Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Proc Natl Acad Sci U S A ; 117(16): 9032-9041, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32253319

ABSTRACT

Lysosomal storage diseases (LSDs) are typically caused by a deficiency in a soluble acid hydrolase and are characterized by the accumulation of undegraded substrates in the lysosome. Determining the role of specific cell types in the pathogenesis of LSDs is a major challenge due to the secretion and subsequent uptake of lysosomal hydrolases by adjacent cells, often referred to as "cross-correction." Here we create and validate a conditional mouse model for cell-autonomous expression of galactocerebrosidase (GALC), the lysosomal enzyme deficient in Krabbe disease. We show that lysosomal membrane-tethered GALC (GALCLAMP1) retains enzyme activity, is able to cleave galactosylsphingosine, and is unable to cross-correct. Ubiquitous expression of GALCLAMP1 fully rescues the phenotype of the GALC-deficient mouse (Twitcher), and widespread deletion of GALCLAMP1 recapitulates the Twitcher phenotype. We demonstrate the utility of this model by deleting GALCLAMP1 specifically in myelinating Schwann cells in order to characterize the peripheral neuropathy seen in Krabbe disease.


Subject(s)
Galactosylceramidase/metabolism , Leukodystrophy, Globoid Cell/pathology , Lysosomes/enzymology , Recombinant Fusion Proteins/metabolism , Schwann Cells/pathology , Animals , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts , Galactosylceramidase/genetics , Gene Knockdown Techniques , Humans , Intracellular Membranes/metabolism , Leukodystrophy, Globoid Cell/diagnosis , Leukodystrophy, Globoid Cell/genetics , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Male , Mice , Mice, Transgenic , Primary Cell Culture , Recombinant Fusion Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 116(40): 20097-20103, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31527255

ABSTRACT

Infantile globoid cell leukodystrophy (GLD, Krabbe disease) is a fatal demyelinating disorder caused by a deficiency in the lysosomal enzyme galactosylceramidase (GALC). GALC deficiency leads to the accumulation of the cytotoxic glycolipid, galactosylsphingosine (psychosine). Complementary evidence suggested that psychosine is synthesized via an anabolic pathway. Here, we show instead that psychosine is generated catabolically through the deacylation of galactosylceramide by acid ceramidase (ACDase). This reaction uncouples GALC deficiency from psychosine accumulation, allowing us to test the long-standing "psychosine hypothesis." We demonstrate that genetic loss of ACDase activity (Farber disease) in the GALC-deficient mouse model of human GLD (twitcher) eliminates psychosine accumulation and cures GLD. These data suggest that ACDase could be a target for substrate reduction therapy (SRT) in Krabbe patients. We show that pharmacological inhibition of ACDase activity with carmofur significantly decreases psychosine accumulation in cells from a Krabbe patient and prolongs the life span of the twitcher (Twi) mouse. Previous SRT experiments in the Twi mouse utilized l-cycloserine, which inhibits an enzyme several steps upstream of psychosine synthesis, thus altering the balance of other important lipids. Drugs that directly inhibit ACDase may have a more acceptable safety profile due to their mechanistic proximity to psychosine biogenesis. In total, these data clarify our understanding of psychosine synthesis, confirm the long-held psychosine hypothesis, and provide the impetus to discover safe and effective inhibitors of ACDase to treat Krabbe disease.


Subject(s)
Acid Ceramidase/genetics , Gene Deletion , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/metabolism , Psychosine/metabolism , Animals , Cell Line, Tumor , Cytokines/metabolism , DNA Methylation , Disease Models, Animal , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Leukodystrophy, Globoid Cell/drug therapy
3.
Proc Natl Acad Sci U S A ; 114(29): E5920-E5929, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28673981

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL, or CLN1 disease) is an inherited neurodegenerative storage disorder caused by a deficiency of the lysosomal enzyme palmitoyl protein thioesterase 1 (PPT1). It was widely believed that the pathology associated with INCL was limited to the brain, but we have now found unexpectedly profound pathology in the human INCL spinal cord. Similar pathological changes also occur at every level of the spinal cord of PPT1-deficient (Ppt1-/- ) mice before the onset of neuropathology in the brain. Various forebrain-directed gene therapy approaches have only had limited success in Ppt1-/- mice. Targeting the spinal cord via intrathecal administration of an adeno-associated virus (AAV) gene transfer vector significantly prevented pathology and produced significant improvements in life span and motor function in Ppt1-/- mice. Surprisingly, forebrain-directed gene therapy resulted in essentially no PPT1 activity in the spinal cord, and vice versa. This leads to a reciprocal pattern of histological correction in the respective tissues when comparing intracranial with intrathecal injections. However, the characteristic pathological features of INCL were almost completely absent in both the brain and spinal cord when intracranial and intrathecal injections of the same AAV vector were combined. Targeting both the brain and spinal cord also produced dramatic and synergistic improvements in motor function with an unprecedented increase in life span. These data show that spinal cord pathology significantly contributes to the clinical progression of INCL and can be effectively targeted therapeutically. This has important implications for the delivery of therapies in INCL, and potentially in other similar disorders.


Subject(s)
Brain/pathology , Genetic Therapy/methods , Membrane Proteins/pharmacology , Neuronal Ceroid-Lipofuscinoses/therapy , Spinal Cord/pathology , Thiolester Hydrolases/pharmacology , Animals , Brain/drug effects , Child , Disease Models, Animal , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Humans , Injections, Intraventricular/methods , Injections, Spinal , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Mutant Strains , Neuroglia/pathology , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/pathology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Spinal Cord/drug effects , Thiolester Hydrolases/administration & dosage , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism
4.
Mol Genet Metab ; 117(2): 210-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26597320

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL, Infantile Batten disease) is an invariably fatal neurodegenerative pediatric disorder caused by an inherited mutation in the PPT1 gene. Patients with INCL lack the lysosomal enzyme palmitoyl protein thioesterase-1 (PPT1, EC 3.1.2.22), resulting in intracellular accumulation of autofluorescent storage material and subsequent neuropathology. The Ppt1(-/-) mouse is deficient in PPT1 activity and represents a useful animal model of INCL that recapitulates most of the clinical and pathological aspects of the disease. Preclinical therapeutic experiments performed in the INCL mouse include CNS-directed gene therapy and recombinant enzyme replacement therapy; both seek to re-establish therapeutic levels of the deficient enzyme. We present a novel method for the histochemical localization of PPT1 activity in the Ppt1(-/-) mouse. By utilizing the substrate CUS-9235, tissues known to be positive for PPT1 activity turn varying intensities of blue. Presented here are histochemistry data showing the staining pattern in Ppt1(-/-), wild type, and Ppt1(-/-) mice treated with enzyme replacement therapy or AAV2/9-PPT1-mediated gene therapy. Results are paired with quantitative biochemistry data that confirm the ability of CUS-9235 to detect and localize PPT1 activity. This new method complements the current tools for the study of INCL and evaluation of effective therapies.


Subject(s)
Thiolester Hydrolases/metabolism , Animals , Female , Genetic Therapy , Humans , Kidney/enzymology , Liver/enzymology , Male , Mice, Knockout , Myocardium/enzymology , Neuronal Ceroid-Lipofuscinoses/therapy , Organ Specificity , Thiolester Hydrolases/genetics
5.
J Neurosci ; 34(17): 5800-15, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760840

ABSTRACT

Nicotinamide adenine dinucleotide (NAD(+)) is an enzyme cofactor or cosubstrate in many essential biological pathways. To date, the primary source of neuronal NAD(+) has been unclear. NAD(+) can be synthesized from several different precursors, among which nicotinamide is the substrate predominantly used in mammals. The rate-limiting step in the NAD(+) biosynthetic pathway from nicotinamide is performed by nicotinamide phosphoribosyltransferase (Nampt). Here, we tested the hypothesis that neurons use intracellular Nampt-mediated NAD(+) biosynthesis by generating and evaluating mice lacking Nampt in forebrain excitatory neurons (CaMKIIαNampt(-/-) mice). CaMKIIαNampt(-/-) mice showed hippocampal and cortical atrophy, astrogliosis, microgliosis, and abnormal CA1 dendritic morphology by 2-3 months of age. Importantly, these histological changes occurred with altered intrahippocampal connectivity and abnormal behavior; including hyperactivity, some defects in motor skills, memory impairment, and reduced anxiety, but in the absence of impaired sensory processes or long-term potentiation of the Schaffer collateral pathway. These results clearly demonstrate that forebrain excitatory neurons mainly use intracellular Nampt-mediated NAD(+) biosynthesis to mediate their survival and function. Studying this particular NAD(+) biosynthetic pathway in these neurons provides critical insight into their vulnerability to pathophysiological stimuli and the development of therapeutic and preventive interventions for their preservation.


Subject(s)
Cerebral Cortex/metabolism , Cognition/physiology , Hippocampus/metabolism , Neurons/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Animals , Atrophy/genetics , Atrophy/metabolism , Atrophy/pathology , Behavior, Animal/physiology , Cerebral Cortex/pathology , Gliosis/metabolism , Gliosis/pathology , Hippocampus/pathology , Memory/physiology , Mice , Mice, Knockout , Motor Activity/physiology , Nerve Net/metabolism , Nerve Net/pathology , Neurons/pathology , Nicotinamide Phosphoribosyltransferase/genetics
6.
J Neurosci ; 34(21): 7281-92, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24849360

ABSTRACT

Apolipoprotein E (apoE) is the strongest known genetic risk factor for late onset Alzheimer's disease (AD). It influences amyloid-ß (Aß) clearance and aggregation, which likely contributes in large part to its role in AD pathogenesis. We recently found that HJ6.3, a monoclonal antibody against apoE, significantly reduced Aß plaque load when given to APPswe/PS1ΔE9 (APP/PS1) mice starting before the onset of plaque deposition. To determine whether the anti-apoE antibody HJ6.3 affects Aß plaques, neuronal network function, and behavior in APP/PS1 mice after plaque onset, we administered HJ6.3 (10 mg/kg/week) or PBS intraperitoneally to 7-month-old APP/PS1 mice for 21 weeks. HJ6.3 mildly improved spatial learning performance in the water maze, restored resting-state functional connectivity, and modestly reduced brain Aß plaque load. There was no effect of HJ6.3 on total plasma cholesterol or cerebral amyloid angiopathy. To investigate the underlying mechanisms of anti-apoE immunotherapy, HJ6.3 was applied to the brain cortical surface and amyloid deposition was followed over 2 weeks using in vivo imaging. Acute exposure to HJ6.3 affected the course of amyloid deposition in that it prevented the formation of new amyloid deposits, limited their growth, and was associated with occasional clearance of plaques, a process likely associated with direct binding to amyloid aggregates. Topical application of HJ6.3 for only 14 d also decreased the density of amyloid plaques assessed postmortem. Collectively, these studies suggest that anti-apoE antibodies have therapeutic potential when given before or after the onset of Aß pathology.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Antibodies/therapeutic use , Apolipoproteins E/immunology , Brain/metabolism , Alzheimer Disease/blood , Alzheimer Disease/genetics , Amyloid beta-Peptides/drug effects , Amyloid beta-Protein Precursor/genetics , Amyloidosis/drug therapy , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Brain/drug effects , Cholesterol/blood , Disease Models, Animal , Female , Hemorrhage/drug therapy , Hemorrhage/etiology , Lameness, Animal/drug therapy , Lameness, Animal/etiology , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics
7.
J Neurosci ; 34(39): 13077-82, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25253854

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL) is an inherited neurodegenerative lysosomal storage disease (LSD) caused by a deficiency in palmitoyl protein thioesterase-1 (PPT1). Studies in Ppt1(-/-) mice demonstrate that glial activation is central to the pathogenesis of INCL. Astrocyte activation precedes neuronal loss, while cytokine upregulation associated with microglial reactivity occurs before and concurrent with neurodegeneration. Therefore, we hypothesized that cytokine cascades associated with neuroinflammation are important therapeutic targets for the treatment of INCL. MW01-2-151SRM (MW151) is a blood-brain barrier penetrant, small-molecule anti-neuroinflammatory that attenuates glial cytokine upregulation in models of neuroinflammation such as traumatic brain injury, Alzheimer's disease, and kainic acid toxicity. Thus, we used MW151, alone and in combination with CNS-directed, AAV-mediated gene therapy, as a possible treatment for INCL. MW151 alone decreased seizure susceptibility. When combined with AAV-mediated gene therapy, treated INCL mice had increased life spans, improved motor performance, and eradication of seizures. Combination-treated INCL mice also had decreased brain atrophy, astrocytosis, and microglial activation, as well as intermediary effects on cytokine upregulation. These data suggest that MW151 can attenuate seizure susceptibility but is most effective when used in conjunction with a therapy that targets the primary genetic defect.


Subject(s)
Blood-Brain Barrier/metabolism , Genetic Therapy , Microglia/metabolism , Neuronal Ceroid-Lipofuscinoses/therapy , Thiolester Hydrolases/genetics , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Blood-Brain Barrier/drug effects , Cytokines/genetics , Cytokines/metabolism , Dependovirus/genetics , Locomotion , Mice , Mice, Inbred C57BL , Microglia/drug effects , Pyridazines/pharmacokinetics , Pyridazines/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Seizures/therapy , Thiolester Hydrolases/metabolism
8.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37104037

ABSTRACT

Although a disease-modifying therapy for classic late infantile neuronal ceroid lipofuscinosis (CLN2 disease) exists, poor understanding of cellular pathophysiology has hampered the development of more effective and persistent therapies. Here, we investigated the nature and progression of neurological and underlying neuropathological changes in Cln2R207X mice, which carry one of the most common pathogenic mutations in human patients but are yet to be fully characterized. Long-term electroencephalography recordings revealed progressive epileptiform abnormalities, including spontaneous seizures, providing a robust, quantifiable, and clinically relevant phenotype. These seizures were accompanied by the loss of multiple cortical neuron populations, including those stained for interneuron markers. Further histological analysis revealed early localized microglial activation months before neuron loss started in the thalamocortical system and spinal cord, which was accompanied by astrogliosis. This pathology was more pronounced and occurred in the cortex before the thalamus or spinal cord and differed markedly from the staging seen in mouse models of other forms of neuronal ceroid lipofuscinosis. Neonatal administration of adeno-associated virus serotype 9-mediated gene therapy ameliorated the seizure and gait phenotypes and prolonged the life span of Cln2R207X mice, attenuating most pathological changes. Our findings highlight the importance of clinically relevant outcome measures for judging preclinical efficacy of therapeutic interventions for CLN2 disease.


Subject(s)
Neurons , Seizures , Animals , Humans , Mice , Neurons/pathology , Seizures/genetics , Seizures/therapy , Seizures/pathology , Gliosis/pathology , Interneurons/pathology , Thalamus/pathology , Disease Models, Animal
9.
Sci Rep ; 12(1): 11286, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35789177

ABSTRACT

Cannabidiol (CBD) has gained attention as a therapeutic agent and is purported to have immunomodulatory, neuroprotective, and anti-seizure effects. Here, we determined the effects of chronic CBD administration in a mouse model of CLN1 disease (Cln1-/-) that simultaneously exhibits neuroinflammation, neurodegeneration, and spontaneous seizures. Proteomic analysis showed that putative CBD receptors are expressed at similar levels in the brains of Cln1-/- mice compared to normal animals. Cln1-/- mice received an oral dose (100 mg/kg/day) of CBD for six months and were evaluated for changes in pathological markers of disease and seizures. Chronic cannabidiol administration was well-tolerated, high levels of CBD were detected in the brain, and markers of astrocytosis and microgliosis were reduced. However, CBD had no apparent effect on seizure frequency or neuron survival. These data are consistent with CBD having immunomodulatory effects. It is possible that a higher dose of CBD could also reduce neurodegeneration and seizure frequency.


Subject(s)
Cannabidiol , Graft vs Host Disease , Animals , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Disease Models, Animal , Graft vs Host Disease/drug therapy , Mice , Neuroinflammatory Diseases , Neuronal Ceroid-Lipofuscinoses , Proteomics
10.
J Clin Invest ; 132(20)2022 10 17.
Article in English | MEDLINE | ID: mdl-36040802

ABSTRACT

CLN1 disease, also called infantile neuronal ceroid lipofuscinosis (NCL) or infantile Batten disease, is a fatal neurodegenerative lysosomal storage disorder resulting from mutations in the CLN1 gene encoding the soluble lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT1). Therapies for CLN1 disease have proven challenging because of the aggressive disease course and the need to treat widespread areas of the brain and spinal cord. Indeed, gene therapy has proven less effective for CLN1 disease than for other similar lysosomal enzyme deficiencies. We therefore tested the efficacy of enzyme replacement therapy (ERT) by administering monthly infusions of recombinant human PPT1 (rhPPT1) to PPT1-deficient mice (Cln1-/-) and CLN1R151X sheep to assess how to potentially scale up for translation. In Cln1-/- mice, intracerebrovascular (i.c.v.) rhPPT1 delivery was the most effective route of administration, resulting in therapeutically relevant CNS levels of PPT1 activity. rhPPT1-treated mice had improved motor function, reduced disease-associated pathology, and diminished neuronal loss. In CLN1R151X sheep, i.c.v. infusions resulted in widespread rhPPT1 distribution and positive treatment effects measured by quantitative structural MRI and neuropathology. This study demonstrates the feasibility and therapeutic efficacy of i.c.v. rhPPT1 ERT. These findings represent a key step toward clinical testing of ERT in children with CLN1 disease and highlight the importance of a cross-species approach to developing a successful treatment strategy.


Subject(s)
Neuronal Ceroid-Lipofuscinoses , Animals , Child , Disease Models, Animal , Enzyme Replacement Therapy , Humans , Mice , Mutation , Neuronal Ceroid-Lipofuscinoses/drug therapy , Neuronal Ceroid-Lipofuscinoses/genetics , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL