Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Neurobiol Dis ; 152: 105279, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33516873

RESUMO

Sialidosis is a neuropathic lysosomal storage disease caused by a deficiency in the NEU1 gene-encoding lysosomal neuraminidase and characterized by abnormal accumulation of undigested sialyl-oligoconjugates in systemic organs including brain. Although patients exhibit neurological symptoms, the underlying neuropathological mechanism remains unclear. Here, we generated induced pluripotent stem cells (iPSCs) from skin fibroblasts with sialidosis and induced the differentiation into neural progenitor cells (NPCs) and neurons. Sialidosis NPCs and neurons mimicked the disease-like phenotypes including reduced neuraminidase activity, accumulation of sialyl-oligoconjugates and lysosomal expansions. Functional analysis also revealed that sialidosis neurons displayed two distinct abnormalities, defective exocytotic glutamate release and augmented α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR)-mediated Ca2+ influx. These abnormalities were restored by overexpression of the wild-type NEU1 gene, demonstrating causative role of neuraminidase deficiency in functional impairments of disease neurons. Comprehensive proteomics analysis revealed the significant reduction of SNARE proteins and glycolytic enzymes in synaptosomal fraction, with downregulation of ATP production. Bypassing the glycolysis by treatment of pyruvate, which is final metabolite of glycolysis pathway, improved both the synaptsomal ATP production and the exocytotic function. We also found that upregulation of AMPAR and L-type voltage dependent Ca2+ channel (VDCC) subunits in disease neurons, with the restoration of AMPAR-mediated Ca2+ over-load by treatment of antagonists for the AMPAR and L-type VDCC. Our present study provides new insights into both the neuronal pathophysiology and potential therapeutic strategy for sialidosis.


Assuntos
Sinalização do Cálcio/fisiologia , Mucolipidoses/fisiopatologia , Neurônios/patologia , Neurônios/fisiologia , Exocitose/fisiologia , Glicólise/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas , Sinapses/patologia , Sinapses/fisiologia
2.
J Proteome Res ; 16(8): 2802-2810, 2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28628327

RESUMO

Niemann-Pick disease type C (NPC) is an autosomal recessive lipid storage disorder, and the majority of cases are caused by mutations in the NPC1 gene. In this study, we clarified how a single gene mutation in the NPC1 gene impacts the cellular glycome by analyzing the total glycomic expression profile of Chinese hamster ovary cell mutants defective in the Npc1 gene (Npc1 KO CHO cells). A number of glycomic alterations were identified, including increased expression of lactosylceramide, GM1, GM2, GD1, various neolacto-series glycosphingolipids, and sialyl-T (O-glycan), which was found to be the major sialylated protein-bound glycan, as well as various N-glycans, which were commonly both fucosylated and sialylated. We also observed significant increases in the total amounts of free oligosaccharides (fOSs), especially in the unique complex- and hybrid-type fOSs. Treatment of Npc1 KO CHO cells with 2-hydroxypropyl-ß-cyclodextrin (HPBCD), which can reduce cholesterol and glycosphingolipid (GSL) storage, did not affect the glycomic alterations observed in the GSL-, N-, and O-glycans of Npc1 KO CHO cells. However, HPBCD treatment corrected the glycomic alterations observed in fOSs to levels observed in wild-type cells.


Assuntos
Glicômica , Mutação , Doença de Niemann-Pick Tipo C/genética , Animais , Antígenos CD/metabolismo , Células CHO , Cricetulus , Glicoesfingolipídeos/metabolismo , Lactosilceramidas/metabolismo , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/genética , Polissacarídeos/análise , beta-Ciclodextrinas/farmacologia
3.
Stem Cells ; 33(4): 1075-88, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25522247

RESUMO

Niemann-Pick disease type C (NPC) is a lysosomal storage disease characterized by abnormal accumulation of free cholesterol and glycolipids. Here, we established induced pluripotent stem cell (iPSC) lines from NPC patients. Hepatocyte-like cells (HLCs) and neural progenitors derived from the iPSC lines accumulated cholesterol and displayed impaired autophagy and ATP production. A molecular signature related to lipid metabolism was also impaired in the NPC-iPSC-derived HLCs. These findings indicate that iPSC-derived cells can phenocopy human NPC. We also newly found that 2-hydroxypropyl-γ-cyclodextrin (HPGCD) could reduce the cholesterol accumulation and restore the functional and molecular abnormalities in the NPC patient-derived cells, and do so more effectively than 2-hydroxypropyl-ß-cyclodextrin treatment. In addition, NPC model mice showed an improved liver status and prolonged survival with HPGCDs. Thus, iPSC lines derived from patient cells are powerful tools to study cellular models of NPC, and HPGCD is a potential new drug candidate for future treatment of this disease.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Doença de Niemann-Pick Tipo C/patologia , beta-Ciclodextrinas/farmacologia , gama-Ciclodextrinas/farmacologia , 2-Hidroxipropil-beta-Ciclodextrina , Animais , Técnicas de Cocultura , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Doença de Niemann-Pick Tipo C/metabolismo , Resultado do Tratamento , beta-Ciclodextrinas/uso terapêutico , gama-Ciclodextrinas/uso terapêutico
4.
ACS Appl Bio Mater ; 5(5): 2377-2388, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35506864

RESUMO

Niemann-Pick disease type C (NPC) is characterized by the accumulation of glycolipids such as free cholesterol, sphingomyelin, and gangliosides in late endosomes/lysosomes (endolysosomes) due to abnormalities in the membrane proteins NPC1 or NPC2. The main symptoms of NPC caused by free cholesterol accumulation in various tissues vary depending on the time of onset, but hepatosplenomegaly and neurological symptoms accompanied by decreased motor, cognitive, and mental functions are observed in all age groups. However, the efficacy of NPC treatment remains limited. Herein, we have fabricated lactose-appended hydroxypropyl-ß-cyclodextrin (Lac-HPßCD) and evaluated its lowering effects on cholesterol accumulation in NPC model mice. We reveal that Lac-HPßCD lowers cholesterol accumulation in the liver and spleen by reducing the amount of free cholesterol. Moreover, Lac-HPßCD reduces the amount of free cholesterol in the cerebrum and slightly alleviates motor dysfunction. These results suggest that Lac-HPßCD has potential for the treatment of NPC.


Assuntos
Doença de Niemann-Pick Tipo C , 2-Hidroxipropil-beta-Ciclodextrina/farmacologia , Animais , Colesterol/metabolismo , Endossomos/metabolismo , Lactose/metabolismo , Camundongos , Doença de Niemann-Pick Tipo C/tratamento farmacológico
5.
Sci Rep ; 11(1): 3982, 2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33597637

RESUMO

Mutations in the NPHS1 gene, which encodes NEPHRIN, cause congenital nephrotic syndrome, resulting from impaired slit diaphragm (SD) formation in glomerular podocytes. We previously reported NEPHRIN and SD abnormalities in the podocytes of kidney organoids generated from patient-derived induced pluripotent stem cells (iPSCs) with an NPHS1 missense mutation (E725D). However, the mechanisms underlying the disease may vary depending on the mutations involved, and thus generation of iPSCs from multiple patients is warranted. Here we established iPSCs from two additional patients with different NPHS1 mutations and examined the podocyte abnormalities in kidney organoids derived from these cells. One patient had truncating mutations, and NEPHRIN was undetectable in the resulting organoids. The other patient had a missense mutation (R460Q), and the mutant NEPHRIN in the organoids failed to accumulate on the podocyte surface to form SD precursors. However, the same mutant protein behaved normally when overexpressed in heterologous cells, suggesting that NEPHRIN localization is cell context-dependent. The localization of another SD-associated protein, PODOCIN, was impaired in both types of mutant organoids in a cell domain-specific manner. Thus, the new iPSC lines and resultant kidney organoids will be useful resources for dissecting the disease mechanisms, as well as for drug development for therapies.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Mutantes/metabolismo , Síndrome Nefrótica/fisiopatologia , Organoides/metabolismo , Sequência de Aminoácidos , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Rim , Masculino , Proteínas de Membrana/genética , Proteínas Mutantes/genética , Mutação de Sentido Incorreto
6.
Sci Rep ; 11(1): 1552, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33452387

RESUMO

Moyamoya disease (MMD) is characterized by progressive bilateral stenotic changes in the terminal portion of the internal carotid arteries. Although RNF213 was identified as a susceptibility gene for MMD, the exact pathogenesis remains unknown. Immunohistochemical analysis of autopsy specimens from a patient with MMD revealed marked accumulation of hyaluronan and chondroitin sulfate (CS) in the thickened intima of occlusive lesions of MMD. Hyaluronan synthase 2 was strongly expressed in endothelial progenitor cells in the thickened intima. Furthermore, MMD lesions showed minimal staining for CS and hyaluronan in the endothelium, in contrast to control endothelium showing positive staining for both. Glycosaminoglycans of endothelial cells derived from MMD and control induced pluripotent stem cells demonstrated a decreased amount of CS, especially sulfated CS, in MMD. A computational fluid dynamics model showed highest wall shear stress values in the terminal portion of the internal carotid artery, which is the predisposing region in MMD. Because the peri-endothelial extracellular matrix plays an important role in protection, cell adhesion and migration, an altered peri-endothelial matrix in MMD may contribute to endothelial vulnerability to wall shear stress. Invading endothelial progenitor cells repairing endothelial injury would produce excessive hyaluronan and CS in the intima, and cause vascular stenosis.


Assuntos
Células Endoteliais/metabolismo , Doença de Moyamoya/fisiopatologia , Adenosina Trifosfatases/metabolismo , Adolescente , Idoso , Fenômenos Biomecânicos/fisiologia , Artéria Carótida Interna/patologia , Espessura Intima-Media Carotídea , Sulfatos de Condroitina/análise , Células Endoteliais/fisiologia , Endotélio/metabolismo , Feminino , Predisposição Genética para Doença , Humanos , Ácido Hialurônico/análise , Hidrodinâmica , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Doença de Moyamoya/metabolismo , Resistência ao Cisalhamento/fisiologia , Estresse Mecânico , Ubiquitina-Proteína Ligases/metabolismo
7.
Neuroscience ; 414: 128-140, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31283907

RESUMO

Tay-Sachs disease (TSD) is a GM2 gangliosidosis lysosomal storage disease caused by a loss of lysosomal hexosaminidase-A (HEXA) activity and characterized by progressive neurodegeneration due to the massive accumulation of GM2 ganglioside in the brain. Here, we generated iPSCs derived from patients with TSD, and found similar potential for neural differentiation between TSD-iPSCs and normal iPSCs, although neural progenitor cells (NPCs) derived from the TSD-iPSCs exhibited enlarged lysosomes and upregulation of the lysosomal marker, LAMP1, caused by the accumulation of GM2 ganglioside. The NPCs derived from TSD-iPSCs also had an increased incidence of oxidative stress-induced cell death. TSD-iPSC-derived neurons showed a decrease in exocytotic activity with the accumulation of GM2 ganglioside, suggesting deficient neurotransmission in TSD. Our findings demonstrated that NPCs and mature neurons derived from TSD-iPSCs are potentially useful cellular models of TSD and are useful for investigating the efficacy of drug candidates in the future.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Terminações Pré-Sinápticas/fisiologia , Doença de Tay-Sachs/fisiopatologia , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Células-Tronco Neurais/fisiologia , Neuritos/fisiologia , Sinapsinas/metabolismo , Doença de Tay-Sachs/metabolismo , Regulação para Cima/fisiologia
8.
PLoS One ; 13(7): e0200790, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30044827

RESUMO

Mesenchymal stem cells (MSCs) isolated from adult human tissues are capable of proliferating in vitro and maintaining their multipotency, making them attractive cell sources for regenerative medicine. However, the availability and capability of self-renewal under current preparation regimes are limited. Induced pluripotent stem cells (iPSCs) now offer an alternative, similar cell source to MSCs. Herein, we established new methods for differentiating hiPSCs into MSCs via mesoderm-like and neuroepithelium-like cells. Both derived MSC populations exhibited self-renewal and multipotency, as well as therapeutic potential in mouse models of skin wounds, pressure ulcers, and osteoarthritis. Interestingly, the therapeutic effects differ between the two types of MSCs in the disease models, suggesting that the therapeutic effect depends on the cell origin. Our results provide valuable basic insights for the clinical application of such cells.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Medicina Regenerativa/métodos , Adipócitos/citologia , Animais , Diferenciação Celular , Modelos Animais de Doenças , Humanos , Masculino , Mesoderma/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese , Pele/metabolismo
9.
Sci Rep ; 8(1): 1634, 2018 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-29374279

RESUMO

Recently, we reported that bacterial incorporation induces cellular transdifferentiation of human fibroblasts. However, the bacterium-intrinsic cellular- transdifferentiation factor remained unknown. Here, we found that cellular transdifferentiation is caused by ribosomes. Ribosomes, isolated from both prokaryotic and eukaryotic cells, induce the formation of embryoid body-like cell clusters. Numerous ribosomes are incorporated into both the cytoplasm and nucleus through trypsin-activated endocytosis, which leads to cell-cluster formation. Although ribosome-induced cell clusters (RICs) express several stemness markers and differentiate into derivatives of all three germ layers in heterogeneous cell populations, RICs fail to proliferate, alter the methylation states of pluripotent genes, or contribute to teratoma or chimera formation. However, RICs express markers of epithelial-mesenchymal transition without altering the cell cycle, despite their proliferation obstruction. These findings demonstrate that incorporation of ribosomes into host cells induces cell transdifferentiation and alters cellular plasticity.


Assuntos
Transdiferenciação Celular , Fibroblastos/fisiologia , Ribossomos/metabolismo , Bactérias/metabolismo , Células Cultivadas , Endocitose , Humanos
10.
PLoS One ; 9(12): e113052, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25479600

RESUMO

Induced pluripotent stem cells (iPSCs) are potentially valuable cell sources for disease models and future therapeutic applications; however, inefficient generation and the presence of integrated transgenes remain as problems limiting their current use. Here, we developed a new Sendai virus vector, TS12KOS, which has improved efficiency, does not integrate into the cellular DNA, and can be easily eliminated. TS12KOS carries KLF4, OCT3/4, and SOX2 in a single vector and can easily generate iPSCs from human blood cells. Using TS12KOS, we established iPSC lines from chimpanzee blood, and used DNA array analysis to show that the global gene-expression pattern of chimpanzee iPSCs is similar to those of human embryonic stem cell and iPSC lines. These results demonstrated that our new vector is useful for generating iPSCs from the blood cells of both human and chimpanzee. In addition, the chimpanzee iPSCs are expected to facilitate unique studies into human physiology and disease.


Assuntos
Diferenciação Celular/genética , Vetores Genéticos , Vírus Sendai/genética , Transdução Genética , Animais , Linhagem Celular , Reprogramação Celular/genética , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator 4 Semelhante a Kruppel , Pan troglodytes , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA