Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(1): e2210442120, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36574647

RESUMO

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.


Assuntos
Doença de Gaucher , Doença de Parkinson , Progranulinas , Animais , Camundongos , Encéfalo/metabolismo , Doença de Gaucher/genética , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Lisossomos/metabolismo , Mutação , Doença de Parkinson/genética , Progranulinas/genética , Camundongos Knockout
2.
Nature ; 543(7643): 108-112, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28225753

RESUMO

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.


Assuntos
Proteínas do Sistema Complemento/imunologia , Doença de Gaucher/imunologia , Doença de Gaucher/patologia , Glucosilceramidas/imunologia , Glucosilceramidas/metabolismo , Inflamação/imunologia , Inflamação/patologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Autoanticorpos/imunologia , Ativação do Complemento , Complemento C5a/biossíntese , Complemento C5a/imunologia , Proteínas do Sistema Complemento/biossíntese , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Feminino , Doença de Gaucher/metabolismo , Doença de Gaucher/prevenção & controle , Glucosilceramidase/antagonistas & inibidores , Glucosilceramidase/deficiência , Glucosilceramidase/genética , Glucosiltransferases/biossíntese , Glucosiltransferases/metabolismo , Humanos , Imunoglobulina G/imunologia , Inflamação/metabolismo , Inflamação/prevenção & controle , Masculino , Camundongos , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/imunologia , Receptor da Anafilatoxina C5a/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia
3.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-35054815

RESUMO

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.


Assuntos
Envelhecimento/patologia , Biomarcadores/metabolismo , Doenças Neurodegenerativas/metabolismo , Progranulinas/deficiência , Peptídeos beta-Amiloides/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Autofagia , Encéfalo/patologia , Encéfalo/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Lipofuscina/metabolismo , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Progranulinas/metabolismo , Agregados Proteicos , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
4.
Hum Mol Genet ; 28(20): 3406-3421, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31373366

RESUMO

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.


Assuntos
Doença de Gaucher/metabolismo , Doença de Gaucher/terapia , Glucosilceramidase/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Infusões Intravenosas , Integrina alfa4beta1/metabolismo , Camundongos , Células-Tronco Neurais/citologia , beta-Glucosidase/metabolismo
5.
Hum Mol Genet ; 25(23): 5126-5141, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27655403

RESUMO

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.


Assuntos
Dantroleno/administração & dosagem , Doença de Gaucher/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Animais , Sinalização do Cálcio/genética , Modelos Animais de Doenças , Doença de Gaucher/genética , Doença de Gaucher/fisiopatologia , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
6.
Hum Mol Genet ; 24(24): 7031-48, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26420838

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Doença de Gaucher/genética , Imino Piranoses/uso terapêutico , MicroRNAs/metabolismo , Chaperonas Moleculares/uso terapêutico , RNA Mensageiro/metabolismo , Animais , Axônios/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Encefalite/metabolismo , Encefalite/patologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Doença de Gaucher/tratamento farmacológico , Doença de Gaucher/metabolismo , Doença de Gaucher/patologia , Perfilação da Expressão Gênica , Glucosilceramidas/metabolismo , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Chaperonas Moleculares/metabolismo , Neuroglia/patologia , Neurônios/patologia , Fenótipo , Psicosina/análogos & derivados , Psicosina/metabolismo , Transdução de Sinais , Transmissão Sináptica , Serina-Treonina Quinases TOR/metabolismo
7.
Hum Mol Genet ; 23(15): 3943-57, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24599400

RESUMO

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.


Assuntos
Doença de Gaucher/genética , Regulação da Expressão Gênica , Mitocôndrias/metabolismo , Neurônios/metabolismo , beta-Glucosidase/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Doença de Gaucher/metabolismo , Doença de Gaucher/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Inositol/análogos & derivados , Inositol/farmacologia , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Neurônios/patologia , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Agregação Patológica de Proteínas , Substância Negra/metabolismo , Substância Negra/patologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , beta-Glucosidase/antagonistas & inibidores , beta-Glucosidase/metabolismo
8.
J Biol Chem ; 289(43): 30063-74, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25202012

RESUMO

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.


Assuntos
Doença de Gaucher/enzimologia , Glucosilceramidase/química , Glucosilceramidase/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Doenças Neurodegenerativas/enzimologia , Receptores Depuradores/metabolismo , Alanina/genética , Aminas/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Ligação Competitiva , Doença de Gaucher/patologia , Glucosilceramidase/deficiência , Proteínas de Fluorescência Verde/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Imunoprecipitação , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Mutação/genética , Doenças Neurodegenerativas/patologia , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Transfecção
9.
Mol Genet Metab ; 112(3): 229-41, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24837159

RESUMO

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.


Assuntos
Doença de Wolman/metabolismo , Doença de Wolman/patologia , Animais , Peso Corporal , Modelos Animais de Doenças , Terapia de Reposição de Enzimas , Feminino , Metabolismo dos Lipídeos , Lipídeos/sangue , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão , Fenótipo , Esterol Esterase/administração & dosagem , Resultado do Tratamento , Doença de Wolman/tratamento farmacológico , Doença de Wolman/genética , Doença de Wolman/mortalidade , Doença de Wolman
10.
J Biol Chem ; 287(6): 4275-87, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22167193

RESUMO

Isofagomine (IFG) is an acid ß-glucosidase (GCase) active site inhibitor that acts as a pharmacological chaperone. The effect of IFG on GCase function was investigated in GCase mutant fibroblasts and mouse models. IFG inhibits GCase with K(i) ∼30 nM for wild-type and mutant enzymes (N370S and V394L). Fibroblasts treated with IFG at µM concentrations showed enhancement of WT and mutant GCase activities and protein levels. Administration of IFG (30 mg/kg/day) to the mice homozygous for GCase mutations (V394L, D409H, or D409V) led to increased GCase activity in visceral tissues and brain extracts. IFG effects on GCase stability and substrate levels were evaluated in a mouse model (hG/4L/PS-NA) that has doxycycline-controlled human WT GCase (hGCase) expression driven by a liver-specific promoter and is also homozygous for the IFG-responsive V394L GCase. Both human and mouse GCase activity and protein levels were increased in IFG-treated mice. The liver-secreted hGCase in serum was stabilized, and its effect on the lung and spleen involvement was enhanced by IFG treatment. In 8-week IFG-treated mice, the accumulated glucosylceramide and glucosylsphingosine were reduced by 75 and 33%, respectively. Decreases of storage cells were correlated with >50% reductions in substrate levels. These results indicate that IFG stabilizes GCase in tissues and serum and can reduce visceral substrates in vivo.


Assuntos
Doença de Gaucher/enzimologia , Glucosilceramidase/metabolismo , Glucosilceramidas/metabolismo , Imino Piranoses/farmacologia , Psicosina/análogos & derivados , Substituição de Aminoácidos , Animais , Domínio Catalítico , Células Cultivadas , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Doença de Gaucher/tratamento farmacológico , Doença de Gaucher/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucosilceramidase/antagonistas & inibidores , Glucosilceramidase/genética , Glucosilceramidas/genética , Humanos , Camundongos , Mutação de Sentido Incorreto , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Psicosina/genética , Psicosina/metabolismo
11.
Mol Ther Methods Clin Dev ; 29: 185-201, 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37063480

RESUMO

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.

12.
Hum Mol Genet ; 19(6): 1088-97, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20047948

RESUMO

Gaucher disease is caused by defective acid beta-glucosidase (GCase) function. Saposin C is a lysosomal protein needed for optimal GCase activity. To test the in vivo effects of saposin C on GCase, saposin C deficient mice (C-/-) were backcrossed to point mutated GCase (V394L/V394L) mice. The resultant mice (4L;C*) began to exhibit CNS abnormalities approximately 30 days: first as hindlimb paresis, then progressive tremor and ataxia. Death occurred approximately 48 days due to neurological deficits. Axonal degeneration was evident in brain stem, spinal cord and white matter of cerebellum accompanied by increasing infiltration of the brain stem, cortex and thalamus by CD68 positive microglial cells and activation of astrocytes. Electron microscopy showed inclusion bodies in neuronal processes and degenerating cells. Accumulation of p62 and Lamp2 were prominent in the brain suggesting the impairment of autophagosome/lysosome function. This phenotype was different from either V394L/V394L or C-/- alone. Relative to V394L/V394L mice, 4L;C* mice had diminished GCase protein and activity. Marked increases (20- to 30-fold) of glucosylsphingosine (GS) and moderate elevation (1.5- to 3-fold) of glucosylceramide (GC) were in 4L;C* brains. Visceral tissues had increases of GS and GC, but no storage cells were found. Neuronal cells in thick hippocampal slices from 4L;C* mice had significantly attenuated long-term potentiation, presumably resulting from substrate accumulation. The 4L;C* mouse mimics the CNS phenotype and biochemistry of some type 3 (neuronopathic) variants of Gaucher disease and is a unique model suitable for testing pharmacological chaperone and substrate reduction therapies, and investigating the mechanisms of neuronopathic Gaucher disease.


Assuntos
Doença de Gaucher/enzimologia , Glucosilceramidase/genética , Glucosilceramidas/metabolismo , Proteínas Mutantes/metabolismo , Doenças do Sistema Nervoso/complicações , Psicosina/análogos & derivados , Saposinas/deficiência , Substituição de Aminoácidos/genética , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/ultraestrutura , Cromatografia Líquida , Modelos Animais de Doenças , Doença de Gaucher/genética , Doença de Gaucher/patologia , Doença de Gaucher/fisiopatologia , Inflamação/complicações , Inflamação/patologia , Potenciação de Longa Duração/fisiologia , Longevidade , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Espectrometria de Massas , Camundongos , Doenças do Sistema Nervoso/enzimologia , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/fisiopatologia , Fenótipo , Psicosina/metabolismo , Saposinas/metabolismo
13.
Sci Transl Med ; 14(636): eabl9945, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35294256

RESUMO

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.


Assuntos
Encefalopatias , Transplante de Células-Tronco Hematopoéticas , Animais , Células da Medula Óssea , Encéfalo , Sistema Nervoso Central , Camundongos , Microglia
14.
Cells ; 10(9)2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34571934

RESUMO

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.


Assuntos
Autofagia , Doença de Gaucher/tratamento farmacológico , Glucosilceramidase/antagonistas & inibidores , Inibidores de Glicosídeo Hidrolases/farmacologia , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Doença de Gaucher/metabolismo , Doença de Gaucher/patologia , Glucosilceramidase/fisiologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neurônios/metabolismo , Neurônios/patologia , Serina-Treonina Quinases TOR/genética
15.
EBioMedicine ; 55: 102735, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32279952

RESUMO

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.


Assuntos
Barreira Hematoencefálica/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Doença de Gaucher/terapia , Glucosilceramidase/administração & dosagem , Fosfatidilserinas/química , Saposinas/química , Animais , Transporte Biológico , Modelos Animais de Doenças , Estabilidade de Medicamentos , Terapia de Reposição de Enzimas/métodos , Feminino , Doença de Gaucher/enzimologia , Doença de Gaucher/genética , Doença de Gaucher/mortalidade , Glucosilceramidase/deficiência , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Nanoestruturas/administração & dosagem , Nanoestruturas/química , Permeabilidade , Desempenho Psicomotor/efeitos dos fármacos , Desempenho Psicomotor/fisiologia , Análise de Sobrevida , Resultado do Tratamento
16.
ACS Chem Neurosci ; 11(20): 3464-3473, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33035424

RESUMO

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.


Assuntos
Doença de Gaucher , Animais , Inibidores Enzimáticos/farmacologia , Doença de Gaucher/tratamento farmacológico , Glucosiltransferases , Camundongos , Pirrolidinas/farmacologia
17.
Mol Genet Metab ; 97(1): 65-74, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19217815

RESUMO

The hydrolysis of glucosylceramide by acid beta-glucosidase proceeds via a two-step, double displacement mechanism that includes cleavage of the O-beta-glucosidic bond, enzyme-glucosylation and, then, enzyme-deglucosylation. Two residues that may impact this cycle are N370 and E235. The N370S mutant enzyme is very common in Gaucher disease type 1 patients. Homology and crystal data predictions suggested that E235 is the acid/base catalyst in the hydrolytic reaction. Here, the roles of N370 and E235 in hydrolysis were explored using mutant proteins with selected amino acid substitutions. Heterologously expressed enzymes were characterized using inhibitors, activators, and alternative substrates to gain insight into the effects on the glucosylation (single turnover) and deglucosylation (transglucosylation) steps in catalysis. Specific substitutions at N370 selectively altered only the glucosylation step whereas N370S altered this and the deglucosylation steps. To provide functional data to support E235 as the acid/base catalyst, progress curves with poor substrates with more acidic leaving groups were used in the presence and absence of azide as an exogenous nucleophile. The restoration of E235G activity to nearly wild-type levels was achieved using azide with 2,4-dinitrophenyl-beta-glucoside as substrate. The loss of the acidic arm of the pH optimum activity curve of E235G provided additional functional support for E235 as the acid/base in catalysis. This study provides insight into the function of these residues in acid beta-glucosidase active site function.


Assuntos
Asparagina/metabolismo , Biocatálise , Doença de Gaucher/enzimologia , Glucosilceramidase/deficiência , Glucosilceramidase/metabolismo , Glutamina/metabolismo , 2,4-Dinitrofenol , Substituição de Aminoácidos/efeitos dos fármacos , Azidas/farmacologia , Biocatálise/efeitos dos fármacos , Domínio Catalítico , Ativação Enzimática/efeitos dos fármacos , Glucosilceramidase/antagonistas & inibidores , Glicosilação/efeitos dos fármacos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Hidrólise/efeitos dos fármacos , Proteínas Mutantes/metabolismo , Fosfatidilserinas/farmacologia , Relação Estrutura-Atividade , Especificidade por Substrato/efeitos dos fármacos , Ácido Taurocólico/farmacologia
18.
Sci Rep ; 9(1): 5571, 2019 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-30944381

RESUMO

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.


Assuntos
Doença de Gaucher/genética , Glucosilceramidase/genética , Mutação/genética , Saposinas/deficiência , beta-Glucosidase/genética , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Glucosilceramidas/genética , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
19.
Neurotoxicology ; 64: 256-266, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28595912

RESUMO

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.


Assuntos
Adenosina Trifosfatases/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Manganês/toxicidade , Proteínas de Membrana/metabolismo , Adenosina Trifosfatases/genética , Animais , Comportamento Animal , Feminino , Masculino , Manganês/metabolismo , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , ATPases Translocadoras de Prótons , alfa-Sinucleína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA