Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
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.
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.
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
7.
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
8.
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
9.
Arterioscler Thromb Vasc Biol ; 38(5): 1191-1201, 2018 05.
Article in English | MEDLINE | ID: mdl-29599133

ABSTRACT

OBJECTIVE: To explore the role of LAL (lysosomal acid lipase) in macrophage cholesterol efflux and whole-body reverse cholesterol transport. APPROACH AND RESULTS: Immortalized peritoneal macrophages from lal-/- mice showed reduced expression of ABCA1 (ATP-binding cassette transporter A1) and ABCG1 (ATP-binding cassette transporter G1), reduced production of the regulatory oxysterol 27-hydroxycholesterol, and impaired suppression of cholesterol synthesis on exposure to acetylated low-density lipoprotein when compared with lal+/+ macrophages. LAL-deficient mice also showed reduced hepatic ABCG5 (ATP-binding cassette transporter G5) and ABCG8 (ATP-binding cassette transporter G8) expression compared with lal+/+ mice. LAL-deficient macrophages loaded with [3H]-cholesteryl oleate-labeled acetylated low-density lipoprotein showed impaired efflux of released [3H]-cholesterol to apoA-I (apolipoprotein A-I), with normalization of [3H]-cholesteryl ester levels and partial correction of ABCA1 expression and cholesterol efflux to apoA-I when treated with exogenous rhLAL (recombinant human LAL protein). LAL-deficient mice injected intraperitoneally with lal-/- macrophages cholesterol loaded and labeled in the same way exhibited only 1.55±0.35% total injected [3H]-cholesterol counts appearing in the feces for 48 h (n=30), compared with 5.38±0.92% in lal+/+ mice injected with labeled lal+/+ macrophages (n=27), P<0.001. To mimic the therapeutic condition of delivery of supplemental LAL in vivo, injection of labeled lal-/- macrophages into lal+/+ mice resulted in a significant increase in reverse cholesterol transport (2.60±0.46% of 3H-cholesterol counts in feces at 48 hours [n=19]; P<0.001 when compared with injection into lal-/- mice). CONCLUSIONS: These results indicate a critical role for LAL in promoting both macrophage and whole-body reverse cholesterol transport and the ability of supplemental LAL to be taken up and correct reverse cholesterol transport in vivo.


Subject(s)
Cholesterol/metabolism , Macrophages, Peritoneal/enzymology , Sterol Esterase/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Animals , Apolipoprotein A-I/metabolism , Biological Transport , Cell Line , Cholesterol/blood , Feces/chemistry , Lipoproteins/genetics , Lipoproteins/metabolism , Liver/metabolism , Mice, 129 Strain , Mice, Knockout , Sterol Esterase/deficiency , Sterol Esterase/genetics
10.
EBioMedicine ; 28: 251-260, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29396296

ABSTRACT

We recently reported that progranulin (PGRN) is a novel regulator of glucocerebrosidase and its deficiency associates with Gaucher Diseases (GD) (Jian et al., 2016a; Jian et al., 2018). To isolate the relevant downstream molecules, we performed a whole genome microarray and mass spectrometry analysis, which led to the isolation of Chitinase-3-like-1 (CHI3L1) as one of the up-regulated genes in PGRN null mice. Elevated levels of CHI3L1 were confirmed by immunoblotting and immunohistochemistry. In contrast, treatment with recombinant Pcgin, a derivative of PGRN, as well as imigluerase, significantly reduced the expressions of CHI3L1 in both PGRN null GD model and the fibroblasts from GD patients. Serum levels of CHIT1, a clinical biomarker for GD, were significantly higher in GD patients than healthy controls (51.16±2.824ng/ml vs 35.07±2.099ng/ml, p<0.001). Similar to CHIT1, serum CHI3L1 was also significantly increased in GD patients compared with healthy controls (1736±152.1pg/ml vs 684.7±68.20pg/ml, p<0.001). Whereas the PGRN level is significantly reduced in GD patients as compared to the healthy control (91.56±3.986ng/ml vs 150.6±4.501, p<0.001). Collectively, these results indicate that CHI3L1 may be a previously unrecognized biomarker for diagnosing GD and for evaluating the therapeutic effects of new GD drug(s).


Subject(s)
Chitinase-3-Like Protein 1/metabolism , Gaucher Disease/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Chitinase-3-Like Protein 1/blood , Chitinases/blood , Disease Models, Animal , Gaucher Disease/blood , Granulins , Humans , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Progranulins , Up-Regulation
11.
Semin Immunol ; 37: 30-42, 2018 06.
Article in English | MEDLINE | ID: mdl-29478824

ABSTRACT

The complement system is well appreciated for its role as an important effector of innate immunity that is activated by the classical, lectin or alternative pathway. C5a is one important mediator of the system that is generated in response to canonical and non-canonical C5 cleavage by circulating or cell-derived proteases. In addition to its function as a chemoattractant for neutrophils and other myeloid effectors, C5a and its sister molecule C3a have concerted roles in cell homeostasis and surveillance. Through activation of their cognate G protein coupled receptors, C3a and C5a regulate multiple intracellular pathways within the mitochondria and the lysosomal compartments that harbor multiple enzymes critical for protein, carbohydrate and lipid metabolism. Genetic mutations of such lysosomal enzymes or their receptors can result in the compartmental accumulation of specific classes of substrates in this organelle summarized as lysosomal storage diseases (LSD). A frequent LSD is Gaucher disease (GD), caused by autosomal recessively inherited mutations in GBA1, resulting in functional defects of the encoded enzyme, acid ß-glucosidase (glucocerebrosidase, GCase). Such mutations promote excessive accumulation of ß-glucosylceramide (GC or GL1) in innate and adaptive immune cells frequently associated with chronic inflammation. Recently, we uncovered an unexpected link between the C5a and C5a receptor 1 (C5aR1) axis and the accumulation of GL1 in experimental and clinical GD. Here, we will review the pathways of complement activation in GD, its role as a mediator of the inflammatory response, and its impact on glucosphingolipid metabolism. Further, we will discuss the potential role of the C5a/C5aR1 axis in GL1-specific autoantibody formation and as a novel therapeutic target in GD.


Subject(s)
Complement C5a/metabolism , Gaucher Disease/immunology , Glucosylceramidase/genetics , Inflammation/immunology , Lysosomal Storage Diseases/immunology , Animals , Autoantibodies/metabolism , Gaucher Disease/genetics , Glucosylceramides/metabolism , Humans , Receptor, Anaphylatoxin C5a/metabolism
12.
Am J Pathol ; 188(2): 525-538, 2018 02.
Article in English | MEDLINE | ID: mdl-29154769

ABSTRACT

Lipid accumulation is a key characteristic of advancing atherosclerotic lesions. Herein, we analyzed the ultrastructure of the accumulated lipids in endarterectomized human carotid atherosclerotic plaques using three-dimensional (3D) electron microscopy, a method never used in this context before. 3D electron microscopy revealed intracellular lipid droplets and extracellular lipoprotein particles. Most of the particles were aggregated, and some connected to needle-shaped or sheet-like cholesterol crystals. Proteomic analysis of isolated extracellular lipoprotein particles revealed that apolipoprotein B is their main protein component, indicating their origin from low-density lipoprotein, intermediate-density lipoprotein, very-low-density lipoprotein, lipoprotein (a), or chylomicron remnants. The particles also contained small exchangeable apolipoproteins, complement components, and immunoglobulins. Lipidomic analysis revealed differences between plasma lipoproteins and the particles, thereby indicating involvement of lipolytic enzymes in their generation. Incubation of human monocyte-derived macrophages with the isolated extracellular lipoprotein particles or with plasma lipoproteins that had been lipolytically modified in vitro induced intracellular lipid accumulation and triggered inflammasome activation in them. Taken together, extracellular lipids accumulate in human carotid plaques as distinct 3D structures that include aggregated and fused lipoprotein particles and cholesterol crystals. The particles originate from plasma lipoproteins, show signs of lipolytic modifications, and associate with cholesterol crystals. By inducing intracellular cholesterol accumulation (ie, foam cell formation) and inflammasome activation, the extracellular lipoprotein particles may actively enhance atherogenesis.


Subject(s)
Carotid Arteries/metabolism , Carotid Artery Diseases/metabolism , Inflammation Mediators/metabolism , Lipid Metabolism/physiology , Carotid Arteries/ultrastructure , Carotid Artery Diseases/pathology , Carotid Artery Diseases/surgery , Cells, Cultured , Cholesterol/metabolism , Endarterectomy, Carotid , Extracellular Space/metabolism , Humans , Imaging, Three-Dimensional/methods , Inflammasomes/metabolism , Lipolysis/physiology , Lipoproteins/metabolism , Macrophages/metabolism , Microscopy, Electron, Transmission/methods
13.
Semin Pediatr Neurol ; 24(3): 207-213, 2017 08.
Article in English | MEDLINE | ID: mdl-29103428

ABSTRACT

Neuroinflammation is an intrinsic component of the neurodegeneration of inborn errors of neurometabolic diseases. Diseases resulting in lysosomal, peroxisomal, and autophagocytic disruption lead to neuroinflammation by different mechanisms relating to accumulated substrates and/or downstream deficiencies that cause presymptomatic microglial activation, axonal instabilities and/or direct hyperactivation of intrinsic inflammatory mechanisms. Only in selected diseases is the blood-brain barrier (BBB) breached, thereby permitting peripheral adaptive immune mechanisms to amplify intrinsic immune reactions in the central nervous system. These result in evoking several different programmed cell death pathways, including apoptosis, necroptosis, and pyroptosis, with the subsequent neuronal death of specific types and in selected regions of the brain or spinal cord. In addition to correction of the primary genetic or metabolic defects, successful therapeutic interventions require greater molecular understanding of the specific neuroinflammatory components of neurometabolic diseases to permit identification of significant targets for intervention.


Subject(s)
Central Nervous System/immunology , Central Nervous System/pathology , Inflammation/etiology , Metabolic Diseases/complications , Metabolic Diseases/pathology , Humans
14.
SLAS Discov ; 22(10): 1218-1228, 2017 12.
Article in English | MEDLINE | ID: mdl-28714776

ABSTRACT

To better understand regional brain glycosphingolipid (GSL) accumulation in Gaucher disease (GD) and its relationship to neuropathology, a feasibility study using mass spectrometry and immunohistochemistry was conducted using brains derived from a GD mouse model (4L/PS/NA) homozygous for a mutant GCase (V394L [4L]) and expressing a prosaposin hypomorphic (PS-NA) transgene. Whole brains from GD and control animals were collected using one hemisphere for MALDI FTICR IMS analysis and the other for quantitation by LC-ESI-MS/MS. MALDI IMS detected several HexCers across the brains. Comparison with the brain hematoxylin and eosin (H&E) revealed differential signal distributions in the midbrain, brain stem, and CB of the GD brain versus the control. Quantitation of serial brain sections with LC-ESI-MS/MS supported the imaging results, finding the overall HexCer levels in the 4L/PS-NA brains to be four times higher than the control. LC-ESI-MS/MS also confirmed that the elevated hexosyl isomers were glucosylceramides rather than galactosylceramides. MALDI imaging also detected differential analyte distributions of lactosylceramide species and gangliosides in the 4L/PS-NA brain, which was validated by LC-ESI-MS/MS. Immunohistochemistry revealed regional inflammation, altered autophagy, and defective protein degradation correlating with regions of GSL accumulation, suggesting that specific GSLs may have distinct neuropathological effects.


Subject(s)
Brain/metabolism , Gaucher Disease/metabolism , Glycosphingolipids/metabolism , Imaging, Three-Dimensional , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Brain/pathology , Chromatography, Liquid , Mice, Inbred C57BL , Organ Specificity , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry
15.
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
16.
Mol Genet Metab ; 120(1-2): 47-56, 2017.
Article in English | MEDLINE | ID: mdl-28040394

ABSTRACT

In Gaucher disease (GD), deficiency of lysosomal acid ß-glucosidase results in a broad phenotypic spectrum that is classified into three types based on the absence (type 1 [GD1]) or presence and severity of primary central nervous system involvement (type 2 [GD2], the fulminant neuronopathic form, and type 3 [GD3], the milder chronic neuronopathic form). Enzyme replacement therapy (ERT) with imiglucerase ameliorates and prevents hematological and visceral manifestations in GD1, but data in GD3 are limited to small, single-center series. The effects of imiglucerase ERT on hematological, visceral and growth outcomes (note: ERT is not expected to directly impact neurologic outcomes) were evaluated during the first 5years of treatment in 253 children and adolescents (<18years of age) with GD3 enrolled in the International Collaborative Gaucher Group (ICGG) Gaucher Registry. The vast majority of GBA mutations in this diverse global population consisted of only 2 mutations: L444P (77%) and D409H (7%). At baseline, GD3 patients exhibited early onset of severe hematological and visceral disease and growth failure. During the first year of imiglucerase treatment, hemoglobin levels and platelet counts increased and liver and spleen volumes decreased, leading to marked decreases in the number of patients with moderate or severe anemia, thrombocytopenia, and hepatosplenomegaly. These improvements were maintained through Year 5. There was also acceleration in linear growth as evidenced by increasing height Z-scores. Despite devastating disease at baseline, the probability of surviving for at least 5years after starting imiglucerase was 92%. In this large, multinational cohort of pediatric GD3 patients, imiglucerase ERT provided a life-saving and life-prolonging benefit for patients with GD3, suggesting that, with proper treatment, many such severely affected patients can lead productive lives and contribute to society.


Subject(s)
Gaucher Disease/drug therapy , Glucosylceramidase/genetics , Mutation , Adolescent , Child , Child, Preschool , Enzyme Replacement Therapy , Female , Gaucher Disease/classification , Gaucher Disease/genetics , Glucosylceramidase/therapeutic use , Humans , Male , Registries , Survival Analysis , Treatment Outcome
17.
Mol Genet Metab ; 120(1-2): 1-7, 2017.
Article in English | MEDLINE | ID: mdl-27866832

ABSTRACT

To celebrate the research visions and accomplishments of the late Roscoe O. Brady (1923-2016), remembrance commentaries were requested from several of his postdoctoral research fellows and colleagues. These commentaries not only reflect on the accomplishments of Dr. Brady, but they also share some of the backstories and experiences working in the Brady laboratory. They provide insights and perspectives on Brady's research activities, and especially on his efforts to develop an effective treatment for patients with Type 1 Gaucher disease. These remembrances illuminate Brady's efforts to implement the latest scientific advances with an outstanding team of young co-investigators to develop and demonstrate the safety and effectiveness of the first enzyme replacement therapy for a lysosomal storage disease. Brady's pursuit and persistence in accomplishing his research objectives provide insights into this remarkably successful physician scientist who paved the way for the development of treatments for patients with other lysosomal storage diseases.


Subject(s)
Enzyme Replacement Therapy/history , Lysosomal Storage Diseases/drug therapy , Enzyme Replacement Therapy/methods , Gaucher Disease/drug therapy , History, 20th Century , History, 21st Century , Humans , Research Personnel
18.
Mol Genet Metab ; 120(1-2): 38-46, 2017.
Article in English | MEDLINE | ID: mdl-27923545

ABSTRACT

The advent of the first effective specific therapy for a lysosomal storage disease (LSDs), Gaucher disease type 1, by Roscoe O. Brady was foundational for development of additional treatments for this group of rare diseases. The past 26years, since the approval of enzyme therapy for Gaucher disease type 1, have witnessed a burgeoning understanding of LSDs at genetic, molecular, biochemical, cell biologic, and clinical levels. Simultaneously, this expansion of knowledge has exposed our incomplete understanding of the individual pathophysiologies of LSDs as well as difficult challenges for improvement in therapy and therapeutic outcomes for afflicted individuals. Here, 10 such challenges/problems representing major impediments, which need to be overcome, to move forward toward the goals of more effective and complete therapies for these devastating diseases.


Subject(s)
Enzyme Replacement Therapy/methods , Lysosomal Storage Diseases/drug therapy , Disease Progression , Gaucher Disease/drug therapy , Gaucher Disease/genetics , Humans , Lysosomal Storage Diseases/genetics , Mutation , Treatment Outcome
19.
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
20.
SELECTION OF CITATIONS
SEARCH DETAIL
...