1
|
Wei X, Hormel TT, Renner L, Neuringer M, Jia Y. Wide-field OCT angiography for non-human primate retinal imaging. BIOMEDICAL OPTICS EXPRESS 2024; 15:4642-4654. [PMID: 39346973 PMCID: PMC11427193 DOI: 10.1364/boe.525839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 10/01/2024]
Abstract
Optical coherence tomography (OCT) is a well-established research tool for vision research in animal models capable of providing in vivo imaging of the retina. Structural OCT can be enhanced using OCT angiography (OCTA) processing in order to provide simultaneously acquired, automatically co-registered vascular information. Currently available OCT. Currently available OCTA lack either large field of view or high resolution. In this study we developed a wide-field (60-degree), high-resolution (10.5-µm optical transverse) and high-sensitivity (104-dB) OCTA-enabled system for non-human primate imaging and with it imaged multiple disease models, including models of age-related macular degeneration (AMD), Bardet-Biedl Syndrome (BBS), and the CLN7 variant of Batten disease. We demonstrate clear visualization of features including drusen, ellipsoid zone loss, vascular retinopathy, and retinal thinning in these eyes.
Collapse
Affiliation(s)
- Xiang Wei
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Tristan T Hormel
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Laurie Renner
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Martha Neuringer
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Yali Jia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
2
|
Mhlanga-Mutangadura T, Bullock G, Cerda-Gonzalez S, Katz ML. Neuronal Ceroid Lipofuscinosis in a Mixed-Breed Dog with a Splice Site Variant in CLN6. Genes (Basel) 2024; 15:661. [PMID: 38927597 PMCID: PMC11203140 DOI: 10.3390/genes15060661] [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: 04/30/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
A 23-month-old neutered male dog of unknown ancestry presented with a history of progressive neurological signs that included anxiety, cognitive impairment, tremors, seizure activity, ataxia, and pronounced visual impairment. The clinical signs were accompanied by global brain atrophy. Due to progression in the severity of disease signs, the dog was euthanized at 26 months of age. An examination of the tissues collected at necropsy revealed dramatic intracellular accumulations of autofluorescent inclusions in the brain, retina, and cardiac muscle. The inclusions were immunopositive for subunit c of mitochondrial ATP synthase, and their ultrastructural appearances were similar to those of lysosomal storage bodies that accumulate in some neuronal ceroid lipofuscinosis (NCL) diseases. The dog also exhibited widespread neuroinflammation. Based on these findings, the dog was deemed likely to have suffered from a form of NCL. A whole genome sequence analysis of the proband's DNA revealed a homozygous C to T substitution that altered the intron 3-exon 4 splice site of CLN6. Other mutations in CLN6 cause NCL diseases in humans and animals, including dogs. The CLN6 protein was undetectable with immunolabeling in the tissues of the proband. Based on the clinical history, fluorescence and electron-microscopy, immunohistochemistry, and molecular genetic findings, the disorder in this dog was classified as an NCL resulting from the absence of the CLN6 protein. Screening the dog's genome for a panel of breed-specific polymorphisms indicated that its ancestry included numerous breeds, with no single breed predominating. This suggests that the CLN6 disease variant is likely to be present in other mixed-breed dogs and at least some ancestral breeds, although it is likely to be rare since other cases have not been reported to date.
Collapse
Affiliation(s)
- Tendai Mhlanga-Mutangadura
- Canine Genetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (T.M.-M.); (G.B.)
| | - Garrett Bullock
- Canine Genetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (T.M.-M.); (G.B.)
| | | | - Martin L. Katz
- Canine Genetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (T.M.-M.); (G.B.)
- Neurodegenerative Diseases Research Laboratory, Department of Ophthalmology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
3
|
Koster KP, Fyke Z, Nguyen TTA, Niqula A, Noriega-González LY, Woolfrey KM, Dell’Acqua ML, Cologna SM, Yoshii A. Akap5 links synaptic dysfunction to neuroinflammatory signaling in a mouse model of infantile neuronal ceroid lipofuscinosis. Front Synaptic Neurosci 2024; 16:1384625. [PMID: 38798824 PMCID: PMC11116793 DOI: 10.3389/fnsyn.2024.1384625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Palmitoylation and depalmitoylation represent dichotomic processes by which a labile posttranslational lipid modification regulates protein trafficking and degradation. The depalmitoylating enzyme, palmitoyl-protein thioesterase 1 (PPT1), is associated with the devastating pediatric neurodegenerative condition, infantile neuronal ceroid lipofuscinosis (CLN1). CLN1 is characterized by the accumulation of autofluorescent lysosomal storage material (AFSM) in neurons and robust neuroinflammation. Converging lines of evidence suggest that in addition to cellular waste accumulation, the symptomology of CLN1 corresponds with disruption of synaptic processes. Indeed, loss of Ppt1 function in cortical neurons dysregulates the synaptic incorporation of the GluA1 AMPA receptor (AMPAR) subunit during a type of synaptic plasticity called synaptic scaling. However, the mechanisms causing this aberration are unknown. Here, we used the Ppt1-/- mouse model (both sexes) to further investigate how Ppt1 regulates synaptic plasticity and how its disruption affects downstream signaling pathways. To this end, we performed a palmitoyl-proteomic screen, which provoked the discovery that Akap5 is excessively palmitoylated at Ppt1-/- synapses. Extending our previous data, in vivo induction of synaptic scaling, which is regulated by Akap5, caused an excessive upregulation of GluA1 in Ppt1-/- mice. This synaptic change was associated with exacerbated disease pathology. Furthermore, the Akap5- and inflammation-associated transcriptional regulator, nuclear factor of activated T cells (NFAT), was sensitized in Ppt1-/- cortical neurons. Suppressing the upstream regulator of NFAT activation, calcineurin, with the FDA-approved therapeutic FK506 (Tacrolimus) modestly improved neuroinflammation in Ppt1-/- mice. These findings indicate that the absence of depalmitoylation stifles synaptic protein trafficking and contributes to neuroinflammation via an Akap5-associated mechanism.
Collapse
Affiliation(s)
- Kevin P. Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zach Fyke
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Thu T. A. Nguyen
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Amanda Niqula
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Kevin M. Woolfrey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Stephanie M. Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Akira Yoshii
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, United States
- Department of Neurology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
4
|
Keller SH, Johnson GS, Bullock G, Mhlanga-Mutangadura T, Schwartz M, Pattridge SG, Guo J, Kortz GD, Katz ML. Homozygous CNP Mutation and Neurodegeneration in Weimaraners: Myelin Abnormalities and Accumulation of Lipofuscin-like Inclusions. Genes (Basel) 2024; 15:246. [PMID: 38397235 PMCID: PMC10888007 DOI: 10.3390/genes15020246] [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: 01/25/2024] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
A progressive neurological disorder was observed in a male neutered Weimaraner. Clinical signs included fecal incontinence, lethargy, moderate paraparesis, proprioceptive pelvic limb ataxia, falling, cognitive decline, incoordination, decreased interest in food, changes in posture, and episodes of trance-like behavior. Neurologic signs were first observed at approximately 4 years, 10 months of age and progressed slowly. Magnetic resonance imaging showed generalized brain atrophy with areas of white matter pathology. Humane euthanasia was elected at 6 years, 7 months of age due to increasing severity of the neurological signs. Autofluorescent intracellular granules were observed in the cerebral and cerebellar cortexes, optic nerve, and cardiac muscle of the affected dog. These abnormal inclusions in the cerebral cortex and cardiac muscle immunolabeled with antibodies to mitochondrial ATP synthase subunit c protein, like that observed in the neuronal ceroid lipofuscinosis group of lysosomal storage diseases. Immunolabeling also demonstrated pronounced neuroinflammation in brain tissues. The ultrastructural appearances of the disease-related inclusion bodies in the brain and optic nerve were quite variable. The ultrastructure and locations of many of the inclusions in the nervous tissues suggested that they were derived, at least in part, from the myelin surrounding axons. The storage bodies in the cardiac muscle were located in mitochondria-rich regions and consisted of parallel arrays of membrane-like components interspersed with electron-dense flocculent material. The disease was characterized by pronounced abnormalities in the myelin of the brain and optic nerve consisting of distinctive areas of ballooning between the layers of myelin. The whole genome sequence generated from the affected dog contained a homozygous G-to-A missense mutation in CNP, which encodes proteins with CNPase enzyme activity and a structural role in myelin. The mutation predicts a Thr42Met amino acid sequence substitution. Genotyping of archived Weimaraner DNA samples identified an additional G > A variant homozygote with a clinical history and brain lesions similar to those of the proband. Of 304 Weimaraners and over 4000 other dogs of various breeds, the proband and the other Weimaraner that exhibited similar signs were the only two that were homozygous for the CNP missense variant. CNPase immunolabeling was widespread in brain tissues from normal dogs but was undetectable in the same tissues from the proband. Based on the clinical history, fluorescence and electron-microscopy, immunohistochemistry, and molecular genetic findings, the late-onset Weimaraner disorder likely results from the missense mutation that results in CNPase deficiency, leading to myelin abnormalities, accumulation of lysosomal storage bodies, and brain atrophy. Similar disorders have been associated with different CNP variants in Dalmatians and in human subjects.
Collapse
Affiliation(s)
- Stefan H. Keller
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (S.H.K.); (G.B.); (T.M.-M.); (S.G.P.); (J.G.)
| | - Gary S. Johnson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (S.H.K.); (G.B.); (T.M.-M.); (S.G.P.); (J.G.)
| | - Garrett Bullock
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (S.H.K.); (G.B.); (T.M.-M.); (S.G.P.); (J.G.)
| | - Tendai Mhlanga-Mutangadura
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (S.H.K.); (G.B.); (T.M.-M.); (S.G.P.); (J.G.)
| | - Malte Schwartz
- Summit Veterinary Referral Center, Tacoma, WA 98409, USA;
| | - Savannah G. Pattridge
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (S.H.K.); (G.B.); (T.M.-M.); (S.G.P.); (J.G.)
| | - Juyuan Guo
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (S.H.K.); (G.B.); (T.M.-M.); (S.G.P.); (J.G.)
| | - Gregg D. Kortz
- VCA Sacramento Veterinary Referral Center, Sacramento, CA 95827, USA;
| | - Martin L. Katz
- Neurodegenerative Diseases Research Laboratory, Department of Ophthalmology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
5
|
Burlina AP, Manara R, Gueraldi D. Lysosomal storage diseases. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:147-172. [PMID: 39322377 DOI: 10.1016/b978-0-323-99209-1.00008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Lysosomal storage disorders (LSDs) are a group of inherited metabolic diseases caused by dysfunction of the lysosomal system, with subsequent progressive accumulation of macromolecules, activation of inflammatory response, and cell death. Neurologic damage is almost always present, and it is usually degenerative. White matter (WM) involvement may be primary or secondary. Diseases with primary WM involvement are leukodystrophies, demyelinating (Krabbe disease and metachromatic leukodystrophy), and hypomyelinating leukodystrophies (free sialic acid storage disease, fucosidosis, and mucolipidosis type IV). LSDs with secondary WM involvement are classified as leukoencephalopathies and include gangliosidosis, mucopolysaccharidosis (MPS), ceroid neuronal lipofuscinosis, multiple sulfatase deficiency, alpha-mannosidosis, Pompe disease, and Fabry disease. Neurologic manifestations may overlap among LSDs and include developmental delays, motor, cognitive and speech impairments, seizures, visual failure, ataxia, and extrapyramidal signs. Most of LSDs are typically present in early or late infancy, but juvenile and adult forms also exist and are associated with predominantly neuropsychiatric and behavioral symptoms. The outcome of these disorders is generally poor and specific treatments (enzyme replacement therapy, hematopoietic stem cell transplantation, or gene therapy) are only available in a small number of them.
Collapse
Affiliation(s)
| | - Renzo Manara
- Neuroradiology Unit, Department of Neurosciences, University Hospital of Padova, Padova, Italy
| | - Daniela Gueraldi
- Division of Inherited Metabolic Diseases, University Hospital of Padova, Padova, Italy
| |
Collapse
|
6
|
Rus CM, Polla DL, Di Bucchianico S, Fischer S, Hartkamp J, Hartmann G, Alpagu Y, Cozma C, Zimmermann R, Bauer P. Neuronal progenitor cells-based metabolomics study reveals dysregulated lipid metabolism and identifies putative biomarkers for CLN6 disease. Sci Rep 2023; 13:18550. [PMID: 37899458 PMCID: PMC10613621 DOI: 10.1038/s41598-023-45789-7] [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: 03/14/2023] [Accepted: 10/24/2023] [Indexed: 10/31/2023] Open
Abstract
Neuronal ceroid lipofuscinosis 6 (CLN6) is a rare and fatal autosomal recessive disease primarily affecting the nervous system in children. It is caused by a pathogenic mutation in the CLN6 gene for which no therapy is available. Employing an untargeted metabolomics approach, we analyzed the metabolic changes in CLN6 subjects to see if this system could potentially yield biomarkers for diagnosis and monitoring disease progression. Neuronal-like cells were derived from human fibroblast lines from CLN6-affected subjects (n = 3) and controls (wild type, n = 3). These were used to assess the potential of a neuronal-like cell-based metabolomics approach to identify CLN6 distinctive and specific biomarkers. The most impacted metabolic profile is associated with sphingolipids, glycerophospholipids metabolism, and calcium signaling. Over 2700 spectral features were screened, and fifteen metabolites were identified that differed significantly between both groups, including the sphingolipids C16 GlcCer, C24 GlcCer, C24:1 GlcCer and glycerophospholipids PG 40:6 and PG 40:7. Of note, these fifteen metabolites were downregulated in the CLN6 disease group. This study is the first to analyze the metabolome of neuronal-like cells with a pathogenic mutation in the CLN6 gene and to provide insights into their metabolomic alterations. This could allow for the development of novel biomarkers for monitoring CLN6 disease.
Collapse
Affiliation(s)
- Corina-Marcela Rus
- Centogene GmbH, Am Strande 7, 18057, Rostock, Germany.
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, Albert-Einstein Straße 27, 18059, Rostock, Germany.
| | | | - Sebastiano Di Bucchianico
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, Albert-Einstein Straße 27, 18059, Rostock, Germany
- Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Department Life, Light & Matter, University of Rostock, Albert-Einstein Straße 25, 18059, Rostock, Germany
| | | | - Jörg Hartkamp
- Centogene GmbH, Am Strande 7, 18057, Rostock, Germany
| | | | - Yunus Alpagu
- Centogene GmbH, Am Strande 7, 18057, Rostock, Germany
| | - Claudia Cozma
- Centogene GmbH, Am Strande 7, 18057, Rostock, Germany
| | - Ralf Zimmermann
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, Albert-Einstein Straße 27, 18059, Rostock, Germany
- Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Department Life, Light & Matter, University of Rostock, Albert-Einstein Straße 25, 18059, Rostock, Germany
| | - Peter Bauer
- Centogene GmbH, Am Strande 7, 18057, Rostock, Germany
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| |
Collapse
|
7
|
Munesue Y, Ageyama N, Kimura N, Takahashi I, Nakayama S, Okabayashi S, Katakai Y, Koie H, Yagami KI, Ishii K, Tamaoka A, Yasutomi Y, Shimozawa N. Cynomolgus macaque model of neuronal ceroid lipofuscinosis type 2 disease. Exp Neurol 2023; 363:114381. [PMID: 36918063 DOI: 10.1016/j.expneurol.2023.114381] [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: 07/04/2022] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are autosomal-recessive fatal neurodegenerative diseases that occur in children and young adults, with symptoms including ataxia, seizures and visual impairment. We report the discovery of cynomolgus macaques carrying the CLN2/TPP1 variant and our analysis of whether the macaques could be a new non-human primate model for NCL type 2 (CLN2) disease. Three cynomolgus macaques presented progressive neuronal clinical symptoms such as limb tremors and gait disturbance after about 2 years of age. Morphological analyses using brain MRI at the endpoint of approximately 3 years of age revealed marked cerebellar and cerebral atrophy of the gray matter, with sulcus dilation, gyrus thinning, and ventricular enlargement. Histopathological analyses of three affected macaques revealed severe neuronal loss and degeneration in the cerebellar and cerebral cortices, accompanied by glial activation and/or changes in axonal morphology. Neurons observed throughout the central nervous system contained autofluorescent cytoplasmic pigments, which were identified as ceroid-lipofuscin based on staining properties, and the cerebral cortex examined by transmission electron microscopy had curvilinear profiles, the typical ultrastructural pattern of CLN2. These findings are commonly observed in all forms of NCL. DNA sequencing analysis identified a homozygous single-base deletion (c.42delC) of the CLN2/TPP1 gene, resulting in a frameshifted premature stop codon. Immunohistochemical analysis showed that tissue from the affected macaques lacked a detectable signal against TPP1, the product of the CLN2/TPP1 gene. Analysis for transmission of the CLN2/TPP1 mutated gene revealed that 47 (49.5%) and 48 (50.5%) of the 95 individuals genotyped in the CLN2-affected macaque family were heterozygous carriers and homozygous wild-type individuals, respectively. Thus, we identified cynomolgus macaques as a non-human primate model of CLN2 disease. The CLN2 macaques reported here could become a useful resource for research and the development of drugs and methods for treating CLN2 disease, which involves severe symptoms in humans.
Collapse
Affiliation(s)
- Yoshiko Munesue
- Division of Clinical Medicine, Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Naohide Ageyama
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Nobuyuki Kimura
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan; Department of Veterinary Associated Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime 794-8555, Japan
| | - Ichiro Takahashi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Shunya Nakayama
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan; Laboratory of Veterinary Physiology/Pathophysiology, Nihon University, College of Bioresource Science, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Sachi Okabayashi
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0843, Japan
| | - Yuko Katakai
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0843, Japan
| | - Hiroshi Koie
- Laboratory of Veterinary Physiology/Pathophysiology, Nihon University, College of Bioresource Science, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Ken-Ichi Yagami
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuhiro Ishii
- Division of Clinical Medicine, Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Akira Tamaoka
- Division of Clinical Medicine, Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan; Department of Molecular and Experimental Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Nobuhiro Shimozawa
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan.
| |
Collapse
|
8
|
Ikari N, Arakawa H. Identification of a mitochondrial targeting sequence in cathepsin D and its localization in mitochondria. Biochem Biophys Res Commun 2023; 655:25-34. [PMID: 36921448 DOI: 10.1016/j.bbrc.2023.03.016] [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: 02/21/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/10/2023]
Abstract
Cathepsin D (CTSD) is a major lysosomal protease harboring an N-terminal signal peptide (amino acids 1-20) to enable vesicular transport from endoplasmic reticulum to lysosomes. Here, we report the possibility of a mitochondrial targeting sequence and mitochondrial localization of CTSD in cells. Live-cell imaging analysis with C-terminal enhanced green fluorescent protein-tagged CTSD (EGFP-CTSD) indicated that CTSD localizes to mitochondria. CTSD amino acids 21-35 are responsible for its mitochondrial localization, which exhibit typical features of mitochondrial targeting sequences, and are evolutionarily conserved. A proteinase K protection assay and sucrose gradient analysis showed that a small population of endogenous CTSD molecules exists in mitochondria. These results suggest that CTSD is a dual-targeted protein that may localize in both lysosomes and mitochondria.
Collapse
Affiliation(s)
- Naoki Ikari
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Hirofumi Arakawa
- Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan.
| |
Collapse
|
9
|
Vallender EJ, Hotchkiss CE, Lewis AD, Rogers J, Stern JA, Peterson SM, Ferguson B, Sayers K. Nonhuman primate genetic models for the study of rare diseases. Orphanet J Rare Dis 2023; 18:20. [PMID: 36721163 PMCID: PMC9887761 DOI: 10.1186/s13023-023-02619-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/15/2023] [Indexed: 02/01/2023] Open
Abstract
Pre-clinical research and development relies heavily upon translationally valid models of disease. A major difficulty in understanding the biology of, and developing treatments for, rare disease is the lack of animal models. It is important that these models not only recapitulate the presentation of the disease in humans, but also that they share functionally equivalent underlying genetic causes. Nonhuman primates share physiological, anatomical, and behavioral similarities with humans resulting from close evolutionary relationships and high genetic homology. As the post-genomic era develops and next generation sequencing allows for the resequencing and screening of large populations of research animals, naturally occurring genetic variation in nonhuman primates with clinically relevant phenotypes is regularly emerging. Here we review nonhuman primate models of multiple rare genetic diseases with a focus on the similarities and differences in manifestation and etiologies across species. We discuss how these models are being developed and how they can offer new tools and opportunities for researchers interested in exploring novel therapeutics for these and other genetic diseases. Modeling human genetic diseases in translationally relevant nonhuman primates presents new prospects for development of therapeutics and a better understanding of rare diseases. The post-genomic era offers the opportunity for the discovery and further development of more models like those discussed here.
Collapse
Affiliation(s)
- Eric J. Vallender
- University of Mississippi Medical Center, Jackson, MS USA
- Tulane National Primate Research Center, Covington, LA USA
| | - Charlotte E. Hotchkiss
- University of Washington, Seattle, WA USA
- Washington National Primate Research Center, Seattle, WA USA
| | - Anne D. Lewis
- Oregon Health and Sciences University, Beaverton, OR USA
- Oregon National Primate Research Center, Beaverton, OR USA
| | - Jeffrey Rogers
- Baylor College of Medicine, Houston, TX USA
- Wisconsin National Primate Research Center, Madison, WI USA
| | - Joshua A. Stern
- University of California-Davis, Davis, CA USA
- California National Primate Research Center, Davis, CA USA
| | - Samuel M. Peterson
- Oregon Health and Sciences University, Beaverton, OR USA
- Oregon National Primate Research Center, Beaverton, OR USA
| | - Betsy Ferguson
- Oregon Health and Sciences University, Beaverton, OR USA
- Oregon National Primate Research Center, Beaverton, OR USA
| | - Ken Sayers
- Texas Biomedical Research Institute, San Antonio, TX USA
- Southwest National Primate Research Center, San Antonio, TX USA
| |
Collapse
|
10
|
Holmes AD, White KA, Pratt MA, Johnson TB, Likhite S, Meyer K, Weimer JM. Sex-split analysis of pathology and motor-behavioral outcomes in a mouse model of CLN8-Batten disease reveals an increased disease burden and trajectory in female Cln8 mnd mice. Orphanet J Rare Dis 2022; 17:411. [PMID: 36369162 PMCID: PMC9652919 DOI: 10.1186/s13023-022-02564-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 10/23/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND CLN8-Batten disease (CLN8 disease) is a rare neurodegenerative disorder characterized phenotypically by progressive deterioration of motor and cognitive abilities, visual symptoms, epileptic seizures, and premature death. Mutations in CLN8 results in characteristic Batten disease symptoms and brain-wide pathology including accumulation of lysosomal storage material, gliosis, and neurodegeneration. Recent investigations of other subforms of Batten disease (CLN1, CLN3, CLN6) have emphasized the influence of biological sex on disease and treatment outcomes; however, little is known about sex differences in the CLN8 subtype. To determine the impact of sex on CLN8 disease burden and progression, we utilized a Cln8mnd mouse model to measure the impact and progression of histopathological and behavioral outcomes between sexes. RESULTS Several notable sex differences were observed in the presentation of brain pathology, including Cln8mnd female mice consistently presenting with greater GFAP+ astrocytosis and CD68+ microgliosis in the somatosensory cortex, ventral posteromedial/ventral posterolateral nuclei of the thalamus, striatum, and hippocampus when compared to Cln8mnd male mice. Furthermore, sex differences in motor-behavioral assessments revealed Cln8mnd female mice experience poorer motor performance and earlier death than their male counterparts. Cln8mnd mice treated with an AAV9-mediated gene therapy were also examined to assess sex differences on therapeutics outcomes, which revealed no appreciable differences between the sexes when responding to the therapy. CONCLUSIONS Taken together, our results provide further evidence of biologic sex as a modifier of Batten disease progression and outcome, thus warranting consideration when conducting investigations and monitoring therapeutic impact.
Collapse
Affiliation(s)
- Andrew D. Holmes
- grid.430154.70000 0004 5914 2142Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60Th St N, Sioux Falls, SD USA ,grid.267169.d0000 0001 2293 1795Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD USA
| | - Katherine A. White
- grid.430154.70000 0004 5914 2142Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60Th St N, Sioux Falls, SD USA
| | - Melissa A. Pratt
- grid.430154.70000 0004 5914 2142Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60Th St N, Sioux Falls, SD USA
| | - Tyler B. Johnson
- grid.430154.70000 0004 5914 2142Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60Th St N, Sioux Falls, SD USA
| | - Shibi Likhite
- grid.240344.50000 0004 0392 3476The Research Institute at Nationwide Children’s Hospital, Columbus, OH USA
| | - Kathrin Meyer
- grid.240344.50000 0004 0392 3476The Research Institute at Nationwide Children’s Hospital, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Pediatrics, The Ohio State University, Columbus, OH USA
| | - Jill M. Weimer
- grid.430154.70000 0004 5914 2142Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60Th St N, Sioux Falls, SD USA ,grid.267169.d0000 0001 2293 1795Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD USA
| |
Collapse
|
11
|
Thirumal Kumar D, Shaikh N, Udhaya Kumar S, George Priya Doss C. Computational and structural investigation of Palmitoyl-Protein Thioesterase 1 (PPT1) protein causing Neuronal Ceroid Lipofuscinoses (NCL). ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:89-109. [PMID: 36088080 DOI: 10.1016/bs.apcsb.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The Neuronal Ceroid Lipofuscinoses (NCL) are a group of progressive neurodegenerative disorders, associated with 14 Ceroid Lipofuscinosis Neuronal genes (CLN1-14). The mutations in the Palmitoyl-Protein Thioesterase 1 (PPT1) protein serve as one of the major reasons for the causative of NCL. The PPT1 involves degrading and modifying cysteine residues in proteins or peptides by removing thioester-linked fatty acyl groups like palmitate prefers acyl chains of 14-18 carbons in length. In this study, we have analyzed the impact of PPT1 mutations on the deleteriousness, stability, conservative nature of amino acid, and impact of mutations on the protein structure. We have also used molecular dynamics simulations using GROMACS to perceive the alteration in the dynamic behavior of the PPT1 at the residual level. In this study, we have retrieved 23 PPT1 mutations from the UniProt database, and these were subjected to a series of analyses using varied computer algorithms. From these analyses, out of 23 mutations, 16 mutations were identified as deleterious. Among 16, eight mutations were identified to destabilize the protein structure, and finally, two mutations (W38C and L222P) were found to be positioned in the highly conserved region. The structural impact study observed that the mutant proline could disrupt the alpha helix formed by the leucine at position 222. Finally, from the molecular dynamics simulations, we observed that due to the mutations (W38C and L222P), the protein had experienced higher deviation, fluctuation, and lower compactness. These structural changes elucidate that these mutations can impact the structure and function of the PPT1 protein.
Collapse
Affiliation(s)
- D Thirumal Kumar
- Faculty of Allied Health Sciences, Meenakshi Academy of Higher Education and Research, Chennai, Tamil Nadu, India.
| | - Nishaat Shaikh
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - S Udhaya Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
12
|
Doccini S, Marchese M, Morani F, Gammaldi N, Mero S, Pezzini F, Soliymani R, Santi M, Signore G, Ogi A, Rocchiccioli S, Kanninen KM, Simonati A, Lalowski MM, Santorelli FM. Lysosomal Proteomics Links Disturbances in Lipid Homeostasis and Sphingolipid Metabolism to CLN5 Disease. Cells 2022; 11:1840. [PMID: 35681535 PMCID: PMC9180748 DOI: 10.3390/cells11111840] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 12/01/2022] Open
Abstract
CLN5 disease (MIM: 256731) represents a rare late-infantile form of neuronal ceroid lipofuscinosis (NCL), caused by mutations in the CLN5 gene that encodes the CLN5 protein (CLN5p), whose physiological roles stay unanswered. No cure is currently available for CLN5 patients and the opportunities for therapies are lagging. The role of lysosomes in the neuro-pathophysiology of CLN5 disease represents an important topic since lysosomal proteins are directly involved in the primary mechanisms of neuronal injury occurring in various NCL forms. We developed and implemented a lysosome-focused, label-free quantitative proteomics approach, followed by functional validations in both CLN5-knockout neuronal-like cell lines and Cln5-/- mice, to unravel affected pathways and modifying factors involved in this disease scenario. Our results revealed a key role of CLN5p in lipid homeostasis and sphingolipid metabolism and highlighted mutual NCL biomarkers scored with high lysosomal confidence. A newly generated cln5 knockdown zebrafish model recapitulated most of the pathological features seen in NCL disease. To translate the findings from in-vitro and preclinical models to patients, we evaluated whether two FDA-approved drugs promoting autophagy via TFEB activation or inhibition of the glucosylceramide synthase could modulate in-vitro ROS and lipid overproduction, as well as alter the locomotor phenotype in zebrafish. In summary, our data advance the general understanding of disease mechanisms and modifying factors in CLN5 disease, which are recurring in other NCL forms, also stimulating new pharmacological treatments.
Collapse
Affiliation(s)
- Stefano Doccini
- Molecular Medicine–IRCCS Stella Maris, 56128 Pisa, Italy; (M.M.); (N.G.); (S.M.); (A.O.)
| | - Maria Marchese
- Molecular Medicine–IRCCS Stella Maris, 56128 Pisa, Italy; (M.M.); (N.G.); (S.M.); (A.O.)
| | - Federica Morani
- Department of Biology, University of Pisa, 56126 Pisa, Italy;
| | - Nicola Gammaldi
- Molecular Medicine–IRCCS Stella Maris, 56128 Pisa, Italy; (M.M.); (N.G.); (S.M.); (A.O.)
- Ph.D. Program in Neuroscience, University of Florence, 50121 Florence, Italy
| | - Serena Mero
- Molecular Medicine–IRCCS Stella Maris, 56128 Pisa, Italy; (M.M.); (N.G.); (S.M.); (A.O.)
| | - Francesco Pezzini
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (F.P.); (A.S.)
| | - Rabah Soliymani
- HiLIFE, Meilahti Clinical Proteomics Core Facility, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Melissa Santi
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, 56127 Pisa, Italy;
| | | | - Asahi Ogi
- Molecular Medicine–IRCCS Stella Maris, 56128 Pisa, Italy; (M.M.); (N.G.); (S.M.); (A.O.)
| | | | - Katja M. Kanninen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Alessandro Simonati
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (F.P.); (A.S.)
| | - Maciej M. Lalowski
- HiLIFE, Meilahti Clinical Proteomics Core Facility, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Institute of Bioorganic Chemistry, PAS, Department of Biomedical Proteomics, 61-704 Poznan, Poland
| | - Filippo M. Santorelli
- Molecular Medicine–IRCCS Stella Maris, 56128 Pisa, Italy; (M.M.); (N.G.); (S.M.); (A.O.)
| |
Collapse
|
13
|
Recent Insight into the Genetic Basis, Clinical Features, and Diagnostic Methods for Neuronal Ceroid Lipofuscinosis. Int J Mol Sci 2022; 23:ijms23105729. [PMID: 35628533 PMCID: PMC9145894 DOI: 10.3390/ijms23105729] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are a group of rare, inherited, neurodegenerative lysosomal storage disorders that affect children and adults. They are traditionally grouped together, based on shared clinical symptoms and pathological ground. To date, 13 autosomal recessive gene variants, as well as one autosomal dominant gene variant, of NCL have been described. These genes encode a variety of proteins, whose functions have not been fully defined; most are lysosomal enzymes, transmembrane proteins of the lysosome, or other organelles. Common symptoms of NCLs include the progressive loss of vision, mental and motor deterioration, epileptic seizures, premature death, and, in rare adult-onset cases, dementia. Depending on the mutation, these symptoms can vary, with respect to the severity and onset of symptoms by age. Currently, all forms of NCL are fatal, and no curative treatments are available. Herein, we provide an overview to summarize the current knowledge regarding the pathophysiology, genetics, and clinical manifestation of these conditions, as well as the approach to diagnosis.
Collapse
|
14
|
Meiman EJ, Kick GR, Jensen CA, Coates JR, Katz ML. Characterization of neurological disease progression in a canine model of CLN5 neuronal ceroid lipofuscinosis. Dev Neurobiol 2022; 82:326-344. [PMID: 35427439 PMCID: PMC9119968 DOI: 10.1002/dneu.22878] [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: 12/09/2021] [Revised: 01/14/2022] [Accepted: 02/25/2022] [Indexed: 11/08/2022]
Abstract
Golden Retriever dogs with a frameshift variant in CLN5 (c.934_935delAG) suffer from a progressive neurodegenerative disorder analogous to the CLN5 form of neuronal ceroid lipofuscinosis (NCL). Five littermate puppies homozygous for the deletion allele were identified prior to the onset of disease signs. Studies were performed to characterize the onset and progression of the disease in these dogs. Neurological signs that included restlessness, unwillingness to cooperate with the handlers, and proprioceptive deficits first became apparent at approximately 12 months of age. The neurological signs progressed over time and by 21 to 23 months of age included general proprioceptive ataxia, menace response deficits, aggressive behaviors, cerebellar ataxia, intention tremors, decreased visual tracking, seizures, cognitive decline, and impaired prehension. Due to the severity of these signs, the dogs were euthanized between 21 and 23 months of age. Magnetic resonance imaging revealed pronounced progressive global brain atrophy with a more than sevenfold increase in the volume of the ventricular system between 9.5 and 22.5 months of age. Accompanying this atrophy were pronounced accumulations of autofluorescent inclusions throughout the brain and spinal cord. Ultrastructurally, the contents of these inclusions were found to consist primarily of membrane‐like aggregates. Inclusions with similar fluorescence properties were present in cardiac muscle. Similar to other forms of NCL, the affected dogs had low plasma carnitine concentrations, suggesting impaired carnitine biosynthesis. These data on disease progression will be useful in future studies using the canine model for therapeutic intervention studies.
Collapse
Affiliation(s)
- Elizabeth J. Meiman
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine University of Missouri Columbia MO 65211 USA
| | - Grace Robinson Kick
- Neurodegenerative Diseases Research Laboratory University of Missouri Columbia MO 65212 USA
| | - Cheryl A. Jensen
- Neurodegenerative Diseases Research Laboratory University of Missouri Columbia MO 65212 USA
| | - Joan R. Coates
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine University of Missouri Columbia MO 65211 USA
| | - Martin L. Katz
- Neurodegenerative Diseases Research Laboratory University of Missouri Columbia MO 65212 USA
| |
Collapse
|
15
|
Barry LA, Kay GW, Mitchell NL, Murray SJ, Jay NP, Palmer DN. Aggregation chimeras provide evidence of in vivo intercellular correction in ovine CLN6 neuronal ceroid lipofuscinosis (Batten disease). PLoS One 2022; 17:e0261544. [PMID: 35404973 PMCID: PMC9000108 DOI: 10.1371/journal.pone.0261544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/29/2022] [Indexed: 11/29/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs; Batten disease) are fatal, mainly childhood, inherited neurodegenerative lysosomal storage diseases. Sheep affected with a CLN6 form display progressive regionally defined glial activation and subsequent neurodegeneration, indicating that neuroinflammation may be causative of pathogenesis. In this study, aggregation chimeras were generated from homozygous unaffected normal and CLN6 affected sheep embryos, resulting in seven chimeric animals with varied proportions of normal to affected cells. These sheep were classified as affected-like, recovering-like or normal-like, based on their cell-genotype ratios and their clinical and neuropathological profiles. Neuropathological examination of the affected-like animals revealed intense glial activation, prominent storage body accumulation and severe neurodegeneration within all cortical brain regions, along with vision loss and decreasing intracranial volumes and cortical thicknesses consistent with ovine CLN6 disease. In contrast, intercellular communication affecting pathology was evident at both the gross and histological level in the normal-like and recovering-like chimeras, resulting in a lack of glial activation and rare storage body accumulation in only a few cells. Initial intracranial volumes of the recovering-like chimeras were below normal but progressively recovered to about normal by two years of age. All had normal cortical thicknesses, and none went blind. Extended neurogenesis was evident in the brains of all the chimeras. This study indicates that although CLN6 is a membrane bound protein, the consequent defect is not cell intrinsic. The lack of glial activation and inflammatory responses in the normal-like and recovering-like chimeras indicate that newly generated cells are borne into a microenvironment conducive to maturation and survival.
Collapse
Affiliation(s)
- Lucy Anne Barry
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Graham William Kay
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Nadia Lesley Mitchell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
- Department of Radiology, University of Otago, Christchurch, Canterbury, New Zealand
| | - Samantha Jane Murray
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Nigel P. Jay
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - David Norris Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
- Department of Radiology, University of Otago, Christchurch, Canterbury, New Zealand
- * E-mail:
| |
Collapse
|
16
|
A Novel, Apparently Silent Variant in MFSD8 Causes Neuronal Ceroid Lipofuscinosis with Marked Intrafamilial Variability. Int J Mol Sci 2022; 23:ijms23042271. [PMID: 35216386 PMCID: PMC8877174 DOI: 10.3390/ijms23042271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Variants in MFSD8 can cause neuronal ceroid lipofuscinoses (NCLs) as well as nonsyndromic retinopathy. The mutation spectrum includes mainly missense and stop variants, but splice sites and frameshift variants have also been reported. To date, apparently synonymous substitutions have not been shown to cause MFSD8-associated diseases. We report two closely related subjects from a consanguineous Turkish family who presented classical features of NCLs but demonstrated marked intrafamilial variability in age at the onset and severity of symptoms. In fact, the difference in the onset of first neurologic symptoms was 15 years and that of ophthalmologic symptoms was 12 years. One subject presented an intellectual disability and a considerable cerebellar ataxia syndrome, while the other subject showed no intellectual disability and only a mild atactic syndrome. The diagnostic genetic testing of both subjects based on genome sequencing prioritized a novel, apparently synonymous variant in MFSD8, which was found in homozygosity in both subjects. The variant was not located within an integral part of the splice site consensus sequences. However, the bioinformatic analyses suggested that the mutant allele is more likely to cause exon skipping due to an altered ratio of exonic splice enhancer and silencer motifs. Exon skipping was confirmed in vitro by minigene assays and in vivo by RNA analysis from patient lymphocytes. The mutant transcript is predicted to result in a frameshift and, if translated, in a truncated protein. Synonymous variants are often given a low priority in genetic diagnostics because of their expected lack of functional impact. This study highlights the importance of investigating the impact of synonymous variants on splicing.
Collapse
|
17
|
Liu J, Bassal M, Schlichting S, Braren I, Di Spiezio A, Saftig P, Bartsch U. Intravitreal gene therapy restores the autophagy-lysosomal pathway and attenuates retinal degeneration in cathepsin D-deficient mice. Neurobiol Dis 2022; 164:105628. [PMID: 35033660 DOI: 10.1016/j.nbd.2022.105628] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
Loss of vision due to progressive retinal degeneration is a hallmark of neuronal ceroid lipofuscinoses (NCL), a group of fatal neurodegenerative lysosomal storage diseases. Enzyme substitution therapies represent promising treatment options for NCLs caused by dysfunctions of soluble lysosomal enzymes. Here, we compared the efficacy of a cell-based enzyme substitution strategy and a gene therapy approach to attenuate the retinal pathology in cathepsin D- (CTSD) deficient mice, an animal model of CLN10 disease. Levels of enzymatically active CTSD in mutant retinas were significantly higher after an adeno-associated virus vector-mediated CTSD transfer to retinal glial cells and retinal pigment epithelial cells than after intravitreal transplantations of a CTSD overexpressing clonal neural stem cell line. In line with this finding, the gene therapy treatment restored the disrupted autophagy-lysosomal pathway more effectively than the cell-based approach, as indicated by a complete clearance of storage, significant attenuation of lysosomal hypertrophy, and normalized levels of the autophagy marker sequestosome 1/p62 and microtubule-associated protein 1 light chain 3-II. While the cell-based treatment did not prevent the rapidly progressing loss of various retinal cell types, the gene therapy approach markedly attenuated retinal degeneration as demonstrated by a pronounced rescue of photoreceptor cells and rod bipolar cells.
Collapse
Affiliation(s)
- Junling Liu
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mahmoud Bassal
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefanie Schlichting
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ingke Braren
- Vector Facility, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
18
|
Domowicz MS, Chan WC, Claudio-Vázquez P, Gonzalez T, Schwartz NB. Brain transcriptome analysis of a CLN2 mouse model as a function of disease progression. J Neuroinflammation 2021; 18:262. [PMID: 34749772 PMCID: PMC8576919 DOI: 10.1186/s12974-021-02302-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Neuronal ceroid lipofuscinoses, (NCLs or Batten disease) are a group of inherited, early onset, fatal neurodegenerative diseases associated with mutations in 13 genes. All forms of the disease are characterized by lysosomal accumulation of fluorescent storage material, as well as profound neurodegeneration, but the relationship of the various genes’ function to a single biological process is not obvious. In this study, we used a well-characterized mouse model of classical late infantile NCL (cLINCL) in which the tripeptidyl peptidase 1 (Tpp1) gene is disrupted by gene targeting, resulting in loss of detectable TPP1 activity and leading to progressive neurological phenotypes including ataxia, increased motor deficiency, and early death. Methods In order to identify genes and pathways that may contribute to progression of the neurodegenerative process, we analyzed forebrain/midbrain and cerebellar transcriptional differences at 1, 2, 3 and 4 months of age in control and TPP1-deficient mice by global RNA-sequencing. Results Progressive neurodegenerative inflammatory responses involving microglia, astrocytes and endothelial cells were observed, accompanied by activation of leukocyte extravasation signals and upregulation of nitric oxide production and reactive oxygen species. Several astrocytic (i.e., Gfap, C4b, Osmr, Serpina3n) and microglial (i.e., Ctss, Itgb2, Itgax, Lyz2) genes were identified as strong markers for assessing disease progression as they showed increased levels of expression in vivo over time. Furthermore, transient increased expression of choroid plexus genes was observed at 2 months in the lateral and fourth ventricle, highlighting an early role for the choroid plexus and cerebrospinal fluid in the disease pathology. Based on these gene expression changes, we concluded that neuroinflammation starts, for the most part, after 2 months in the Tpp1−/− brain and that activation of microglia and astrocytes occur more rapidly in cerebellum than in the rest of the brain; confirming increased severity of inflammation in this region. Conclusions These findings have led to a better understanding of cLINCL pathological onset and progression, which may aid in development of future therapeutic treatments for this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02302-z.
Collapse
Affiliation(s)
- Miriam S Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.
| | - Wen-Ching Chan
- Center for Research Informatics, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Patricia Claudio-Vázquez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Tatiana Gonzalez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Nancy B Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.,Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
19
|
Walkley SU. Rethinking lysosomes and lysosomal disease. Neurosci Lett 2021; 762:136155. [PMID: 34358625 DOI: 10.1016/j.neulet.2021.136155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/14/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022]
Abstract
Lysosomal storage diseases were recognized and defined over a century ago as a class of disorders affecting mostly children and causing systemic disease often accompanied by major neurological consequences. Since their discovery, research focused on understanding their causes has been an important driver of our ever-expanding knowledge of cell biology and the central role that lysosomes play in cell function. Today we recognize over 50 so-called storage diseases, with most understood at the level of gene, protein and pathway involvement, but few fully clarified in terms of how the defective lysosomal function causes brain disease; even fewer have therapies that can effectively rescue brain function. Importantly, we also recognize that storage diseases are not simply a class of lysosomal disorders all by themselves, as increasingly a critical role for the greater lysosomal system with its endosomal, autophagosomal and salvage streams has also emerged in a host of neurodevelopmental and neurodegenerative diseases. Despite persistent challenges across all aspects of these complex disorders, and as reflected in this and other articles focused on lysosomal storage diseases in this special issue of Neuroscience Letters, the progress and promise to both understand and effectively treat these conditions has never been greater.
Collapse
Affiliation(s)
- Steven U Walkley
- Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
20
|
Shiro Y, Yamashita A, Watanabe K, Yamazaki T. CLN6's luminal tail-mediated functional interference between CLN6 mutants as a novel pathomechanism for the neuronal ceroid lipofuscinoses. Biomed Res 2021; 42:129-138. [PMID: 34380921 DOI: 10.2220/biomedres.42.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
CLN6 (Ceroid Lipofuscinosis, Neuronal, 6) is a 311-amino acid protein spanning the endoplasmic reticulum membrane. Mutations in CLN6 are linked to CLN6 disease, a hereditary neurodegenerative disorder categorized into the neuronal ceroid lipofuscinoses. CLN6 disease is an autosomal recessive disorder and individuals affected with this disease have two identical (homozygous) or two distinct (compound heterozygous) CLN6 mutant alleles. Little has been known about CLN6's physiological roles and the disease mechanism. We recently found that CLN6 prevents protein aggregate formation, pointing to impaired CLN6's anti-aggregate activity as a cause for the disease. To comprehensively understand the pathomechanism, overall anti-aggregate activity derived from two different CLN6 mutants needs to be investigated, considering patients compound heterozygous for CLN6 alleles. We focused on mutant combinations involving the S132CfsX18 (132fsX) prematurely terminated protein, produced from the most frequent mutation in CLN6. The 132fsX mutant nullified anti-aggregate activity of the P299L CLN6 missense mutant but not of wild-type CLN6. Wild-type CLN6's resistance to the 132fsX mutant was abolished by replacement of amino acids 297-301, including Pro297 and Pro299, with five alanine residues. Given that removal of CLN6's C-terminal fifteen amino acids 297-311 (luminal tail) did not affect the resistance, we suggested that CLN6's luminal tail, when unleashed from Pro297/299-mediated conformational constraints, is improperly positioned by the 132fsX mutant, thereby blocking the induction of anti- aggregate activity. We here reveal a novel mechanism for dissipating CLN6 mutants' residual functions, providing an explanation for the compound heterozygosity-driven pathogenesis.
Collapse
Affiliation(s)
- Yuki Shiro
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University
| | - Arisa Yamashita
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University
| | - Kana Watanabe
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University
| | - Tetsuo Yamazaki
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University
| |
Collapse
|
21
|
Terryn J, Verfaillie CM, Van Damme P. Tweaking Progranulin Expression: Therapeutic Avenues and Opportunities. Front Mol Neurosci 2021; 14:713031. [PMID: 34366786 PMCID: PMC8343103 DOI: 10.3389/fnmol.2021.713031] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 01/01/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease, leading to behavioral changes and language difficulties. Heterozygous loss-of-function mutations in progranulin (GRN) induce haploinsufficiency of the protein and are associated with up to one-third of all genetic FTD cases worldwide. While the loss of GRN is primarily associated with neurodegeneration, the biological functions of the secreted growth factor-like protein are more diverse, ranging from wound healing, inflammation, vasculogenesis, and metabolic regulation to tumor cell growth and metastasis. To date, no disease-modifying treatments exist for FTD, but different therapeutic approaches to boost GRN levels in the central nervous system are currently being developed (including AAV-mediated GRN gene delivery as well as anti-SORT1 antibody therapy). In this review, we provide an overview of the multifaceted regulation of GRN levels and the corresponding therapeutic avenues. We discuss the opportunities, advantages, and potential drawbacks of the diverse approaches. Additionally, we highlight the therapeutic potential of elevating GRN levels beyond patients with loss-of-function mutations in GRN.
Collapse
Affiliation(s)
- Joke Terryn
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Interdepartmental Stem Cell Institute, KU Leuven-University of Leuven, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Bassal M, Liu J, Jankowiak W, Saftig P, Bartsch U. Rapid and Progressive Loss of Multiple Retinal Cell Types in Cathepsin D-Deficient Mice-An Animal Model of CLN10 Disease. Cells 2021; 10:696. [PMID: 33800998 PMCID: PMC8003850 DOI: 10.3390/cells10030696] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/11/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
Vision loss is among the characteristic symptoms of neuronal ceroid lipofuscinosis (NCL), a fatal neurodegenerative lysosomal storage disorder. Here, we performed an in-depth analysis of retinal degeneration at the molecular and cellular levels in mice lacking the lysosomal aspartyl protease cathepsin D, an animal model of congenital CLN10 disease. We observed an early-onset accumulation of storage material as indicated by elevated levels of saposin D and subunit C of the mitochondrial ATP synthase. The accumulation of storage material was accompanied by reactive astrogliosis and microgliosis, elevated expression of the autophagy marker sequestosome 1/p62 and a dysregulated expression of several lysosomal proteins. The number of cone photoreceptor cells was reduced as early as at postnatal day 5. At the end stage of the disease, the outer nuclear layer was almost atrophied, and all cones were lost. A significant loss of rod and cone bipolar cells, amacrine cells and ganglion cells was found at advanced stages of the disease. Results demonstrate that cathepsin D deficiency results in an early-onset and rapidly progressing retinal dystrophy that involves all retinal cell types. Data of the present study will serve as a reference for studies aimed at developing treatments for retinal degeneration in CLN10 disease.
Collapse
Affiliation(s)
- Mahmoud Bassal
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.B.); (J.L.); (W.J.)
| | - Junling Liu
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.B.); (J.L.); (W.J.)
| | - Wanda Jankowiak
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.B.); (J.L.); (W.J.)
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany;
| | - Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.B.); (J.L.); (W.J.)
| |
Collapse
|
23
|
Iwan K, Clayton R, Mills P, Csanyi B, Gissen P, Mole SE, Palmer DN, Mills K, Heywood WE. Urine proteomics analysis of patients with neuronal ceroid lipofuscinoses. iScience 2021; 24:102020. [PMID: 33532713 PMCID: PMC7822952 DOI: 10.1016/j.isci.2020.102020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/11/2020] [Accepted: 12/29/2020] [Indexed: 01/18/2023] Open
Abstract
The neuronal ceroid lipofuscinoses (NCL) are a group of 13 rare neurodegenerative disorders characterized by accumulation of cellular storage bodies. There are few therapeutic options, and existing tests do not monitor disease progression and treatment response. However, urine biomarkers could address this need. Proteomic analysis of CLN2 patient urine revealed activation of immune response pathways and pathways associated with the unfolded protein response. Analysis of CLN5 and CLN6 sheep model urine showed subtle changes. To confirm and investigate the relevance of candidate biomarkers a targeted LC-MS/MS proteomic assay was created. We applied this assay to additional CLN2 samples as well as other patients with NCL (CLN1, CLN3, CLN5, CLN6, and CLN7) and demonstrated that hexosaminidase-A, aspartate aminotransferase-1, and LAMP1 are increased in NCL samples and betaine-homocysteine S-methyltransferase-1 was specifically increased in patients with CLN2. These proteins could be used to monitor the effectiveness of future therapies aimed at treating systemic NCL disease. The urine proteome is altered in humans and animals with NCL Hexosaminidase A and LAMP1 are increased in patients with NCL Betaine-homocysteine S-methyltransferase 1 is elevated in CLN2 patients Proteins altered in CLN5 and CLN6 sheep models are not affected in humans
Collapse
Affiliation(s)
- Katharina Iwan
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Robert Clayton
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Philippa Mills
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | | | - Paul Gissen
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK.,Great Ormond Street Hospital for Children, London, UK
| | - Sara E Mole
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.,MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - David N Palmer
- Department of Molecular Biosciences, Agriculture and Life Sciences Faculty, University Lincoln 7647, Canterbury, New Zealand
| | - Kevin Mills
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Wendy E Heywood
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| |
Collapse
|
24
|
Best HL, Clare AJ, McDonald KO, Wicky HE, Hughes SM. An altered secretome is an early marker of the pathogenesis of CLN6 Batten disease. J Neurochem 2021; 157:764-780. [PMID: 33368303 DOI: 10.1111/jnc.15285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/20/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are a group of inherited childhood neurodegenerative disorders. In addition to the accumulation of auto-fluorescent storage material in lysosomes, NCLs are largely characterised by region-specific neuroinflammation that can predict neuron loss. These phenotypes suggest alterations in the extracellular environment-making the secretome an area of significant interest. This study investigated the secretome in the CLN6 (ceroid-lipofuscinosis neuronal protein 6) variant of NCL. To investigate the CLN6 secretome, we co-cultured neurons and glia isolated from Cln6nclf or Cln6± mice, and utilised mass spectrometry to compare protein constituents of conditioned media. The significant changes noted in cathepsin enzymes, were investigated further via western blotting and enzyme activity assays. Viral-mediated gene therapy was used to try and rescue the wild-type phenotype and restore the secretome-both in vitro in co-cultures and in vivo in mouse plasma. In Cln6nclf cells, proteomics revealed a marked increase in catabolic and cytoskeletal-associated proteins-revealing new similarities between the pathogenic signatures of NCLs with other neurodegenerative disorders. These changes were, in part, corrected by gene therapy intervention, suggesting these proteins as candidate in vitro biomarkers. Importantly, these in vitro changes show promise for in vivo translation, with Cathepsin L (CTSL) activity reduced in both co-cultures and Cln6nclf plasma samples post gene-therapy. This work suggests the secretome plays a role in CLN6 pathogenesis and highlights its potential use as an in vitro model. Proteomic changes present a list of candidate biomarkers for monitoring disease and assessing potential therapeutics in future studies.
Collapse
Affiliation(s)
- Hannah L Best
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Alison J Clare
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Kirstin O McDonald
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Hollie E Wicky
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
25
|
Behnke V, Langmann T. [Neuroinflammation in neuronal ceroid lipofuscinosis]. Ophthalmologe 2021; 118:98-105. [PMID: 33411040 DOI: 10.1007/s00347-020-01301-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Retinal degeneration and neuroinflammation are often early hallmarks of different subtypes of neuronal ceroid lipofuscinosis (NCL) in patients and genetic animal models. OBJECTIVE This article gives a summary of recently published research articles and novel concepts in the field of NCL-related neuroinflammation. MATERIAL AND METHODS A search was carried out in PubMed for relevant publications and the results as well as own NCL-related research are discussed. RESULTS Microglia and other glial cells are chronically activated and show various dysfunctions in the central nervous system (CNS) and retina of NCL patients and animal models. This is accompanied by significant changes in the transcriptome and proteome. In NCL there is also involvement of the adaptive immune response, as demonstrated by the influx of autoantibodies and activated T cells. CONCLUSION A deeper understanding of the molecular processes that contribute to neuroinflammation and ultimately lead to neuronal cell death is an important basis for the discovery of possible biomarkers and the development of immunotherapies in NCL.
Collapse
Affiliation(s)
- V Behnke
- Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde, Medizinische Fakultät und Uniklinik Köln, Joseph-Stelzmann-Str. 9, 50931, Köln, Deutschland
| | - T Langmann
- Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde, Medizinische Fakultät und Uniklinik Köln, Joseph-Stelzmann-Str. 9, 50931, Köln, Deutschland. .,Zentrum für Molekulare Medizin, Köln, Deutschland.
| |
Collapse
|
26
|
Abstract
Neuronal ceroid lipofuscinosis (NCLs) is a group of inherited neurodegenerative lysosomal storage diseases that together represent the most common cause of dementia in children. Phenotypically, patients have visual impairment, cognitive and motor decline, epilepsy, and premature death. A primary challenge is to halt and/or reverse these diseases, towards which developments in potential effective therapies are encouraging. Many treatments, including enzyme replacement therapy (for CLN1 and CLN2 diseases), stem-cell therapy (for CLN1, CLN2, and CLN8 diseases), gene therapy vector (for CLN1, CLN2, CLN3, CLN5, CLN6, CLN7, CLN10, and CLN11 diseases), and pharmacological drugs (for CLN1, CLN2, CLN3, and CLN6 diseases) have been evaluated for safety and efficacy in pre-clinical and clinical studies. Currently, cerliponase alpha for CLN2 disease is the only approved therapy for NCL. Lacking is any study of potential treatments for CLN4, CLN9, CLN12, CLN13 or CLN14 diseases. This review provides an overview of genetics for each CLN disease, and we discuss the current understanding from pre-clinical and clinical study of potential therapeutics. Various therapeutic interventions have been studied in many experimental animal models. Combination of treatments may be useful to slow or even halt disease progression; however, few therapies are unlikely to even partially reverse the disease and a complete reversal is currently improbable. Early diagnosis to allow initiation of therapy, when indicated, during asymptomatic stages is more important than ever.
Collapse
|
27
|
Neuroinflammation and progressive myoclonus epilepsies: from basic science to therapeutic opportunities. Expert Rev Mol Med 2020; 22:e4. [PMID: 32938505 PMCID: PMC7520540 DOI: 10.1017/erm.2020.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progressive myoclonus epilepsies (PMEs) are a group of genetic neurological disorders characterised by the occurrence of epileptic seizures, myoclonus and progressive neurological deterioration including cerebellar involvement and dementia. The primary cause of PMEs is variable and alterations in the corresponding mutated genes determine the progression and severity of the disease. In most cases, they lead to the death of the patient after a period of prolonged disability. PMEs also share poor information on the pathophysiological bases and the lack of a specific treatment. Recent reports suggest that neuroinflammation is a common trait under all these conditions. Here, we review similarities and differences in neuroinflammatory response in several PMEs and discuss the window of opportunity of using anti-inflammatory drugs in the treatment of several of these conditions.
Collapse
|
28
|
Werner G, Damme M, Schludi M, Gnörich J, Wind K, Fellerer K, Wefers B, Wurst W, Edbauer D, Brendel M, Haass C, Capell A. Loss of TMEM106B potentiates lysosomal and FTLD-like pathology in progranulin-deficient mice. EMBO Rep 2020; 21:e50241. [PMID: 32929860 PMCID: PMC7534633 DOI: 10.15252/embr.202050241] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/05/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) in TMEM106B encoding the lysosomal type II transmembrane protein 106B increase the risk for frontotemporal lobar degeneration (FTLD) of GRN (progranulin gene) mutation carriers. Currently, it is unclear if progranulin (PGRN) and TMEM106B are synergistically linked and if a gain or a loss of function of TMEM106B is responsible for the increased disease risk of patients with GRN haploinsufficiency. We therefore compare behavioral abnormalities, gene expression patterns, lysosomal activity, and TDP‐43 pathology in single and double knockout animals. Grn−/−/Tmem106b−/− mice show a strongly reduced life span and massive motor deficits. Gene expression analysis reveals an upregulation of molecular signature characteristic for disease‐associated microglia and autophagy. Dysregulation of maturation of lysosomal proteins as well as an accumulation of ubiquitinated proteins and widespread p62 deposition suggest that proteostasis is impaired. Moreover, while single Grn−/− knockouts only occasionally show TDP‐43 pathology, the double knockout mice exhibit deposition of phosphorylated TDP‐43. Thus, a loss of function of TMEM106B may enhance the risk for GRN‐associated FTLD by reduced protein turnover in the lysosomal/autophagic system.
Collapse
Affiliation(s)
- Georg Werner
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Damme
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Martin Schludi
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Karin Wind
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katrin Fellerer
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Haass
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anja Capell
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
29
|
Nelvagal HR, Lange J, Takahashi K, Tarczyluk-Wells MA, Cooper JD. Pathomechanisms in the neuronal ceroid lipofuscinoses. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165570. [DOI: 10.1016/j.bbadis.2019.165570] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 12/22/2022]
|
30
|
Kinarivala N, Morsy A, Patel R, Carmona AV, Sajib MS, Raut S, Mikelis CM, Al-Ahmad A, Trippier PC. An iPSC-Derived Neuron Model of CLN3 Disease Facilitates Small Molecule Phenotypic Screening. ACS Pharmacol Transl Sci 2020; 3:931-947. [PMID: 33073192 DOI: 10.1021/acsptsci.0c00077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Indexed: 02/06/2023]
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are a family of rare lysosomal storage disorders. The most common form of NCL occurs in children harboring a mutation in the CLN3 gene. This form is lethal with no existing cure or treatment beyond symptomatic relief. The pathophysiology of CLN3 disease is complex and poorly understood, with current in vivo and in vitro models failing to identify pharmacological targets for therapeutic intervention. This study reports the characterization of the first CLN3 patient-specific induced pluripotent stem cell (iPSC)-derived model of the blood-brain barrier and establishes the suitability of an iPSC-derived neuron model of the disease to facilitate compound screening. Upon differentiation, hallmarks of CLN3 disease are apparent, including lipofuscin and subunit c of mitochondrial ATP synthase accumulation, mitochondrial dysfunction, and attenuated Bcl-2 expression. The model led to the identification of small molecules that cleared subunit c accumulation by mTOR-independent modulation of autophagy, conferred protective effects through induction of Bcl-2 and rescued mitochondrial dysfunction.
Collapse
Affiliation(s)
- Nihar Kinarivala
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Ahmed Morsy
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ronak Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Angelica V Carmona
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Md Sanaullah Sajib
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Snehal Raut
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States.,UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
31
|
Katz ML, Buckley RM, Biegen V, O'Brien DP, Johnson GC, Warren WC, Lyons LA. Neuronal Ceroid Lipofuscinosis in a Domestic Cat Associated with a DNA Sequence Variant That Creates a Premature Stop Codon in CLN6. G3 (BETHESDA, MD.) 2020; 10:2741-2751. [PMID: 32518081 PMCID: PMC7407459 DOI: 10.1534/g3.120.401407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/06/2020] [Indexed: 01/04/2023]
Abstract
A neutered male domestic medium-haired cat presented at a veterinary neurology clinic at 20 months of age due to progressive neurological signs that included visual impairment, focal myoclonus, and frequent severe generalized seizures that were refractory to treatment with phenobarbital. Magnetic resonance imaging revealed diffuse global brain atrophy. Due to the severity and frequency of its seizures, the cat was euthanized at 22 months of age. Microscopic examination of the cerebellum, cerebral cortex and brainstem revealed pronounced intracellular accumulations of autofluorescent storage material and inflammation in all 3 brain regions. Ultrastructural examination of the storage material indicated that it consisted almost completely of tightly-packed membrane-like material. The clinical signs and neuropathology strongly suggested that the cat suffered from a form of neuronal ceroid lipofuscinosis (NCL). Whole exome sequence analysis was performed on genomic DNA from the affected cat. Comparison of the sequence data to whole exome sequence data from 39 unaffected cats and whole genome sequence data from an additional 195 unaffected cats revealed a homozygous variant in CLN6 that was unique to the affected cat. This variant was predicted to cause a stop gain in the transcript due to a guanine to adenine transition (ENSFCAT00000025909:c.668G > A; XM_003987007.5:c.668G > A) and was the sole loss of function variant detected. CLN6 variants in other species, including humans, dogs, and sheep, are associated with the CLN6 form of NCL. Based on the affected cat's clinical signs, neuropathology and molecular genetic analysis, we conclude that the cat's disorder resulted from the loss of function of CLN6. This study is only the second to identify the molecular genetic basis of a feline NCL. Other cats exhibiting similar signs can now be screened for the CLN6 variant. This could lead to establishment of a feline model of CLN6 disease that could be used in therapeutic intervention studies.
Collapse
Affiliation(s)
- Martin L Katz
- Neurodegenerative Diseases Research Laboratory and Department of Ophthalmology,
| | | | | | | | | | - Wesley C Warren
- Life Sciences Center, University of Missouri, Columbia, MO and
| | | |
Collapse
|
32
|
Zhong Y, Mohan K, Liu J, Al-Attar A, Lin P, Flight RM, Sun Q, Warmoes MO, Deshpande RR, Liu H, Jung KS, Mitov MI, Lin N, Butterfield DA, Lu S, Liu J, Moseley HNB, Fan TWM, Kleinman ME, Wang QJ. Loss of CLN3, the gene mutated in juvenile neuronal ceroid lipofuscinosis, leads to metabolic impairment and autophagy induction in retinal pigment epithelium. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165883. [PMID: 32592935 DOI: 10.1016/j.bbadis.2020.165883] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022]
Abstract
Juvenile neuronal ceroid lipofuscinosis (JNCL, aka. juvenile Batten disease or CLN3 disease) is a lysosomal storage disease characterized by progressive blindness, seizures, cognitive and motor failures, and premature death. JNCL is caused by mutations in the Ceroid Lipofuscinosis, Neuronal 3 (CLN3) gene, whose function is unclear. Although traditionally considered a neurodegenerative disease, CLN3 disease displays eye-specific effects: Vision loss not only is often one of the earliest symptoms of JNCL, but also has been reported in non-syndromic CLN3 disease. Here we described the roles of CLN3 protein in maintaining healthy retinal pigment epithelium (RPE) and normal vision. Using electroretinogram, fundoscopy and microscopy, we showed impaired visual function, retinal autofluorescent lesions, and RPE disintegration and metaplasia/hyperplasia in a Cln3 ~ 1 kb-deletion mouse model [1] on C57BL/6J background. Utilizing a combination of biochemical analyses, RNA-Seq, Seahorse XF bioenergetic analysis, and Stable Isotope Resolved Metabolomics (SIRM), we further demonstrated that loss of CLN3 increased autophagic flux, suppressed mTORC1 and Akt activities, enhanced AMPK activity, and up-regulated gene expression of the autophagy-lysosomal system in RPE-1 cells, suggesting autophagy induction. This CLN3 deficiency induced autophagy induction coincided with decreased mitochondrial oxygen consumption, glycolysis, the tricarboxylic acid (TCA) cycle, and ATP production. We also reported for the first time that loss of CLN3 led to glycogen accumulation despite of impaired glycogen synthesis. Our comprehensive analyses shed light on how loss of CLN3 affect autophagy and metabolism. This work suggests possible links among metabolic impairment, autophagy induction and lysosomal storage, as well as between RPE atrophy/degeneration and vision loss in JNCL.
Collapse
Affiliation(s)
- Yu Zhong
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Kabhilan Mohan
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Ahmad Al-Attar
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Penghui Lin
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Robert M Flight
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Qiushi Sun
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Marc O Warmoes
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Rahul R Deshpande
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Huijuan Liu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Kyung Sik Jung
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States
| | - Mihail I Mitov
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | | | - D Allan Butterfield
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Department of Chemistry, University of Kentucky, Lexington, KY, United States
| | - Shuyan Lu
- Pfizer Inc., San Diego, CA, United States
| | - Jinze Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Department of Computer Science, University of Kentucky, Lexington, KY, United States; Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Hunter N B Moseley
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States; Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Teresa W M Fan
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, United States
| | - Mark E Kleinman
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States
| | - Qing Jun Wang
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States; Markey Cancer Center, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
33
|
Domowicz MS, Chan WC, Claudio-Vázquez P, Henry JG, Ware CB, Andrade J, Dawson G, Schwartz NB. Global Brain Transcriptome Analysis of a Tpp1 Neuronal Ceroid Lipofuscinoses Mouse Model. ASN Neuro 2020; 11:1759091419843393. [PMID: 31003587 PMCID: PMC6475859 DOI: 10.1177/1759091419843393] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In humans, homozygous mutations in the TPP1 gene results in loss
of tripeptidyl peptidase 1 (TPP1) enzymatic activity, leading to late infantile
neuronal ceroid lipofuscinoses disease. Using a mouse model that targets the
Tpp1 gene and recapitulates the pathology and clinical
features of the human disease, we analyzed end-stage (4 months) transcriptional
changes associated with lack of TPP1 activity. Using RNA sequencing technology,
Tpp1 expression changes in the forebrain/midbrain and
cerebellum of 4-month-old homozygotes were compared with strain-related
controls. Transcriptional changes were found in 510 and 1,550 gene transcripts
in forebrain/midbrain and cerebellum, respectively, from
Tpp1-deficient brain tissues when compared with age-matched
controls. Analysis of the differentially expressed genes using the Ingenuity™
pathway software, revealed increased neuroinflammation activity in microglia and
astrocytes that could lead to neuronal dysfunction, particularly in the
cerebellum. We also observed upregulation in the production of nitric oxide and
reactive oxygen species; activation of leukocyte extravasation signals and
complement pathways; and downregulation of major transcription factors involved
in control of circadian rhythm. Several of these expression changes were
confirmed by independent quantitative polymerase chain reaction and histological
analysis by mRNA in situ hybridization, which allowed for an
in-depth anatomical analysis of the pathology and provided independent
confirmation of at least two of the major networks affected in this model. The
identification of differentially expressed genes has revealed new lines of
investigation for this complex disorder that may lead to novel therapeutic
targets.
Collapse
Affiliation(s)
- Miriam S Domowicz
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Wen-Ching Chan
- 2 Center for Research Informatics, Biological Sciences Division, The University of Chicago, IL, USA
| | | | - Judith G Henry
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Christopher B Ware
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Jorge Andrade
- 2 Center for Research Informatics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Glyn Dawson
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Nancy B Schwartz
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA.,3 Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, IL, USA
| |
Collapse
|
34
|
Yamashita A, Shiro Y, Hiraki Y, Yujiri T, Yamazaki T. Implications of graded reductions in CLN6's anti-aggregate activity for the development of the neuronal ceroid lipofuscinoses. Biochem Biophys Res Commun 2020; 525:883-888. [PMID: 32171521 DOI: 10.1016/j.bbrc.2020.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 01/07/2023]
Abstract
CLN6, spanning the endoplasmic reticulum membrane, is a protein of unknown function. Mutations in the CLN6 gene are linked to an autosomal recessively inherited disorder termed CLN6 disease, classified as a form of the neuronal ceroid lipofuscinoses (NCL). The pathogenesis of CLN6 disease remains poorly understood due to a lack of information about physiological roles CLN6 plays. We previously demonstrated that CLN6 has the ability to prevent protein aggregate formation, and thus hypothesized that the abrogation of CLN6's anti-aggregate activity underlies the development of CLN6 disease. To test this hypothesis, we narrowed down the region vital for CLN6's anti-aggregate activity, and subsequently investigated if pathogenic mutations within the region attenuate CLN6's anti-aggregate activity toward four aggregation-prone αB-crystallin (αBC) mutants. None of the four αBC mutants was prevented from aggregating by the Arg106ProfsX truncated CLN6 mutant, the human counterpart of the nclf mutant identified in a naturally occurring mouse model of late infantile-onset CLN6 disease. In contrast, the Arg149Cys and the Arg149His CLN6 mutants, both associated with adult-onset CLN6 disease, blocked aggregation of two out of and all of the four αBC mutants, respectively, indicating that CLN6's anti-aggregate activity is differentially modulated according to the substitution pattern at the same amino acid position. Collectively, we here propose that the graded reduction in CLN6's anti-aggregate activity governs the clinical course of late infantile- and adult-onset NCL.
Collapse
Affiliation(s)
- Arisa Yamashita
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan
| | - Yuki Shiro
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan
| | - Yuri Hiraki
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan
| | - Takatoshi Yujiri
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan
| | - Tetsuo Yamazaki
- Department of Molecular Cell Biology and Medicine, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan.
| |
Collapse
|
35
|
Rosenberg JB, Chen A, Kaminsky SM, Crystal RG, Sondhi D. Advances in the Treatment of Neuronal Ceroid Lipofuscinosis. Expert Opin Orphan Drugs 2019; 7:473-500. [PMID: 33365208 PMCID: PMC7755158 DOI: 10.1080/21678707.2019.1684258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCL) represent a class of neurodegenerative disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders, typically characterized by observation of cognitive and visual impairments, epileptic seizures, ataxia, and deterioration of motor skills. Recent success of a biologic (Brineura®) for the treatment of neurologic manifestations of the central nervous system (CNS) has led to renewed interest in therapeutics for NCL, with the goal of ablating or reversing the impact of these devastating disorders. Despite complex challenges associated with CNS therapy, many treatment modalities have been evaluated, including enzyme replacement therapy, gene therapy, stem cell therapy, and small molecule pharmacotherapy. Because the clinical endpoints for the evaluation of candidate therapies are complex and often reliant on subjective clinical scales, the development of quantitative biomarkers for NCLs has become an apparent necessity for the validation of potential treatments. We will discuss the latest findings in the search for relevant biomarkers for assessing disease progression. For this review, we will focus primarily on recent pre-clinical and clinical developments for treatments to halt or cure these NCL diseases. Continued development of current therapies and discovery of newer modalities will be essential for successful therapeutics for NCL. AREAS COVERED The reader will be introduced to the NCL subtypes, natural histories, experimental animal models, and biomarkers for NCL progression; challenges and different therapeutic approaches, and the latest pre-clinical and clinical research for therapeutic development for the various NCLs. This review corresponds to the literatures covering the years from 1968 to mid-2019, but primarily addresses pre-clinical and clinical developments for the treatment of NCL disease in the last decade and as a follow-up to our 2013 review of the same topic in this journal. EXPERT OPINION Much progress has been made in the treatment of neurologic diseases, such as the NCLs, including better animal models and improved therapeutics with better survival outcomes. Encouraging results are being reported at symposiums and in the literature, with multiple therapeutics reaching the clinical trial stage for the NCLs. The potential for a cure could be at hand after many years of trial and error in the preclinical studies. The clinical development of enzyme replacement therapy (Brineura® for CLN2), immunosuppression (CellCept® for CLN3), and gene therapy vectors (for CLN1, CLN2, CLN3, and CLN6) are providing encouragement to families that have a child afflicted with NCL. We believe that successful therapies in the future may involve the combination of two or more therapeutic modalities to provide therapeutic benefit especially as the patients grow older.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Alvin Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
36
|
Mice deficient in the lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1) display a complex retinal phenotype. Sci Rep 2019; 9:14185. [PMID: 31578378 PMCID: PMC6775149 DOI: 10.1038/s41598-019-50726-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/13/2019] [Indexed: 01/09/2023] Open
Abstract
Neuronal ceroid lipofuscinosis (NCL) type 1 (CLN1) is a neurodegenerative storage disorder caused by mutations in the gene encoding the lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1). CLN1 patients suffer from brain atrophy, mental and motor retardation, seizures, and retinal degeneration ultimately resulting in blindness. Here, we performed an in-depth analysis of the retinal phenotype of a PPT1-deficient mouse, an animal model of this condition. Reactive astrogliosis and microgliosis were evident in mutant retinas prior to the onset of retinal cell loss. Progressive accumulation of storage material, a pronounced dysregulation of various lysosomal proteins, and accumulation of sequestosome/p62-positive aggregates in the inner nuclear layer also preceded retinal degeneration. At advanced stages of the disease, the mutant retina was characterized by a significant loss of ganglion cells, rod and cone photoreceptor cells, and rod and cone bipolar cells. Results demonstrate that PPT1 dysfunction results in early-onset pathological alterations in the mutant retina, followed by a progressive degeneration of various retinal cell types at relatively late stages of the disease. Data will serve as a reference for future work aimed at developing therapeutic strategies for the treatment of retinal degeneration in CLN1 disease.
Collapse
|
37
|
Kauss V, Dambrova M, Medina DL. Pharmacological approaches to tackle NCLs. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165553. [PMID: 31521819 DOI: 10.1016/j.bbadis.2019.165553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 01/06/2023]
Abstract
Neuronal ceroid lipofuscinoses, also collectively known as Batten disease, are a group of rare monogenic disorders caused by mutations in at least 13 different genes. They are characterized by the accumulation of lysosomal storage material and progressive neurological deterioration with dementia, epilepsy, retinopathy, motor disturbances, and early death [1]. Although the identification of disease-causing genes provides an important step for understanding the molecular mechanisms underlying neuronal ceroid lipofuscinoses, compared to other diseases, obstacles to the development of therapies for these rare diseases include less extensive physiopathology knowledge, limited number of patients to test treatments, and poor commercial interest from the industry. Current therapeutic strategies include enzyme replacement therapies, gene therapies targeting the brain and the eye, cell therapies, and pharmacological drugs that could modulate defective molecular pathways. In this review, we will focus in the emerging therapies based in the identification of small-molecules. Recent advances in high- throughput and high-content screening (HTS and HCS) using relevant cell-based assays and applying automation and imaging analysis algorithms, will allow the screening of a large number of compounds in lesser time. These approaches are particularly useful for drug repurposing for Batten disease, that takes the advantage to search for compounds that have already been tested in humans, thereby reducing significantly the resources needed for translation to clinics.
Collapse
Affiliation(s)
- Valerjans Kauss
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia; Riga Stradins University, Dzirciema 16, Riga LV-1007, Latvia
| | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia; Riga Stradins University, Dzirciema 16, Riga LV-1007, Latvia
| | - 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
|
38
|
Tarczyluk-Wells MA, Salzlechner C, Najafi AR, Lim MJ, Smith D, Platt FM, Williams BP, Cooper JD. Combined Anti-inflammatory and Neuroprotective Treatments Have the Potential to Impact Disease Phenotypes in Cln3 -/- Mice. Front Neurol 2019; 10:963. [PMID: 31572287 PMCID: PMC6749847 DOI: 10.3389/fneur.2019.00963] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022] Open
Abstract
Batten disease, or juvenile NCL, is a fatal neurodegenerative disorder that occurs due to mutations in the CLN3 gene. Because the function of CLN3 remains unclear, experimental therapies for JNCL have largely concentrated upon the targeting of downstream pathomechanisms. Neuron loss is preceded by localized glial activation, and in this proof-of-concept study we have investigated whether targeting this innate immune response with ibuprofen in combination with the neuroprotective agent lamotrigine improves the previously documented beneficial effects of immunosuppressants alone. Drugs were administered daily to symptomatic Cln3 -/- mice over a 3 month period, starting at 6 months of age, and their impact was assessed using both behavioral and neuropathological outcome measures. During the treatment period, the combination of ibuprofen and lamotrigine significantly improved the performance of Cln3 -/- mice on the vertical pole test, slowing the disease-associated decline, but had less of an impact upon their rotarod performance. There were also moderate and regionally dependent effects upon astrocyte activation that were most pronounced for ibuprofen alone, but there was no overt effect upon microglial activation. Administering such treatments for longer periods will enable testing for any impact upon the neuron loss that occurs later in disease progression. Given the partial efficacy of these treatments, it will be important to test further drugs of this type in order to find more effective combinations.
Collapse
Affiliation(s)
- Marta A. Tarczyluk-Wells
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Christoph Salzlechner
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Allison R. Najafi
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Ming J. Lim
- Guy's and St. Thomas' NHS Foundation Trust, King's Health Partners Academic Health Science Centre, Evelina London Children's Hospital, London, United Kingdom
- Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - David Smith
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Frances M. Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Brenda P. Williams
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Jonathan D. Cooper
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
39
|
Koster KP, Yoshii A. Depalmitoylation by Palmitoyl-Protein Thioesterase 1 in Neuronal Health and Degeneration. Front Synaptic Neurosci 2019; 11:25. [PMID: 31555119 PMCID: PMC6727029 DOI: 10.3389/fnsyn.2019.00025] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022] Open
Abstract
Protein palmitoylation is the post-translational, reversible addition of a 16-carbon fatty acid, palmitate, to proteins. Protein palmitoylation has recently garnered much attention, as it robustly modifies the localization and function of canonical signaling molecules and receptors. Protein depalmitoylation, on the other hand, is the process by which palmitic acid is removed from modified proteins and contributes, therefore, comparably to palmitoylated-protein dynamics. Palmitoylated proteins also require depalmitoylation prior to lysosomal degradation, demonstrating the significance of this process in protein sorting and turnover. Palmitoylation and depalmitoylation serve as particularly crucial regulators of protein function in neurons, where a specialized molecular architecture and cholesterol-rich membrane microdomains contribute to synaptic transmission. Three classes of depalmitoylating enzymes are currently recognized, the acyl protein thioesterases, α/β hydrolase domain-containing 17 proteins (ABHD17s), and the palmitoyl-protein thioesterases (PPTs). However, a clear picture of depalmitoylation has not yet emerged, in part because the enzyme-substrate relationships and specific functions of depalmitoylation are only beginning to be uncovered. Further, despite the finding that loss-of-function mutations affecting palmitoyl-protein thioesterase 1 (PPT1) function cause a severe pediatric neurodegenerative disease, the role of PPT1 as a depalmitoylase has attracted relatively little attention. Understanding the role of depalmitoylation by PPT1 in neuronal function is a fertile area for ongoing basic science and translational research that may have broader therapeutic implications for neurodegeneration. Here, we will briefly introduce the rapidly growing field surrounding protein palmitoylation and depalmitoylation, then will focus on the role of PPT1 in development, health, and neurological disease.
Collapse
Affiliation(s)
- Kevin P Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Akira Yoshii
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States.,Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, United States.,Department of Neurology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
40
|
Gardner E, Bailey M, Schulz A, Aristorena M, Miller N, Mole SE. Mutation update: Review of TPP1 gene variants associated with neuronal ceroid lipofuscinosis CLN2 disease. Hum Mutat 2019; 40:1924-1938. [PMID: 31283065 PMCID: PMC6851559 DOI: 10.1002/humu.23860] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/25/2019] [Accepted: 06/28/2019] [Indexed: 01/03/2023]
Abstract
Neuronal ceroid lipofuscinosis type 2 (CLN2 disease) is an autosomal recessive condition caused by variants in the TPP1 gene, leading to deficient activity of the lysosomal enzyme tripeptidyl peptidase I (TPP1). We update on the spectrum of TPP1 variants associated with CLN2 disease, comprising 131 unique variants from 389 individuals (717 alleles) collected from the literature review, public databases, and laboratory communications. Previously unrecorded individuals were added to the UCL TPP1‐specific database. Two known pathogenic variants, c.509–1 G>C and c.622 C>T (p.(Arg208*)), collectively occur in 60% of affected individuals in the sample, and account for 50% of disease‐associated alleles. At least 86 variants (66%) are private to single families. Homozygosity occurs in 45% of individuals where both alleles are known (87% of reported individuals). Atypical CLN2 disease, TPP1 enzyme deficiency with disease onset and/or progression distinct from classic late‐infantile CLN2, represents 13% of individuals recorded with associated phenotype. NCBI ClinVar currently holds records for 37% of variants collected here. Effective CLN2 disease management requires early diagnosis; however, irreversible neurodegeneration occurs before a diagnosis is typically reached at age 5. Timely classification and public reporting of TPP1 variants is essential as molecular testing increases in use as a first‐line diagnostic test for pediatric‐onset neurological disease.
Collapse
Affiliation(s)
- Emily Gardner
- UCL MRC Laboratory for Molecular Cell Biology and UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Mitch Bailey
- Global Scientific Affairs, BioMarin Pharmaceutical Inc, Novato, California
| | - Angela Schulz
- Department of Paediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mikel Aristorena
- UCL MRC Laboratory for Molecular Cell Biology and UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Nicole Miller
- Global Scientific Affairs, BioMarin Pharmaceutical Inc, Novato, California
| | - Sara E Mole
- UCL MRC Laboratory for Molecular Cell Biology and UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
41
|
Ella A, Barrière DA, Adriaensen H, Palmer DN, Melzer TR, Mitchell NL, Keller M. The development of brain magnetic resonance approaches in large animal models for preclinical research. Anim Front 2019; 9:44-51. [PMID: 32002261 PMCID: PMC6951960 DOI: 10.1093/af/vfz024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Arsène Ella
- Physiologie de la Reproduction & des Comportements, INRA/CNRS/Université de Tours, France.,MRC Cognition and Brain Science Unit, University of Cambridge, UK
| | - David A Barrière
- Physiologie de la Reproduction & des Comportements, INRA/CNRS/Université de Tours, France.,Neurospin, CEA, France
| | - Hans Adriaensen
- Physiologie de la Reproduction & des Comportements, INRA/CNRS/Université de Tours, France
| | - David N Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, New Zealand
| | - Tracy R Melzer
- Department of Medicine, University of Otago, Christchurch, and New Zealand Brain Research Institute, New Zealand
| | - Nadia L Mitchell
- Faculty of Agriculture and Life Sciences, Lincoln University, New Zealand.,Department of Radiology, University of Otago, Christchurch, New Zealand
| | - Matthieu Keller
- Physiologie de la Reproduction & des Comportements, INRA/CNRS/Université de Tours, France
| |
Collapse
|
42
|
Carraro M, Checchetto V, Szabó I, Bernardi P. F‐ATPsynthase and the permeability transition pore: fewer doubts, more certainties. FEBS Lett 2019; 593:1542-1553. [DOI: 10.1002/1873-3468.13485] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Michela Carraro
- Department of Biomedical Sciences University of Padova Italy
| | | | - Ildikó Szabó
- Department of Biology University of Padova Italy
| | - Paolo Bernardi
- Department of Biomedical Sciences University of Padova Italy
| |
Collapse
|
43
|
Kline RA, Wishart TM, Mills K, Heywood WE. Applying modern Omic technologies to the Neuronal Ceroid Lipofuscinoses. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165498. [PMID: 31207290 DOI: 10.1016/j.bbadis.2019.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/30/2019] [Accepted: 06/07/2019] [Indexed: 11/27/2022]
Abstract
The Neuronal Ceroid Lipofuscinoses are a group of severe and progressive neurodegenerative disorders, which generally present during childhood. With new treatments emerging on the horizon, there is a growing need to understand the specific disease mechanisms as well as identify prospective biomarkers for use to stratify patients and monitor treatment. The use of Omics technologies to NCLs has the potential to address this need. We discuss the recent use and outcomes of Omics to various forms of NCL including identification of interactomes, affected biological pathways and potential biomarker candidates. We also identify common pathways affected in NCL across the reviewed studies.
Collapse
Affiliation(s)
- Rachel A Kline
- Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Edinburgh, UK; The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas M Wishart
- Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Edinburgh, UK; The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Kevin Mills
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK; NIHR Great Ormond Street Biomedical Research Centre, Great Ormond Street Hospital, UCL Great Ormond Street Institute of Child Health, UK
| | - Wendy E Heywood
- Inborn Errors of Metabolism Section, Genetics & Genomic Medicine Unit, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK; NIHR Great Ormond Street Biomedical Research Centre, Great Ormond Street Hospital, UCL Great Ormond Street Institute of Child Health, UK.
| |
Collapse
|
44
|
Schmidtke C, Tiede S, Thelen M, Käkelä R, Jabs S, Makrypidi G, Sylvester M, Schweizer M, Braren I, Brocke-Ahmadinejad N, Cotman SL, Schulz A, Gieselmann V, Braulke T. Lysosomal proteome analysis reveals that CLN3-defective cells have multiple enzyme deficiencies associated with changes in intracellular trafficking. J Biol Chem 2019; 294:9592-9604. [PMID: 31040178 DOI: 10.1074/jbc.ra119.008852] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/21/2019] [Indexed: 12/25/2022] Open
Abstract
Numerous lysosomal enzymes and membrane proteins are essential for the degradation of proteins, lipids, oligosaccharides, and nucleic acids. The CLN3 gene encodes a lysosomal membrane protein of unknown function, and CLN3 mutations cause the fatal neurodegenerative lysosomal storage disorder CLN3 (Batten disease) by mechanisms that are poorly understood. To define components critical for lysosomal homeostasis that are affected by this disease, here we quantified the lysosomal proteome in cerebellar cell lines derived from a CLN3 knock-in mouse model of human Batten disease and control cells. We purified lysosomes from SILAC-labeled, and magnetite-loaded cerebellar cells by magnetic separation and analyzed them by MS. This analysis identified 70 proteins assigned to the lysosomal compartment and 3 lysosomal cargo receptors, of which most exhibited a significant differential abundance between control and CLN3-defective cells. Among these, 28 soluble lysosomal proteins catalyzing the degradation of various macromolecules had reduced levels in CLN3-defective cells. We confirmed these results by immunoblotting and selected protease and glycosidase activities. The reduction of 11 lipid-degrading lysosomal enzymes correlated with reduced capacity for lipid droplet degradation and several alterations in the distribution and composition of membrane lipids. In particular, levels of lactosylceramides and glycosphingolipids were decreased in CLN3-defective cells, which were also impaired in the recycling pathway of the exocytic transferrin receptor. Our findings suggest that CLN3 has a crucial role in regulating lysosome composition and their function, particularly in degrading of sphingolipids, and, as a consequence, in membrane transport along the recycling endosome pathway.
Collapse
Affiliation(s)
- Carolin Schmidtke
- From the Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 20246
| | - Stephan Tiede
- From the Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 20246
| | - Melanie Thelen
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany D-53115
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland 00014
| | - Sabrina Jabs
- Leibniz-Institut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany 13125
| | - Georgia Makrypidi
- From the Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 20246
| | - Marc Sylvester
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany D-53115
| | - Michaela Schweizer
- the Department of Electron Microscopy, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 20251
| | - Ingke Braren
- Vector Core Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 20251
| | | | - Susan L Cotman
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Angela Schulz
- From the Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 20246
| | - Volkmar Gieselmann
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany D-53115
| | - Thomas Braulke
- From the Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 20246,
| |
Collapse
|
45
|
Poppens MJ, Cain JT, Johnson TB, White KA, Davis SS, Laufmann R, Kloth AD, Weimer JM. Tracking sex-dependent differences in a mouse model of CLN6-Batten disease. Orphanet J Rare Dis 2019; 14:19. [PMID: 30665444 PMCID: PMC6341540 DOI: 10.1186/s13023-019-0994-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 01/07/2019] [Indexed: 02/08/2023] Open
Abstract
Background CLN6-Batten disease is a rare neurodevelopmental disorder characterized pathologically by the accumulation of lysosomal storage material, glial activation and neurodegeneration, and phenotypically by loss of vision, motor coordination, and cognitive ability, with premature death occurring in the second decade of life. In this study, we investigate whether sex differences in a mouse model of CLN6-Batten disease impact disease onset and progression. Results A number of noteworthy differences were observed including elevated accumulation of mitochondrial ATP synthase subunit C in the thalamus and cortex of female Cln6 mutant mice at 2 months of age. Moreover, female mutant mice showed more severe behavioral deficits. Beginning at 9 months of age, female mice demonstrated learning and memory deficits and suffered a more severe decline in motor coordination. Further, compared to their male counterparts, female animals succumbed to the disease at a slightly younger age, indicating an accelerated disease progression. Conversely, males showed a marked increase in microglial activation at 6 months of age in the cortex relative to females. Conclusions Thus, as female Cln6 mutant mice exhibit cellular and behavioral deficits that precede similar pathologies in male mutant mice, our findings suggest the need for consideration of sex-based differences in CLN6 disease progression during development of preclinical and clinical studies. Electronic supplementary material The online version of this article (10.1186/s13023-019-0994-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- McKayla J Poppens
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jacob T Cain
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Tyler B Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Samantha S Davis
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Rachel Laufmann
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | | | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA. .,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA.
| |
Collapse
|
46
|
Mole SE, Anderson G, Band HA, Berkovic SF, Cooper JD, Kleine Holthaus SM, McKay TR, Medina DL, Rahim AA, Schulz A, Smith AJ. Clinical challenges and future therapeutic approaches for neuronal ceroid lipofuscinosis. Lancet Neurol 2019; 18:107-116. [PMID: 30470609 DOI: 10.1016/s1474-4422(18)30368-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 12/24/2022]
Abstract
Treatment of the neuronal ceroid lipofuscinoses, also known as Batten disease, is at the start of a new era because of diagnostic and therapeutic advances relevant to this group of inherited neurodegenerative and life-limiting disorders that affect children. Diagnosis has improved with the use of comprehensive DNA-based tests that simultaneously screen for many genes. The identification of disease-causing mutations in 13 genes provides a basis for understanding the molecular mechanisms underlying neuronal ceroid lipofuscinoses, and for the development of targeted therapies. These targeted therapies include enzyme replacement therapies, gene therapies targeting the brain and the eye, cell therapies, and pharmacological drugs that could modulate defective molecular pathways. Such therapeutic developments have the potential to enable earlier diagnosis and better targeted therapeutic management. The first approved treatment is an intracerebroventricularly administered enzyme for neuronal ceroid lipofuscinosis type 2 disease that delays symptom progression. Efforts are underway to make similar progress for other forms of the disorder.
Collapse
Affiliation(s)
- Sara E Mole
- Medical Research Council Laboratory for Molecular Cell Biology and UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Glenn Anderson
- Department of Histopathology, Great Ormond Street Hospital, London, UK
| | | | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, Austin Health & Northern Health, University of Melbourne, Melbourne, VIC, Australia
| | - Jonathan D Cooper
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | | | - Tristan R McKay
- Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London, UK
| | - Angela Schulz
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander J Smith
- UCL Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
47
|
Mathavarajah S, O'Day DH, Huber RJ. Neuronal Ceroid Lipofuscinoses: Connecting Calcium Signalling through Calmodulin. Cells 2018; 7:cells7110188. [PMID: 30380624 PMCID: PMC6262527 DOI: 10.3390/cells7110188] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/24/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
Despite the increased focus on the role of calcium in the neuronal ceroid lipofuscinoses (NCLs, also known as Batten disease), links between calcium signalling and the proteins associated with the disease remain to be identified. A central protein in calcium signalling is calmodulin (CaM), which regulates many of the same cellular processes affected in the NCLs. In this study, we show that 11 of the 13 NCL proteins contain putative CaM-binding domains (CaMBDs). Many of the missense mutations documented from NCL patients overlap with the predicted CaMBDs and are often key residues of those domains. The two NCL proteins lacking such domains, CLN7 and CLN11, share a commonality in undergoing proteolytic processing by cathepsin L, which contains a putative CaMBD. Since CaM appears to have both direct and indirect roles in the NCLs, targeting it may be a valid therapeutic approach for treating the disease.
Collapse
Affiliation(s)
| | - Danton H O'Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| | - Robert J Huber
- Department of Biology, Trent University, Peterborough, ON K9L 0G2, Canada.
| |
Collapse
|
48
|
von Eisenhart-Rothe P, Grubman A, Greferath U, Fothergill LJ, Jobling AI, Phipps JA, White AR, Fletcher EL, Vessey KA. Failure of Autophagy–Lysosomal Pathways in Rod Photoreceptors Causes the Early Retinal Degeneration Phenotype Observed inCln6nclfMice. ACTA ACUST UNITED AC 2018; 59:5082-5097. [DOI: 10.1167/iovs.18-24757] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
| | - Alexandra Grubman
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| | - Linda J. Fothergill
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew I. Jobling
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| | - Joanna A. Phipps
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anthony R. White
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Erica L. Fletcher
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kirstan A. Vessey
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Lange J, Haslett LJ, Lloyd-Evans E, Pocock JM, Sands MS, Williams BP, Cooper JD. Compromised astrocyte function and survival negatively impact neurons in infantile neuronal ceroid lipofuscinosis. Acta Neuropathol Commun 2018; 6:74. [PMID: 30089511 PMCID: PMC6081811 DOI: 10.1186/s40478-018-0575-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 01/28/2023] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are the most common cause of childhood dementia and are invariably fatal. Early localized glial activation occurs in these disorders, and accurately predicts where neuronal loss is most pronounced. Recent evidence suggests that glial dysfunction may contribute to neuron loss, and we have now explored this possibility in infantile NCL (INCL, CLN1 disease). We grew primary cultures of astrocytes, microglia, and neurons derived from Ppt1 deficient mice (Ppt1−/−) and assessed their properties compared to wildtype (WT) cultures, before co-culturing them in different combinations (astrocytes with microglia, astrocytes or microglia with neurons, all three cell types together). These studies revealed that both Ppt1−/− astrocytes and microglia exhibit a more activated phenotype under basal unstimulated conditions, as well as alterations to their protein expression profile following pharmacological stimulation. Ppt1- /− astrocytes also displayed abnormal calcium signalling and an elevated cytoplasmic Ca2+ level, and a profound defect in their survival. Ppt1−/− neurons displayed decreased neurite outgrowth, altered complexity, a reduction in cell body size, and impaired neuron survival with prolonged time in culture. In co-cultures, the presence of both astrocytes and microglia from Ppt1−/− mice further impaired the morphology of both wild type and Ppt1−/− neurons. This negative influence was more pronounced for Ppt1−/− microglia, which appeared to trigger increased Ppt1−/− neuronal death. In contrast, wild type glial cells, especially astrocytes, ameliorated some of the morphological defects observed in Ppt1−/− neurons. These findings suggest that both Ppt1−/− microglia and astrocytes are dysfunctional and may contribute to the neurodegeneration observed in CLN1 disease. However, the dysfunctional phenotypes of Ppt1−/− glia are different from those present in CLN3 disease, suggesting that the pathogenic role of glia may differ between NCLs.
Collapse
|
50
|
McBride JL, Neuringer M, Ferguson B, Kohama SG, Tagge IJ, Zweig RC, Renner LM, McGill TJ, Stoddard J, Peterson S, Su W, Sherman LS, Domire JS, Ducore RM, Colgin LM, Lewis AD. Discovery of a CLN7 model of Batten disease in non-human primates. Neurobiol Dis 2018; 119:65-78. [PMID: 30048804 PMCID: PMC6200145 DOI: 10.1016/j.nbd.2018.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/23/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
We have identified a natural Japanese macaque model of the childhood neurodegenerative disorder neuronal ceroid lipofuscinosis, commonly known as Batten Disease, caused by a homozygous frameshift mutation in the CLN7 gene (CLN7−/−). Affected macaques display progressive neurological deficits including visual impairment, tremor, incoordination, ataxia and impaired balance. Imaging, functional and pathological studies revealed that CLN7−/− macaques have reduced retinal thickness and retinal function early in disease, followed by profound cerebral and cerebellar atrophy that progresses over a five to six-year disease course. Histological analyses showed an accumulation of cerebral, cerebellar and cardiac storage material as well as degeneration of neurons, white matter fragmentation and reactive gliosis throughout the brain of affected animals. This novel CLN7−/− macaque model recapitulates key behavioral and neuropathological features of human Batten Disease and provides novel insights into the pathophysiology linked to CLN7 mutations. These animals will be invaluable for evaluating promising therapeutic strategies for this devastating disease.
Collapse
Affiliation(s)
- Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.
| | - Martha Neuringer
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States; Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Betsy Ferguson
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States; Division of Genetics, Oregon National Primate Research Center, Beaverton, OR, United States; Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Ian J Tagge
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Robert C Zweig
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Laurie M Renner
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Trevor J McGill
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States; Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Jonathan Stoddard
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Samuel Peterson
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Weiping Su
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, United States
| | - Jacqueline S Domire
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Rebecca M Ducore
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Lois M Colgin
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Anne D Lewis
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, United States
| |
Collapse
|