Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 29: 185-201, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37063480

ABSTRACT

Mutations in GBA1, encoding the lysosomal acid ß-glucosidase (GCase), cause neuronopathic Gaucher disease (nGD) and promote Parkinson disease (PD). The mutations on GBA1 include deletion and missense mutations that are pathological and lead to GCase deficiency in Gaucher disease. Both nGD and PD lack disease-modifying treatments and are critical unmet medical needs. In this study, we evaluated a cell therapy treatment using mouse iPSC-derived neural precursor cells (NPCs) engineered to overexpress GCase (termed hGBA1-NPCs). The hGBA1-NPCs secreted GCase that was taken up by adjacent mouse Gba -/- neurons and improved GCase activity, reduced GCase substrate accumulation, and improved mitochondrial function. Short-term in vivo effects were evaluated in 9H/PS-NA mice, an nGD mouse model exhibiting neuropathology and α-synuclein aggregation, the typical PD phenotypes. Intravenously administrated hGBA1-NPCs were engrafted throughout the brain and differentiated into neural lineages. GCase activity was increased in various brain regions of treated 9H/PS-NA mice. Compared with vehicle, hGBA1-NPC-transplanted mice showed ∼50% reduction of α-synuclein aggregates in the substantia nigra, significant reduction of neuroinflammation and neurodegeneration in the regions of NPC migration, and increased expression of neurotrophic factors that support neural cell function. Together, these results support the therapeutic benefit of intravenous delivery of iPSC-derived NPCs overexpressing GCase in mitigating nGD and PD phenotypes and establish the feasibility of combined cell and gene therapy for GBA1-associated PD.

2.
Proc Natl Acad Sci U S A ; 120(1): e2210442120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574647

ABSTRACT

Mutations in GBA1, encoding glucocerebrosidase (GCase), cause Gaucher disease (GD) and are also genetic risks in developing Parkinson's disease (PD). Currently, the approved therapies are only effective for directly treating visceral symptoms, but not for primary neuronopathic involvement in GD (nGD). Progranulin (PGRN), encoded by GRN, is a novel modifier of GCase, but the impact of PGRN in GBA1 mutation-associated pathologies in vivo remains unknown. Herein, Grn-/- mice crossed into Gba9v/9v mice, a Gba1 mutant line homozygous for the Gba1 D409V mutation, generating Grn-/-Gba9v/9v (PG9V) mice. PG9V mice exhibited neurobehavioral deficits, early onset, and more severe GD phenotypes compared to Grn-/- and Gba9v/9v mice. Moreover, PG9V mice also displayed PD-like phenotype. Mechanistic analysis revealed that PGRN deficiency caused severe neuroinflammation with microgliosis and astrogliosis, along with impaired autophagy associated with the Gba1 mutation. A PGRN-derived peptide, termed ND7, ameliorated the disease phenotype in GD patient fibroblasts ex vivo. Unexpectedly, ND7 penetrated the blood-brain barrier (BBB) and effectively ameliorated the nGD manifestations and PD pathology in Gba9v/null and PG9V mice. Collectively, this study not only provides the first line of in vivo but also ex vivo evidence demonstrating the crucial role of PGRN in GBA1/Gba1 mutation-related pathologies, as well as a clinically relevant mouse model for mechanistic and potential therapeutics studies for nGD and PD. Importantly, a BBB penetrant PGRN-derived biologic was developed that may provide treatment for rare lysosomal storage diseases and common neurodegenerative disorders, particularly nGD and PD.


Subject(s)
Gaucher Disease , Parkinson Disease , Progranulins , Animals , Mice , Brain/metabolism , Gaucher Disease/genetics , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Lysosomes/metabolism , Mutation , Parkinson Disease/genetics , Progranulins/genetics , Mice, Knockout
3.
Sci Transl Med ; 14(636): eabl9945, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35294256

ABSTRACT

Hematopoietic cell transplantation after myeloablative conditioning has been used to treat various genetic metabolic syndromes but is largely ineffective in diseases affecting the brain presumably due to poor and variable myeloid cell incorporation into the central nervous system. Here, we developed and characterized a near-complete and homogeneous replacement of microglia with bone marrow cells in mice without the need for genetic manipulation of donor or host. The high chimerism resulted from a competitive advantage of scarce donor cells during microglia repopulation rather than enhanced recruitment from the periphery. Hematopoietic stem cells, but not immediate myeloid or monocyte progenitor cells, contained full microglia replacement potency equivalent to whole bone marrow. To explore its therapeutic potential, we applied microglia replacement to a mouse model for Prosaposin deficiency, which is characterized by a progressive neurodegeneration phenotype. We found a reduction of cerebellar neurodegeneration and gliosis in treated brains, improvement of motor and balance impairment, and life span extension even with treatment started in young adulthood. This proof-of-concept study suggests that efficient microglia replacement may have therapeutic efficacy for a variety of neurological diseases.


Subject(s)
Brain Diseases , Hematopoietic Stem Cell Transplantation , Animals , Bone Marrow Cells , Brain , Central Nervous System , Mice , Microglia
4.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35054815

ABSTRACT

Neurodegenerative diseases are debilitating impairments that affect millions of people worldwide and are characterized by progressive degeneration of structure and function of the central or peripheral nervous system. Effective biomarkers for neurodegenerative diseases can be used to improve the diagnostic workup in the clinic as well as facilitate the development of effective disease-modifying therapies. Progranulin (PGRN) has been reported to be involved in various neurodegenerative disorders. Hence, in the current study we systematically compared the inflammation and accumulation of typical neurodegenerative disease markers in the brain tissue between PGRN knockout (PGRN KO) and wildtype (WT) mice. We found that PGRN deficiency led to significant neuron loss as well as activation of microglia and astrocytes in aged mice. Several characteristic neurodegenerative markers, including α-synuclein, TAR DNA-binding protein 43 (TDP-43), Tau, and ß-amyloid, were all accumulated in the brain of PGRN-deficient mice as compared to WT mice. Moreover, higher aggregation of lipofuscin was observed in the brain tissue of PGRN-deficient mice compared with WT mice. In addition, the autophagy was also defective in the brain of PGRN-deficient mice, indicated by the abnormal expression level of autophagy marker LC3-II. Collectively, comprehensive assays support the idea that PGRN plays an important role during the development of neurodegenerative disease, indicating that PGRN might be a useful biomarker for neurodegenerative diseases in clinical settings.


Subject(s)
Aging/pathology , Biomarkers/metabolism , Neurodegenerative Diseases/metabolism , Progranulins/deficiency , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Autophagy , Brain/pathology , Brain/ultrastructure , DNA-Binding Proteins/metabolism , Lipofuscin/metabolism , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Progranulins/metabolism , Protein Aggregates , alpha-Synuclein/metabolism , tau Proteins/metabolism
5.
Cells ; 10(9)2021 09 02.
Article in English | MEDLINE | ID: mdl-34571934

ABSTRACT

Substrate reduction therapy (SRT) in clinic adequately manages the visceral manifestations in Gaucher disease (GD) but has no direct effect on brain disease. To understand the molecular basis of SRT in GD treatment, we evaluated the efficacy and underlying mechanism of SRT in an immortalized neuronal cell line derived from a Gba knockout (Gba-/-) mouse model. Gba-/- neurons accumulated substrates, glucosylceramide, and glucosylsphingosine. Reduced cell proliferation was associated with altered lysosomes and autophagy, decreased mitochondrial function, and activation of the mTORC1 pathway. Treatment of the Gba-/- neurons with venglustat analogue GZ452, a central nervous system-accessible SRT, normalized glucosylceramide levels in these neurons and their isolated mitochondria. Enlarged lysosomes were reduced in the treated Gba-/- neurons, accompanied by decreased autophagic vacuoles. GZ452 treatment improved mitochondrial membrane potential and oxygen consumption rate. Furthermore, GZ452 diminished hyperactivity of selected proteins in the mTORC1 pathway and improved cell proliferation of Gba-/- neurons. These findings reinforce the detrimental effects of substrate accumulation on mitochondria, autophagy, and mTOR in neurons. A novel rescuing mechanism of SRT was revealed on the function of mitochondrial and autophagy-lysosomal pathways in GD. These results point to mitochondria and the mTORC1 complex as potential therapeutic targets for treatment of GD.


Subject(s)
Autophagy , Gaucher Disease/drug therapy , Glucosylceramidase/antagonists & inhibitors , Glycoside Hydrolase Inhibitors/pharmacology , Mitochondria/drug effects , Neurons/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Gaucher Disease/metabolism , Gaucher Disease/pathology , Glucosylceramidase/physiology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , TOR Serine-Threonine Kinases/genetics
6.
ACS Chem Neurosci ; 11(20): 3464-3473, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33035424

ABSTRACT

There remain no approved therapies for rare but devastating neuronopathic glyocosphingolipid storage diseases, such as Sandhoff, Tay-Sachs, and Gaucher disease type 3. We previously reported initial optimization of the scaffold of eliglustat, an approved therapy for the peripheral symptoms of Gaucher disease type 1, to afford 2, which effected modest reductions in brain glucosylceramide (GlcCer) in normal mice at 60 mg/kg. The relatively poor pharmacokinetic properties and high Pgp-mediated efflux of 2 prompted further optimization of the scaffold. With a general objective of reducing topological polar surface area, and guided by multiple metabolite identification studies, we were successful at identifying 17 (CCG-222628), which achieves remarkably greater brain exposure in mice than 2. After demonstrating an over 60-fold improvement in potency over 2 at reducing brain GlcCer in normal mice, we compared 17 with Sanofi clinical candidate venglustat (Genz-682452) in the CBE mouse model of Gaucher disease type 3. At doses of 10 mg/kg, 17 and venglustat effected comparable reductions in both brain GlcCer and glucosylsphingosine. Importantly, 17 achieved these equivalent pharmacodynamic effects at significantly lower brain exposure than venglustat.


Subject(s)
Gaucher Disease , Animals , Enzyme Inhibitors/pharmacology , Gaucher Disease/drug therapy , Glucosyltransferases , Mice , Pyrrolidines/pharmacology
7.
EBioMedicine ; 55: 102735, 2020 May.
Article in English | MEDLINE | ID: mdl-32279952

ABSTRACT

BACKGROUND: Enzyme replacement therapy (ERT) can positively affect the visceral manifestations of lysosomal storage diseases (LSDs). However, the exclusion of the intravenous ERT agents from the central nervous system (CNS) prevents direct therapeutic effects. METHODS: Using a neuronopathic Gaucher disease (nGD) mouse model, CNS-ERT was created using a systemic, non-invasive, and CNS-selective delivery system based on nanovesicles of saposin C (SapC) and dioleoylphosphatidylserine (DOPS) to deliver to CNS cells and tissues the corrective, functional acid ß-glucosidase (GCase). FINDINGS: Compared to free GCase, human GCase formulated with SapC-DOPS nanovesicles (SapC-DOPS-GCase) was more stable in serum, taken up into cells, mostly by a mannose receptor-independent pathway, and resulted in higher activity in GCase-deficient cells. In contrast to free GCase, SapC-DOPS-GCase nanovesicles penetrated through the blood-brain barrier into the CNS. The CNS targeting was mediated by surface phosphatidylserine (PS) of blood vessel and brain cells. Increased GCase activity and reduced GCase substrate levels were found in the CNS of SapC-DOPS-GCase-treated nGD mice, which showed profound improvement in brain inflammation and neurological phenotypes. INTERPRETATION: This first-in-class CNS-ERT approach provides considerable promise of therapeutic benefits for neurodegenerative diseases. FUNDING: This study was supported by the National Institutes of Health grants R21NS 095047 to XQ and YS, R01NS 086134 and UH2NS092981 in part to YS; Cincinnati Children's Hospital Medical Center Research Innovation/Pilot award to YS and XQ; Gardner Neuroscience Institute/Neurobiology Research Center Pilot award to XQ and YS, Hematology-Oncology Programmatic Support from University of Cincinnati and New Drug State Key Project grant 009ZX09102-205 to XQ.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Gaucher Disease/therapy , Glucosylceramidase/administration & dosage , Phosphatidylserines/chemistry , Saposins/chemistry , Animals , Biological Transport , Disease Models, Animal , Drug Stability , Enzyme Replacement Therapy/methods , Female , Gaucher Disease/enzymology , Gaucher Disease/genetics , Gaucher Disease/mortality , Glucosylceramidase/deficiency , Humans , Male , Mice , Mice, Transgenic , Nanostructures/administration & dosage , Nanostructures/chemistry , Permeability , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Survival Analysis , Treatment Outcome
8.
Hum Mol Genet ; 28(20): 3406-3421, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31373366

ABSTRACT

Gaucher disease (GD) is caused by GBA1 mutations leading to functional deficiency of acid-ß-glucosidase (GCase). No effective treatment is available for neuronopathic GD (nGD). A subclass of neural stem and precursor cells (NPCs) expresses VLA4 (integrin α4ß1, very late antigen-4) that facilitates NPC entry into the brain following intravenous (IV) infusion. Here, the therapeutic potential of IV VLA4+NPCs was assessed for nGD using wild-type mouse green fluorescent protein (GFP)-positive multipotent induced pluripotent stem cell (iPSC)-derived VLA4+NPCs. VLA4+NPCs successfully engrafted in the nGD (4L;C*) mouse brain. GFP-positive cells differentiated into neurons, astrocytes and oligodendrocytes in the brainstem, midbrain and thalamus of the transplanted mice and significantly improved sensorimotor function and prolonged life span compared to vehicle-treated 4L;C* mice. VLA4+NPC transplantation significantly decreased levels of CD68 and glial fibrillary acidic protein, as well as TNFα mRNA levels in the brain, indicating reduced neuroinflammation. Furthermore, decreased Fluoro-Jade C and NeuroSilver staining suggested inhibition of neurodegeneration. VLA4+NPC-engrafted 4L;C* midbrains showed 35% increased GCase activity, reduced substrate [glucosylceramide (GC, -34%) and glucosylsphingosine (GS, -11%)] levels and improved mitochondrial oxygen consumption rates in comparison to vehicle-4L;C* mice. VLA4+NPC engraftment in 4L;C* brain also led to enhanced expression of neurotrophic factors that have roles in neuronal survival and the promotion of neurogenesis. This study provides evidence that iPSC-derived NPC transplantation has efficacy in an nGD mouse model and provides proof of concept for autologous NPC therapy in nGD.


Subject(s)
Gaucher Disease/metabolism , Gaucher Disease/therapy , Glucosylceramidase/metabolism , Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Animals , Cell- and Tissue-Based Therapy/methods , Induced Pluripotent Stem Cells/cytology , Infusions, Intravenous , Integrin alpha4beta1/metabolism , Mice , Neural Stem Cells/cytology , beta-Glucosidase/metabolism
9.
Sci Rep ; 9(1): 5571, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944381

ABSTRACT

Gaucher disease is caused by mutations in GBA1 encoding acid ß-glucosidase (GCase). Saposin C enhances GCase activity and protects GCase from intracellular proteolysis. Structure simulations indicated that the mutant GCases, N370S (0 S), V394L (4L) and D409V(9V)/H(9H), had altered function. To investigate the in vivo function of Gba1 mutants, mouse models were generated by backcrossing the above homozygous mutant GCase mice into Saposin C deficient (C*) mice. Without saposin C, the mutant GCase activities in the resultant mouse tissues were reduced by ~50% compared with those in the presence of Saposin C. In contrast to 9H and 4L mice that have normal histology and life span, the 9H;C* and 4L;C* mice had shorter life spans. 9H;C* mice developed significant visceral glucosylceramide (GC) and glucosylsphingosine (GS) accumulation (GC¼GS) and storage macrophages, but lesser GC in the brain, compared to 4L;C* mice that presents with a severe neuronopathic phenotype and accumulated GC and GS primarily in the brain. Unlike 9V mice that developed normally for over a year, 9V;C* pups had a lethal skin defect as did 0S;C* mice resembled that of 0S mice. These variant Gaucher disease mouse models presented a mutation specific phenotype and underscored the in vivo role of Saposin C in the modulation of Gaucher disease.


Subject(s)
Gaucher Disease/genetics , Glucosylceramidase/genetics , Mutation/genetics , Saposins/deficiency , beta-Glucosidase/genetics , Animals , Brain/pathology , Disease Models, Animal , Glucosylceramides/genetics , Mice , Mice, Inbred C57BL , Phenotype
10.
Neurotoxicology ; 64: 256-266, 2018 01.
Article in English | MEDLINE | ID: mdl-28595912

ABSTRACT

Loss of function mutations in the P5-ATPase ATP13A2 are associated with Kufor-Rakeb Syndrome and Neuronal Ceroid Lipofuscinosis. While the function of ATP13A2 is unclear, in vitro studies suggest it is a lysosomal protein that interacts with the metals manganese (Mn) and zinc and the presynaptic protein alpha-synuclein. Loss of ATP13A2 function in mice causes sensorimotor deficits, enhanced autofluorescent storage material, and accumulation of alpha-synuclein. The present study sought to determine the effect of Mn administration on these same outcomes in ATP13A2-deficient mice. Wildtype and ATP13A2-deficient mice received saline or Mn at 5-9 or 12-19 months for 45days. Sensorimotor function was assessed starting at day 30. Autofluorescence was quantified in multiple brain regions and alpha-synuclein protein levels were determined in the ventral midbrain. Brain Mn, iron, zinc, and copper concentrations were measured in 5-9 month old mice. The results show Mn enhanced sensorimotor function, increased autofluorescence in the substantia nigra, and increased insoluble alpha-synuclein in the ventral midbrain in older ATP13A2-deficient mice. In addition, the Mn regimen used increased Mn concentration in the brain and levels were higher in Mn-treated mutants than controls. These results indicate loss of ATP13A2 function leads to increased sensitivity to Mn in vivo.


Subject(s)
Adenosine Triphosphatases/metabolism , Brain/drug effects , Brain/metabolism , Manganese/toxicity , Membrane Proteins/metabolism , Adenosine Triphosphatases/genetics , Animals , Behavior, Animal , Female , Male , Manganese/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Proton-Translocating ATPases , alpha-Synuclein/metabolism
11.
Nature ; 543(7643): 108-112, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28225753

ABSTRACT

Gaucher disease is caused by mutations in GBA1, which encodes the lysosomal enzyme glucocerebrosidase (GCase). GBA1 mutations drive extensive accumulation of glucosylceramide (GC) in multiple innate and adaptive immune cells in the spleen, liver, lung and bone marrow, often leading to chronic inflammation. The mechanisms that connect excess GC to tissue inflammation remain unknown. Here we show that activation of complement C5a and C5a receptor 1 (C5aR1) controls GC accumulation and the inflammatory response in experimental and clinical Gaucher disease. Marked local and systemic complement activation occurred in GCase-deficient mice or after pharmacological inhibition of GCase and was associated with GC storage, tissue inflammation and proinflammatory cytokine production. Whereas all GCase-inhibited mice died within 4-5 weeks, mice deficient in both GCase and C5aR1, and wild-type mice in which GCase and C5aR were pharmacologically inhibited, were protected from these adverse effects and consequently survived. In mice and humans, GCase deficiency was associated with strong formation of complement-activating GC-specific IgG autoantibodies, leading to complement activation and C5a generation. Subsequent C5aR1 activation controlled UDP-glucose ceramide glucosyltransferase production, thereby tipping the balance between GC formation and degradation. Thus, extensive GC storage induces complement-activating IgG autoantibodies that drive a pathway of C5a generation and C5aR1 activation that fuels a cycle of cellular GC accumulation, innate and adaptive immune cell recruitment and activation in Gaucher disease. As enzyme replacement and substrate reduction therapies are expensive and still associated with inflammation, increased risk of cancer and Parkinson disease, targeting C5aR1 may serve as a treatment option for patients with Gaucher disease and, possibly, other lysosomal storage diseases.


Subject(s)
Complement System Proteins/immunology , Gaucher Disease/immunology , Gaucher Disease/pathology , Glucosylceramides/immunology , Glucosylceramides/metabolism , Inflammation/immunology , Inflammation/pathology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Autoantibodies/immunology , Complement Activation , Complement C5a/biosynthesis , Complement C5a/immunology , Complement System Proteins/biosynthesis , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Female , Gaucher Disease/metabolism , Gaucher Disease/prevention & control , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/deficiency , Glucosylceramidase/genetics , Glucosyltransferases/biosynthesis , Glucosyltransferases/metabolism , Humans , Immunoglobulin G/immunology , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Hum Mol Genet ; 25(23): 5126-5141, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27655403

ABSTRACT

Neuronopathic Gaucher disease (nGD) manifests as severe neurological symptoms in patients with no effective treatment available. Ryanodine receptors (Ryrs) are a family of calcium release channels on intracellular stores. The goal of this study is to determine if Ryrs are potential targets for nGD treatment. A nGD cell model (CBE-N2a) was created by inhibiting acid ß-glucosidase (GCase) in N2a cells with conduritol B epoxide (CBE). Enhanced cytosolic calcium in CBE-N2a cells was blocked by either ryanodine or dantrolene, antagonists of Ryrs and by Genz-161, a glucosylceramide synthase inhibitor, suggesting substrate-mediated ER-calcium efflux occurs through ryanodine receptors. In the brain of a nGD (4L;C*) mouse model, expression of Ryrs was normal at 13 days of age, but significantly decreased below the wild type level in end-stage 4L;C* brains at 40 days. Treatment with dantrolene in 4L;C* mice starting at postnatal day 5 delayed neurological pathology and prolonged survival. Compared to untreated 4L;C* mice, dantrolene treatment significantly improved gait, reduced LC3-II levels, improved mitochondrial ATP production and reduced inflammation in the brain. Dantrolene treatment partially normalized Ryr expression and its potential regulators, CAMK IV and calmodulin. Furthermore, dantrolene treatment increased residual mutant GCase activity in 4L;C* brains. These data demonstrate that modulating Ryrs has neuroprotective effects in nGD through mechanisms that protect the mitochondria, autophagy, Ryr expression and enhance GCase activity. This study suggests that calcium signalling stabilization, e.g. with dantrolene, could be a potential disease modifying therapy for nGD.


Subject(s)
Dantrolene/administration & dosage , Gaucher Disease/drug therapy , Mitochondria/drug effects , Ryanodine Receptor Calcium Release Channel/genetics , Animals , Calcium Signaling/genetics , Disease Models, Animal , Gaucher Disease/genetics , Gaucher Disease/physiopathology , Humans , Mice , Mitochondria/genetics , Mitochondria/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/administration & dosage , Ryanodine Receptor Calcium Release Channel/metabolism
13.
PLoS One ; 11(9): e0162367, 2016.
Article in English | MEDLINE | ID: mdl-27598339

ABSTRACT

To study the neuronal deficits in neuronopathic Gaucher Disease (nGD), the chronological behavioral profiles and the age of onset of brain abnormalities were characterized in a chronic nGD mouse model (9V/null). Progressive accumulation of glucosylceramide (GC) and glucosylsphingosine (GS) in the brain of 9V/null mice were observed at as early as 6 and 3 months of age for GC and GS, respectively. Abnormal accumulation of α-synuclein was present in the 9V/null brain as detected by immunofluorescence and Western blot analysis. In a repeated open-field test, the 9V/null mice (9 months and older) displayed significantly less environmental habituation and spent more time exploring the open-field than age-matched WT group, indicating the onset of short-term spatial memory deficits. In the marble burying test, the 9V/null group had a shorter latency to initiate burying activity at 3 months of age, whereas the latency increased significantly at ≥12 months of age; 9V/null females buried significantly more marbles to completion than the WT group, suggesting an abnormal response to the instinctive behavior and an abnormal activity in non-associative anxiety-like behavior. In the conditional fear test, only the 9V/null males exhibited a significant decrease in response to contextual fear, but both genders showed less response to auditory-cued fear compared to age- and gender-matched WT at 12 months of age. These results indicate hippocampus-related emotional memory defects. Abnormal gait emerged in 9V/null mice with wider front-paw and hind-paw widths, as well as longer stride in a gender-dependent manner with different ages of onset. Significantly higher liver- and spleen-to-body weight ratios were detected in 9V/null mice with different ages of onsets. These data provide temporal evaluation of neurobehavioral dysfunctions and brain pathology in 9V/null mice that can be used for experimental designs to evaluate novel therapies for nGD.


Subject(s)
Aging/pathology , Gaucher Disease/physiopathology , Glucosylceramidase/genetics , Hippocampus/physiopathology , Memory Disorders/physiopathology , Acoustic Stimulation , Aging/genetics , Animals , Behavior, Animal , Conditioning, Psychological/physiology , Disease Models, Animal , Disease Progression , Exploratory Behavior/physiology , Fear/physiology , Female , Gait/physiology , Gaucher Disease/metabolism , Gaucher Disease/pathology , Glucosylceramidase/deficiency , Glucosylceramides/biosynthesis , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Psychosine/analogs & derivatives , Psychosine/biosynthesis , Sex Factors , Spatial Memory/physiology , alpha-Synuclein/biosynthesis
14.
Hum Mol Genet ; 24(24): 7031-48, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26420838

ABSTRACT

Defective lysosomal acid ß-glucosidase (GCase) in Gaucher disease causes accumulation of glucosylceramide (GC) and glucosylsphingosine (GS) that distress cellular functions. To study novel pathological mechanisms in neuronopathic Gaucher disease (nGD), a mouse model (4L;C*), an analogue to subacute human nGD, was investigated for global profiles of differentially expressed brain mRNAs (DEGs) and miRNAs (DEmiRs). 4L;C* mice displayed accumulation of GC and GS, activated microglial cells, reduced number of neurons and aberrant mitochondrial function in the brain followed by deterioration in motor function. DEGs and DEmiRs were characterized from sequencing of mRNA and miRNA from cerebral cortex, brain stem, midbrain and cerebellum of 4L;C* mice. Gene ontology enrichment and pathway analysis showed preferential mitochondrial dysfunction in midbrain and uniform inflammatory response and identified novel pathways, axonal guidance signaling, synaptic transmission, eIF2 and mammalian target of rapamycin (mTOR) signaling potentially involved in nGD. Similar analyses were performed with mice treated with isofagomine (IFG), a pharmacologic chaperone for GCase. IFG treatment did not alter the GS and GC accumulation significantly but attenuated the progression of the disease and altered numerous DEmiRs and target DEGs to their respective normal levels in inflammation, mitochondrial function and axonal guidance pathways, suggesting its regulation on miRNA and the associated mRNA that underlie the neurodegeneration in nGD. These analyses demonstrate that the neurodegenerative phenotype in 4L;C* mice was associated with dysregulation of brain mRNAs and miRNAs in axonal guidance, synaptic plasticity, mitochondria function, eIF2 and mTOR signaling and inflammation and provides new insights for the nGD pathological mechanism.


Subject(s)
Brain/metabolism , Gaucher Disease/genetics , Imino Pyranoses/therapeutic use , MicroRNAs/metabolism , Molecular Chaperones/therapeutic use , RNA, Messenger/metabolism , Animals , Axons/metabolism , Brain/pathology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Eukaryotic Initiation Factor-2/metabolism , Gaucher Disease/drug therapy , Gaucher Disease/metabolism , Gaucher Disease/pathology , Gene Expression Profiling , Glucosylceramides/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Molecular Chaperones/metabolism , Neuroglia/pathology , Neurons/pathology , Phenotype , Psychosine/analogs & derivatives , Psychosine/metabolism , Signal Transduction , Synaptic Transmission , TOR Serine-Threonine Kinases/metabolism
15.
PLoS One ; 10(3): e0118771, 2015.
Article in English | MEDLINE | ID: mdl-25822147

ABSTRACT

Gaucher disease (GD) is caused by insufficient activity of acid ß-glucosidase (GCase) resulting from mutations in GBA1. To understand the pathogenesis of the neuronopathic GD, induced pluripotent stem cells (iPSCs) were generated from fibroblasts isolated from three GD type 2 (GD2) and 2 unaffected (normal and GD carrier) individuals. The iPSCs were converted to neural precursor cells (NPCs) which were further differentiated into neurons. Parental GD2 fibroblasts as well as iPSCs, NPCs, and neurons had similar degrees of GCase deficiency. Lipid analyses showed increases of glucosylsphingosine and glucosylceramide in the GD2 cells. In addition, GD2 neurons showed increased α-synuclein protein compared to control neurons. Whole cell patch-clamping of the GD2 and control iPSCs-derived neurons demonstrated excitation characteristics of neurons, but intriguingly, those from GD2 exhibited consistently less negative resting membrane potentials with various degree of reduction in action potential amplitudes, sodium and potassium currents. Culture of control neurons in the presence of the GCase inhibitor (conduritol B epoxide) recapitulated these findings, providing a functional link between decreased GCase activity in GD and abnormal neuronal electrophysiological properties. To our knowledge, this study is first to report abnormal electrophysiological properties in GD2 iPSC-derived neurons that may underlie the neuropathic phenotype in Gaucher disease.


Subject(s)
Fibroblasts/cytology , Gaucher Disease/pathology , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/cytology , Cells, Cultured , Fibroblasts/metabolism , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Glucosylceramides/metabolism , Humans , Membrane Potentials , Neural Stem Cells/metabolism , Neurogenesis , Neurons/metabolism , Neurons/physiology , Psychosine/analogs & derivatives , Psychosine/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
16.
J Biol Chem ; 289(43): 30063-74, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25202012

ABSTRACT

The acid ß-glucosidase (glucocerbrosidase (GCase)) binding sequence to LIMP-2 (lysosomal integral membrane protein 2), the receptor for intracellular GCase trafficking to the lysosome, has been identified. Heterologous expression of deletion constructs, the available GCase crystal structures, and binding and co-localization of identified peptides or mutant GCases were used to identify and characterize a highly conserved 11-amino acid sequence, DSPIIVDITKD, within human GCase. The binding to LIMP-2 is not dependent upon a single amino acid, but the interactions of GCase with LIMP-2 are heavily influenced by Asp(399) and the di-isoleucines, Ile(402) and Ile(403). A single alanine substitution at any of these decreases GCase binding to LIMP-2 and alters its pH-dependent binding as well as diminishing the trafficking of GCase to the lysosome and significantly increasing GCase secretion. Enterovirus 71 also binds to LIMP-2 (also known as SCARB2) on the external surface of the plasma membrane. However, the LIMP-2/SCARB2 binding sequences for enterovirus 71 and GCase are not similar, indicating that LIMP-2/SCARB2 may have multiple or overlapping binding sites with differing specificities. These findings have therapeutic implications for the production of GCase and the distribution of this enzyme that is delivered to various organs.


Subject(s)
Gaucher Disease/enzymology , Glucosylceramidase/chemistry , Glucosylceramidase/metabolism , Lysosomal Membrane Proteins/metabolism , Neurodegenerative Diseases/enzymology , Receptors, Scavenger/metabolism , Alanine/genetics , Amines/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding, Competitive , Gaucher Disease/pathology , Glucosylceramidase/deficiency , Green Fluorescent Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Immunoprecipitation , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutation/genetics , Neurodegenerative Diseases/pathology , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Transfection
17.
Mol Genet Metab ; 112(3): 229-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24837159

ABSTRACT

Lysosomal acid lipase (LAL) is an essential enzyme that hydrolyzes triglycerides (TG) and cholesteryl esters (CE) in lysosomes. Mutations of the LIPA gene lead to Wolman disease (WD) and cholesterol ester storage disease (CESD). The disease hallmarks include hepatosplenomegaly and extensive storage of CE and/or TG. The effects of intravenous investigational enzyme therapy (ET) on survival and efficacy were evaluated in Lipa knock out, lal-/- mice with advanced disease using recombinant human LAL (rhLAL). Comparative ET was conducted with lower doses (weekly, 0.8 and 3.2mg/kg) beginning at 16 weeks (study 1), and with higher dose (10mg/kg) in early (8-weeks), middle (16-weeks) and late (24-weeks) disease stages (study 2). In study 1, rhLAL extended the life span of lal-/- mice in a dose dependent manner by 52 (0.8 mg/kg) or 94 (3.2mg/kg) days. This was accompanied by partial correction of cholesterol and TG levels in spleen and liver. In study 2, the high dose resulted in a significant improvement in organ size (liver, spleen and small intestine) and tissue histology as well as significant decreases in cholesterol and TG in all three groups. In the treated livers and spleens the cholesterol and TG levels were reduced to below treatment initiation levels indicating a reversal of disease manifestations, even in advanced disease. ET diminished liver fibrosis and macrophage proliferation. These results show that LAL deficiency can be improved biochemically and histopathologically by various dosages of ET, even in advanced disease.


Subject(s)
Wolman Disease/metabolism , Wolman Disease/pathology , Animals , Body Weight , Disease Models, Animal , Enzyme Replacement Therapy , Female , Lipid Metabolism , Lipids/blood , Male , Mice , Mice, Knockout , Organ Size , Phenotype , Sterol Esterase/administration & dosage , Treatment Outcome , Wolman Disease/drug therapy , Wolman Disease/genetics , Wolman Disease/mortality , Wolman Disease
18.
Hum Mol Genet ; 23(15): 3943-57, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24599400

ABSTRACT

Gaucher disease, a prevalent lysosomal storage disease (LSD), is caused by insufficient activity of acid ß-glucosidase (GCase) and the resultant glucosylceramide (GC)/glucosylsphingosine (GS) accumulation in visceral organs (Type 1) and the central nervous system (Types 2 and 3). Recent clinical and genetic studies implicate a pathogenic link between Gaucher and neurodegenerative diseases. The aggregation and inclusion bodies of α-synuclein with ubiquitin are present in the brains of Gaucher disease patients and mouse models. Indirect evidence of ß-amyloid pathology promoting α-synuclein fibrillation supports these pathogenic proteins as a common feature in neurodegenerative diseases. Here, multiple proteins are implicated in the pathogenesis of chronic neuronopathic Gaucher disease (nGD). Immunohistochemical and biochemical analyses showed significant amounts of ß-amyloid and amyloid precursor protein (APP) aggregates in the cortex, hippocampus, stratum and substantia nigra of the nGD mice. APP aggregates were in neuronal cells and colocalized with α-synuclein signals. A majority of APP co-localized with the mitochondrial markers TOM40 and Cox IV; a small portion co-localized with the autophagy proteins, P62/LC3, and the lysosomal marker, LAMP1. In cultured wild-type brain cortical neural cells, the GCase-irreversible inhibitor, conduritol B epoxide (CBE), reproduced the APP/α-synuclein aggregation and the accumulation of GC/GS. Ultrastructural studies showed numerous larger-sized and electron-dense mitochondria in nGD cerebral cortical neural cells. Significant reductions of mitochondrial adenosine triphosphate production and oxygen consumption (28-40%) were detected in nGD brains and in CBE-treated neural cells. These studies implicate defective GCase function and GC/GS accumulation as risk factors for mitochondrial dysfunction and the multi-proteinopathies (α-synuclein-, APP- and Aß-aggregates) in nGD.


Subject(s)
Gaucher Disease/genetics , Gene Expression Regulation , Mitochondria/metabolism , Neurons/metabolism , beta-Glucosidase/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gaucher Disease/metabolism , Gaucher Disease/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Inositol/analogs & derivatives , Inositol/pharmacology , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/pathology , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Aggregation, Pathological , Substantia Nigra/metabolism , Substantia Nigra/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , beta-Glucosidase/antagonists & inhibitors , beta-Glucosidase/metabolism
19.
PLoS One ; 9(12): e116023, 2014.
Article in English | MEDLINE | ID: mdl-25551612

ABSTRACT

Gaucher disease is a lysosomal storage disease caused by defective activity of acid ß-glucosidase (GCase), which leads to the accumulation of its major substrates, glucosylceramide (GlcCer) and glucosylsphingosine (GlcSph) in many cells. To modulate cellular substrate concentration in viable mouse models of Gaucher disease (Gba1 mutants), a novel mouse model was created with enhanced glycosphingolipid biosynthesis. This was accomplished by cross-breeding Gba1 mutant mice with mice expressing a transgene (GCStg) containing the mouse glucosylceramide synthase (GCS, Ugcg) cDNA driven by the ROSA promoter, yielding GCStg/Gba1 mice. The GCStg rescued Ugcg null mice from embryonic lethality. GCStg/Gba1 mice showed 2-3 fold increases in tissue GCS activity as well as accelerated GlcCer accumulation and the appearance of lipid-laden CD68 positive macrophages in visceral organs. Although GlcCer/GlcSph concentrations were elevated in the brain, there was no neurodegenerative phenotype up to 1 yr of age conceivably due to the greater residual GCase hydrolytic activity in the brains than in the visceral tissues of 9V/null mice. These studies provide 'proof of principle' for threshold substrate flux that modifies phenotypic development in Gaucher disease and other lysosomal storage diseases.


Subject(s)
Gaucher Disease/enzymology , Glucosylceramidase/genetics , Glucosylceramides/biosynthesis , Glucosyltransferases/genetics , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Disease Models, Animal , Gaucher Disease/pathology , Glucosylceramidase/biosynthesis , Glucosylceramides/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics
20.
PLoS One ; 8(3): e57560, 2013.
Article in English | MEDLINE | ID: mdl-23520473

ABSTRACT

Gaucher disease results from GBA1 mutations that lead to defective acid ß-glucosidase (GCase) mediated cleavage of glucosylceramide (GC) and glucosylsphingosine as well as heterogeneous manifestations in the viscera and CNS. The mutation, tissue, and age-dependent accumulations of different GC species were characterized in mice with Gba1 missense mutations alone or in combination with isolated saposin C deficiency (C*). Gba1 heteroallelism for D409V and null alleles (9V/null) led to GC excesses primarily in the visceral tissues with preferential accumulations of lung GC24∶0, but not in liver, spleen, or brain. Age-dependent increases of different GC species were observed. The combined saposin C deficiency (C*) with V394L homozygosity (4L;C*) showed major GC18:0 degradation defects in the brain, whereas the analogous mice with D409H homozygosity and C* (9H;C*) led to all GC species accumulating in visceral tissues. Glucosylsphingosine was poorly degraded in brain by V394L and D409H GCases and in visceral tissues by D409V GCase. The neonatal lethal N370S/N370S genotype had insignificant substrate accumulations in any tissue. These results demonstrate age, organ, and mutation-specific quantitative differences in GC species and glucosylsphingosine accumulations that can have influence in the tissue/regional expression of Gaucher disease phenotypes.


Subject(s)
Aging/metabolism , Brain/enzymology , Gaucher Disease/enzymology , Glucosylceramidase/metabolism , Glucosylceramides/metabolism , Mutation, Missense , Psychosine/analogs & derivatives , Aging/genetics , Aging/pathology , Amino Acid Substitution , Animals , Brain/pathology , Disease Models, Animal , Gaucher Disease/genetics , Gaucher Disease/pathology , Glucosylceramidase/genetics , Glucosylceramides/genetics , Humans , Mice , Mice, Mutant Strains , Organ Specificity/genetics , Psychosine/genetics , Psychosine/metabolism , Saposins/genetics , Saposins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...