 2023; 24:34. [PMID: 37344788 DOI: 10.1186/s12863-023-01133-6] [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: 10/29/2022] [Accepted: 05/22/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Evidence shows that genetic factors play important roles in the severity of coronavirus disease 2019 (COVID-19). Sulfatase modifying factor 1 (SUMF1) gene is involved in alveolar damage and systemic inflammatory response. Therefore, we speculate that it may play a key role in COVID-19. RESULTS We found that rs794185 was significantly associated with COVID-19 severity in Chinese population, under the additive model after adjusting for gender and age (for C allele = 0.62, 95% CI = 0.44-0.88, P = 0.0073, logistic regression). And this association was consistent with this in European population Genetics Of Mortality In Critical Care (GenOMICC: OR for C allele = 0.94, 95% CI = 0.90-0.98, P = 0.0037). Additionally, we also revealed a remarkable association between rs794185 and the prothrombin activity (PTA) in subjects (P = 0.015, Generalized Linear Model). CONCLUSIONS In conclusion, our study for the first time identified that rs794185 in SUMF1 gene was associated with the severity of COVID-19.
Collapse
Affiliation(s)
- Shaohui Liang
- Department of Respiratory, Hebei Chest Hospital, Shijiazhuang, 050000, Hebei, China
| | - Huixia Gao
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, 050021, Hebei, China
| | - Tongxin He
- College of Plant Protection, Hunan Agricultural University, Changsha, 410128, Hunan, China
| | - Li Li
- Intensive Care Unit, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, 050021, Hebei, China
| | - Xin Zhang
- Department of Tuberculosis, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, 050021, Hebei, China
| | - Lei Zhao
- The Second Internal Medicine, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, 050021, Hebei, China
| | - Jie Chen
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yanyan Xie
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Jie Bao
- Department of Respiratory, Hebei Chest Hospital, Shijiazhuang, 050000, Hebei, China
| | - Yong Gao
- Department of Respiratory, Hebei Chest Hospital, Shijiazhuang, 050000, Hebei, China
| | - Erhei Dai
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, 050021, Hebei, China.
| | - Yuling Wang
- Department of Tuberculosis, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, 050021, Hebei, China.
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
9
|
Schlotawa L, Tyka K, Kettwig M, Ahrens‐Nicklas RC, Baud M, Berulava T, Brunetti‐Pierri N, Gagne A, Herbst ZM, Maguire JA, Monfregola J, Pena T, Radhakrishnan K, Schröder S, Waxman EA, Ballabio A, Dierks T, Fischer A, French DL, Gelb MH, Gärtner J. Drug screening identifies tazarotene and bexarotene as therapeutic agents in multiple sulfatase deficiency. EMBO Mol Med 2023; 15:e14837. [PMID: 36789546 PMCID: PMC9994482 DOI: 10.15252/emmm.202114837] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/09/2022] [Accepted: 01/09/2023] [Indexed: 02/16/2023] Open
Abstract
Multiple sulfatase deficiency (MSD, MIM #272200) results from pathogenic variants in the SUMF1 gene that impair proper function of the formylglycine-generating enzyme (FGE). FGE is essential for the posttranslational activation of cellular sulfatases. MSD patients display reduced or absent sulfatase activities and, as a result, clinical signs of single sulfatase disorders in a unique combination. Up to date therapeutic options for MSD are limited and mostly palliative. We performed a screen of FDA-approved drugs using immortalized MSD patient fibroblasts. Recovery of arylsulfatase A activity served as the primary readout. Subsequent analysis confirmed that treatment of primary MSD fibroblasts with tazarotene and bexarotene, two retinoids, led to a correction of MSD pathophysiology. Upon treatment, sulfatase activities increased in a dose- and time-dependent manner, reduced glycosaminoglycan content decreased and lysosomal position and size normalized. Treatment of MSD patient derived induced pluripotent stem cells (iPSC) differentiated into neuronal progenitor cells (NPC) resulted in a positive treatment response. Tazarotene and bexarotene act to ultimately increase the stability of FGE variants. The results lay the basis for future research on the development of a first therapeutic option for MSD patients.
Collapse
Affiliation(s)
- Lars Schlotawa
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| | - Karolina Tyka
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| | - Matthias Kettwig
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| | - Rebecca C Ahrens‐Nicklas
- Division of Human Genetics and MetabolismThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Matthias Baud
- School of Chemistry and Institute for Life SciencesUniversity of SouthamptonSouthamptonUK
| | - Tea Berulava
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
| | - Nicola Brunetti‐Pierri
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational MedicineUniversity of Naples Federico IINaplesItaly
| | - Alyssa Gagne
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | | | - Jean A Maguire
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Jlenia Monfregola
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational MedicineUniversity of Naples Federico IINaplesItaly
| | - Tonatiuh Pena
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
- Bioinformatics UnitGerman Centre for Neurodegenerative DiseasesGöttingenGermany
| | | | - Sophie Schröder
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
| | - Elisa A Waxman
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Andrea Ballabio
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational MedicineUniversity of Naples Federico IINaplesItaly
- Department of Molecular and Human Genetics and Neurological Research InstituteBaylor College of MedicineHoustonTXUSA
| | - Thomas Dierks
- Faculty of Chemistry, Biochemistry IBielefeld UniversityBielefeldGermany
| | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
- Department of Psychiatry and PsychotherapyUniversity Medical Center GöttingenGöttingenGermany
- Multiscale Bioimaging Cluster of Excellence, University Medical Center GöttingenUniversity of GöttingenGöttingenGermany
| | - Deborah L French
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Michael H Gelb
- Department of ChemistryUniversity of WashingtonSeattleWAUSA
| | - Jutta Gärtner
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| |
Collapse
|
10
|
Sorrentino NC, Presa M, Attanasio S, Cacace V, Sofia M, Zuberi A, Ryan J, Ray S, Petkovic I, Radhakrishnan K, Schlotawa L, Ballabio A, Lutz C, Brunetti-Pierri N. New mouse models with hypomorphic SUMF1 variants mimic attenuated forms of multiple sulfatase deficiency. J Inherit Metab Dis 2023; 46:335-347. [PMID: 36433920 PMCID: PMC10832386 DOI: 10.1002/jimd.12577] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/25/2022] [Accepted: 11/23/2022] [Indexed: 11/28/2022]
Abstract
Multiple sulfatase deficiency (MSD) is an ultrarare lysosomal storage disorder due to deficiency of all known sulfatases. MSD is caused by mutations in the Sulfatase Modifying Factor 1 (SUMF1) gene encoding the enzyme responsible for the post-translational modification and activation of all sulfatases. Most MSD patients carry hypomorph SUMF1 variants resulting in variable degrees of residual sulfatase activities. In contrast, Sumf1 null mice with complete deficiency in all sulfatase enzyme activities, have very short lifespan with significant pre-wean lethality, owing to a challenging preclinical model. To overcome this limitation, we genetically engineered and characterized in mice two commonly identified patient-based SUMF1 pathogenic variants, namely p.Ser153Pro and p.Ala277Val. These pathogenic missense variants correspond to variants detected in patients with attenuated MSD presenting with partial-enzyme deficiency and relatively less severe disease. These novel MSD mouse models have a longer lifespan and show biochemical and pathological abnormalities observed in humans. In conclusion, mice harboring the p.Ser153Pro or the p.Ala277Val variant mimic the attenuated MSD and are attractive preclinical models for investigation of pathogenesis and treatments for MSD.
Collapse
Affiliation(s)
- Nicolina Cristina Sorrentino
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Sergio Attanasio
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Vincenzo Cacace
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Martina Sofia
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | | | | | | | - Igor Petkovic
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | | | - Lars Schlotawa
- Department of Paediatrics and Adolescent Medicine, University Medical Centre Göttingen, Göttingen, Germany
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Pozzuoli (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
- Department of Translational Medicine, Federico II University, Naples, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| | | | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
- Department of Translational Medicine, Federico II University, Naples, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Abstract
Ca2+ is a universal second messenger that plays a wide variety of fundamental roles in cellular physiology. Thus, to warrant selective responses and to allow rapid mobilization upon specific stimuli, Ca2+ is accumulated in organelles to keep it at very low levels in the cytoplasm during resting conditions. Major Ca2+ storage organelles include the endoplasmic reticulum (ER), the mitochondria, and as recently demonstrated, the lysosome (Xu and Ren, Annu Rev Physiol 77:57-80, 2015). The importance of Ca2+ signaling deregulation in human physiology is underscored by its involvement in several human diseases, including lysosomal storage disorders, neurodegenerative disease and cancer (Shen et al., Nat Commun 3:731, 2012; Bae et al., J Neurosci 34:11485-11503, 2014). Recent evidence strongly suggests that lysosomal Ca2+ plays a major role in the regulation of lysosomal adaptation to nutrient availability through a lysosomal signaling pathway involving the lysosomal Ca2+ channel TRPML1 and the transcription factor TFEB, a master regulator for lysosomal function and autophagy (Sardiello et al., Science 325:473-477, 2009; Settembre et al., Science 332:1429-1433, 2011; Medina et al., Nat Cell Biol 17:288-299, 2015; Di Paola et al., Cell Calcium 69:112-121, 2018). Due to the tight relationship of this lysosomal Ca2+ channel and TFEB, in this chapter, we will focus on the role of the TRPML1/TFEB pathway in the regulation of lysosomal function and autophagy.
Collapse
Affiliation(s)
- Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.
| |
Collapse
|
12
|
Zhou Z, Zhou J, Liao J, Chen Z, Zheng Y. The Emerging Role of Astrocytic Autophagy in Central Nervous System Disorders. Neurochem Res 2022; 47:3697-3708. [PMID: 35960484 DOI: 10.1007/s11064-022-03714-w] [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: 03/13/2022] [Revised: 07/15/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Astrocytes act as "housekeeping cells" for maintaining cerebral homeostasis and play an important role in many disorders. Recent studies further highlight the contribution of autophagy to astrocytic functions, including astrogenesis, the astrocytic removal of neurotoxins or stressors, and astrocytic polarization. More importantly, genetic and pharmacological approaches have provided evidence that outlines the contributions of astrocytic autophagy to several brain disorders, including neurodegeneration, cerebral ischemia, and depression. In this study, we summarize the emerging role of autophagy in regulating astrocytic functions and discuss the contributions of astrocytic autophagy to different CNS disorders.
Collapse
Affiliation(s)
- Zhuchen Zhou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing Zhou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jie Liao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Asiimwe N, Al Mazid MF, Jeong YT, Lee J, Lee JS. The discovery of penta-peptides inhibiting the activity of the formylglycine-generating enzyme and their potential antibacterial effects against Mycobacterium tuberculosis. RSC Adv 2022; 12:18884-18888. [PMID: 35873338 PMCID: PMC9241360 DOI: 10.1039/d2ra03379h] [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: 05/31/2022] [Accepted: 06/16/2022] [Indexed: 11/21/2022] Open
Abstract
The formylglycine-generating enzyme is a key regulator that converts sulfatase into an active form. Despite its key role in many diseases, enzyme activity inhibitors have not yet been reported. In this study, we investigated penta-peptide ligands for FGE activity inhibition and discovered two hit peptides. In addition, the lead peptides also showed potential antibacterial effects in a Mycobacterium tuberculosis model.
Collapse
Affiliation(s)
| | | | - Yong Taek Jeong
- Department of Pharmacology, Korea University College of Medicine South Korea
| | - Juyong Lee
- Department of Chemistry, Kangwon National University South Korea
| | - Jun-Seok Lee
- Department of Pharmacology, Korea University College of Medicine South Korea
| |
Collapse
|
14
|
Fleming A, Xuan LZ, Sanchez-Elexpuru G, Williams SV, Windell D, Gelb MH, Herbst ZM, Schlotawa L, Rubinsztein DC. Unexpected Phenotype Reversion and Survival in a Zebrafish Model of Multiple Sulfatase Deficiency. Front Cell Dev Biol 2022; 10:843079. [PMID: 35721514 PMCID: PMC9203071 DOI: 10.3389/fcell.2022.843079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sulfatase deficiency (MSD) is a rare recessively inherited Mendelian disorder that manifests with developmental delay, neurodegeneration, skeletal deformities, facial dysmorphism, congenital growth retardation, and other clinical signs. The disorder is caused by mutations in the SUMF1 gene, which encodes the formylglycine-generating enzyme (FGE), and responsible for the activation of sulfatases. Mutations in SUMF1 result in reduced or absent FGE function with consequent compromised activities of its client sulfatases. This leads to an accumulation of enzyme substrates, such as glycosaminoglycans and sulfolipids, within lysosomes and subsequently impaired lysosome function and cellular pathology. Currently, there are no disease modifying therapeutic options for MSD patients, hence the need for more suitable animal models to investigate the disorder. Here, we describe the characterisation of a sumf1 null zebrafish model, which has negligible sulfatase activity. Our sumf1 -/- zebrafish model successfully recapitulates the pathology of MSD such as cranial malformation, altered bone development, an enlarged population of microglia, and growth retardation during early development but lacks early lethality of mouse Sumf1 -/- models. Notably, we provide evidence of recovery in MSD pathology during later developmental stages, resulting in homozygous mutants that are viable. Hence, our data suggest the possibility of a unique compensatory mechanism that allows the sumf1 -/- null zebrafish to survive better than human MSD patients and mouse Sumf1 -/- models.
Collapse
Affiliation(s)
- Angeleen Fleming
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom,UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom,*Correspondence: Angeleen Fleming, ; David C. Rubinsztein,
| | - Low Zhe Xuan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Gentzane Sanchez-Elexpuru
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom,UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Sarah V. Williams
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom,UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Dylan Windell
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom,UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Zackary M. Herbst
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Lars Schlotawa
- Department of Paediatrics and Adolescent Medicine, University Medical Centre Goettingen, Goettingen, Germany
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom,UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom,*Correspondence: Angeleen Fleming, ; David C. Rubinsztein,
| |
Collapse
|
15
|
DeChellis-Marks MR, Wei Y, Ding Y, Wolfe CM, Krivinko JM, MacDonald ML, Lopez OL, Sweet RA, Kofler J. Psychosis in Alzheimer's Disease Is Associated With Increased Excitatory Neuron Vulnerability and Post-transcriptional Mechanisms Altering Synaptic Protein Levels. Front Neurol 2022; 13:778419. [PMID: 35309563 PMCID: PMC8925864 DOI: 10.3389/fneur.2022.778419] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/04/2022] [Indexed: 12/01/2022] Open
Abstract
Alzheimer's disease with psychosis (AD+P) is a heritable phenotypic variant of the disease which is associated with more rapid cognitive deterioration compared to Alzheimer's disease without psychosis (AD-P). Cognitive decline in AD correlates with synapse loss, and our previous studies suggest that those with AD+P have a differentially affected synaptic proteome relative to those with AD-P. In this study, we utilized RNA-sequencing of dorsolateral prefrontal cortex (DLPFC) in a cohort of 80 AD cases to evaluate novel transcriptomic signatures that may confer risk of psychosis in AD. We found that AD+P was associated with a 9% reduction in excitatory neuron proportion compared to AD-P [Mean (SD) AD+P 0.295 (0.061); AD-P 0.324 (0.052), p = 0.026]. mRNA levels contributed only modestly to altered synaptic proteins in AD+P relative to AD-P. Instead, network analysis identified altered expression of gene modules from protein ubiquitination, unfolded protein response, eukaryotic initiation factor 2 (EIF2) signaling and endoplasmic reticulum stress pathways in AD+P. We previously found that neuropathologies account for ~18% of the variance in the occurrence of psychosis in AD. Further inclusion of cell type proportions and differentially expressed modules increased the percent of the variance in psychosis occurrence accounted for in our AD cohort to 67.5%.
Collapse
Affiliation(s)
| | - Yue Wei
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, United States
| | - Ying Ding
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, United States
| | - Cody M. Wolfe
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, United States
| | - Joshua M. Krivinko
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Matthew L. MacDonald
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Oscar L. Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
16
|
Maroofian R, Gubas A, Kaiyrzhanov R, Scala M, Hundallah K, Severino M, Abdel-Hamid MS, Rosenfeld JA, Ebrahimi-Fakhari D, Ali Z, Rahim F, Houlden H, Tooze SA, Alsaleh NS, Zaki MS. Homozygous missense WIPI2 variants cause a congenital disorder of autophagy with neurodevelopmental impairments of variable clinical severity and disease course. Brain Commun 2021; 3:fcab183. [PMID: 34557665 PMCID: PMC8453401 DOI: 10.1093/braincomms/fcab183] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 12/27/2022] Open
Abstract
WIPI2 is a member of the human WIPI protein family (seven-bladed b-propeller proteins binding phosphatidylinositols, PROPPINs), which play a pivotal role in autophagy and has been implicated in the pathogenesis of several neurological conditions. The homozygous WIPI2 variant c.745G>A; p.(Val249Met) (NM_015610.4) has recently been associated with a neurodevelopmental disorder in a single family. Using exome sequencing and Sanger segregation analysis, here, two novel homozygous WIPI2 variants [c.551T>G; p.(Val184Gly) and c.724C>T; p.(Arg242Trp) (NM_015610.4)] were identified in four individuals of two consanguineous families. Additionally, follow-up clinical data were sought from the previously reported family. Three non-ambulant affected siblings of the first family harbouring the p.(Val184Gly) missense variant presented with microcephaly, profound global developmental delay/intellectual disability, refractory infantile/childhood-onset epilepsy, progressive tetraplegia with joint contractures and dyskinesia. In contrast, the proband of the second family carrying the p.(Arg242Trp) missense variant, similar to the initially reported WIPI2 cases, presented with a milder phenotype, encompassing moderate intellectual disability, speech and visual impairment, autistic features, and an ataxic gait. Brain MR imaging in five patients showed prominent white matter involvement with a global reduction in volume, posterior corpus callosum hypoplasia, abnormal dentate nuclei and hypoplasia of the inferior cerebellar vermis. To investigate the functional impact of these novel WIPI2 variants, we overexpressed both in WIPI2-knockout HEK293A cells. In comparison to wildtype, expression of the Val166Gly WIPI2b mutant resulted in a deficient rescue of LC3 lipidation whereas Arg224Trp mutant increased LC3 lipidation, in line with the previously reported Val231Met variant. These findings support a dysregulation of the early steps of the autophagy pathway. Collectively, our findings provide evidence that biallelic WIPI2 variants cause a neurodevelopmental disorder of variable severity and disease course. Our report expands the clinical spectrum and establishes WIPI2-related disorder as a congenital disorders of autophagy.
Collapse
Grants
- P50 HD105351 NICHD NIH HHS
- MR/S005021/1 Medical Research Council
- FC001187 Arthritis Research UK
- MR/S01165X/1 Medical Research Council
- Wellcome Trust
- G0601943 Medical Research Council
- R01 NS107428 NINDS NIH HHS
- Wellcome Trust [WT093205MA, WT104033AIA and the Synaptopathies Strategic Award, 165908]
- University of Maryland
- National Institute of Neurological Disorders and Stroke (NINDS) (R01NS107428) (to S.R.)
- Medical Research Council (MR/S01165X/1, MR/S005021/1, G0601943)
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, Rosetree Trust, Ataxia UK, Multiple System Atrophy Trust, Brain Research United Kingdom, Sparks Great Ormond Street Hospital Charity, Muscular Dystrophy United Kingdom (MDUK)
- Muscular Dystrophy Association (MDA USA)
- Cancer Research United Kingdom (FC001187)
- United Kingdom Medical Research Council (FC001187) and the Wellcome Trust (FC001187)
Collapse
Affiliation(s)
- Reza Maroofian
- Department of Neuromuscular Diseases, University College London, Queen Square, Institute of Neurology, London, UK
| | - Andrea Gubas
- Goethe University Medical School, University Hospital, 60590 Frankfurt am Main, Germany
| | - Rauan Kaiyrzhanov
- Department of Neuromuscular Diseases, University College London, Queen Square, Institute of Neurology, London, UK
| | - Marcello Scala
- Department of Neuromuscular Diseases, University College London, Queen Square, Institute of Neurology, London, UK
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini, Genoa, Italy
| | - Khalid Hundallah
- Division of Neurology, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | | | - Mohamed S Abdel-Hamid
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Baylor Genetics Laboratories, Houston, TX, USA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zahir Ali
- Laboratory for Genome Engineering, Division of Biological Sciences, 4700 King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia
| | - Fazal Rahim
- Department of Physiology, Bacha Khan Medical College, Mardan, Pakistan
| | - Henry Houlden
- Department of Neuromuscular Diseases, University College London, Queen Square, Institute of Neurology, London, UK
| | - Sharon A Tooze
- The Francis Crick Institute, Molecular Cell Biology of Autophagy, NW1 1AT London, UK
| | - Norah S Alsaleh
- Division of Medical Genetics and Metabolic Medicine, Department of Pediatrics, Prince Sultan Military Medical City, 11159 Riyadh, Saudi Arabia
| | - Maha S Zaki
- Human Genetics and Genome Research Division, Clinical Genetics Department, National Research Centre, 12311 Cairo, Egypt
| |
Collapse
|
17
|
Liaw K, Reddy R, Sharma A, Li J, Chang M, Sharma R, Salazar S, Kannan S, Kannan RM. Targeted systemic dendrimer delivery of CSF-1R inhibitor to tumor-associated macrophages improves outcomes in orthotopic glioblastoma. Bioeng Transl Med 2021; 6:e10205. [PMID: 34027092 PMCID: PMC8126814 DOI: 10.1002/btm2.10205] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/04/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma is the most common and aggressive form of primary brain cancer, with median survival of 16-20 months and a 5-year survival rates of <5%. Recent advances in immunotherapies have shown that addressing the tumor immune profile by targeting the colony-stimulating factor 1 (CSF-1) signaling pathway of tumor-associated macrophages (TAMs) has the potential to improve glioblastoma therapy. However, such therapies have shown limited successes in clinical translation partially due to lack of specific cell targeting in solid tumors and systemic toxicity. In this study, we present a novel hydroxyl dendrimer-mediated immunotherapy to deliver CSF-1R inhibitor BLZ945 (D-BLZ) from systemic administration selectively to TAMs in glioblastoma brain tumors to repolarize the tumor immune environment in a localized manner. We show that conjugation of BLZ945 to dendrimers enables sustained release in intracellular and intratumor conditions. We demonstrate that a single systemic dose of D-BLZ targeted to TAMs decreases pro-tumor expression in TAMs and promotes cytotoxic T cell infiltration, resulting in prolonged survival and ameliorated disease burden compared to free BLZ945. Our results demonstrate that dendrimer-drug conjugates can facilitate specific, localized manipulation of tumor immune responses from systemic administration by delivering immunotherapies selectively to TAMs, thereby improving therapeutic efficacy while reducing off-target effects.
Collapse
Affiliation(s)
- Kevin Liaw
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Center for Nanomedicine, Department of OphthalmologyJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Rajsekhar Reddy
- Center for Nanomedicine, Department of OphthalmologyJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Anjali Sharma
- Center for Nanomedicine, Department of OphthalmologyJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Jiangyu Li
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Michelle Chang
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Rishi Sharma
- Center for Nanomedicine, Department of OphthalmologyJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Sebastian Salazar
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Sujatha Kannan
- Anesthesiology and Critical Care MedicineJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Rangaramanujam M. Kannan
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Center for Nanomedicine, Department of OphthalmologyJohns Hopkins MedicineBaltimoreMarylandUSA
| |
Collapse
|
18
|
Arsenault EJ, McGill CM, Barth BM. Sphingolipids as Regulators of Neuro-Inflammation and NADPH Oxidase 2. Neuromolecular Med 2021; 23:25-46. [PMID: 33547562 PMCID: PMC9020407 DOI: 10.1007/s12017-021-08646-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
Neuro-inflammation accompanies numerous neurological disorders and conditions where it can be associated with a progressive neurodegenerative pathology. In a similar manner, alterations in sphingolipid metabolism often accompany or are causative features in degenerative neurological conditions. These include dementias, motor disorders, autoimmune conditions, inherited metabolic disorders, viral infection, traumatic brain and spinal cord injury, psychiatric conditions, and more. Sphingolipids are major regulators of cellular fate and function in addition to being important structural components of membranes. Their metabolism and signaling pathways can also be regulated by inflammatory mediators. Therefore, as certain sphingolipids exert distinct and opposing cellular roles, alterations in their metabolism can have major consequences. Recently, regulation of bioactive sphingolipids by neuro-inflammatory mediators has been shown to activate a neuronal NADPH oxidase 2 (NOX2) that can provoke damaging oxidation. Therefore, the sphingolipid-regulated neuronal NOX2 serves as a mechanistic link between neuro-inflammation and neurodegeneration. Moreover, therapeutics directed at sphingolipid metabolism or the sphingolipid-regulated NOX2 have the potential to alleviate neurodegeneration arising out of neuro-inflammation.
Collapse
Affiliation(s)
- Emma J Arsenault
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, USA
| | - Colin M McGill
- Department of Chemistry, University of Alaska Anchorage, Anchorage, AK, 99508, USA
| | - Brian M Barth
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, USA.
| |
Collapse
|
19
|
Al-Hazmi NA, Alhazzazi TY, Bukhary SMN, Weekes D, McDonald F, Hill P, Grigoriadis A, Al-Dabbagh RA. Mechanisms Involved in Ceramide-Induced Autophagy in Osteoblasts. ARCHIVES OF PHARMACY PRACTICE 2021. [DOI: 10.51847/b8jia53a5q] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
20
|
Yan W, Sun W, Fan J, Wang H, Han S, Li J, Yin Y. Sirt1-ROS-TRAF6 Signaling-Induced Pyroptosis Contributes to Early Injury in Ischemic Mice. Neurosci Bull 2020; 36:845-859. [PMID: 32253651 PMCID: PMC7410906 DOI: 10.1007/s12264-020-00489-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/04/2019] [Indexed: 01/01/2023] Open
Abstract
Stroke is an acute cerebro-vascular disease with high incidence and poor prognosis, most commonly ischemic in nature. In recent years, increasing attention has been paid to inflammatory reactions as symptoms of a stroke. However, the role of inflammation in stroke and its underlying mechanisms require exploration. In this study, we evaluated the inflammatory reactions induced by acute ischemia and found that pyroptosis occurred after acute ischemia both in vivo and in vitro, as determined by interleukin-1β, apoptosis-associated speck-like protein, and caspase-1. The early inflammation resulted in irreversible ischemic injury, indicating that it deserves thorough investigation. Meanwhile, acute ischemia decreased the Sirtuin 1 (Sirt1) protein levels, and increased the TRAF6 (TNF receptor associated factor 6) protein and reactive oxygen species (ROS) levels. In further exploration, both Sirt1 suppression and TRAF6 activation were found to contribute to this pyroptosis. Reduced Sirt1 levels were responsible for the production of ROS and increased TRAF6 protein levels after ischemic exposure. Moreover, N-acetyl-L-cysteine, an ROS scavenger, suppressed the TRAF6 accumulation induced by oxygen-glucose deprivation via suppression of ROS bursts. These phenomena indicate that Sirt1 is upstream of ROS, and ROS bursts result in increased TRAF6 levels. Further, the activation of Sirt1 during the period of ischemia reduced ischemia-induced injury after 72 h of reperfusion in mice with middle cerebral artery occlusion. In sum, these results indicate that pyroptosis-dependent machinery contributes to the neural injury during acute ischemia via the Sirt1-ROS-TRAF6 signaling pathway. We propose that inflammatory reactions occur soon after oxidative stress and are detrimental to neuronal survival; this provides a promising therapeutic target against ischemic injuries such as a stroke.
Collapse
Affiliation(s)
- Weijie Yan
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Wei Sun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jiahui Fan
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Haiqing Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Song Han
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Junfa Li
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Yanling Yin
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
21
|
Schlotawa L, Adang LA, Radhakrishnan K, Ahrens-Nicklas RC. Multiple Sulfatase Deficiency: A Disease Comprising Mucopolysaccharidosis, Sphingolipidosis, and More Caused by a Defect in Posttranslational Modification. Int J Mol Sci 2020; 21:E3448. [PMID: 32414121 PMCID: PMC7279497 DOI: 10.3390/ijms21103448] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
Multiple sulfatase deficiency (MSD, MIM #272200) is an ultra-rare disease comprising pathophysiology and clinical features of mucopolysaccharidosis, sphingolipidosis and other sulfatase deficiencies. MSD is caused by impaired posttranslational activation of sulfatases through the formylglycine generating enzyme (FGE) encoded by the sulfatase modifying factor 1 (SUMF1) gene, which is mutated in MSD. FGE is a highly conserved, non-redundant ER protein that activates all cellular sulfatases by oxidizing a conserved cysteine in the active site of sulfatases that is necessary for full catalytic activity. SUMF1 mutations result in unstable, degradation-prone FGE that demonstrates reduced or absent catalytic activity, leading to decreased activity of all sulfatases. As the majority of sulfatases are localized to the lysosome, loss of sulfatase activity induces lysosomal storage of glycosaminoglycans and sulfatides and subsequent cellular pathology. MSD patients combine clinical features of all single sulfatase deficiencies in a systemic disease. Disease severity classifications distinguish cases based on age of onset and disease progression. A genotype- phenotype correlation has been proposed, biomarkers like excreted storage material and residual sulfatase activities do not correlate well with disease severity. The diagnosis of MSD is based on reduced sulfatase activities and detection of mutations in SUMF1. No therapy exists for MSD yet. This review summarizes the unique FGE/ sulfatase physiology, pathophysiology and clinical aspects in patients and their care and outlines future perspectives in MSD.
Collapse
Affiliation(s)
- Lars Schlotawa
- Department of Paediatrics and Adolescent Medicine, University Medical Centre Goettingen, 37075 Goettingen, Germany
| | - Laura A. Adang
- Division of Child Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | | | - Rebecca C. Ahrens-Nicklas
- Division of Human Genetics and Metabolism, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
Favret JM, Weinstock NI, Feltri ML, Shin D. Pre-clinical Mouse Models of Neurodegenerative Lysosomal Storage Diseases. Front Mol Biosci 2020; 7:57. [PMID: 32351971 PMCID: PMC7174556 DOI: 10.3389/fmolb.2020.00057] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
There are over 50 lysosomal hydrolase deficiencies, many of which cause neurodegeneration, cognitive decline and death. In recent years, a number of broad innovative therapies have been proposed and investigated for lysosomal storage diseases (LSDs), such as enzyme replacement, substrate reduction, pharmacologic chaperones, stem cell transplantation, and various forms of gene therapy. Murine models that accurately reflect the phenotypes observed in human LSDs are critical for the development, assessment and implementation of novel translational therapies. The goal of this review is to summarize the neurodegenerative murine LSD models available that recapitulate human disease, and the pre-clinical studies previously conducted. We also describe some limitations and difficulties in working with mouse models of neurodegenerative LSDs.
Collapse
Affiliation(s)
| | | | | | - Daesung Shin
- Hunter James Kelly Research Institute, Department of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
23
|
Beerepoot S, Nierkens S, Boelens JJ, Lindemans C, Bugiani M, Wolf NI. Peripheral neuropathy in metachromatic leukodystrophy: current status and future perspective. Orphanet J Rare Dis 2019; 14:240. [PMID: 31684987 PMCID: PMC6829806 DOI: 10.1186/s13023-019-1220-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 10/09/2019] [Indexed: 11/23/2022] Open
Abstract
Metachromatic leukodystrophy (MLD) is an autosomal recessively inherited metabolic disease characterized by deficient activity of the lysosomal enzyme arylsulfatase A. Its deficiency results in accumulation of sulfatides in neural and visceral tissues, and causes demyelination of the central and peripheral nervous system. This leads to a broad range of neurological symptoms and eventually premature death. In asymptomatic patients with juvenile and adult MLD, treatment with allogeneic hematopoietic stem cell transplantation (HCT) provides a symptomatic and survival benefit. However, this treatment mainly impacts brain white matter, whereas the peripheral neuropathy shows no or only limited response. Data about the impact of peripheral neuropathy in MLD patients are currently lacking, although in our experience peripheral neuropathy causes significant morbidity due to neuropathic pain, foot deformities and neurogenic bladder disturbances. Besides, the reasons for residual and often progressive peripheral neuropathy after HCT are not fully understood. Preliminary studies suggest that peripheral neuropathy might respond better to gene therapy due to higher enzyme levels achieved than with HCT. However, histopathological and clinical findings also suggest a role of neuroinflammation in the pathology of peripheral neuropathy in MLD. In this literature review, we discuss clinical aspects, pathological findings, distribution of mutations, and treatment approaches in MLD with particular emphasis on peripheral neuropathy. We believe that future therapies need more emphasis on the management of peripheral neuropathy, and additional research is needed to optimize care strategies.
Collapse
Affiliation(s)
- Shanice Beerepoot
- Department of Child Neurology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.,Pediatric Blood and Marrow Transplantation Program, Princess Máxima Center and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jaap Jan Boelens
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Pediatrics, Stem Cell Transplant and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caroline Lindemans
- Pediatric Blood and Marrow Transplantation Program, Princess Máxima Center and University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative medicine institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands.
| |
Collapse
|
24
|
Schlotawa L, Dierks T, Christoph S, Cloppenburg E, Ohlenbusch A, Korenke GC, Gärtner J. Severe neonatal multiple sulfatase deficiency presenting with hydrops fetalis in a preterm birth patient. JIMD Rep 2019; 49:48-52. [PMID: 31497481 PMCID: PMC6718111 DOI: 10.1002/jmd2.12074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 01/30/2023] Open
Abstract
Multiple sulfatase deficiency (MSD) is an ultra-rare lysosomal storage disorder (LSD). Mutations in the SUMF1 gene encoding the formylglycine generating enzyme (FGE) result in an unstable FGE protein with reduced enzymatic activity, thereby affecting the posttranslational activation of newly synthesized sulfatases. Complete absence of FGE function results in the most severe clinical form of MSD with neonatal onset and rapid deterioration. We report on a preterm infant presenting with hydrops fetalis, lung hypoplasia, and dysmorphism as major clinical signs. The patient died after 6 days from an intraventricular hemorrhage followed by multi-organ failure. MSD was caused by a homozygous SUMF1 stop mutation (c.191C>A, p.Ser64Ter). FGE protein and sulfatase activities were absent in patient fibroblasts. Hydrops fetalis is a rare symptom of LSDs and should be considered in the differential diagnosis in combination with dysmorphism. The diagnostic set up should include measurements of glycosaminoglycan excretion and lysosomal enzyme activities, among them at least two sulfatases, and molecular confirmation.
Collapse
Affiliation(s)
- Lars Schlotawa
- Department of Paediatrics and Adolescent MedicineUniversity Medical Center GöttingenGöttingenGermany
| | - Thomas Dierks
- Department of Chemistry, Biochemistry IBielefeld UniversityBielefeldGermany
| | - Sophie Christoph
- Department of Child Neurology and Metabolic Disorders, Medical Centre OldenburgUniversity Children's Hospital OldenburgOldenburgGermany
| | - Eva Cloppenburg
- Department of Neonatology, Intensive Care Medicine and Paediatric Cardiology, Medical Centre OldenburgUniversity Children's Hospital OldenburgOldenburgGermany
| | - Andreas Ohlenbusch
- Department of Paediatrics and Adolescent MedicineUniversity Medical Center GöttingenGöttingenGermany
| | - G. Christoph Korenke
- Department of Child Neurology and Metabolic Disorders, Medical Centre OldenburgUniversity Children's Hospital OldenburgOldenburgGermany
| | - Jutta Gärtner
- Department of Paediatrics and Adolescent MedicineUniversity Medical Center GöttingenGöttingenGermany
| |
Collapse
|
25
|
Expression, activity and localization of lysosomal sulfatases in Chronic Obstructive Pulmonary Disease. Sci Rep 2019; 9:1991. [PMID: 30760748 PMCID: PMC6374378 DOI: 10.1038/s41598-018-37958-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/05/2018] [Indexed: 12/31/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of death world-wide. Recently, we showed that COPD is associated with gene polymorphisms in SUMF1, a master regulator of sulfatases. Sulfatases are involved in extracellular matrix remodeling and activated by SUMF1, but their role in the lung is poorly described. We aimed to examine how sulfatases are affected in the airways of patients with COPD compared to ever smokers and never smokers. We observed that mRNA expression of the sulfatases GALNS, GNS and IDS was increased, while protein expression of many sulfatases was decreased in COPD fibroblasts. Several sulfatases, including GALNS, IDS, and SGSH, showed increased activity in COPD fibroblasts. Examination of different sulfatases by immunofluorescence showed that IDS, ARSB, GNS and SGSH in fibroblasts were localized to sites other than their reported destination. Using a master panel from different organs, RNA expression of all sulfatases could be observed in lung tissue. Additionally, immunohistochemistry on lung biopsies indicated differing expression of sulfatases in COPD patients. In conclusion, mRNA, protein expression, sulfatase activity levels, and localization of sulfatases are altered in lung fibroblasts and lung tissue from COPD patients and may be mechanistically important in COPD pathogenesis. This could contribute to the understanding of the disease mechanism in COPD and in the long run, to lead to more individualized therapies.
Collapse
|
26
|
Grassi S, Chiricozzi E, Mauri L, Sonnino S, Prinetti A. Sphingolipids and neuronal degeneration in lysosomal storage disorders. J Neurochem 2018; 148:600-611. [PMID: 29959861 DOI: 10.1111/jnc.14540] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 12/27/2022]
Abstract
Ceramide, sphingomyelin, and glycosphingolipids (both neutral and acidic) are characterized by the presence in the lipid moiety of an aliphatic base known as sphingosine. Altogether, they are called sphingolipids and are particularly abundant in neuronal plasma membranes, where, via interactions with the other membrane lipids and membrane proteins, they play a specific role in modulating the cell signaling processes. The metabolic pathways determining the plasma membrane sphingolipid composition are thus the key point for functional changes of the cell properties. Unnatural changes of the neuronal properties are observed in sphingolipidoses, lysosomal storage diseases occurring when a lysosomal sphingolipid hydrolase is not working, leading to the accumulation of the substrate and to its distribution to all the cell membranes interacting with lysosomes. Moreover, secondary accumulation of sphingolipids is a common trait of other lysosomal storage diseases. This article is part of the Special Issue "Lysosomal Storage Disorders".
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Chiricozzi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
27
|
Sambri I, D'Alessio R, Ezhova Y, Giuliano T, Sorrentino NC, Cacace V, De Risi M, Cataldi M, Annunziato L, De Leonibus E, Fraldi A. Lysosomal dysfunction disrupts presynaptic maintenance and restoration of presynaptic function prevents neurodegeneration in lysosomal storage diseases. EMBO Mol Med 2017; 9:112-132. [PMID: 27881461 PMCID: PMC5210158 DOI: 10.15252/emmm.201606965] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Lysosomal storage disorders (LSDs) are inherited diseases characterized by lysosomal dysfunction and often showing a neurodegenerative course. There is no cure to treat the central nervous system in LSDs. Moreover, the mechanisms driving neuronal degeneration in these pathological conditions remain largely unknown. By studying mouse models of LSDs, we found that neurodegeneration develops progressively with profound alterations in presynaptic structure and function. In these models, impaired lysosomal activity causes massive perikaryal accumulation of insoluble α‐synuclein and increased proteasomal degradation of cysteine string protein α (CSPα). As a result, the availability of both α‐synuclein and CSPα at nerve terminals strongly decreases, thus inhibiting soluble NSF attachment receptor (SNARE) complex assembly and synaptic vesicle recycling. Aberrant presynaptic SNARE phenotype is recapitulated in mice with genetic ablation of one allele of both CSPα and α‐synuclein. The overexpression of CSPα in the brain of a mouse model of mucopolysaccharidosis type IIIA, a severe form of LSD, efficiently re‐established SNARE complex assembly, thereby ameliorating presynaptic function, attenuating neurodegenerative signs, and prolonging survival. Our data show that neurodegenerative processes associated with lysosomal dysfunction may be presynaptically initiated by a concomitant reduction in α‐synuclein and CSPα levels at nerve terminals. They also demonstrate that neurodegeneration in LSDs can be slowed down by re‐establishing presynaptic functions, thus identifying synapse maintenance as a novel potentially druggable target for brain treatment in LSDs.
Collapse
Affiliation(s)
- Irene Sambri
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Rosa D'Alessio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Yulia Ezhova
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Teresa Giuliano
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | | | - Vincenzo Cacace
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Maria De Risi
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy.,Institute of Genetics and Biophysics, National Research Council, Naples, Italy
| | - Mauro Cataldi
- Department of Neuroscience, Reproductive and Odontostomatological Sciences Federico II University, Naples, Italy
| | - Lucio Annunziato
- Department of Neuroscience, Reproductive and Odontostomatological Sciences Federico II University, Naples, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy.,Institute of Genetics and Biophysics, National Research Council, Naples, Italy
| | | |
Collapse
|
28
|
Papy-Garcia D, Albanese P. Heparan sulfate proteoglycans as key regulators of the mesenchymal niche of hematopoietic stem cells. Glycoconj J 2017; 34:377-391. [PMID: 28577070 DOI: 10.1007/s10719-017-9773-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 12/21/2022]
Abstract
The complex microenvironment that surrounds hematopoietic stem cells (HSCs) in the bone marrow niche involves different coordinated signaling pathways. The stem cells establish permanent interactions with distinct cell types such as mesenchymal stromal cells, osteoblasts, osteoclasts or endothelial cells and with secreted regulators such as growth factors, cytokines, chemokines and their receptors. These interactions are mediated through adhesion to extracellular matrix compounds also. All these signaling pathways are important for stem cell fates such as self-renewal, proliferation or differentiation, homing and mobilization, as well as for remodeling of the niche. Among these complex molecular cues, this review focuses on heparan sulfate (HS) structures and functions and on the role of enzymes involved in their biosynthesis and turnover. HS associated to core protein, constitute the superfamily of heparan sulfate proteoglycans (HSPGs) present on the cell surface and in the extracellular matrix of all tissues. The key regulatory effects of major medullar HSPGs are described, focusing on their roles in the interactions between hematopoietic stem cells and their endosteal niche, and on their ability to interact with Heparin Binding Proteins (HBPs). Finally, according to the relevance of HS moieties effects on this complex medullar niche, we describe recent data that identify HS mimetics or sulfated HS signatures as new glycanic tools and targets, respectively, for hematopoietic and mesenchymal stem cell based therapeutic applications.
Collapse
Affiliation(s)
- Dulce Papy-Garcia
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France
| | - Patricia Albanese
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France.
| |
Collapse
|
29
|
Weidner J, Jarenbäck L, de Jong K, Vonk JM, van den Berge M, Brandsma CA, Boezen HM, Sin D, Bossé Y, Nickle D, Ankerst J, Bjermer L, Postma DS, Faiz A, Tufvesson E. Sulfatase modifying factor 1 (SUMF1) is associated with Chronic Obstructive Pulmonary Disease. Respir Res 2017; 18:77. [PMID: 28464818 PMCID: PMC5414362 DOI: 10.1186/s12931-017-0562-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Background It has been observed that mice lacking the sulfatase modifying factor (Sumf1) developed an emphysema-like phenotype. However, it is unknown if SUMF1 may play a role in Chronic Obstructive Pulmonary Disease (COPD) in humans. The aim was to investigate if the expression and genetic regulation of SUMF1 differs between smokers with and without COPD. Methods SUMF1 mRNA was investigated in sputum cells and whole blood from controls and COPD patients (all current or former smokers). Expression quantitative trait loci (eQTL) analysis was used to investigate if single nucleotide polymorphisms (SNPs) in SUMF1 were significantly associated with SUMF1 expression. The association of SUMF1 SNPs with COPD was examined in a population based cohort, Lifelines. SUMF1 mRNA from sputum cells, lung tissue, and lung fibroblasts, as well as lung function parameters, were investigated in relation to genotype. Results Certain splice variants of SUMF1 showed a relatively high expression in lung tissue compared to many other tissues. SUMF1 Splice variant 2 and 3 showed lower levels in sputum cells from COPD patients as compared to controls. Twelve SNPs were found significant by eQTL analysis and overlapped with the array used for genotyping of Lifelines. We found alterations in mRNA expression in sputum cells and lung fibroblasts associated with SNP rs11915920 (top hit in eQTL), which validated the results of the lung tissue eQTL analysis. Of the twelve SNPs, two SNPs, rs793391 and rs308739, were found to be associated with COPD in Lifelines. The SNP rs793391 was also confirmed to be associated with lung function changes. Conclusions We show that SUMF1 expression is affected in COPD patients compared to controls, and that SNPs in SUMF1 are associated with an increased risk of COPD. Certain COPD-associated SNPs have effects on either SUMF1 gene expression or on lung function. Collectively, this study shows that SUMF1 is associated with an increased risk of developing COPD. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0562-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julie Weidner
- Respiratory Medicine and Allergology, Department of Clinical Sciences Lund, BMC, D12, Lund University, Skåne University Hospital, 221 84, Lund, Sweden
| | - Linnea Jarenbäck
- Respiratory Medicine and Allergology, Department of Clinical Sciences Lund, BMC, D12, Lund University, Skåne University Hospital, 221 84, Lund, Sweden
| | - Kim de Jong
- University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Department of Epidemiology, University of Groningen, Groningen, The Netherlands
| | - Judith M Vonk
- University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Department of Epidemiology, University of Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- University Medical Center Groningen, Department of Pulmonology, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- University Medical Center Groningen, Department of Pulmonology, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, The Netherlands
| | - H Marike Boezen
- University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Department of Epidemiology, University of Groningen, Groningen, The Netherlands
| | - Don Sin
- Department of Medicine (Respirology), University of British Columbia, Centre for Heart Lung Innovation, Vancouver, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Québec, Canada
| | - David Nickle
- Genetics and Pharmacogenomics (GpGx), Merck Research Laboratories, Boston, MA, USA
| | - Jaro Ankerst
- Respiratory Medicine and Allergology, Department of Clinical Sciences Lund, BMC, D12, Lund University, Skåne University Hospital, 221 84, Lund, Sweden
| | - Leif Bjermer
- Respiratory Medicine and Allergology, Department of Clinical Sciences Lund, BMC, D12, Lund University, Skåne University Hospital, 221 84, Lund, Sweden
| | - Dirkje S Postma
- University Medical Center Groningen, Department of Pulmonology, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, The Netherlands
| | - Alen Faiz
- University Medical Center Groningen, Department of Pulmonology, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, The Netherlands
| | - Ellen Tufvesson
- Respiratory Medicine and Allergology, Department of Clinical Sciences Lund, BMC, D12, Lund University, Skåne University Hospital, 221 84, Lund, Sweden.
| |
Collapse
|
30
|
Independent Maternal and Fetal Genetic Effects on Midgestational Circulating Levels of Environmental Pollutants. G3-GENES GENOMES GENETICS 2017; 7:1287-1299. [PMID: 28235828 PMCID: PMC5386877 DOI: 10.1534/g3.117.039784] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Maternal exposure to environmental pollutants could affect fetal brain development and increase autism spectrum disorder (ASD) risk in conjunction with differential genetic susceptibility. Organohalogen congeners measured in maternal midpregnancy blood samples have recently shown significant, but negative associations with offspring ASD outcome. We report the first large-scale maternal and fetal genetic study of the midpregnancy serum levels of a set of 21 organohalogens in a subset of 790 genotyped women and 764 children collected in California by the Early Markers for Autism (EMA) Project. Levels of PCB (polychlorinated biphenyl) and PBDE (polybrominated diphenyl ether) congeners showed high maternal and fetal estimated SNP-based heritability (h2g) accounting for 39–99% of the total variance. Genome-wide association analyses identified significant maternal loci for p,p′-DDE (P = 7.8 × 10−11) in the CYP2B6 gene and for BDE-28 (P = 3.2 × 10−8) near the SH3GL2 gene, both involved in xenobiotic and lipid metabolism. Fetal genetic loci contributed to the levels of BDE-100 (P = 4.6 × 10−8) and PCB187 (P = 2.8 × 10−8), near the potential metabolic genes LOXHD1 and PTPRD, previously implicated in neurodevelopment. Negative associations were observed for BDE-100, BDE153, and the sum of PBDEs with ASD, partly explained by genome-wide additive genetic effects that predicted PBDE levels. Our results support genetic control of midgestational biomarkers for environmental exposures by nonoverlapping maternal and fetal genetic determinants, suggesting that future studies of environmental risk factors should take genetic variation into consideration. The independent influence of fetal genetics supports previous hypotheses that fetal genotypes expressed in placenta can influence maternal physiology and the transplacental transfer of organohalogens.
Collapse
|
31
|
Dawson PA, Richard K, Perkins A, Zhang Z, Simmons DG. Review: Nutrient sulfate supply from mother to fetus: Placental adaptive responses during human and animal gestation. Placenta 2017; 54:45-51. [PMID: 28089504 DOI: 10.1016/j.placenta.2017.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 01/20/2023]
Abstract
Nutrient sulfate has numerous roles in mammalian physiology and is essential for healthy fetal growth and development. The fetus has limited capacity to generate sulfate and relies on sulfate supplied from the maternal circulation via placental sulfate transporters. The placenta also has a high sulfate requirement for numerous molecular and cellular functions, including sulfate conjugation (sulfonation) to estrogen and thyroid hormone which leads to their inactivation. Accordingly, the ratio of sulfonated (inactive) to unconjugated (active) hormones modulates endocrine function in fetal, placental and maternal tissues. During pregnancy, there is a marked increase in the expression of genes involved in transport and generation of sulfate in the mouse placenta, in line with increasing fetal and placental demands for sulfate. The maternal circulation also provides a vital reservoir of sulfate for the placenta and fetus, with maternal circulating sulfate levels increasing by 2-fold from mid-gestation. However, despite evidence from animal studies showing the requirement of maternal sulfate supply for placental and fetal physiology, there are no routine clinical measurements of sulfate or consideration of dietary sulfate intake in pregnant women. This is also relevant to certain xenobiotics or pharmacological drugs which when taken by the mother use significant quantities of circulating sulfate for detoxification and clearance, and thereby have the potential to decrease sulfonation capacity in the placenta and fetus. This article will review the physiological adaptations of the placenta for maintaining sulfate homeostasis in the fetus and placenta, with a focus on pathophysiological outcomes in animal models of disturbed sulfate homeostasis.
Collapse
Affiliation(s)
- P A Dawson
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia.
| | - K Richard
- Conjoint Endocrine Laboratory, Chemical Pathology, Pathology Queensland, Queensland Health, Herston, Australia
| | - A Perkins
- School of Medical Science, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia
| | - Z Zhang
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - D G Simmons
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
32
|
Langford R, Hurrion E, Dawson PA. Genetics and pathophysiology of mammalian sulfate biology. J Genet Genomics 2017; 44:7-20. [DOI: 10.1016/j.jgg.2016.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 12/23/2022]
|
33
|
Thibert KA, Raymond GV, Tolar J, Miller WP, Orchard PJ, Lund TC. Cerebral Spinal Fluid levels of Cytokines are elevated in Patients with Metachromatic Leukodystrophy. Sci Rep 2016; 6:24579. [PMID: 27079147 PMCID: PMC4832325 DOI: 10.1038/srep24579] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/30/2016] [Indexed: 01/08/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disease resulting from a deficiency of arylsulfatase A causing an accumulation of cerebroside sulfate, a lipid normally abundant in myelin. Sulfatide accumulation is associated with progressive demyelination and a clinical presentation in severe disease forms that is dominated by motor manifestations. Cerebral inflammation may contribute to the pathophysiology of MLD. To date, cytokine levels in the cerebral spinal fluid of MLD patients have not previously been reported. The objective of this study was to evaluate the concentration of inflammatory cytokines in the CSF of patients with MLD and to compare these levels to unaffected controls. Of 22 cytokines evaluated, we documented significant elevations of MCP-1, IL-1Ra, IL-8, MIP-1b and VEGF in the MLD patients compared to unaffected controls. The elevated cytokines identified in this study may play a significant role in the pathophysiology of MLD. Better understanding of the inflammatory and neurodegenerative process of MLD may lead to improved targeted therapies.
Collapse
Affiliation(s)
- Kathryn A Thibert
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Gerald V Raymond
- Division of Pediatric Neurology, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Jakub Tolar
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Weston P Miller
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| | - Troy C Lund
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, 55455, US
| |
Collapse
|
34
|
Sabourdy F, Mourey L, Le Trionnaire E, Bednarek N, Caillaud C, Chaix Y, Delrue MA, Dusser A, Froissart R, Garnotel R, Guffon N, Megarbane A, Ogier de Baulny H, Pédespan JM, Pichard S, Valayannopoulos V, Verloes A, Levade T. Natural disease history and characterisation of SUMF1 molecular defects in ten unrelated patients with multiple sulfatase deficiency. Orphanet J Rare Dis 2015; 10:31. [PMID: 25885655 PMCID: PMC4375846 DOI: 10.1186/s13023-015-0244-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 02/23/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Multiple sulfatase deficiency is a rare inherited metabolic disorder caused by mutations in the SUMF1 gene. The disease remains poorly known, often leading to a late diagnosis. This study aimed to provide improved knowledge of the disease, through complete clinical, biochemical, and molecular descriptions of a cohort of unrelated patients. The main objective was to identify prognostic markers, both phenotypic and genotypic, to accelerate the diagnosis and improve patient care. METHODS The phenotypes of ten unrelated patients were fully documented at the clinical and biochemical levels. The long-term follow-up of each patient allowed correlations of the phenotypes to the disease outcomes. Each patient's molecular defects were also identified. Site-directed mutagenesis was used to individually express the mutants and assess their stability. Characterisation of the protein mutants was completed by in silico analyses based on sequence comparisons and structural models. RESULTS The most severe cases were characterised by the presence of non-neurological symptoms as well as the occurrence of psychomotor regression before 2 years of age. Nine novel SUMF1 mutations were identified. Clinically severe forms were often associated with SUMF1 mutations that strongly affected the protein stability and/or catalytic function as predicted from in silico and western blot analyses. CONCLUSIONS This detailed clinical description and follow-up of a cohort of patients, together with the molecular characterisation of their underlying defects, contribute to improved knowledge of multiple sulfatase deficiency. Predictors of a bad prognosis were the presence of several non-neurological symptoms and the onset of psychomotor regression before 2 years of age. No strict correlation existed between in vitro residual sulfatase activity and disease severity. Genotype-phenotype correlations related to previously reported mutants were strengthened. These and previous observations allow not only improved prediction of the disease outcome but also provision of appropriate care for patients, in the expectation of specific treatment development.
Collapse
Affiliation(s)
- Frédérique Sabourdy
- Laboratoire de Biochimie Métabolique, IFB, CHU Purpan, Toulouse, France. .,INSERM UMR 1037, CRCT, Université Paul Sabatier Toulouse-III, Toulouse, France.
| | - Lionel Mourey
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Centre National de la Recherche Scientifique (CNRS), Toulouse, France. .,Université de Toulouse, Université Paul Sabatier, IPBS, Toulouse, France.
| | | | - Nathalie Bednarek
- Service de Néonatologie, Alix de Champagne, CHU de Reims, Reims, France.
| | - Catherine Caillaud
- Laboratoire de Biochimie, Métabolomique et Protéomique, Hôpital Necker Enfants Malades, Paris, France.
| | - Yves Chaix
- Hôpital des Enfants, CHU Purpan, and INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, Université Paul Sabatier Toulouse-III, CHU Purpan, Toulouse, France.
| | - Marie-Ange Delrue
- Service de Génétique Médicale, CHU Pellegrin, et laboratoire Maladies Rares, Génétique et Métabolisme, Université Bordeaux 2, Bordeaux, France.
| | - Anne Dusser
- Service de Neuropédiatrie, CHU de Bicêtre, Kremlin-Bicêtre, France.
| | - Roseline Froissart
- Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France.
| | - Roselyne Garnotel
- Laboratoire de biologie et de recherche pédiatriques, American Memorial Hospital, CHU Reims, Reims, France.
| | - Nathalie Guffon
- Centre de Référence des Maladies Héréditaires du Métabolisme, Lyon, France.
| | - André Megarbane
- Unité de Génétique Médicale et Laboratoire Associé INSERM UMR_S910, Université Saint-Joseph, Beirut, Lebanon. .,Institut Jérôme Lejeune, Paris, France.
| | - Hélène Ogier de Baulny
- Centre Référence des Maladies Héréditaires du Métabolisme, CHU Robert Debré, APHP, Paris, France.
| | | | - Samia Pichard
- Centre Référence des Maladies Héréditaires du Métabolisme, CHU Robert Debré, APHP, Paris, France.
| | | | - Alain Verloes
- Service de Génétique Clinique, CHU Robert Debré, Paris, France.
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, IFB, CHU Purpan, Toulouse, France. .,INSERM UMR 1037, CRCT, Université Paul Sabatier Toulouse-III, Toulouse, France.
| |
Collapse
|
35
|
A non-conserved miRNA regulates lysosomal function and impacts on a human lysosomal storage disorder. Nat Commun 2014; 5:5840. [PMID: 25524633 DOI: 10.1038/ncomms6840] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 11/13/2014] [Indexed: 12/18/2022] Open
Abstract
Sulfatases are key enzymatic regulators of sulfate homeostasis with several biological functions including degradation of glycosaminoglycans (GAGs) and other macromolecules in lysosomes. In a severe lysosomal storage disorder, multiple sulfatase deficiency (MSD), global sulfatase activity is deficient due to mutations in the sulfatase-modifying factor 1 (SUMF1) gene, encoding the essential activator of all sulfatases. We identify a novel regulatory layer of sulfate metabolism mediated by a microRNA. miR-95 depletes SUMF1 protein levels and suppresses sulfatase activity, causing the disruption of proteoglycan catabolism and lysosomal function. This blocks autophagy-mediated degradation, causing cytoplasmic accumulation of autophagosomes and autophagic substrates. By targeting miR-95 in cells from MSD patients, we can effectively increase residual SUMF1 expression, allowing for reactivation of sulfatase activity and increased clearance of sulfated GAGs. The identification of this regulatory mechanism opens the opportunity for a unique therapeutic approach in MSD patients where the need for exogenous enzyme replacement is circumvented.
Collapse
|
36
|
Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR, Vogel P. High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res 2014; 2:14034. [PMID: 26273529 PMCID: PMC4472125 DOI: 10.1038/boneres.2014.34] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Screening gene function in vivo is a powerful approach to discover novel drug targets. We present high-throughput screening (HTS) data for 3 762 distinct global gene knockout (KO) mouse lines with viable adult homozygous mice generated using either gene-trap or homologous recombination technologies. Bone mass was determined from DEXA scans of male and female mice at 14 weeks of age and by microCT analyses of bones from male mice at 16 weeks of age. Wild-type (WT) cagemates/littermates were examined for each gene KO. Lethality was observed in an additional 850 KO lines. Since primary HTS are susceptible to false positive findings, additional cohorts of mice from KO lines with intriguing HTS bone data were examined. Aging, ovariectomy, histomorphometry and bone strength studies were performed and possible non-skeletal phenotypes were explored. Together, these screens identified multiple genes affecting bone mass: 23 previously reported genes (Calcr, Cebpb, Crtap, Dcstamp, Dkk1, Duoxa2, Enpp1, Fgf23, Kiss1/Kiss1r, Kl (Klotho), Lrp5, Mstn, Neo1, Npr2, Ostm1, Postn, Sfrp4, Slc30a5, Slc39a13, Sost, Sumf1, Src, Wnt10b), five novel genes extensively characterized (Cldn18, Fam20c, Lrrk1, Sgpl1, Wnt16), five novel genes with preliminary characterization (Agpat2, Rassf5, Slc10a7, Slc26a7, Slc30a10) and three novel undisclosed genes coding for potential osteoporosis drug targets.
Collapse
Affiliation(s)
| | - Jeff Liu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | | | | | | | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| |
Collapse
|
37
|
Settembre C, Ballabio A. Lysosomal adaptation: how the lysosome responds to external cues. Cold Spring Harb Perspect Biol 2014; 6:cshperspect.a016907. [PMID: 24799353 DOI: 10.1101/cshperspect.a016907] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent evidence indicates that the importance of the lysosome in cell metabolism and organism physiology goes far beyond the simple disposal of cellular garbage. This dynamic organelle is situated at the crossroad of the most important cellular pathways and is involved in sensing, signaling, and transcriptional mechanisms that respond to environmental cues, such as nutrients. Two main mediators of these lysosomal adaptation mechanisms are the mTORC1 kinase complex and the transcription factor EB (TFEB). These two factors are linked in a lysosome-to-nucleus signaling pathway that provides the lysosome with the ability to adapt to extracellular cues and control its own biogenesis. Modulation of lysosomal function by acting on TFEB has a profound impact on cellular clearance and energy metabolism and is a promising therapeutic target for a large variety of disease conditions.
Collapse
Affiliation(s)
- Carmine Settembre
- Telethon Institute of Genetics and Medicine, 80131 Naples, Italy Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030 Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030 Medical Genetics, Department of Translational Medicine, Federico II University, 80131 Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, 80131 Naples, Italy Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030 Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030 Medical Genetics, Department of Translational Medicine, Federico II University, 80131 Naples, Italy
| |
Collapse
|
38
|
Autophagy impairment aggravates the inhibitory effects of high glucose on osteoblast viability and function. Biochem J 2014; 455:329-37. [PMID: 23981124 DOI: 10.1042/bj20130562] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Autophagy is a highly regulated homoeostatic process involved in the lysosomal degradation of damaged cell organelles and proteins. This process is considered an important pro-survival mechanism under diverse stress conditions. A diabetic milieu is known to hamper osteoblast viability and function. In the present study, we explored the putative protective role of autophagy in osteoblastic cells exposed to an HG (high glucose) medium. HG was found to increase protein oxidation and triggered autophagy by a mechanism dependent on reactive oxygen species overproduction in osteoblastic MC3T3-E1 cells. MC3T3-E1 cell survival was impaired by HG and worsened by chemical or genetic inhibition of autophagy. These findings were mimicked by H2O2-induced oxidative stress in these cells. Autophagy impairment led to both defective mitochondrial morphology and decreased bioenergetic machinery and inhibited further osteoblast differentiation in MC3T3-E1 cells upon exposure to HG. These novel findings indicate that autophagy is an essential mechanism to maintain osteoblast viability and function in an HG environment.
Collapse
|
39
|
Shapiro IM, Layfield R, Lotz M, Settembre C, Whitehouse C. Boning up on autophagy: the role of autophagy in skeletal biology. Autophagy 2013; 10:7-19. [PMID: 24225636 DOI: 10.4161/auto.26679] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
From an evolutionary perspective, the major function of bone is to provide stable sites for muscle attachment and affording protection of vital organs, especially the heart and lungs (ribs) and spinal cord (vertebrae and intervertebral discs). However, bone has a considerable number of other functions: serving as a store for mineral ions, providing a site for blood cell synthesis and participating in a complex system-wide endocrine system. Not surprisingly, bone and cartilage cell homeostasis is tightly controlled, as is the maintenance of tissue structure and mass. While a great deal of new information is accruing concerning skeletal cell homeostasis, one relatively new observation is that the cells of bone (osteoclasts osteoblasts and osteocytes) and cartilage (chondrocytes) exhibit autophagy. The focus of this review is to examine the significance of this process in terms of the functional demands of the skeleton in health and during growth and to provide evidence that dysregulation of the autophagic response is involved in the pathogenesis of diseases of bone (Paget disease of bone) and cartilage (osteoarthritis and the mucopolysaccharidoses). Delineation of molecular changes in the autophagic process is uncovering new approaches for the treatment of diseases that affect the axial and appendicular skeleton.
Collapse
Affiliation(s)
- Irving M Shapiro
- Jefferson Medical College; Thomas Jefferson University; Philadelphia, PA USA
| | - Robert Layfield
- School of Life Sciences; University of Nottingham Medical School; Nottingham UK
| | - Martin Lotz
- Arthritis Research; The Scripps Research Institute; La Jolla, CA USA
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM); Naples, Italy; Department of Molecular and Human Genetics; Baylor College of Medicine, Houston, TXUSA and Jan and Dan Duncan Neurological Research Institute; Texas Children's Hospital; Houston, TX USA; Medical Genetics; Department of Translational and Medical Science; Federico II University; Naples, Italy
| | - Caroline Whitehouse
- Department of Medical and Molecular Genetics; Kings College London; London UK
| |
Collapse
|
40
|
Abstract
Sulphate contributes to numerous processes in mammalian physiology, particularly during development. Sulphotransferases mediate the sulphate conjugation (sulphonation) of numerous compounds, including steroids, glycosaminoglycans, proteins, neurotransmitters and xenobiotics, transforming their biological activities. Importantly, the ratio of sulphonated to unconjugated molecules plays a significant physiological role in many of the molecular events that regulate mammalian growth and development. In humans, the fetus is unable to generate its own sulphate and therefore relies on sulphate being supplied from maternal circulation via the placenta. To meet the gestational needs of the growing fetus, maternal blood sulphate concentrations double from mid-gestation. Maternal hyposulphataemia has been linked to fetal sulphate deficiency and late gestational fetal loss in mice. Disorders of sulphonation have also been linked to a number of developmental disorders in humans, including skeletal dysplasias and premature adrenarche. While recognised as an important nutrient in mammalian physiology, sulphate is largely unappreciated in clinical settings. In part, this may be due to technical challenges in measuring sulphate with standard pathology equipment and hence the limited findings of perturbed sulphate homoeostasis affecting human health. This review article is aimed at highlighting the importance of sulphate in mammalian development, with basic science research being translated through animal models and linkage to human disorders.
Collapse
|
41
|
Patil SA, Maegawa GHB. Developing therapeutic approaches for metachromatic leukodystrophy. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:729-45. [PMID: 23966770 PMCID: PMC3743609 DOI: 10.2147/dddt.s15467] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metachromatic leukodystrophy (MLD) is an autosomal recessive lysosomal disorder caused by the deficiency of arylsulfatase A (ASA), resulting in impaired degradation of sulfatide, an essential sphingolipid of myelin. The clinical manifestations of MLD are characterized by progressive demyelination and subsequent neurological symptoms resulting in severe debilitation. The availability of therapeutic options for treating MLD is limited but expanding with a number of early stage clinical trials already in progress. In the development of therapeutic approaches for MLD, scientists have been facing a number of challenges including blood–brain barrier (BBB) penetration, safety issues concerning therapies targeting the central nervous system, uncertainty regarding the ideal timing for intervention in the disease course, and the lack of more in-depth understanding of the molecular pathogenesis of MLD. Here, we discuss the current status of the different approaches to developing therapies for MLD. Hematopoietic stem cell transplantation has been used to treat MLD patients, utilizing both umbilical cord blood and bone marrow sources. Intrathecal enzyme replacement therapy and gene therapies, administered locally into the brain or by generating genetically modified hematopoietic stem cells, are emerging as novel strategies. In pre-clinical studies, different cell delivery systems including microencapsulated cells or selectively neural cells have shown encouraging results. Small molecules that are more likely to cross the BBB can be used as enzyme enhancers of diverse ASA mutants, either as pharmacological chaperones, or proteostasis regulators. Specific small molecules may also be used to reduce the biosynthesis of sulfatides, or target different affected downstream pathways secondary to the primary ASA deficiency. Given the progressive neurodegenerative aspects of MLD, also seen in other lysosomal diseases, current and future therapeutic strategies will be complementary, whether used in combination or separately at specific stages of the disease course, to produce better outcomes for patients afflicted with this devastating inherited disorder.
Collapse
Affiliation(s)
- Shilpa A Patil
- McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
42
|
Choi SI, Kim BY, Dadakhujaev S, Oh JY, Kim TI, Kim JY, Kim EK. Impaired autophagy and delayed autophagic clearance of transforming growth factor β-induced protein (TGFBI) in granular corneal dystrophy type 2. Autophagy 2012; 8:1782-97. [PMID: 22995918 DOI: 10.4161/auto.22067] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Granular corneal dystrophy type 2 (GCD2) is an autosomal dominant disease characterized by a progressive age-dependent extracellular accumulation of transforming growth factor β-induced protein (TGFBI). Corneal fibroblasts from GCD2 patients also have progressive degenerative features, but the mechanism underlying this degeneration remains unknown. Here we observed that TGFBI was degraded by autophagy, but not by the ubiquitin/proteasome-dependent pathway. We also found that GCD2 homozygous corneal fibroblasts displayed a greater number of fragmented mitochondria. Most notably, mutant TGFBI (mut-TGFBI) extensively colocalized with microtubule-associated protein 1 light chain 3β (MAP1LC3B, hereafter referred to as LC3)-enriched cytosolic vesicles and CTSD in primary cultured GCD2 corneal fibroblasts. Levels of LC3-II, a marker of autophagy activation, were significantly increased in GCD2 corneal fibroblasts. Nevertheless, levels of SQSTM1/p62 and of polyubiquitinated protein were also significantly increased in GCD2 corneal fibroblasts compared with wild-type (WT) cells. However, LC3-II levels did not differ significantly between WT and GCD2 cells, as assessed by the presence of bafilomycin A 1, the fusion blocker of autophagosomes and lysosomes. Likewise, bafilomycin A 1 caused a similar change in levels of SQSTM1. Thus, the increase in autophagosomes containing mut-TGFBI may be due to inefficient fusion between autophagosomes and lysosomes. Rapamycin, an autophagy activator, decreased mut-TGFBI, whereas inhibition of autophagy increased active caspase-3, poly (ADP-ribose) polymerase 1 (PARP1) and reduced the viability of GCD2 corneal fibroblasts compared with WT controls. These data suggest that defective autophagy may play a critical role in the pathogenesis of GCD2.
Collapse
Affiliation(s)
- Seung-Il Choi
- Corneal Dystrophy Research Institute and Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
43
|
Astrocyte dysfunction triggers neurodegeneration in a lysosomal storage disorder. Proc Natl Acad Sci U S A 2012; 109:E2334-42. [PMID: 22826245 DOI: 10.1073/pnas.1209577109] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The role of astrocytes in neurodegenerative processes is increasingly appreciated. Here we investigated the contribution of astrocytes to neurodegeneration in multiple sulfatase deficiency (MSD), a severe lysosomal storage disorder caused by mutations in the sulfatase modifying factor 1 (SUMF1) gene. Using Cre/Lox mouse models, we found that astrocyte-specific deletion of Sumf1 in vivo induced severe lysosomal storage and autophagy dysfunction with consequential cytoplasmic accumulation of autophagic substrates. Lysosomal storage in astrocytes was sufficient to induce degeneration of cortical neurons in vivo. Furthermore, in an ex vivo coculture assay, we observed that Sumf1(-/-) astrocytes failed to support the survival and function of wild-type cortical neurons, suggesting a non-cell autonomous mechanism for neurodegeneration. Compared with the astrocyte-specific deletion of Sumf1, the concomitant removal of Sumf1 in both neurons and glia in vivo induced a widespread neuronal loss and robust neuroinflammation. Finally, behavioral analysis of mice with astrocyte-specific deletion of Sumf1 compared with mice with Sumf1 deletion in both astrocytes and neurons allowed us to link a subset of neurological manifestations of MSD to astrocyte dysfunction. This study indicates that astrocytes are integral components of the neuropathology in MSD and that modulation of astrocyte function may impact disease course.
Collapse
|
44
|
Luo R, Sanders S, Tian Y, Voineagu I, Huang N, Chu S, Klei L, Cai C, Ou J, Lowe J, Hurles M, Devlin B, State M, Geschwind D. Genome-wide transcriptome profiling reveals the functional impact of rare de novo and recurrent CNVs in autism spectrum disorders. Am J Hum Genet 2012; 91:38-55. [PMID: 22726847 PMCID: PMC3397271 DOI: 10.1016/j.ajhg.2012.05.011] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 04/06/2012] [Accepted: 05/08/2012] [Indexed: 12/15/2022] Open
Abstract
Copy-number variants (CNVs) are a major contributor to the pathophysiology of autism spectrum disorders (ASDs), but the functional impact of CNVs remains largely unexplored. Because brain tissue is not available from most samples, we interrogated gene expression in lymphoblasts from 244 families with discordant siblings in the Simons Simplex Collection in order to identify potentially pathogenic variation. Our results reveal that the overall frequency of significantly misexpressed genes (which we refer to here as outliers) identified in probands and unaffected siblings does not differ. However, in probands, but not their unaffected siblings, the group of outlier genes is significantly enriched in neural-related pathways, including neuropeptide signaling, synaptogenesis, and cell adhesion. We demonstrate that outlier genes cluster within the most pathogenic CNVs (rare de novo CNVs) and can be used for the prioritization of rare CNVs of potentially unknown significance. Several nonrecurrent CNVs with significant gene-expression alterations are identified (these include deletions in chromosomal regions 3q27, 3p13, and 3p26 and duplications at 2p15), suggesting that these are potential candidate ASD loci. In addition, we identify distinct expression changes in 16p11.2 microdeletions, 16p11.2 microduplications, and 7q11.23 duplications, and we show that specific genes within the 16p CNV interval correlate with differences in head circumference, an ASD-relevant phenotype. This study provides evidence that pathogenic structural variants have a functional impact via transcriptome alterations in ASDs at a genome-wide level and demonstrates the utility of integrating gene expression with mutation data for the prioritization of genes disrupted by potentially pathogenic mutations.
Collapse
Affiliation(s)
- Rui Luo
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephan J. Sanders
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Program on Human Genetics and Genomics, Child Study Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yuan Tian
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Interdepartmental PhD Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Irina Voineagu
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ni Huang
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Su H. Chu
- Department of Statistics, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Lambertus Klei
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaochao Cai
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Biostatistics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jing Ou
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jennifer K. Lowe
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Matthew W. State
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Program on Human Genetics and Genomics, Child Study Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daniel H. Geschwind
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
45
|
Lieberman AP, Puertollano R, Raben N, Slaugenhaupt S, Walkley SU, Ballabio A. Autophagy in lysosomal storage disorders. Autophagy 2012; 8:719-30. [PMID: 22647656 PMCID: PMC3378416 DOI: 10.4161/auto.19469] [Citation(s) in RCA: 306] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Lysosomes are ubiquitous intracellular organelles that have an acidic internal pH, and play crucial roles in cellular clearance. Numerous functions depend on normal lysosomes, including the turnover of cellular constituents, cholesterol homeostasis, downregulation of surface receptors, inactivation of pathogenic organisms, repair of the plasma membrane and bone remodeling. Lysosomal storage disorders (LSDs) are characterized by progressive accumulation of undigested macromolecules within the cell due to lysosomal dysfunction. As a consequence, many tissues and organ systems are affected, including brain, viscera, bone and cartilage. The progressive nature of phenotype development is one of the hallmarks of LSDs. In recent years biochemical and cell biology studies of LSDs have revealed an ample spectrum of abnormalities in a variety of cellular functions. These include defects in signaling pathways, calcium homeostasis, lipid biosynthesis and degradation and intracellular trafficking. Lysosomes also play a fundamental role in the autophagic pathway by fusing with autophagosomes and digesting their content. Considering the highly integrated function of lysosomes and autophagosomes it was reasonable to expect that lysosomal storage in LSDs would have an impact upon autophagy. The goal of this review is to provide readers with an overview of recent findings that have been obtained through analysis of the autophagic pathway in several types of LSDs, supporting the idea that LSDs could be seen primarily as "autophagy disorders."
Collapse
Affiliation(s)
- Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI USA
| | | | | | | | | | | |
Collapse
|
46
|
Arteaga-Solis E, Settembre C, Ballabio A, Karsenty G. Sulfatases are determinants of alveolar formation. Matrix Biol 2012; 31:253-60. [PMID: 22366163 DOI: 10.1016/j.matbio.2012.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 02/07/2012] [Accepted: 02/10/2012] [Indexed: 10/28/2022]
Abstract
Alveolar formation or alveolarization is orchestrated by a finely regulated and complex interaction between growth factors and extracellular matrix proteins. The lung parenchyma contains various extracellular matrix proteins including proteoglycans, which are composed of glycosaminoglycans (GAGs) linked to a protein core. Although GAGs are known to regulate growth factor distribution and activity according to their degree of sulfation the role of sulfated GAG in the respiratory system is not well understood. The degree of sulfation of GAGs is regulated in part, by sulfatases that remove sulfate groups. In vertebrates, the enzyme Sulfatase-Modifying Factor 1 (Sumf1) activates all sulfatases. Here we utilized mice lacking Sumf1(-/-) to study the importance of proteoglycan desulfation in lung development. The Sumf1(-/-) mice have normal lungs up until the onset of alveolarization at post-natal day 5 (P5). We detected increased deposition of sulfated GAG throughout the lung parenchyma and a decrease in alveolar septa formation. Moreover, stereological analysis showed that the alveolar volume is 20% larger in Sumf1(-/-) as compared to wild type (WT) mice at P10 and P30. Additionally, pulmonary function test was consistent with increased alveolar volume. Genetic experiments demonstrate that in Sumf1(-/-) mice arrest of alveolarization is independent of fibroblast growth factor signaling. In turn, the Sumf1(-/-) mice have increased transforming growth factor β (TGFβ) signaling and in vivo injection of TGFβ neutralizing antibody leads to normalization of alveolarization. Thus, absence of sulfatase activity increases sulfated GAG deposition in the lungs causing deregulation of TGFβ signaling and arrest of alveolarization.
Collapse
Affiliation(s)
- Emilio Arteaga-Solis
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | | | | | | |
Collapse
|
47
|
de Pablo-Latorre R, Saide A, Polishhuck EV, Nusco E, Fraldi A, Ballabio A. Impaired parkin-mediated mitochondrial targeting to autophagosomes differentially contributes to tissue pathology in lysosomal storage diseases. Hum Mol Genet 2012; 21:1770-81. [PMID: 22215441 PMCID: PMC3313794 DOI: 10.1093/hmg/ddr610] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Dysfunctional mitochondria are a well-known disease hallmark. The accumulation of aberrant mitochondria can alter cell homeostasis, thus resulting in tissue degeneration. Lysosomal storage disorders (LSDs) are a group of inherited diseases characterized by the buildup of undegraded material inside the lysosomes that leads to autophagic-lysosomal dysfunction. In LSDs, autophagic stress has been associated to mitochondrial accumulation and dysfunction. However, the mechanisms underlying mitochondrial aberrations and how these are involved in tissue pathogenesis remain largely unexplored. In normal conditions, mitochondrial clearance occurs by mitophagy, a selective form of autophagy, which relies on a parkin-mediated mitochondrial priming and subsequent sequestration by autophagosomes. Here, we performed a detailed analysis of key steps of mitophagy in a mouse model of multiple sulfatase deficiency (MSD), a severe type of LSD characterized by both neurological and systemic involvement. We demonstrated that in MSD liver reduced parkin levels resulted in inefficient mitochondrial priming, thus contributing to the accumulation of giant mitochondria that are located outside autophagic vesicles ultimately leading to cytochrome c release and apoptotic cell death. Morphological and functional changes were also observed in mitochondria from MSD brain but these were not directly associated with neuronal cell loss, suggesting a secondary contribution of mitochondria to neurodegeneration. Together, these data shed new light on the mechanisms underlying mitochondrial dysfunction in LSDs and on their tissue-specific differential contribution to the pathogenesis of this group of metabolic disorders.
Collapse
|
48
|
Medina DL, Fraldi A, Bouche V, Annunziata F, Mansueto G, Spampanato C, Puri C, Pignata A, Martina JA, Sardiello M, Palmieri M, Polishchuk R, Puertollano R, Ballabio A. Transcriptional activation of lysosomal exocytosis promotes cellular clearance. Dev Cell 2011; 21:421-30. [PMID: 21889421 PMCID: PMC3173716 DOI: 10.1016/j.devcel.2011.07.016] [Citation(s) in RCA: 553] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 05/09/2011] [Accepted: 07/28/2011] [Indexed: 11/30/2022]
Abstract
Lysosomes are cellular organelles primarily involved in degradation and recycling processes. During lysosomal exocytosis, a Ca2+-regulated process, lysosomes are docked to the cell surface and fuse with the plasma membrane (PM), emptying their content outside the cell. This process has an important role in secretion and PM repair. Here we show that the transcription factor EB (TFEB) regulates lysosomal exocytosis. TFEB increases the pool of lysosomes in the proximity of the PM and promotes their fusion with PM by raising intracellular Ca2+ levels through the activation of the lysosomal Ca2+ channel MCOLN1. Induction of lysosomal exocytosis by TFEB overexpression rescued pathologic storage and restored normal cellular morphology both in vitro and in vivo in lysosomal storage diseases (LSDs). Our data indicate that lysosomal exocytosis may directly modulate cellular clearance and suggest an alternative therapeutic strategy for disorders associated with intracellular storage.
Collapse
Affiliation(s)
- Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Via P. Castellino 111, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schultz ML, Tecedor L, Chang M, Davidson BL. Clarifying lysosomal storage diseases. Trends Neurosci 2011; 34:401-10. [PMID: 21723623 DOI: 10.1016/j.tins.2011.05.006] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 05/17/2011] [Accepted: 05/27/2011] [Indexed: 11/29/2022]
Abstract
Lysosomal storage diseases (LSDs) are a class of metabolic disorders caused by mutations in proteins critical for lysosomal function. Such proteins include lysosomal enzymes, lysosomal integral membrane proteins, and proteins involved in the post-translational modification and trafficking of lysosomal proteins. There are many recognized forms of LSDs and, although individually rare, their combined prevalence is estimated to be 1 in 8000 births. Over two-thirds of LSDs involve central nervous system (CNS) dysfunction (progressive cognitive and motor decline) and these symptoms are often the most debilitating. Although the genetic basis for these disorders is clear and the biochemistry of the proteins well understood, the cellular mechanisms by which deficiencies in these proteins disrupt neuronal viability remain ambiguous. In this review, we provide an overview of the widespread cellular perturbations occurring in LSDs, how they might be linked and interventions that may specifically or globally correct those defects.
Collapse
Affiliation(s)
- Mark L Schultz
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
50
|
Abstract
Autophagy is a basic cell biological process ongoing under physiologic circumstances in almost all cell types of the human organism and upregulated by various stress conditions including those leading to inflammation. Since autophagy affects the effector cells of innate and adaptive immunity mediating the inflammatory response, its activity in these cells influences the antimicrobial response, the development of an effective cognate immune defense, and the course of the normal sterile inflammatory reactions. The level of autophagic activity may determine whether tissue cells die by apoptosis, necrosis, or through autophagy, and, as a consequence, whether the clearance of these dying cells is a silent process or results in an inflammatory response. Loss or decreased autophagy may lead to necrotic death that can initiate an inflammatory reaction in phagocytes through their surface and cytosolic receptors. Engulfment of certain cells dying through autophagy can activate the inflammasome. The intertwining regulatory connections between inflammation and immunity extend to pathologic conditions including chronic inflammatory diseases, autoimmunity and cancer.
Collapse
Affiliation(s)
- László Fésüs
- Department of Biochemistry and Molecular Biology, The Apoptosis and Genomics Research Group of the Hungarian Academy of Sciences, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | |
Collapse
|