Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 37(11): e23216, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37779422

RESUMO

Glycogen storage disease type Ib (GSD-Ib) is an autosomal recessive disorder caused by a deficiency in the glucose-6-phosphate (G6P) transporter (G6PT) that is responsible for transporting G6P into the endoplasmic reticulum. GSD-Ib is characterized by disturbances in glucose homeostasis, neutropenia, and neutrophil dysfunction. Although some studies have explored neutrophils abnormalities in GSD-Ib, investigations regarding monocytes/macrophages remain limited so far. In this study, we examined the impact of G6PT deficiency on monocyte-to-macrophage differentiation using bone marrow-derived monocytes from G6pt-/- mice as well as G6PT-deficient human THP-1 monocytes. Our findings revealed that G6PT-deficient monocytes exhibited immature differentiation into macrophages. Notably, the impaired differentiation observed in G6PT-deficient monocytes seemed to be associated with abnormal glucose metabolism, characterized by enhanced glucose consumption through glycolysis, even under quiescent conditions with oxidative phosphorylation. Furthermore, we observed a reduced secretion of inflammatory cytokines in G6PT-deficient THP-1 monocytes during the inflammatory response, despite their elevated glucose consumption. In conclusion, this study sheds light on the significance of G6PT in monocyte-to-macrophage differentiation and underscores its importance in maintaining glucose homeostasis and supporting immune response in GSD-Ib. These findings may contribute to a better understanding of the pathogenesis of GSD-Ib and potentially pave the way for the development of targeted therapeutic interventions.


Assuntos
Doença de Depósito de Glicogênio Tipo I , Animais , Camundongos , Humanos , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/metabolismo , Doença de Depósito de Glicogênio Tipo I/terapia , Glucose-6-Fosfatase/metabolismo , Glucose/metabolismo , Macrófagos/metabolismo
2.
Cell Mol Life Sci ; 79(5): 246, 2022 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-35437689

RESUMO

Glycogen storage disease type Ib (GSD-Ib), characterized by impaired glucose homeostasis, neutropenia, and neutrophil dysfunction, is caused by a deficiency in glucose-6-phosphate transporter (G6PT). Neutropenia in GSD-Ib has been known to result from enhanced apoptosis of neutrophils. However, it has also been raised that neutrophil maturation arrest in the bone marrow would contribute to neutropenia. We now show that G6pt-/- mice exhibit severe neutropenia and impaired neutrophil differentiation in the bone marrow. To investigate the role of G6PT in myeloid progenitor cells, the G6PT gene was mutated using CRISPR/Cas9 system, and single cell-derived G6PT-/- human promyelocyte HL-60 cell lines were established. The G6PT-/- HL-60s exhibited impaired neutrophil differentiation, which is associated with two mechanisms: (i) abnormal lipid metabolism causing a delayed metabolic reprogramming and (ii) reduced nuclear transcriptional activity of peroxisome proliferator-activated receptor-γ (PPARγ) in G6PT-/- HL-60s. In this study, we demonstrated that G6PT is essential for neutrophil differentiation of myeloid progenitor cells and regulates PPARγ activity.


Assuntos
Doença de Depósito de Glicogênio Tipo I , Neutropenia , Animais , Antiporters/genética , Antiporters/metabolismo , Glucose/metabolismo , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/metabolismo , Camundongos , Neutropenia/complicações , Neutropenia/metabolismo , Neutrófilos/metabolismo , PPAR gama/genética , PPAR gama/metabolismo
3.
Int J Mol Sci ; 23(17)2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-36077341

RESUMO

Glycogen storage disease type IX (GSD-IX) constitutes nearly a quarter of all GSDs. This ketotic form of GSD is caused by mutations in phosphorylase kinase (PhK), which is composed of four subunits (α, ß, γ, δ). PhK is required for the activation of the liver isoform of glycogen phosphorylase (PYGL), which generates free glucose-1-phosphate monomers to be used as energy via cleavage of the α -(1,4) glycosidic linkages in glycogen chains. Mutations in any of the PhK subunits can negatively affect the regulatory and catalytic activity of PhK during glycogenolysis. To understand the pathogenesis of GSD-IX-beta, we characterized a newly created PHKB knockout (Phkb−/−) mouse model. In this study, we assessed fasting blood glucose and ketone levels, serum metabolite concentrations, glycogen phosphorylase activity, and gene expression of gluconeogenic genes and fibrotic genes. Phkb−/− mice displayed hepatomegaly with lower fasting blood glucose concentrations. Phkb−/− mice showed partial liver glycogen phosphorylase activity and increased sensitivity to pyruvate, indicative of partial glycogenolytic activity and upregulation of gluconeogenesis. Additionally, gene expression analysis demonstrated increased lipid metabolism in Phkb−/− mice. Gene expression analysis and liver histology in the livers of old Phkb−/− mice (>40 weeks) showed minimal profibrogenic features when analyzed with age-matched wild-type (WT) mice. Collectively, the Phkb−/− mouse recapitulates mild clinical features in patients with GSD-IX-beta. Metabolic and molecular analysis confirmed that Phkb−/− mice were capable of sustaining energy homeostasis during prolonged fasting by using partial glycogenolysis, increased gluconeogenesis, and potentially fatty acid oxidation in the liver.


Assuntos
Doença de Depósito de Glicogênio , Glicogenólise , Fosforilase Quinase/metabolismo , Animais , Glicemia/metabolismo , Modelos Animais de Doenças , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/metabolismo , Fígado/metabolismo , Camundongos , Fosforilase Quinase/genética
4.
J Inherit Metab Dis ; 44(1): 118-128, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32474930

RESUMO

Glycogen storage disease type Ia (GSD-Ia) is an inherited metabolic disease caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC) which plays a critical role in blood glucose homeostasis by catalyzing the hydrolysis of glucose-6-phosphate (G6P) to glucose and phosphate in the terminal step of glycogenolysis and gluconeogenesis. Patients with GSD-Ia manifest life-threatening fasting hypoglycemia along with the excessive accumulation of hepatic glycogen and triglycerides which results in hepatomegaly and a risk of long-term complications such as hepatocellular adenoma and carcinoma (HCA/HCC). The etiology of HCA/HCC development in GSD-Ia, however, is unknown. Recent studies have shown that the livers in model animals of GSD-Ia display impairment of autophagy, a cellular recycling process which is critical for energy metabolism and cellular homeostasis. However, molecular mechanisms of autophagy impairment and its involvement in pathogenesis in GSD-Ia are still under investigation. Here, we summarize the latest advances for signaling pathways implicated in hepatic autophagy impairment and the roles of autophagy in hepatic tumorigenesis in GSD-Ia. In addition, recent evidence has illustrated that autophagy plays an important role in hepatic metabolism and liver-directed gene therapy mediated by recombinant adeno-associated virus (rAAV). Therefore, we highlight the possible role of hepatic autophagy in metabolic control and rAAV-mediated gene therapy for GSD-Ia. In this review, we also provide potential therapeutic strategies for GSD-Ia on the basis of molecular mechanisms underlying hepatic autophagy impairment in GSD-Ia.


Assuntos
Autofagia , Carcinoma Hepatocelular/prevenção & controle , Terapia Genética/métodos , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/terapia , Neoplasias Hepáticas/prevenção & controle , Animais , Carcinogênese/patologia , Carcinoma Hepatocelular/enzimologia , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Glucose/metabolismo , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/enzimologia , Homeostase , Humanos , Fígado/enzimologia , Neoplasias Hepáticas/enzimologia , Camundongos , Camundongos Knockout , Transdução de Sinais
5.
Int J Mol Sci ; 23(1)2021 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-35008754

RESUMO

Glycogen storage disease type Ia (GSDIa) is an inherited metabolic disorder caused by mutations in the enzyme glucose-6-phosphatase-α (G6Pase-α). Affected individuals develop renal and liver complications, including the development of hepatocellular adenoma/carcinoma and kidney failure. The purpose of this study was to identify potential biomarkers of the evolution of the disease in GSDIa patients. To this end, we analyzed the expression of exosomal microRNAs (Exo-miRs) in the plasma exosomes of 45 patients aged 6 to 63 years. Plasma from age-matched normal individuals were used as controls. We found that the altered expression of several Exo-miRs correlates with the pathologic state of the patients and might help to monitor the progression of the disease and the development of late GSDIa-associated complications.


Assuntos
Exossomos/genética , Doença de Depósito de Glicogênio Tipo I/genética , Nefropatias/genética , Fígado/lesões , Fígado/metabolismo , MicroRNAs/genética , Adolescente , Adulto , Fatores Etários , Animais , Biomarcadores/metabolismo , Estudos de Casos e Controles , Criança , Pré-Escolar , Estudos de Coortes , Exossomos/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ontologia Genética , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio Tipo I/sangue , Doença de Depósito de Glicogênio Tipo I/patologia , Humanos , Nefropatias/sangue , Nefropatias/patologia , Masculino , Camundongos , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Fatores de Tempo , Adulto Jovem
6.
Biochem Biophys Res Commun ; 522(1): 1-7, 2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-31735334

RESUMO

Hepatocellular adenoma/carcinoma (HCA/HCC) is a long-term complication of the metabolic disorder glycogen storage disease type Ia (GSD-Ia) deficient in glucose-6-phosphatase-α (G6PC or G6Pase-α). We have shown previously that hepatic G6Pase-α deficiency leads to autophagy impairment, mitochondrial dysfunction, enhanced glycolysis, and augmented hexose monophosphate shunt, all of which can contribute to hepatocarcinogenesis. However, the mechanism underlying HCA/HCC development in GSD-Ia remains unclear. We now show that G6Pase-α deficiency-mediated hepatic autophagy impairment leads to sustained accumulation of an autophagy-specific substrate p62 which can activate tumor-promoting pathways including nuclear factor erythroid 2-related factor 2 (Nrf2) and mammalian target of rapamycin complex 1 (mTORC1). Consistently, the HCA/HCC lesions developed in the G6Pase-α-deficient livers display marked accumulation of p62 aggregates and phosphorylated p62 along with activation of Nrf2 and mTORC1 signaling. Furthermore, the HCA/HCC lesions exhibit activation of additional oncogenic pathways, ß-catenin and Yes-associated protein (YAP) which is implicated in autophagy impairment. Intriguingly, hepatic levels of glucose-6-phosphate and glycogen which are accumulated in the G6Pase-α-deficient livers were significantly lower in HCC than those in HCA. Conversely, compared to HCA, the HCC lesion display increased expression of many oncogenes and the M2 isoform of pyruvate kinase (PKM2), a glycolytic enzyme critical for aerobic glycolysis and tumorigenesis. Collectively, our data show that hepatic G6Pase-α-deficiency leads to persistent autophagy impairment and activation of multiple tumor-promoting pathways that contribute to HCA/HCC development in GSD-Ia.


Assuntos
Carcinoma Hepatocelular/etiologia , Doença de Depósito de Glicogênio Tipo I/complicações , Neoplasias Hepáticas/etiologia , Animais , Autofagia , Carcinogênese/metabolismo , Carcinogênese/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio Tipo I/metabolismo , Doença de Depósito de Glicogênio Tipo I/patologia , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais
7.
Biochem Biophys Res Commun ; 524(1): 89-95, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-31980167

RESUMO

Glycogen storage disease type Ib (GSD-Ib), caused by a deficiency in glucose-6-phosphate transporter (G6PT), is characterized by disrupted glucose homeostasis, inflammatory bowel disease, neutropenia, and neutrophil dysfunction. The purpose of this study was to investigate the role of G6PT on macrophage functions and metabolism. Peritoneal macrophages of G6pt-/- mice were lower in number and their effector functions including migration, superoxide production, and phagocytosis were impaired. To investigate the underlying mechanisms of macrophage dysfunction, the G6PT gene was mutated in porcine alveolar macrophage 3D4/31 cells using the CRISPR/Cas9 technology. The G6PT-deficient macrophages exhibited significant decline in cell growth, bactericidal activity, and antiviral response. These phenotypes are associated with the impaired glycolysis and mitochondrial oxidative phosphorylation. We therefore propose that the G6PT-mediated metabolism is essential for effector functions of macrophage, the immune deficiencies observed in GSD-Ib extend beyond neutropenia and neutrophil dysfunction, and future therapeutic targets aimed both the neutrophils and macrophages may be necessary.


Assuntos
Antiporters/genética , Antiporters/metabolismo , Doença de Depósito de Glicogênio Tipo I/metabolismo , Macrófagos/metabolismo , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Linhagem Celular , Proliferação de Células , Glucose/metabolismo , Glicólise , Humanos , Macrófagos/citologia , Camundongos , Mitocôndrias/metabolismo , Modelos Animais , Mutação , Neutrófilos/metabolismo , Oxirredução , Fenótipo , Fosforilação , Suínos
8.
Hum Mol Genet ; 26(22): 4395-4405, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28973635

RESUMO

Glycogen storage disease type-Ib (GSD-Ib), deficient in the glucose-6-phosphate transporter (G6PT), is characterized by impaired glucose homeostasis, myeloid dysfunction, and long-term risk of hepatocellular adenoma (HCA). We examined the efficacy of G6PT gene therapy in G6pt-/- mice using recombinant adeno-associated virus (rAAV) vectors, directed by either the G6PC or the G6PT promoter/enhancer. Both vectors corrected hepatic G6PT deficiency in murine GSD-Ib but the G6PC promoter/enhancer was more efficacious. Over a 78-week study, using dose titration of the rAAV vectors, we showed that G6pt-/- mice expressing 3-62% of normal hepatic G6PT activity exhibited a normalized liver phenotype. Two of the 12 mice expressing < 6% of normal hepatic G6PT activity developed HCA. All treated mice were leaner and more sensitive to insulin than wild-type mice. Mice expressing 3-22% of normal hepatic G6PT activity exhibited higher insulin sensitivity than mice expressing 44-62%. The levels of insulin sensitivity correlated with the magnitudes of hepatic carbohydrate response element binding protein signaling activation. In summary, we established the threshold of hepatic G6PT activity required to prevent tumor formation and showed that mice expressing 3-62% of normal hepatic G6PT activity maintained glucose homeostasis and were protected against age-related obesity and insulin resistance.


Assuntos
Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/terapia , Animais , Antiporters/genética , Antiporters/metabolismo , Modelos Animais de Doenças , Vetores Genéticos , Glucose-6-Fosfatase/genética , Glucose-6-Fosfatase/metabolismo , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Doença de Depósito de Glicogênio Tipo I/metabolismo , Homeostase , Humanos , Resistência à Insulina , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Regiões Promotoras Genéticas
9.
J Inherit Metab Dis ; 42(3): 459-469, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30637773

RESUMO

Hepatocellular adenoma/carcinoma (HCA/HCC) is a long-term complication of glycogen storage disease type-Ia (GSD-Ia), which is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC), a key enzyme in gluconeogenesis. Currently, there is no therapy to address HCA/HCC in GSD-Ia. We have previously shown that a recombinant adeno-associated virus (rAAV) vector-mediated G6PC gene transfer to 2-week-old G6pc-/- mice prevents HCA development. However, it remains unclear whether G6PC gene transfer at the tumor developing stage of GSD-Ia can prevent tumor initiation or abrogate the pre-existing tumors. Using liver-specific G6pc-knockout (L-G6pc-/-) mice that develop HCA/HCC, we now show that treating the mice at the tumor-developing stage with rAAV-G6PC restores hepatic G6Pase-α expression, normalizes glucose homeostasis, and prevents de novo HCA/HCC development. The rAAV-G6PC treatment also normalizes defective hepatic autophagy and corrects metabolic abnormalities in the nontumor liver tissues of both tumor-free and tumor-bearing mice. However, gene therapy cannot restore G6Pase-α expression in the HCA/HCC lesions and fails to abrogate any pre-existing tumors. We show that the expression of 11 ß-hydroxysteroid dehydrogenase type-1 that mediates local glucocorticoid activation is downregulated in HCA/HCC lesions, leading to impairment in glucocorticoid signaling critical for gluconeogenesis activation. This suggests that local glucocorticoid action downregulation in the HCA/HCC lesions may suppress gene therapy mediated G6Pase-α restoration. Collectively, our data show that rAAV-mediated gene therapy can prevent de novo HCA/HCC development in L-G6pc-/- mice at the tumor developing stage, but it cannot reduce any pre-existing tumor burden.


Assuntos
Carcinoma Hepatocelular/prevenção & controle , Terapia Genética/métodos , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/terapia , Neoplasias Hepáticas/prevenção & controle , Animais , Carcinoma Hepatocelular/enzimologia , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Glucose/metabolismo , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/enzimologia , Homeostase , Humanos , Fígado/enzimologia , Neoplasias Hepáticas/enzimologia , Camundongos , Camundongos Knockout
10.
J Inherit Metab Dis ; 41(6): 977-984, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29802554

RESUMO

BACKGROUND: Viral mediated gene therapy has progressed after overcoming early failures, and gene therapy has now been approved for several conditions in Europe and the USA. Glycogen storage disease (GSD) type Ia, caused by a deficiency of glucose-6-phosphatase-α, has been viewed as an outstanding candidate for gene therapy. This follow-up report describes the long-term outcome for the naturally occurring GSD-Ia dogs treated with rAAV-GPE-hG6PC-mediated gene therapy. METHODS: A total of seven dogs were treated with rAAV-GPE-hG6PC-mediated gene therapy. The first four dogs were treated at birth, and three dogs were treated between 2 and 6 months of age to assess the efficacy and safety in animals with mature livers. Blood and urine samples, radiographic studies, histological evaluation, and biodistribution were assessed. RESULTS: Gene therapy improved survival in the GSD-Ia dogs. With treatment, the biochemical studies normalized for the duration of the study (up to 7 years). None of the rAAV-GPE-hG6PC-treated dogs had focal hepatic lesions or renal abnormalities. Dogs treated at birth required a second dose of rAAV after 2-4 months; gene therapy after hepatic maturation resulted in improved efficacy after a single dose. CONCLUSION: rAAV-GPE-hG6PC treatment in GSD-Ia dogs was found to be safe and efficacious. GSD-Ia is an attractive target for human gene therapy since it is a monogenic disorder with limited tissue involvement. Blood glucose and lactate monitoring can be used to assess effectiveness and as a biomarker of success. GSD-Ia can also serve as a model for other hepatic monogenic disorders.


Assuntos
Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo I/terapia , Animais , Glicemia/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Cães , Europa (Continente) , Vetores Genéticos , Glucose-6-Fosfatase/genética , Hipoglicemia/genética , Hipoglicemia/metabolismo , Rim/metabolismo , Fígado/metabolismo
11.
Hum Mol Genet ; 24(18): 5115-25, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26089201

RESUMO

Glycogen storage disease type-Ia (GSD-Ia) is caused by a lack of glucose-6-phosphatase-α (G6Pase-α or G6PC) activity. We have shown that gene therapy mediated by a recombinant adeno-associated virus (rAAV) vector expressing human G6Pase-α normalizes blood glucose homeostasis in the global G6pc knockout (G6pc(-/-)) mice for 70-90 weeks. The treated G6pc(-/-) mice expressing 3-63% of normal hepatic G6Pase-α activity (AAV mice) produce endogenous hepatic glucose levels 61-68% of wild-type littermates, have a leaner phenotype and exhibit fasting blood insulin levels more typical of young adult mice. We now show that unlike wild-type mice, the lean AAV mice have increased caloric intake and do not develop age-related obesity or insulin resistance. Pathway analysis shows that signaling by hepatic carbohydrate response element binding protein that improves glucose tolerance and insulin signaling is activated in AAV mice. In addition, several longevity factors in the calorie restriction pathway, including the NADH shuttle systems, NAD(+) concentrations and the AMP-activated protein kinase/sirtuin 1/peroxisome proliferator-activated receptor-γ coactivator 1α pathway are upregulated in the livers of AAV mice. The finding that partial restoration of hepatic G6Pase-α activity in GSD-Ia mice not only attenuates the phenotype of hepatic G6Pase-α deficiency but also prevents the development of age-related obesity and insulin resistance seen in wild-type mice may suggest relevance of the G6Pase-α enzyme to obesity and diabetes.


Assuntos
Expressão Gênica , Glucose-6-Fosfatase/genética , Resistência à Insulina/genética , Obesidade/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Dependovirus/genética , Modelos Animais de Doenças , Metabolismo Energético/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , NAD/metabolismo , Proteínas Nucleares/metabolismo , Obesidade/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Fatores de Transcrição/metabolismo
12.
Biochem Biophys Res Commun ; 482(4): 569-574, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27864142

RESUMO

Glycogen storage disease type Ib (GSD-Ib), characterized by impaired glucose homeostasis, neutropenia, and neutrophil dysfunction, is an inherited autosomal recessive disorder caused by a deficiency in the glucose-6-phosphate transporter (G6PT). Neutrophils play an essential role in the defense against invading pathogens. The recruitment of neutrophils towards the inflammation sites in response to inflammatory stimuli is a tightly regulated process involving rolling, adhesion, and transmigration. In this study, we investigated the role of G6PT in neutrophil adhesion and migration using in vivo and in vitro models. We showed that the GSD-Ib (G6pt-/-) mice manifested severe neutropenia in both blood and bone marrow, and treating G6pt-/- mice with granulocyte colony-stimulating factor (G-CSF) corrected neutropenia. However, upon thioglycolate challenge, neutrophils from both untreated and G-CSF-treated G6pt-/-mice exhibited decreased ability to migrate to the peritoneal cavity. In vitro migration and cell adhesion of G6PT-deficient neutrophils were also significantly impaired. Defects in cell migration were not due to enhanced apoptosis or altered fMLP receptor expression. Remarkably, the expression of the ß2 integrins CD11a and CD11b, which are critical for cell adhesion, was greatly decreased in G6PT-deficient neutrophils. This study suggests that deficiencies in G6PT cause impairment in neutrophil adhesion and migration via aberrant expression of ß2 integrins, and our finding should facilitate the development of novel therapies for GSD-Ib.


Assuntos
Adesão Celular , Movimento Celular , Doença de Depósito de Glicogênio Tipo I/patologia , Neutropenia/patologia , Neutrófilos/patologia , Animais , Antiporters/genética , Apoptose , Células CACO-2 , Células Cultivadas , Deleção de Genes , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/genética , Humanos , Camundongos , Proteínas de Transporte de Monossacarídeos/genética , Neutropenia/complicações , Neutropenia/genética , Neutrófilos/citologia
13.
Mol Genet Metab ; 120(3): 229-234, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28096054

RESUMO

Glycogen storage disease type Ia (GSD-Ia), characterized by impaired glucose homeostasis and chronic risk of hepatocellular adenoma (HCA) and carcinoma (HCC), is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC). We have previously shown that G6pc-/- mice receiving gene transfer mediated by rAAV-G6PC, a recombinant adeno-associated virus (rAAV) vector expressing G6Pase-α, and expressing 3-63% of normal hepatic G6Pase-α activity maintain glucose homeostasis and do not develop HCA/HCC. However, the threshold of hepatic G6Pase-α activity required to prevent tumor formation remained unknown. In this study, we constructed rAAV-co-G6PC, a rAAV vector expressing a codon-optimized (co) G6Pase-α and showed that rAAV-co-G6PC was more efficacious than rAAV-G6PC in directing hepatic G6Pase-α expression. Over an 88-week study, we showed that both rAAV-G6PC- and rAAV-co-G6PC-treated G6pc-/- mice expressing 3-33% of normal hepatic G6Pase-α activity (AAV mice) maintained glucose homeostasis, lacked HCA/HCC, and were protected against age-related obesity and insulin resistance. Of the eleven rAAV-G6PC/rAAV-co-G6PC-treated G6pc-/- mice harboring 0.9-2.4% of normal hepatic G6Pase-α activity (AAV-low mice), 3 expressing 0.9-1.3% of normal hepatic G6Pase-α activity developed HCA/HCC, while 8 did not (AAV-low-NT). Finally, we showed that the AAV-low-NT mice exhibited a phenotype indistinguishable from that of AAV mice expressing ≥3% of normal hepatic G6Pase-α activity. The results establish the threshold of hepatic G6Pase-α activity required to prevent HCA/HCC and show that GSD-Ia mice harboring <2% of normal hepatic G6Pase-α activity are at risk of tumor development.


Assuntos
Adenoma de Células Hepáticas/prevenção & controle , Carcinoma Hepatocelular/prevenção & controle , Terapia Genética/métodos , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/terapia , Neoplasias Hepáticas/prevenção & controle , Adenoma de Células Hepáticas/enzimologia , Animais , Carcinoma Hepatocelular/enzimologia , Dependovirus/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vetores Genéticos/administração & dosagem , Glucose/metabolismo , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/enzimologia , Homeostase , Humanos , Fígado/enzimologia , Neoplasias Hepáticas/enzimologia , Camundongos
14.
Blood ; 123(18): 2843-53, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24565827

RESUMO

Glycogen storage disease type Ib (GSD-Ib) is an autosomal-recessive syndrome characterized by neutropenia and impaired glucose homeostasis resulting from a deficiency in the glucose-6-phosphate (G6P) transporter (G6PT). The underlying cause of GSD-Ib neutropenia is an enhanced neutrophil apoptosis, but patients also manifest neutrophil dysfunction of unknown etiology. Previously, we showed G6PT interacts with the enzyme glucose-6-phosphatase-ß (G6Pase-ß) to regulate the availability of G6P/glucose in neutrophils. A deficiency in G6Pase-ß activity in neutrophils impairs both their energy homeostasis and function. We now show that G6PT-deficient neutrophils from GSD-Ib patients are similarly impaired. Their energy impairment is characterized by decreased glucose uptake and reduced levels of intracellular G6P, lactate, adenosine triphosphate, and reduced NAD phosphate, whereas functional impairment is reflected in reduced neutrophil respiratory burst, chemotaxis, and calcium mobilization. We further show that the mechanism of neutrophil dysfunction in GSD-Ib arises from activation of the hypoxia-inducible factor-1α/peroxisome-proliferators-activated receptor-γ pathway.


Assuntos
Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/metabolismo , Neutrófilos/metabolismo , Trifosfato de Adenosina/metabolismo , Adolescente , Adulto , Antiporters/deficiência , Antiporters/metabolismo , Criança , Pré-Escolar , Ativação Enzimática , Glucose/metabolismo , Glucose-6-Fosfatase/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imunofenotipagem , Espaço Intracelular/metabolismo , Ácido Láctico/metabolismo , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/metabolismo , NADP/metabolismo , NADPH Oxidases/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , PPAR gama/metabolismo , Fenótipo , Transdução de Sinais , Adulto Jovem
15.
Blood ; 119(17): 4047-55, 2012 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-22246029

RESUMO

Glucose-6-phosphatase-ß (G6Pase-ß or G6PC3) deficiency, also known as severe congenital neutropenia syndrome 4, is characterized not only by neutropenia but also by impaired neutrophil energy homeostasis and functionality. We now show the syndrome is also associated with macrophage dysfunction, with murine G6pc3(-/-) macrophages having impairments in their respiratory burst, chemotaxis, calcium flux, and phagocytic activities. Consistent with a glucose-6-phosphate (G6P) metabolism deficiency, G6pc3(-/-) macrophages also have a lower glucose uptake and lower levels of G6P, lactate, and ATP than wild-type macrophages. Furthermore, the expression of NADPH oxidase subunits and membrane translocation of p47(phox) are down-regulated, and G6pc3(-/-) macrophages exhibit repressed trafficking in vivo both during an inflammatory response and in pregnancy. During pregnancy, the absence of G6Pase-ß activity also leads to impaired energy homeostasis in the uterus and reduced fertility of G6pc3(-/-) mothers. Together these results show that immune deficiencies in this congenital neutropenia syndrome extend beyond neutrophil dysfunction.


Assuntos
Glucose-6-Fosfatase/fisiologia , Glucose-6-Fosfato/metabolismo , Homeostase/fisiologia , Inflamação/patologia , Macrófagos/fisiologia , Neutropenia/congênito , Animais , Apoptose , Western Blotting , Cálcio/metabolismo , Proliferação de Células , Quimiotaxia , Síndrome Congênita de Insuficiência da Medula Óssea , Citocinas/metabolismo , Feminino , Glucose/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 3/genética , Transportador de Glucose Tipo 3/metabolismo , Técnicas Imunoenzimáticas , Inflamação/genética , Inflamação/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Neutropenia/genética , Neutropenia/metabolismo , Neutropenia/patologia , Fagocitose , Gravidez , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Explosão Respiratória , Transdução de Sinais , Síndrome
16.
J Korean Med Sci ; 29(12): 1626-31, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25469061

RESUMO

Depression is an important comorbidity of asthma. However, little information is available about depression and its potential impact on asthma control in Korean adult asthma patients. We aimed to estimate the prevalence and risk factors for depression in Korean adults with persistent asthma. The 127 non-elderly (20-64 yr) and 75 elderly (≥65 yr) patients with asthma were recruited. Demographic and clinical data were extracted, and the patients completed the Asthma Specific Quality of Life (AQOL) questionnaire and asthma control test (ACT). Depression status was defined using the Korean version of the Patient Health Questionnaire-9 (PHQ-9). Depression was more prevalent in non-elderly (18.9%) than in elderly patients with asthma (13.3%). Patients with depression were significantly younger, had lower economic status, shorter disease duration, poorer asthma control, and worse AQOL scores (P<0.05). Within the non-elderly group, younger age and shorter disease duration were significantly associated with depression (P<0.05). Within the elderly group, a higher body mass index and current smoking status were significantly associated with depression (P<0.05). The PHQ-9 score was significantly correlated with worse ACT and AQOL scores in both groups. In conclusion, depression is strongly associated with poor asthma control and quality of life in Korean adult asthma patients. Our results provide important clues that used to target modifiable factors which contribute to development of depression in asthma patients.


Assuntos
Asma/epidemiologia , Asma/psicologia , Depressão/epidemiologia , Depressão/psicologia , Qualidade de Vida/psicologia , Adulto , Distribuição por Idade , Idoso , Idoso de 80 Anos ou mais , Asma/diagnóstico , Causalidade , Comorbidade , Depressão/diagnóstico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , República da Coreia/epidemiologia , Fatores de Risco , Distribuição por Sexo , Adulto Jovem
17.
Mol Genet Metab ; 110(3): 275-80, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23856420

RESUMO

Glycogen storage disease type-Ia (GSD-Ia) patients deficient in glucose-6-phosphatase-α (G6Pase-α or G6PC) manifest impaired glucose homeostasis characterized by fasting hypoglycemia, growth retardation, hepatomegaly, nephromegaly, hyperlipidemia, hyperuricemia, and lactic acidemia. Two efficacious recombinant adeno-associated virus pseudotype 2/8 (rAAV8) vectors expressing human G6Pase-α have been independently developed. One is a single-stranded vector containing a 2864-bp of the G6PC promoter/enhancer (rAAV8-GPE) and the other is a double-stranded vector containing a shorter 382-bp minimal G6PC promoter/enhancer (rAAV8-miGPE). To identify the best construct, a direct comparison of the rAAV8-GPE and the rAAV8-miGPE vectors was initiated to determine the best vector to take forward into clinical trials. We show that the rAAV8-GPE vector directed significantly higher levels of hepatic G6Pase-α expression, achieved greater reduction in hepatic glycogen accumulation, and led to a better toleration of fasting in GSD-Ia mice than the rAAV8-miGPE vector. Our results indicated that additional control elements in the rAAV8-GPE vector outweigh the gains from the double-stranded rAAV8-miGPE transduction efficiency, and that the rAAV8-GPE vector is the current choice for clinical translation in human GSD-Ia.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/genética , Regiões Promotoras Genéticas , Animais , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glucose/metabolismo , Doença de Depósito de Glicogênio Tipo I/metabolismo , Humanos , Fígado/metabolismo , Metaboloma , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Transdução Genética , Transgenes
18.
Hepatology ; 56(5): 1719-29, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22422504

RESUMO

UNLABELLED: Glycogen storage disease type Ia (GSD-Ia), which is characterized by impaired glucose homeostasis and chronic risk of hepatocellular adenoma (HCA), is caused by deficiencies in the endoplasmic reticulum (ER)-associated glucose-6-phosphatase-α (G6Pase-α or G6PC) that hydrolyzes glucose-6-phosphate (G6P) to glucose. G6Pase-α activity depends on the G6P transporter (G6PT) that translocates G6P from the cytoplasm into the ER lumen. The functional coupling of G6Pase-α and G6PT maintains interprandial glucose homeostasis. We have shown previously that gene therapy mediated by AAV-GPE, an adeno-associated virus (AAV) vector expressing G6Pase-α directed by the human G6PC promoter/enhancer (GPE), completely normalizes hepatic G6Pase-α deficiency in GSD-Ia (G6pc(-/-) ) mice for at least 24 weeks. However, a recent study showed that within 78 weeks of gene deletion, all mice lacking G6Pase-α in the liver develop HCA. We now show that gene therapy mediated by AAV-GPE maintains efficacy for at least 70-90 weeks for mice expressing more than 3% of wild-type hepatic G6Pase-α activity. The treated mice displayed normal hepatic fat storage, had normal blood metabolite and glucose tolerance profiles, had reduced fasting blood insulin levels, maintained normoglycemia over a 24-hour fast, and had no evidence of hepatic abnormalities. After a 24-hour fast, hepatic G6PT messenger RNA levels in G6pc(-/-) mice receiving gene therapy were markedly increased. Because G6PT transport is the rate-limiting step in microsomal G6P metabolism, this may explain why the treated G6pc(-/-) mice could sustain prolonged fasts. The low fasting blood insulin levels and lack of hepatic steatosis may explain the absence of HCA. CONCLUSION: These results confirm that AAV-GPE-mediated gene transfer corrects hepatic G6Pase-α deficiency in murine GSD-Ia and prevents chronic HCA formation.


Assuntos
Adenoma/prevenção & controle , Terapia Genética , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/enzimologia , Doença de Depósito de Glicogênio Tipo I/terapia , Neoplasias Hepáticas/prevenção & controle , Fígado/metabolismo , Animais , Antiporters/genética , Antiporters/metabolismo , Glicemia , Índice de Massa Corporal , Peso Corporal , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Terapia Genética/efeitos adversos , Vetores Genéticos , Teste de Tolerância a Glucose , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio Tipo I/genética , Homeostase , Insulina/sangue , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo
19.
Blood ; 117(14): 3881-92, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21292774

RESUMO

G6PC3 (or glucose-6-phosphatase-ß) deficiency underlies a congenital neutropenia syndrome in which neutrophils exhibit enhanced endoplasmic reticulum (ER) stress, increased apoptosis, impaired energy homeostasis, and impaired functionality. Here we show that murine G6pc3(-/-) neutrophils undergoing ER stress activate protein kinase-like ER kinase and phosphatidylinositol 3,4,5-trisphosphate/Akt signaling pathways, and that neutrophil apoptosis is mediated in part by the intrinsic mitochondrial pathway. In G6PC3-deficient patients, granulocyte colony-stimulating factor (G-CSF) improves neutropenia, but its impact on neutrophil apoptosis and dysfunction is unknown. We now show that G-CSF delays neutrophil apoptosis in vitro by modulating apoptotic mediators. However, G6pc3(-/-) neutrophils in culture exhibit accelerated apoptosis compared with wild-type neutrophils both in the presence or absence of G-CSF. Limiting glucose (0.6mM) accelerates apoptosis but is more pronounced for wild-type neutrophils, leading to similar survival profiles for both neutrophil populations. In vivo G-CSF therapy completely corrects neutropenia and normalizes levels of p-Akt, phosphatidylinositol 3,4,5-trisphosphate, and active caspase-3. Neutrophils from in vivo G-CSF-treated G6pc3(-/-) mice exhibit increased glucose uptake and elevated intracellular levels of G6P, lactate, and adenosine-5'-triphosphate, leading to improved functionality. Together, the results strongly suggest that G-CSF improves G6pc3(-/-) neutrophil survival by modulating apoptotic mediators and rectifies function by enhancing energy homeostasis.


Assuntos
Apoptose/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Glucose-6-Fosfatase/genética , Fator Estimulador de Colônias de Granulócitos/farmacologia , Neutrófilos/efeitos dos fármacos , Animais , Apoptose/genética , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Metabolismo Energético/genética , Glucose-6-Fosfatase/fisiologia , Homeostase/efeitos dos fármacos , Homeostase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/fisiologia , Fosfatos de Fosfatidilinositol/metabolismo , Subunidades Proteicas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , eIF-2 Quinase/metabolismo , eIF-2 Quinase/fisiologia
20.
Blood ; 116(15): 2783-92, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20498302

RESUMO

G6PC3 deficiency, characterized by neutropenia and neutrophil dysfunction, is caused by deficiencies in the endoplasmic reticulum (ER) enzyme glucose-6-phosphatase-ß (G6Pase-ß or G6PC3) that converts glucose-6-phosphate (G6P) into glucose, the primary energy source of neutrophils. Enhanced neutrophil ER stress and apoptosis underlie neutropenia in G6PC3 deficiency, but the exact functional role of G6Pase-ß in neutrophils remains unknown. We hypothesized that the ER recycles G6Pase-ß-generated glucose to the cytoplasm, thus regulating the amount of available cytoplasmic glucose/G6P in neutrophils. Accordingly, a G6Pase-ß deficiency would impair glycolysis and hexose monophosphate shunt activities leading to reductions in lactate production, adenosine-5'-triphosphate (ATP) production, and reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity. Using annexin V-depleted neutrophils, we show that glucose transporter-1 translocation is impaired in neutrophils from G6pc3(-/-) mice and G6PC3-deficient patients along with impaired glucose uptake in G6pc3(-/-) neutrophils. Moreover, levels of G6P, lactate, and ATP are markedly lower in murine and human G6PC3-deficient neutrophils, compared with their respective controls. In parallel, the expression of NADPH oxidase subunits and membrane translocation of p47(phox) are down-regulated in murine and human G6PC3-deficient neutrophils. The results establish that in nonapoptotic neutrophils, G6Pase-ß is essential for normal energy homeostasis. A G6Pase-ß deficiency prevents recycling of ER glucose to the cytoplasm, leading to neutrophil dysfunction.


Assuntos
Glucose-6-Fosfatase/metabolismo , Glucose/metabolismo , Doença de Depósito de Glicogênio Tipo I/metabolismo , Neutropenia/congênito , Neutropenia/metabolismo , Neutrófilos/metabolismo , Trifosfato de Adenosina/metabolismo , Adolescente , Animais , Anexina A5/metabolismo , Apoptose , Caspase 3/metabolismo , Criança , Citoplasma/metabolismo , Retículo Endoplasmático/metabolismo , Feminino , Transportador de Glucose Tipo 1/metabolismo , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/patologia , Humanos , Ácido Láctico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidases/metabolismo , Neutropenia/genética , Neutropenia/patologia , Neutrófilos/patologia , Estresse Fisiológico , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA