1
|
Zhao JQ, Feng BY, Ye ZL, Ma XY, Du JZ, Li JM, Wu WL, Gao JJ, Li SJ, Peng SY, Huai JS, Ge LH, Lu CB. Activation of D2-like dopamine receptors improves the neuronal network and cognitive function of PPT1KI mice. Acta Pharmacol Sin 2025; 46:338-352. [PMID: 39284877 PMCID: PMC11747101 DOI: 10.1038/s41401-024-01377-7] [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: 01/25/2024] [Accepted: 08/08/2024] [Indexed: 01/22/2025]
Abstract
Palmitoyl-protein thioesterase 1 (PPT1) is a lysosomal depalmitoylation enzyme that mediates protein posttranslational modifications. Loss-of-function mutation of PPT1 causes a failure of the lysosomal degradation of palmitoylated proteins and results in a congenital disease characterized by progressive neuronal degeneration referred to as infantile neuronal ceroid lipofuscinosis (INCL). A mouse knock-in model of PPT1 (PPT1-KI) was established by introducing the R151X mutation into exon 5 of the PPT1 gene, which exhibited INCL-like pathological lesions. We previously reported that hippocampal γ oscillations were impaired in PPT1 mice. Hippocampal γ oscillations can be enhanced by selective activation of the dopamine D4 receptor (DR4), a dopamine D2-like receptor. In this study, we investigated the changes in DR expression and the effects of dopamine and various DR agonists on neural network activity, cognition and motor function in PPT1KI mice. Cognition and motor defects were evaluated via Y-maze, novel object recognition and rotarod tests. Extracellular field potentials were elicited in hippocampal slices, and neuronal network oscillations in the gamma frequency band (γ oscillations) were induced by perfusion with kainic acid (200 nM). PPT1KI mice displayed progressive impairments in γ oscillations and hippocampus-related memory, as well as abnormal expression profiles of dopamine receptors with preserved expression of DR1 and 3, increased membrane expression of DR4 and decreased DR2 levels. The immunocytochemistry analysis revealed the colocalization of PPT1 with DR4 or DR2 in the soma and large dendrites of both WT and PPT1KI mice. Immunoprecipitation confirmed the interaction between PPT1 and DR4 or DR2. The impaired γ oscillations and cognitive functions were largely restored by the application of exogenous dopamine, the selective DR2 agonist quinpirole or the DR4 agonist A412997. Furthermore, the administration of A412997 (0.5 mg/kg, i.p.) significantly upregulated the activity of CaMKII in the hippocampus of 5-month-old PPT1KI mice. Collectively, these results suggest that the activation of D2-like dopamine receptors improves cognition and network activity in PPT1KI mice and that specific DR subunits may be potential targets for the intervention of neurodegenerative disorders, such as INCL.
Collapse
Affiliation(s)
- Jun-Qiang Zhao
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China
- School of Medical Imaging, Xinxiang Medical University, Xinxiang, 435000, China
| | - Bing-Yan Feng
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China
| | - Zhen-Li Ye
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Xiao-Yin Ma
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Jing-Zhi Du
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Jun-Mei Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Wan-Liu Wu
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China
| | - Jing-Jing Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Song-Ji Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Shi-Yong Peng
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Ji-Sen Huai
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Li-Hao Ge
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China.
| | - Cheng-Biao Lu
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China.
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China.
| |
Collapse
|
2
|
Ziółkowska EA, Jansen MJ, Williams LL, Wang SH, Eultgen EM, Takahashi K, Le SQ, Nelvagal HR, Sharma J, Sardiello M, DeBosch BJ, Dickson PI, Anderson JB, Sax SE, Wright CM, Bradley RP, Whiteman IT, Makita T, Grider JR, Sands MS, Heuckeroth RO, Cooper JD. Gene therapy ameliorates bowel dysmotility and enteric neuron degeneration and extends survival in lysosomal storage disorder mouse models. Sci Transl Med 2025; 17:eadj1445. [PMID: 39813314 DOI: 10.1126/scitranslmed.adj1445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2024] [Accepted: 10/24/2024] [Indexed: 01/18/2025]
Abstract
Children with neurodegenerative disease often have debilitating gastrointestinal symptoms. We hypothesized that this may be due at least in part to underappreciated degeneration of neurons in the enteric nervous system (ENS), the master regulator of bowel function. To test this hypothesis, we evaluated mouse models of neuronal ceroid lipofuscinosis type 1 and 2 (CLN1 and CLN2 disease, respectively), neurodegenerative lysosomal storage disorders caused by deficiencies in palmitoyl protein thioesterase-1 and tripeptidyl peptidase-1, respectively. Both mouse lines displayed slow bowel transit in vivo that worsened with age. Although the ENS appeared to develop normally in these mice, there was a progressive and profound loss of myenteric plexus neurons accompanied by changes in enteric glia in adult mice. Similar pathology was evident in colon autopsy material from a child with CLN1 disease. Neonatal administration of adeno-associated virus-mediated gene therapy prevented bowel transit defects, ameliorated loss of enteric neurons, and extended survival in mice. Treatment after weaning was less effective than treating neonatally but still extended the lifespan of CLN1 disease mice. These data provide proof-of-principle evidence of ENS degeneration in two lysosomal storage diseases and suggest that gene therapy can ameliorate ENS disease, also improving survival.
Collapse
Affiliation(s)
- Ewa A Ziółkowska
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J Jansen
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Letitia L Williams
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Sophie H Wang
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Elizabeth M Eultgen
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Keigo Takahashi
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Steven Q Le
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Hemanth R Nelvagal
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Jaiprakash Sharma
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Marco Sardiello
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Brian J DeBosch
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Patricia I Dickson
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Jessica B Anderson
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sophie E Sax
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christina M Wright
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca P Bradley
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ineka T Whiteman
- Batten Disease Support, Research and Advocacy Foundation (US), P.O. Box 30049, Gahanna, OH 43230, USA
- Batten Disease Support and Research Association (Australia), 74 McLachlan Avenue, Shelly Beach, NSW 2261, Australia
| | - Takako Makita
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John R Grider
- Department of Physiology and Biophysics, Division of Gastroenterology, VCU Program in Enteric Neuromuscular Sciences (VPENS), Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Mark S Sands
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Robert O Heuckeroth
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan D Cooper
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
3
|
Grant MT, Nelvagal HR, Tecos M, Hamed A, Swanson K, Cooper JD, Vrecenak JD. Cellular trafficking and fate mapping of cells within the nervous system after in utero hematopoietic cell transplantation. Commun Biol 2024; 7:1624. [PMID: 39638879 PMCID: PMC11621337 DOI: 10.1038/s42003-024-06847-6] [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: 12/09/2022] [Accepted: 09/05/2024] [Indexed: 12/07/2024] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) utilizes fetal immune tolerance to achieve durable chimerism without conditioning or immunosuppression during a unique window in fetal development. Though donor cells have been observed within the nervous system following in utero injection, the timeline and distribution of cellular trafficking across the blood-brain barrier following IUHCT is not well understood. We injected 20 × 106 adult bone marrow mononuclear cells intravenously at gestational age (GA) 12-17 days and found that donor cells were maximally concentrated in the brain with treatment between GA 13-14. Donor cell engraftment persisted within the brain at every timepoint analyzed and concentrated within the hindbrain with significantly more grafted cells than in the forebrain. Additionally, transplanted cells terminally differentiated into various nervous system cellular morphologies and also populated the enteric nervous system. This study is the first to document the timeline and distribution of donor cell trafficking into the immune-protected nervous system and serves as a foundation for the application of IUHCT to treat neurogenetic diseases.
Collapse
Affiliation(s)
- Matthew T Grant
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Hemanth Ramesh Nelvagal
- Washington University in St. Louis School of Medicine, Department of Pediatrics, Division of Genetics and Genomics, St. Louis, MO, USA
- University College London, School of Pharmacy, Department of Pharmacology, London, UK
| | - Maria Tecos
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Amal Hamed
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Kerry Swanson
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Jonathan D Cooper
- Washington University in St. Louis School of Medicine, Department of Pediatrics, Division of Genetics and Genomics, St. Louis, MO, USA
| | - Jesse D Vrecenak
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA.
| |
Collapse
|
4
|
Massaro G, Geard AF, Nelvagal HR, Gore K, Clemo NK, Waddington SN, Rahim AA. Comparison of different promoters to improve AAV vector-mediated gene therapy for neuronopathic Gaucher disease. Hum Mol Genet 2024; 33:1467-1480. [PMID: 38757200 PMCID: PMC11336133 DOI: 10.1093/hmg/ddae081] [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: 04/01/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
Gaucher Disease (GD) is an inherited metabolic disorder caused by mutations in the GBA1 gene. It can manifest with severe neurodegeneration and visceral pathology. The most acute neuronopathic form (nGD), for which there are no curative therapeutic options, is characterised by devastating neuropathology and death during infancy. In this study, we investigated the therapeutic benefit of systemically delivered AAV9 vectors expressing the human GBA1 gene at two different doses comparing a neuronal-selective promoter with ubiquitous promoters. Our results highlight the importance of a careful evaluation of the promoter sequence used in gene delivery vectors, suggesting a neuron-targeted therapy leading to high levels of enzymatic activity in the brain but lower GCase expression in the viscera, might be the optimal therapeutic strategy for nGD.
Collapse
Affiliation(s)
- Giulia Massaro
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Amy F Geard
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand Medical, School, 7 York Road, Parktown 2193, South Africa
| | - Hemanth R Nelvagal
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Katrina Gore
- Apollo Therapeutics, Stevenage Bioscience Catalyst, 50-60 Station Road, Cambridge, CB1 2JH, United Kingdom
| | - Nadine K Clemo
- Apollo Therapeutics, Stevenage Bioscience Catalyst, 50-60 Station Road, Cambridge, CB1 2JH, United Kingdom
| | - Simon N Waddington
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand Medical, School, 7 York Road, Parktown 2193, South Africa
- UCL EGA Institute for Women's Health, University College London, Medical School Building, 74 Huntley Street, London, WC1E 6AU, United Kingdom
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, 29-38 Brunswick Square, London, WC1N 1AX, United Kingdom
| |
Collapse
|
5
|
Takahashi K, Rensing NR, Eultgen EM, Wang SH, Nelvagal HR, Le SQ, Roberts MS, Doray B, Han EB, Dickson PI, Wong M, Sands MS, Cooper JD. GABAergic interneurons contribute to the fatal seizure phenotype of CLN2 disease mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587276. [PMID: 38585903 PMCID: PMC10996664 DOI: 10.1101/2024.03.29.587276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
GABAergic interneuron deficits have been implicated in the epileptogenesis of multiple neurological diseases. While epileptic seizures are a key clinical hallmark of CLN2 disease, a childhood-onset neurodegenerative lysosomal storage disorder caused by a deficiency of tripeptidyl peptidase 1 (TPP1), the etiology of these seizures remains elusive. Given that Cln2 R207X/R207X mice display fatal spontaneous seizures and an early loss of several cortical interneuron populations, we hypothesized that those two events might be causally related. To address this hypothesis, we first generated an inducible transgenic mouse expressing lysosomal membrane-tethered TPP1 (TPP1LAMP1) on the Cln2 R207X/R207X genetic background to study the cell-autonomous effects of cell-type-specific TPP1 deficiency. We crossed the TPP1LAMP1 mice with Vgat-Cre mice to introduce interneuron-specific TPP1 deficiency. Vgat-Cre ; TPP1LAMP1 mice displayed storage material accumulation in several interneuron populations both in cortex and striatum, and increased susceptibility to die after PTZ-induced seizures. Secondly, to test the role of GABAergic interneuron activity in seizure progression, we selectively activated these cells in Cln2 R207X/R207X mice using Designer Receptor Exclusively Activated by Designer Drugs (DREADDs) in in Vgat-Cre : Cln2 R207X/R207X mice. EEG monitoring revealed that DREADD-mediated activation of interneurons via chronic deschloroclozapine administration accelerated the onset of spontaneous seizures and seizure-associated death in Vgat-Cre : Cln2 R207X/R207X mice, suggesting that modulating interneuron activity can exert influence over epileptiform abnormalities in CLN2 disease. Taken together, these results provide new mechanistic insights into the underlying etiology of seizures and premature death that characterize CLN2 disease.
Collapse
|
6
|
Taherzadeh M, Zhang E, Londono I, De Leener B, Wang S, Cooper JD, Kennedy TE, Morales CR, Chen Z, Lodygensky GA, Pshezhetsky AV. Severe central nervous system demyelination in Sanfilippo disease. Front Mol Neurosci 2023; 16:1323449. [PMID: 38163061 PMCID: PMC10756675 DOI: 10.3389/fnmol.2023.1323449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Chronic progressive neuroinflammation is a hallmark of neurological lysosomal storage diseases, including mucopolysaccharidosis III (MPS III or Sanfilippo disease). Since neuroinflammation is linked to white matter tract pathology, we analyzed axonal myelination and white matter density in the mouse model of MPS IIIC HgsnatP304L and post-mortem brain samples of MPS III patients. Methods Brain and spinal cord tissues of human MPS III patients, 6-month-old HgsnatP304L mice and age- and sex-matching wild type mice were analyzed by immunofluorescence to assess levels of myelin-associated proteins, primary and secondary storage materials, and levels of microgliosis. Corpus callosum (CC) region was studied by transmission electron microscopy to analyze axon myelination and morphology of oligodendrocytes and microglia. Mouse brains were analyzed ex vivo by high-filed MRI using Diffusion Basis Spectrum Imaging in Python-Diffusion tensor imaging algorithms. Results Analyses of CC and spinal cord tissues by immunohistochemistry revealed substantially reduced levels of myelin-associated proteins including Myelin Basic Protein, Myelin Associated Glycoprotein, and Myelin Oligodendrocyte Glycoprotein. Furthermore, ultrastructural analyses revealed disruption of myelin sheath organization and reduced myelin thickness in the brains of MPS IIIC mice and human MPS IIIC patients compared to healthy controls. Oligodendrocytes (OLs) in the CC of MPS IIIC mice were scarce, while examination of the remaining cells revealed numerous enlarged lysosomes containing heparan sulfate, GM3 ganglioside or "zebra bodies" consistent with accumulation of lipids and myelin fragments. In addition, OLs contained swollen mitochondria with largely dissolved cristae, resembling those previously identified in the dysfunctional neurons of MPS IIIC mice. Ex vivo Diffusion Basis Spectrum Imaging revealed compelling signs of demyelination (26% increase in radial diffusivity) and tissue loss (76% increase in hindered diffusivity) in CC of MPS IIIC mice. Discussion Our findings demonstrate an important role for white matter injury in the pathophysiology of MPS III. This study also defines specific parameters and brain regions for MRI analysis and suggests that it may become a crucial non-invasive method to evaluate disease progression and therapeutic response.
Collapse
Affiliation(s)
- Mahsa Taherzadeh
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Erjun Zhang
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Irene Londono
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Benjamin De Leener
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
- NeuroPoly Lab, Institute of Biomedical Engineering, Department of Computer Engineering and Software Engineering, École Polytechnique de Montréal, Montreal, QC, Canada
| | - Sophie Wang
- Pediatric Storage Disorders Laboratory (PSDL), Departments of Pediatrics, Genetics and Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory (PSDL), Departments of Pediatrics, Genetics and Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Timothy E. Kennedy
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Carlos R. Morales
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Zesheng Chen
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Gregory A. Lodygensky
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
| | - Alexey V. Pshezhetsky
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Centre, University of Montreal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
7
|
Sleat DE, Banach-Petrosky W, Larrimore KE, Nemtsova Y, Wiseman JA, Najafi A, Johnson D, Poole TA, Takahashi K, Cooper JD, Lobel P. A mouse mutant deficient in both neuronal ceroid lipofuscinosis-associated proteins CLN3 and TPP1. J Inherit Metab Dis 2023; 46:720-734. [PMID: 37078466 PMCID: PMC10330656 DOI: 10.1002/jimd.12619] [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: 01/25/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Late-infantile neuronal ceroid lipofuscinosis (LINCL) and juvenile neuronal ceroid lipofuscinosis (JNCL) are inherited neurodegenerative diseases caused by mutations in the genes encoding lysosomal proteins tripeptidyl peptidase 1 (TPP1) and CLN3 protein, respectively. TPP1 is well-understood and, aided by animal models that accurately recapitulate the human disease, enzyme replacement therapy has been approved and other promising therapies are emerging. In contrast, there are no effective treatments for JNCL, partly because the function of the CLN3 protein remains unknown but also because animal models have attenuated disease and lack robust survival phenotypes. Mouse models for LINCL and JNCL, with mutations in Tpp1 and Cln3, respectively, have been thoroughly characterized but the phenotype of a double Cln3/Tpp1 mutant remains unknown. We created this double mutant and find that its phenotype is essentially indistinguishable from the single Tpp1-/- mutant in terms of survival and brain pathology. Analysis of brain proteomic changes in the single Tpp1-/- and double Cln3-/- ;Tpp1-/- mutants indicates largely overlapping sets of altered proteins and reinforces earlier studies that highlight GPNMB, LYZ2, and SERPINA3 as promising biomarker candidates in LINCL while several lysosomal proteins including SMPD1 and NPC1 appear to be altered in the Cln3-/- animals. An unexpected finding was that Tpp1 heterozygosity significantly decreased lifespan of the Cln3-/- mouse. The truncated survival of this mouse model makes it potentially useful in developing therapies for JNCL using survival as an endpoint. In addition, this model may also provide insights into CLN3 protein function and its potential functional interactions with TPP1.
Collapse
Affiliation(s)
- David E. Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers University, Piscataway, NJ, United States of America
| | - Whitney Banach-Petrosky
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Katherine E. Larrimore
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Yuliya Nemtsova
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Jennifer A. Wiseman
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Allison Najafi
- The Lundquist Institute (formerly Los Angeles Biomedical Research Institute), Harbor-UCLA Medical Center, and David Geffen School of Medicine, University of California, Los Angeles, Torrance, CA United States of America
| | - Dymonn Johnson
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Timothy A. Poole
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Keigo Takahashi
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Jonathan D. Cooper
- The Lundquist Institute (formerly Los Angeles Biomedical Research Institute), Harbor-UCLA Medical Center, and David Geffen School of Medicine, University of California, Los Angeles, Torrance, CA United States of America
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
- Genetics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
- Neurology, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers University, Piscataway, NJ, United States of America
| |
Collapse
|
8
|
Takahashi K, Eultgen EM, Wang SH, Rensing NR, Nelvagal HR, Dearborn JT, Danos O, Buss N, Sands MS, Wong M, Cooper JD. Gene therapy ameliorates spontaneous seizures associated with cortical neuron loss in a Cln2R207X mouse model. J Clin Invest 2023; 133:e165908. [PMID: 37104037 PMCID: PMC10266778 DOI: 10.1172/jci165908] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/25/2023] [Indexed: 04/28/2023] Open
Abstract
Although a disease-modifying therapy for classic late infantile neuronal ceroid lipofuscinosis (CLN2 disease) exists, poor understanding of cellular pathophysiology has hampered the development of more effective and persistent therapies. Here, we investigated the nature and progression of neurological and underlying neuropathological changes in Cln2R207X mice, which carry one of the most common pathogenic mutations in human patients but are yet to be fully characterized. Long-term electroencephalography recordings revealed progressive epileptiform abnormalities, including spontaneous seizures, providing a robust, quantifiable, and clinically relevant phenotype. These seizures were accompanied by the loss of multiple cortical neuron populations, including those stained for interneuron markers. Further histological analysis revealed early localized microglial activation months before neuron loss started in the thalamocortical system and spinal cord, which was accompanied by astrogliosis. This pathology was more pronounced and occurred in the cortex before the thalamus or spinal cord and differed markedly from the staging seen in mouse models of other forms of neuronal ceroid lipofuscinosis. Neonatal administration of adeno-associated virus serotype 9-mediated gene therapy ameliorated the seizure and gait phenotypes and prolonged the life span of Cln2R207X mice, attenuating most pathological changes. Our findings highlight the importance of clinically relevant outcome measures for judging preclinical efficacy of therapeutic interventions for CLN2 disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Joshua T. Dearborn
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Mark S. Sands
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Jonathan D. Cooper
- Department of Pediatrics
- Department of Neurology, and
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Hughes MP, Nelvagal HR, Coombe-Tennant O, Smith D, Smith C, Massaro G, Poupon-Bejuit L, Platt FM, Rahim AA. A Novel Small NPC1 Promoter Enhances AAV-Mediated Gene Therapy in Mouse Models of Niemann-Pick Type C1 Disease. Cells 2023; 12:1619. [PMID: 37371089 PMCID: PMC10296851 DOI: 10.3390/cells12121619] [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: 03/20/2023] [Revised: 05/22/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Niemann-Pick disease type C1 (NP-C) is a prematurely lethal genetic lysosomal storage disorder with neurological and visceral pathology resulting from mutations in the NPC1 gene encoding the lysosomal transmembrane protein NPC1. There is currently no cure for NP-C, and the only disease modifying treatment, miglustat, slows disease progression but does not significantly attenuate neurological symptoms. AAV-mediated gene therapy is an attractive option for NP-C, but due to the large size of the human NPC1 gene, there may be packaging and truncation issues during vector manufacturing. One option is to reduce the size of DNA regulatory elements that are essential for gene expression, such as the promoter sequence. Here, we describe a novel small truncated endogenous NPC1 promoter that leads to high gene expression both in vitro and in vivo and compare its efficacy to other commonly used promoters. Following neonatal intracerebroventricular (ICV) injection into the CNS, this novel promoter provided optimal therapeutic efficacy compared to all other promoters including increased survival, improved behavioural phenotypes, and attenuated neuropathology in mouse models of NP-C. Taken together, we propose that this novel promoter can be extremely efficient in designing an optimised AAV9 vector for gene therapy for NP-C.
Collapse
Affiliation(s)
- Michael Paul Hughes
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Hemanth Ramesh Nelvagal
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Oliver Coombe-Tennant
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Dave Smith
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK; (D.S.); (C.S.); (F.M.P.)
| | - Claire Smith
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK; (D.S.); (C.S.); (F.M.P.)
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Laura Poupon-Bejuit
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Frances Mary Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK; (D.S.); (C.S.); (F.M.P.)
| | - Ahad Abdul Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| |
Collapse
|
10
|
Ostergaard JR, Nelvagal HR, Cooper JD. Top-down and bottom-up propagation of disease in the neuronal ceroid lipofuscinoses. Front Neurol 2022; 13:1061363. [PMID: 36438942 PMCID: PMC9692088 DOI: 10.3389/fneur.2022.1061363] [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: 10/04/2022] [Accepted: 10/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background The Neuronal Ceroid Lipofuscinoses (NCLs) may be considered distinct neurodegenerative disorders with separate underlying molecular causes resulting from monogenetic mutations. An alternative hypothesis is to consider the NCLs as related diseases that share lipofuscin pathobiology as the common core feature, but otherwise distinguished by different a) initial anatomic location, and b) disease propagation. Methods We have tested this hypothesis by comparing known differences in symptomatology and pathology of the CLN1 phenotype caused by complete loss of PPT1 function (i.e., the classical infantile form) and of the classical juvenile CLN3 phenotype. These two forms of NCL represent early onset and rapidly progressing vs. late onset and slowly progressing disease modalities respectively. Results Despite displaying similar pathological endpoints, the clinical phenotypes and the evidence of imaging and postmortem studies reveal strikingly different time courses and distributions of disease propagation. Data from CLN1 disease are indicative of disease propagation from the body, with early effects within the spinal cord and subsequently within the brainstem, the cerebral hemispheres, cerebellum and retina. In contrast, the retina appears to be the most vulnerable organ in CLN3, and the site where pathology is first present. Pathology subsequently is present in the occipital connectome of the CLN3 brain, followed by a top-down propagation in which cerebral and cerebellar atrophy in early adolescence is followed by involvement of the peripheral nerves in later adolescence/early twenties, with the extrapyramidal system also affected during this time course. Discussion The propagation of disease in these two NCLs therefore has much in common with the “Brain-first” vs. “Body-first” models of alpha-synuclein propagation in Parkinson's disease. CLN1 disease represents a “Body-first” or bottom-up disease propagation and CLN3 disease having a “Brain-first” and top-down propagation. It is noteworthy that the varied phenotypes of CLN1 disease, whether it starts in infancy (infantile form) or later in childhood (juvenile form), still fit with our proposed hypothesis of a bottom-up disease propagation in CLN1. Likewise, in protracted CLN3 disease, where both cognitive and motor declines are delayed, the initial manifestations of disease are also seen in the outer retinal layers, i.e., identical to classical Juvenile NCL disease.
Collapse
Affiliation(s)
- John R. Ostergaard
- Department of Child and Adolescencet, Centre for Rare Diseases, Aarhus, Denmark
- *Correspondence: John R. Ostergaard
| | - Hemanth R. Nelvagal
- Department of Pediatrics, School of Medicine, Washington University in St Louis, St Louis, MO, United States
- UCL School of Pharmacy, University College London, London, United Kingdom
| | - Jonathan D. Cooper
- Department of Pediatrics, School of Medicine, Washington University in St Louis, St Louis, MO, United States
- Department of Genetics, School of Medicine, Washington University in St Louis, St Louis, MO, United States
- Department of Neurology, School of Medicine, Washington University in St Louis, St Louis, MO, United States
| |
Collapse
|
11
|
Nelvagal HR, Eaton SL, Wang SH, Eultgen EM, Takahashi K, Le SQ, Nesbitt R, Dearborn JT, Siano N, Puhl AC, Dickson PI, Thompson G, Murdoch F, Brennan PM, Gray M, Greenhalgh SN, Tennant P, Gregson R, Clutton E, Nixon J, Proudfoot C, Guido S, Lillico SG, Whitelaw CBA, Lu JY, Hofmann SL, Ekins S, Sands MS, Wishart TM, Cooper JD. Cross-species efficacy of enzyme replacement therapy for CLN1 disease in mice and sheep. J Clin Invest 2022; 132:163107. [PMID: 36040802 PMCID: PMC9566914 DOI: 10.1172/jci163107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
CLN1 disease, also called infantile neuronal ceroid lipofuscinosis (NCL) or infantile Batten disease, is a fatal neurodegenerative lysosomal storage disorder resulting from mutations in the CLN1 gene encoding the soluble lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1). Therapies for CLN1 disease have proven challenging because of the aggressive disease course and the need to treat widespread areas of the brain and spinal cord. Indeed, gene therapy has proven less effective for CLN1 disease than for other similar lysosomal enzyme deficiencies. We therefore tested the efficacy of enzyme replacement therapy (ERT) by administering monthly infusions of recombinant human PPT1 (rhPPT1) to PPT1-deficient mice (Cln1-/-) and CLN1R151X sheep to assess how to potentially scale up for translation. In Cln1-/- mice, intracerebrovascular (i.c.v.) rhPPT1 delivery was the most effective route of administration, resulting in therapeutically relevant CNS levels of PPT1 activity. rhPPT1-treated mice had improved motor function, reduced disease-associated pathology, and diminished neuronal loss. In CLN1R151X sheep, i.c.v. infusions resulted in widespread rhPPT1 distribution and positive treatment effects measured by quantitative structural MRI and neuropathology. This study demonstrates the feasibility and therapeutic efficacy of i.c.v. rhPPT1 ERT. These findings represent a key step toward clinical testing of ERT in children with CLN1 disease and highlight the importance of a cross-species approach to developing a successful treatment strategy.
Collapse
Affiliation(s)
- Hemanth R. Nelvagal
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Samantha L. Eaton
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Sophie H. Wang
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth M. Eultgen
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Keigo Takahashi
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Steven Q. Le
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Rachel Nesbitt
- Department of Medicine, Washington University in St. Louis, School of Medicine, St .Louis, Missouri, USA
| | - Joshua T. Dearborn
- Department of Medicine, Washington University in St. Louis, School of Medicine, St .Louis, Missouri, USA
| | - Nicholas Siano
- Discovery Science Division, Amicus Therapeutics Inc., Philadelphia, Pennsylvania, USA
| | - Ana C. Puhl
- Collaborations Pharmaceuticals Inc., Lab 3510, Raleigh, North Carolina, USA
| | - Patricia I. Dickson
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Gerard Thompson
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, Edinburgh, Scotland, United Kingdom
- Department of Clinical Neurosciences, NHS Lothian, Edinburgh, Scotland, United Kingdom
| | - Fraser Murdoch
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Paul M. Brennan
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, Edinburgh, Scotland, United Kingdom
- Department of Clinical Neurosciences, NHS Lothian, Edinburgh, Scotland, United Kingdom
| | - Mark Gray
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Stephen N. Greenhalgh
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Peter Tennant
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Rachael Gregson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Eddie Clutton
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - James Nixon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Chris Proudfoot
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
- The Large Animal Research and Imaging Facility (LARIF), Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Stefano Guido
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Simon G. Lillico
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - C. Bruce A. Whitelaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Jui-Yun Lu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sandra L. Hofmann
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., Lab 3510, Raleigh, North Carolina, USA
| | - Mark S. Sands
- Department of Medicine, Washington University in St. Louis, School of Medicine, St .Louis, Missouri, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Thomas M. Wishart
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Easter Bush, Scotland, United Kingdom
| | - Jonathan D. Cooper
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Effects of chronic cannabidiol in a mouse model of naturally occurring neuroinflammation, neurodegeneration, and spontaneous seizures. Sci Rep 2022; 12:11286. [PMID: 35789177 PMCID: PMC9253004 DOI: 10.1038/s41598-022-15134-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Cannabidiol (CBD) has gained attention as a therapeutic agent and is purported to have immunomodulatory, neuroprotective, and anti-seizure effects. Here, we determined the effects of chronic CBD administration in a mouse model of CLN1 disease (Cln1-/-) that simultaneously exhibits neuroinflammation, neurodegeneration, and spontaneous seizures. Proteomic analysis showed that putative CBD receptors are expressed at similar levels in the brains of Cln1-/- mice compared to normal animals. Cln1-/- mice received an oral dose (100 mg/kg/day) of CBD for six months and were evaluated for changes in pathological markers of disease and seizures. Chronic cannabidiol administration was well-tolerated, high levels of CBD were detected in the brain, and markers of astrocytosis and microgliosis were reduced. However, CBD had no apparent effect on seizure frequency or neuron survival. These data are consistent with CBD having immunomodulatory effects. It is possible that a higher dose of CBD could also reduce neurodegeneration and seizure frequency.
Collapse
|
13
|
Takahashi K, Nelvagal HR, Lange J, Cooper JD. Glial Dysfunction and Its Contribution to the Pathogenesis of the Neuronal Ceroid Lipofuscinoses. Front Neurol 2022; 13:886567. [PMID: 35444603 PMCID: PMC9013902 DOI: 10.3389/fneur.2022.886567] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 01/05/2023] Open
Abstract
While significant efforts have been made in developing pre-clinical treatments for the neuronal ceroid lipofuscinoses (NCLs), many challenges still remain to bring children with NCLs a cure. Devising effective therapeutic strategies for the NCLs will require a better understanding of pathophysiology, but little is known about the mechanisms by which loss of lysosomal proteins causes such devastating neurodegeneration. Research into glial cells including astrocytes, microglia, and oligodendrocytes have revealed many of their critical functions in brain homeostasis and potential contributions to neurodegenerative diseases. Genetically modified mouse models have served as a useful platform to define the disease progression in the central nervous system across NCL subtypes, revealing a wide range of glial responses to disease. The emerging evidence of glial dysfunction questions the traditional “neuron-centric” view of NCLs, and would suggest that directly targeting glia in addition to neurons could lead to better therapeutic outcomes. This review summarizes the most up-to-date understanding of glial pathologies and their contribution to the pathogenesis of NCLs, and highlights some of the associated challenges that require further research.
Collapse
Affiliation(s)
- Keigo Takahashi
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Hemanth R. Nelvagal
- Department of Pharmacology, School of Pharmacy, University College London, London, United Kingdom
| | - Jenny Lange
- Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Genetics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Jonathan D. Cooper
| |
Collapse
|
14
|
Rebiai R, Rue E, Zaldua S, Nguyen D, Scesa G, Jastrzebski M, Foster R, Wang B, Jiang X, Tai L, Brady ST, van Breemen R, Givogri MI, Sands MS, Bongarzone ER. CRISPR-Cas9 Knock-In of T513M and G41S Mutations in the Murine β-Galactosyl-Ceramidase Gene Re-capitulates Early-Onset and Adult-Onset Forms of Krabbe Disease. Front Mol Neurosci 2022; 15:896314. [PMID: 35620447 PMCID: PMC9127972 DOI: 10.3389/fnmol.2022.896314] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
Krabbe Disease (KD) is a lysosomal storage disorder characterized by the genetic deficiency of the lysosomal enzyme β-galactosyl-ceramidase (GALC). Deficit or a reduction in the activity of the GALC enzyme has been correlated with the progressive accumulation of the sphingolipid metabolite psychosine, which leads to local disruption in lipid raft architecture, diffuse demyelination, astrogliosis, and globoid cell formation. The twitcher mouse, the most used animal model, has a nonsense mutation, which limits the study of how different mutations impact the processing and activity of GALC enzyme. To partially address this, we generated two new transgenic mouse models carrying point mutations frequently found in infantile and adult forms of KD. Using CRISPR-Cas9 gene editing, point mutations T513M (infantile) and G41S (adult) were introduced in the murine GALC gene and stable founders were generated. We show that GALC T513M/T513M mice are short lived, have the greatest decrease in GALC activity, have sharp increases of psychosine, and rapidly progress into a severe and lethal neurological phenotype. In contrast, GALC G41S/G41S mice have normal lifespan, modest decreases of GALC, and minimal psychosine accumulation, but develop adult mild inflammatory demyelination and slight declines in coordination, motor skills, and memory. These two novel transgenic lines offer the possibility to study the mechanisms by which two distinct GALC mutations affect the trafficking of mutated GALC and modify phenotypic manifestations in early- vs adult-onset KD.
Collapse
Affiliation(s)
- Rima Rebiai
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Emily Rue
- Department of Pharmaceutical Science, College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Steve Zaldua
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Duc Nguyen
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Giuseppe Scesa
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Martin Jastrzebski
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Robert Foster
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Bin Wang
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Xuntian Jiang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Leon Tai
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Scott T Brady
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Richard van Breemen
- Department of Pharmaceutical Science, College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Maria I Givogri
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States.,Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
15
|
Ostergaard JR. Gait phenotype in Batten disease: A marker of disease progression. Eur J Paediatr Neurol 2021; 35:1-7. [PMID: 34547583 DOI: 10.1016/j.ejpn.2021.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/03/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Gait impairment and its etiologic correlate has not previously been subject of special attention in Batten disease. METHODS In the present review, the clinical picture of gait phenotype during Batten disease course accompanied by descriptions of the known concomitant patho-anatomical changes is presented. RESULTS In CLN1 a non-rhythmic gait is seen around 1-1½ years of age. Shortly after, postural hypotonia and exaggerated tendon reflexes develop. The disease reaches a burnt-out stage during the third year of age and subsequently the children are almost without voluntary movements. The existing literature indicates that gait phenotype in CLN1 is caused by early involvement of the spinal interneurons followed by impact of the cortex and the cortico-spinal tracts. The earliest walking abnormality in children with CLN2 is a clumsy, ataxic, and spastic gait, which is in accordance with the existing imaging and histologic studies showing early involvement of the cerebellum and the cortico-spinal pathways. In CLN3, a reduction in walking speed is present at the age of 7-8 years. It occurs simultaneously with a reduction in the white matter microstructure and brain connectivity networks. Functional impairment of the basal ganglia contributing to a parkinsonian gait phenotype occurs in the mid-teens. In the late teens and early twenties involvement of the peripheral nerves, neurogenic musculoskeletal atrophy, loss of tendon reflexes and postural control are seen. CONCLUSION The progressively impaired gait function in Batten disease is related to timing of damage of distinct areas of the nervous system depending on subtype and is a powerful marker of disease progression.
Collapse
Affiliation(s)
- John R Ostergaard
- Centre for Rare Diseases, Department of Children & Youth, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus N, Denmark.
| |
Collapse
|
16
|
Nelvagal HR, Dearborn JT, Ostergaard JR, Sands MS, Cooper JD. Spinal manifestations of CLN1 disease start during the early postnatal period. Neuropathol Appl Neurobiol 2020; 47:251-267. [PMID: 32841420 PMCID: PMC7867600 DOI: 10.1111/nan.12658] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/29/2020] [Accepted: 08/25/2020] [Indexed: 01/28/2023]
Abstract
Aim To understand the progression of CLN1 disease and develop effective therapies we need to characterize early sites of pathology. Therefore, we performed a comprehensive evaluation of the nature and timing of early CLN1 disease pathology in the spinal cord, which appears especially vulnerable, and how this may affect behaviour. Methods We measured the spinal volume and neuronal number, and quantified glial activation, lymphocyte infiltration and oligodendrocyte maturation, as well as cytokine profile analysis during the early stages of pathology in Ppt1‐deficient (Ppt1−/−) mouse spinal cords. We then performed quantitative gait analysis and open‐field behaviour tests to investigate the behavioural correlates during this period. Results We detected significant microglial activation in Ppt1−/− spinal cords at 1 month. This was followed by astrocytosis, selective interneuron loss, altered spinal volumes and oligodendrocyte maturation at 2 months, before significant storage material accumulation and lymphocyte infiltration at 3 months. The same time course was apparent for inflammatory cytokine expression that was altered as early as one month. There was a transient early period at 2 months when Ppt1−/− mice had a significantly altered gait that resembles the presentation in children with CLN1 disease. This occurred before an anticipated decline in overall locomotor performance across all ages. Conclusion These data reveal disease onset 2 months (25% of life‐span) earlier than expected, while spinal maturation is still ongoing. Our multi‐disciplinary data provide new insights into the spatio‐temporal staging of CLN1 pathogenesis during ongoing postnatal maturation, and highlight the need to deliver therapies during the presymptomatic period.
Collapse
Affiliation(s)
- H R Nelvagal
- Department of Pediatrics, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - J T Dearborn
- Department of Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - J R Ostergaard
- Centre for Rare Diseases, Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - M S Sands
- Department of Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, USA.,Department of Genetics, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - J D Cooper
- Department of Pediatrics, Washington University in St Louis, School of Medicine, St Louis, MO, USA.,Department of Genetics, Washington University in St Louis, School of Medicine, St Louis, MO, USA.,Department of Neurology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| |
Collapse
|