Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Pediatr Neurol ; 157: 118-126, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38914025

RESUMEN

BACKGROUND: Since the initial description of glucose transporter-1 deficiency syndrome (Glut1-DS) the phenotype of the condition has expanded, even leading to the recognition of atypical manifestations. We report on eight patients with Glut1-DS who experienced at least one episode of acute focal neurological deficits. METHODS: We conducted a retrospective analysis, collecting clinical, electrophysiological, neuroradiological, and genetic information. We focused in particular on three well-documented cases. RESULTS: Among 42 patients with Glut1-DS, eight individuals aged between six and 38 years presented with an acute onset of neurological disturbances: dysarthria/aphasia, oral dyskinesia, swallowing difficulties, paresthesia, facial palsy, hemi/monoplegia, vomiting, headache, and behavioral disturbances. When performed, magnetic resonance imaging (MRI) revealed signs of venous congestion and hypoperfusion and electroencephalography showed focal contralateral slowing. Deficits were transient in all patients but one. Four patients (50%) were on a ketogenic diet (KD), and two of these patients had lower than usual ketonemia levels during the episode. In two patients, MRI demonstrated the presence of an ischemic brain lesion. CONCLUSIONS: In Glut1-DS, stroke-like episodes are a recurrent manifestation, particularly during early adulthood, and they were reported in 19% of the patients in our cohort. Stroke mimics should be considered a key feature of Glut1-DS, as other paroxysmal disorders. It remains to be established whether a KD can prevent the recurrence of episodes and, if so, at what level of ketosis. Further observations are needed to confirm the correlation between Glut1-DS and ischemic stroke.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos , Proteínas de Transporte de Monosacáridos , Accidente Cerebrovascular , Humanos , Masculino , Femenino , Niño , Errores Innatos del Metabolismo de los Carbohidratos/complicaciones , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Errores Innatos del Metabolismo de los Carbohidratos/fisiopatología , Adulto , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/genética , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/diagnóstico por imagen , Adolescente , Estudios Retrospectivos , Adulto Joven , Recurrencia , Imagen por Resonancia Magnética , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/genética , Electroencefalografía , Encéfalo/diagnóstico por imagen , Encéfalo/patología
2.
Eur J Neurol ; 31(8): e16325, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38803061

RESUMEN

BACKGROUND AND PURPOSE: Glucose transporter-1 (GLUT1) deficiency syndrome (GLUT1-DS) is a metabolic disorder due to reduced expression of GLUT1, a glucose transporter of the central nervous system. GLUT1-DS is caused by heterozygous SLC2A1 variants that mostly arise de novo. Here, we report a large family with heterogeneous phenotypes related to a novel SLC2A1 variant. METHODS: We present clinical and genetic features of a five-generation family with GLUT1-DS. RESULTS: The 14 (nine living) affected members had heterogeneous phenotypes, including seizures (11/14), behavioral disturbances (5/14), mild intellectual disability (3/14), and/or gait disabilities (2/14). Brain magnetic resonance imaging revealed hippocampal sclerosis in the 8-year-old proband, who also had drug-responsive absences associated with attention-deficit/hyperactivity disorder. His 52-year-old father, who had focal epilepsy since childhood, developed paraparesis related to a reversible myelitis associated with hypoglycorrhachia. Molecular study detected a novel heterozygous missense variant (c.446C>T) in exon 4 of SLC2A1 (NM: 006516.2) that cosegregated with the illness. This variant causes an amino acid replacement (p.Pro149Leu) at the fourth transmembrane segment of GLUT1, an important domain located at its catalytic core. CONCLUSIONS: Our study illustrates the extremely heterogenous phenotypes in familial GLUT1-DS, ranging from milder classic phenotypes to more subtle neurological disorder including paraparesis. This novel SLC2A1 variant (c.446C>T) provides new insight into the pathophysiology of GLUT1-DS.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos , Transportador de Glucosa de Tipo 1 , Linaje , Fenotipo , Humanos , Masculino , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/deficiencia , Femenino , Niño , Persona de Mediana Edad , Errores Innatos del Metabolismo de los Carbohidratos/genética , Adulto , Adolescente , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/deficiencia , Mutación Missense/genética , Preescolar , Imagen por Resonancia Magnética , Adulto Joven
3.
Orphanet J Rare Dis ; 18(1): 63, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36944981

RESUMEN

BACKGROUND: GLUT1 deficiency syndrome is a rare, genetically determined neurological disorder for which Ketogenic Dietary Treatment represents the gold standard and lifelong treatment. Patient registries are powerful tools providing insights and real-world data on rare diseases. OBJECTIVE: To describe the implementation of a national web-based registry for GLUT1-DS. METHODS: This is a retrospective and prospective, multicenter, observational registry developed in collaboration with the Italian GLUT1-DS association and based on an innovative, flexible and configurable cloud computing technology platform, structured according to the most rigorous requirements for the management of patient's sensitive data. The Glut1 Registry collects baseline and follow-up data on the patient's demographics, history, symptoms, genotype, clinical, and instrumental evaluations and therapies. RESULTS: Five Centers in Italy joined the registry, and two more Centers are currently joining. In the first two years of running, data from 67 patients (40 females and 27 males) have been collected. Age at symptom onset was within the first year of life in most (40, 60%) patients. The diagnosis was formulated in infancy in almost half of the cases (34, 51%). Symptoms at onset were mainly paroxysmal (mostly epileptic seizure and paroxysmal ocular movement disorder) or mixed paroxysmal and fixed symptoms (mostly psychomotor delay). Most patients (53, 79%) are currently under Ketogenic dietary treatments. CONCLUSIONS: We describe the principles behind the design, development, and deployment of the web-based nationwide GLUT1-DS registry. It represents a stepping stone towards a more comprehensive understanding of the disease from onset to adulthood. It also represents a virtuous model from a technical, legal, and organizational point of view, thus representing a possible paradigmatic example for other rare disease registry implementation.


Asunto(s)
Transportador de Glucosa de Tipo 1 , Enfermedades Raras , Femenino , Humanos , Masculino , Transportador de Glucosa de Tipo 1/deficiencia , Italia , Estudios Prospectivos , Sistema de Registros , Estudios Retrospectivos , Lactante
5.
Genes (Basel) ; 12(9)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34573360

RESUMEN

Glucose transporter type 1 (GLUT1) is the most important energy carrier of the brain across the blood-brain barrier, and a genetic defect of GLUT1 is known as GLUT1 deficiency syndrome (GLUT1DS). It is characterized by early infantile seizures, developmental delay, microcephaly, ataxia, and various paroxysmal neurological phenomena. In most cases, GLUT1DS is caused by heterozygous single-nucleotide variants (SNVs) in the SLC2A1 gene that provoke complete or severe impairment of the functionality and/or expression of GLUT1 in the brain. Despite the rarity of these diseases, GLUT1DS is of high clinical interest since a very effective therapy, the ketogenic diet, can improve or reverse symptoms, especially if it is started as early as possible. We present a clinical phenotype, biochemical analysis, electroencephalographic and neuropsychological features of an 11-month-old boy with myoclonic seizures, hypogammaglobulinemia, and mildly impaired gross motor development. Using sequence analysis and deletion/duplication testing, deletion of an entire coding sequence in the SLC2A1 gene was detected. Early introduction of a modified Atkins diet maintained a seizure-free period without antiseizure medications and normal cognitive development in the follow-up period. Our report summarizes the clinical features of GLUT1 syndromes and discusses the importance of early identification and molecular confirmation of GLUT1DS as a treatable metabolic disorder.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/dietoterapia , Disfunción Cognitiva/prevención & control , Discapacidades del Desarrollo/prevención & control , Dieta Cetogénica , Proteínas de Transporte de Monosacáridos/deficiencia , Tiempo de Tratamiento , Errores Innatos del Metabolismo de los Carbohidratos/complicaciones , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Errores Innatos del Metabolismo de los Carbohidratos/genética , Desarrollo Infantil , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/genética , Análisis Mutacional de ADN , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/genética , Humanos , Lactante , Masculino , Proteínas de Transporte de Monosacáridos/genética , Factores de Tiempo , Resultado del Tratamiento
6.
Cancer Res ; 81(9): 2345-2357, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33753374

RESUMEN

Neutrophils are the most abundant circulating leucocytes and are essential for innate immunity. In cancer, pro- or antitumor properties have been attributed to tumor-associated neutrophils (TAN). Here, focusing on TAN accumulation within lung tumors, we identify GLUT1 as an essential glucose transporter for their tumor supportive behavior. Compared with normal neutrophils, GLUT1 and glucose metabolism increased in TANs from a mouse model of lung adenocarcinoma. To elucidate the impact of glucose uptake on TANs, we used a strategy with two recombinases, dissociating tumor initiation from neutrophil-specific Glut1 deletion. Loss of GLUT1 accelerated neutrophil turnover in tumors and reduced a subset of TANs expressing SiglecF. In the absence of GLUT1 expression by TANs, tumor growth was diminished and the efficacy of radiotherapy was augmented. Our results demonstrate the importance of GLUT1 in TANs, which may affect their pro- versus antitumor behavior. These results also suggest targeting metabolic vulnerabilities to favor antitumor neutrophils. SIGNIFICANCE: Lung tumor support and radiotherapy resistance depend on GLUT1-mediated glucose uptake in tumor-associated neutrophils, indicating that metabolic vulnerabilities should be considered to target both tumor cells as well as innate immune cells. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/9/2345/F1.large.jpg.


Asunto(s)
Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/radioterapia , Proliferación Celular/genética , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/radioterapia , Neutrófilos/inmunología , Insuficiencia del Tratamiento , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Transportador de Glucosa de Tipo 1/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
7.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33351789

RESUMEN

Paucity of the glucose transporter-1 (Glut1) protein resulting from haploinsufficiency of the SLC2A1 gene arrests cerebral angiogenesis and disrupts brain function to cause Glut1 deficiency syndrome (Glut1 DS). Restoring Glut1 to Glut1 DS model mice prevents disease, but the precise cellular sites of action of the transporter, its temporal requirements, and the mechanisms linking scarcity of the protein to brain cell dysfunction remain poorly understood. Here, we show that Glut1 functions in a cell-autonomous manner in the cerebral microvasculature to affect endothelial tip cells and, thus, brain angiogenesis. Moreover, brain endothelial cell-specific Glut1 depletion not only triggers a severe neuroinflammatory response in the Glut1 DS brain, but also reduces levels of brain-derived neurotrophic factor (BDNF) and causes overt disease. Reduced BDNF correlated with fewer neurons in the Glut1 DS brain. Controlled depletion of the protein demonstrated that brain pathology and disease severity was greatest when Glut1 scarcity was induced neonatally, during brain angiogenesis. Reducing Glut1 at later stages had mild or little effect. Our results suggest that targeting brain endothelial cells during early development is important to ensure proper brain angiogenesis, prevent neuroinflammation, maintain BDNF levels, and preserve neuron numbers. This requirement will be essential for any disease-modifying therapeutic strategy for Glut1 DS.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/metabolismo , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/metabolismo , Proteínas de Transporte de Monosacáridos/deficiencia , Animales , Animales Recién Nacidos , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Errores Innatos del Metabolismo de los Carbohidratos/genética , Errores Innatos del Metabolismo de los Carbohidratos/patología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Técnicas de Silenciamiento del Gen , Transportador de Glucosa de Tipo 1/genética , Haploinsuficiencia , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Neovascularización Fisiológica/genética , Neuronas/metabolismo , Neuronas/patología , Fenotipo
8.
J Cereb Blood Flow Metab ; 41(5): 1026-1038, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32703112

RESUMEN

Isolated brain capillaries are essential for analyzing the changes of protein expressions at the blood-brain barrier (BBB) under pathological conditions. The standard brain capillary isolation methods require the use of at least five mouse brains in order to obtain a sufficient amount and purity of brain capillaries. The purpose of this study was to establish a brain capillary isolation method from a single mouse brain for protein expression analysis. We successfully isolated brain capillaries from a single frozen mouse brain by using a bead homogenizer in the brain homogenization step and combination of cell strainers and glass beads in the purification step. Western blot and proteomic analysis showed that proteins expressed at the BBB in mouse brain capillaries isolated by the developed method were more enriched than those isolated from a pool of five mouse brains by the standard method. By using the developed method, we further verified the changes in expression of BBB proteins in Glut1-deficient mouse. The developed method is useful for the analysis of various mice models with low numbers and enables us to understand, in more detail, the physiology and pathology of BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Capilares/metabolismo , Proteómica/métodos , Animales , Transporte Biológico/fisiología , Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Encéfalo/cirugía , Encéfalo/ultraestructura , Errores Innatos del Metabolismo de los Carbohidratos/metabolismo , Modelos Animales de Enfermedad , Congelación , Ontología de Genes/estadística & datos numéricos , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/metabolismo , Preservación de Órganos/métodos , Proteómica/estadística & datos numéricos
9.
Wiley Interdiscip Rev Syst Biol Med ; 12(4): e1483, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32084302

RESUMEN

Knowledge about metabolism of immune cells increased almost exponentially during the last two decades and thereby created the new area immunometabolism. Increased glucose uptake and glycolysis were identified as one of the major drivers in immune cells for rapid adaptation to changes in the microenvironment or external stimuli. These metabolic switches are crucial to generate macromolecules for immune cell proliferation and activation. Glucose transporter 1 (GLUT1), a ubiquitously expressed glucose transporter, is strongly upregulated after innate and adaptive immune cell activation. Deletion or inhibition of GLUT1 blocked T cell proliferation and effector function, antibody production from B cells and reduced inflammatory responses in macrophages. Increased glucose uptake and GLUT1 expression are not only observed in proinflammatory conditions, but also in murine models of autoimmunity as well as in human patients. Rheumatoid arthritis (RA), the most common autoimmune disease, is characterized by infiltration of immune cells, hyperproliferation of fibroblast-like synoviocytes, and destruction of cartilage and bone. These processes create a hypoxic microenvironment in the synovium. Moreover, synovial samples including fibroblast-like synoviocytes from RA patients showed increased lactate level and upregulate GLUT1. Similar upregulation of GLUT1 is observed in systemic lupus erythematosus and psoriasis patients as well as in murine autoimmune models. Inhibition of GLUT1 using either T cell specific knockouts or small molecule GLUT1/glycolysis inhibitors improved phenotypes of different murine autoimmune disease models like arthritis, lupus, and psoriasis. Thereby the therapeutic potential of immunometabolism and especially interference with glycolysis was proven. This article is categorized under: Biological Mechanisms > Metabolism Translational, Genomic, and Systems Medicine > Translational Medicine Physiology > Mammalian Physiology in Health and Disease.


Asunto(s)
Artritis Reumatoide/patología , Autoinmunidad , Transportador de Glucosa de Tipo 1/metabolismo , Animales , Artritis Reumatoide/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/genética , Glucólisis , Humanos , Sinoviocitos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Hum Brain Mapp ; 41(2): 453-466, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31710770

RESUMEN

Glucose transporter type I deficiency syndrome (GLUT1DS) is an encephalopathic disorder due to a chronic insufficient transport of glucose into the brain. PET studies in GLUT1DS documented a widespread cortico-thalamic hypometabolism and a signal increase in the basal ganglia, regardless of age and clinical phenotype. Herein, we captured the pattern of functional connectivity of distinct striatal, cortical, and cerebellar regions in GLUT1DS (10 children, eight adults) and in healthy controls (HC, 19 children, 17 adults) during rest. Additionally, we explored for regional connectivity differences in GLUT1 children versus adults and according to the clinical presentation. Compared to HC, GLUT1DS exhibited increase connectivity within the basal ganglia circuitries and between the striatal regions with the frontal cortex and cerebellum. The excessive connectivity was predominant in patients with movement disorders and in children compared to adults, suggesting a correlation with the clinical phenotype and age at fMRI study. Our findings highlight the primary role of the striatum in the GLUT1DS pathophysiology and confirm the dependency of symptoms to the patients' chronological age. Despite the reduced chronic glucose uptake, GLUT1DS exhibit increased connectivity changes in regions highly sensible to glycopenia. Our results may portrait the effect of neuroprotective brain strategy to overcome the chronic poor energy supply during vulnerable ages.


Asunto(s)
Ganglios Basales , Encefalopatías Metabólicas Innatas , Cerebelo , Transportador de Glucosa de Tipo 1/deficiencia , Desarrollo Humano , Red Nerviosa , Neuroprotección , Corteza Prefrontal , Adolescente , Adulto , Ganglios Basales/diagnóstico por imagen , Ganglios Basales/metabolismo , Ganglios Basales/fisiopatología , Encefalopatías Metabólicas Innatas/diagnóstico por imagen , Encefalopatías Metabólicas Innatas/genética , Encefalopatías Metabólicas Innatas/metabolismo , Encefalopatías Metabólicas Innatas/fisiopatología , Cerebelo/diagnóstico por imagen , Cerebelo/metabolismo , Cerebelo/fisiopatología , Niño , Enfermedad Crónica , Epilepsia/diagnóstico por imagen , Epilepsia/etiología , Epilepsia/metabolismo , Epilepsia/fisiopatología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Trastornos del Movimiento/diagnóstico por imagen , Trastornos del Movimiento/etiología , Trastornos del Movimiento/metabolismo , Trastornos del Movimiento/fisiopatología , Red Nerviosa/diagnóstico por imagen , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Adulto Joven
12.
Nutrients ; 11(10)2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31581549

RESUMEN

Diseases involving inflammation and oxidative stress can be exacerbated by high blood glucose levels. Due to tight metabolic regulation, safely reducing blood glucose can prove difficult. The ketogenic diet (KD) reduces absolute glucose and insulin, while increasing fatty acid oxidation, ketogenesis, and circulating levels of ß-hydroxybutyrate (ßHB), acetoacetate (AcAc), and acetone. Compliance to KD can be difficult, so alternative therapies that help reduce glucose levels are needed. Exogenous ketones provide an alternative method to elevate blood ketone levels without strict dietary requirements. In this study, we tested the changes in blood glucose and ketone (ßHB) levels in response to acute, sub-chronic, and chronic administration of various ketogenic compounds in either a post-exercise or rested state. WAG/Rij (WR) rats, a rodent model of human absence epilepsy, GLUT1 deficiency syndrome mice (GLUT1D), and wild type Sprague Dawley rats (SPD) were assessed. Non-pathological animals were also assessed across different age ranges. Experimental groups included KD, standard diet (SD) supplemented with water (Control, C) or with exogenous ketones: 1, 3-butanediol (BD), ßHB mineral salt (KS), KS with medium chain triglyceride/MCT (KSMCT), BD acetoacetate diester (KE), KE with MCT (KEMCT), and KE with KS (KEKS). In rested WR rats, the KE, KS, KSMCT groups had lower blood glucose level after 1 h of treatment, and in KE and KSMCT groups after 24 h. After exercise, the KE, KSMCT, KEKS, and KEMCT groups had lowered glucose levels after 1 h, and in the KEKS and KEMCT groups after 7 days, compared to control. In GLUT1D mice without exercise, only KE resulted in significantly lower glucose levels at week 2 and week 6 during a 10 weeks long chronic feeding study. In 4-month and 1-year-old SPD rats in the post-exercise trials, blood glucose was significantly lower in KD and KE, and in KEMCT groups, respectively. After seven days, the KSMCT group had the most significantly reduced blood glucose levels, compared to control. These results indicate that exogenous ketones were efficacious in reducing blood glucose levels within and outside the context of exercise in various rodent models of different ages, with and without pathology.


Asunto(s)
Ácido 3-Hidroxibutírico/farmacología , Acetoacetatos/farmacología , Glucemia/efectos de los fármacos , Butileno Glicoles/farmacología , Errores Innatos del Metabolismo de los Carbohidratos/terapia , Dieta Cetogénica , Suplementos Dietéticos , Epilepsia Tipo Ausencia/terapia , Proteínas de Transporte de Monosacáridos/deficiencia , Animales , Biomarcadores , Glucemia/metabolismo , Errores Innatos del Metabolismo de los Carbohidratos/sangre , Errores Innatos del Metabolismo de los Carbohidratos/genética , Errores Innatos del Metabolismo de los Carbohidratos/fisiopatología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Epilepsia Tipo Ausencia/sangre , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/fisiopatología , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/genética , Masculino , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/sangre , Proteínas de Transporte de Monosacáridos/genética , Esfuerzo Físico , Ratas Sprague-Dawley , Descanso , Factores de Tiempo
15.
FASEB J ; 33(7): 7810-7821, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30913395

RESUMEN

Wingless/integrated (Wnt) signaling has emerged as a major mechanism for promoting bone formation and a target pathway for developing bone anabolic agents against osteoporosis. However, the downstream events mediating the potential therapeutic effect of Wnt proteins are not fully understood. Previous studies have indicated that increased glycolysis is associated with osteoblast differentiation in response to Wnt signaling, but direct genetic evidence for the importance of glucose metabolism in Wnt-induced bone formation is lacking. Here, we have generated compound transgenic mice to overexpress Wnt family member 7B (Wnt7b) transiently in the osteoblast lineage of postnatal mice, with or without concurrent deletion of the glucose transporter 1 (Glut1), also known as solute carrier family 2, facilitated glucose transporter member 1. Overexpression of Wnt7b in 1-mo-old mice for 1 wk markedly stimulated bone formation, but the effect was essentially abolished without Glut1, even though transient deletion of Glut1 itself did not affect normal bone accrual. Consistent with the in vivo results, Wnt7b increased Glut1 expression and glucose consumption in the primary culture of osteoblast lineage cells, and deletion of Glut1 diminished osteoblast differentiation in vitro. Thus, Wnt7b promotes bone formation in part through stimulating glucose metabolism in osteoblast lineage cells.-Chen, H., Ji, X., Lee, W.-C., Shi, Y., Li, B., Abel, E. D., Jiang, D., Huang, W., Long, F. Increased glycolysis mediates Wnt7b-induced bone formation.


Asunto(s)
Transportador de Glucosa de Tipo 1/fisiología , Glucosa/metabolismo , Glucólisis , Osteoblastos/metabolismo , Osteogénesis/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Wnt/fisiología , Animales , Linaje de la Célula , Células Cultivadas , Fémur/crecimiento & desarrollo , Fémur/ultraestructura , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/genética , Ratones , Ratones Transgénicos , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Recombinantes/metabolismo , Tamoxifeno/farmacología , Tibia/crecimiento & desarrollo , Tibia/ultraestructura , Proteínas Wnt/genética
16.
J Immunol ; 202(4): 1265-1286, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30659108

RESUMEN

Macrophages (MΦs) are heterogeneous and metabolically flexible, with metabolism strongly affecting immune activation. A classic response to proinflammatory activation is increased flux through glycolysis with a downregulation of oxidative metabolism, whereas alternative activation is primarily oxidative, which begs the question of whether targeting glucose metabolism is a viable approach to control MΦ activation. We created a murine model of myeloid-specific glucose transporter GLUT1 (Slc2a1) deletion. Bone marrow-derived MΦs (BMDM) from Slc2a1M-/- mice failed to uptake glucose and demonstrated reduced glycolysis and pentose phosphate pathway activity. Activated BMDMs displayed elevated metabolism of oleate and glutamine, yet maximal respiratory capacity was blunted in MΦ lacking GLUT1, demonstrating an incomplete metabolic reprogramming. Slc2a1M-/- BMDMs displayed a mixed inflammatory phenotype with reductions of the classically activated pro- and anti-inflammatory markers, yet less oxidative stress. Slc2a1M-/- BMDMs had reduced proinflammatory metabolites, whereas metabolites indicative of alternative activation-such as ornithine and polyamines-were greatly elevated in the absence of GLUT1. Adipose tissue MΦs of lean Slc2a1M-/- mice had increased alternative M2-like activation marker mannose receptor CD206, yet lack of GLUT1 was not a critical mediator in the development of obesity-associated metabolic dysregulation. However, Ldlr-/- mice lacking myeloid GLUT1 developed unstable atherosclerotic lesions. Defective phagocytic capacity in Slc2a1M-/- BMDMs may have contributed to unstable atheroma formation. Together, our findings suggest that although lack of GLUT1 blunted glycolysis and the pentose phosphate pathway, MΦ were metabolically flexible enough that inflammatory cytokine release was not dramatically regulated, yet phagocytic defects hindered MΦ function in chronic diseases.


Asunto(s)
Modelos Animales de Enfermedad , Transportador de Glucosa de Tipo 1/metabolismo , Macrófagos/metabolismo , Animales , Transportador de Glucosa de Tipo 1/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
17.
Nat Commun ; 9(1): 4831, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30446646

RESUMEN

Much of the mammalian skeleton originates from a cartilage template eventually replaced by bone via endochondral ossification. Despite much knowledge about growth factors and nuclear proteins in skeletal development, little is understood about the role of metabolic regulation. Here we report that genetic deletion of the glucose transporter Glut1 (Slc2a1), either before or after the onset of chondrogenesis in the limb, severely impairs chondrocyte proliferation and hypertrophy, resulting in dramatic shortening of the limbs. The cartilage defects are reminiscent to those caused by deficiency in Bmp signaling. Importantly, deletion of Bmpr1a in chondrocytes markedly reduces Glut1 levels in vivo, whereas recombinant BMP2 increases Glut1 mRNA and protein levels, boosting glucose metabolism in primary chondrocytes. Biochemical studies identify a Bmp-mTORC1-Hif1a signaling cascade resulting in upregulation of Glut1 in chondrocytes. The results therefore uncover a hitherto unknown connection between Bmp signaling and glucose metabolism in the regulation of cartilage development.


Asunto(s)
Proteína Morfogenética Ósea 1/genética , Huesos/metabolismo , Transportador de Glucosa de Tipo 1/genética , Glucosa/metabolismo , Osteogénesis/genética , Transducción de Señal/genética , Animales , Animales Recién Nacidos , Proteína Morfogenética Ósea 1/deficiencia , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/deficiencia , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Huesos/citología , Cartílago/citología , Cartílago/crecimiento & desarrollo , Cartílago/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrogénesis/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Transportador de Glucosa de Tipo 1/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Cultivo Primario de Células
18.
BMC Anesthesiol ; 18(1): 85, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-30021521

RESUMEN

BACKGROUND: Ketogenic diet (KD) and exogenous ketone supplements can evoke sustained ketosis, which may modulate sleep and sleep-like effects. However, no studies have been published examining the effect of ketosis on the onset of general isoflurane induced anesthesia. Therefore, we investigated the effect of the KD and different exogenous ketogenic supplements on the onset of akinesia induced by inhalation of isoflurane. METHODS: We used a high fat, medium protein and low carbohydrate diet (KD) chronically (10 weeks) in the glucose transporter 1 (GLUT1) deficiency (G1D) syndrome mice model and sub-chronically (7 days) in Sprague-Dawley (SPD) rats. To investigate the effect of exogenous ketone supplements on anesthetic induction we also provided either 1) a standard rodent chow diet (SD) mixed with 20% ketone salt supplement (KS), or 2) SD mixed with 20% ketone ester supplement (KE; 1,3 butanediol-acetoacetate diester) to G1D mice for 10 weeks. Additionally, SPD rats and Wistar Albino Glaxo Rijswijk (WAG/Rij) rats were fed the SD, which was supplemented by oral gavage of KS or KE for 7 days (SPD rats: 5 g/kg body weight/day; WAG/Rij rats: 2.5 g/kg body weight/day). After these treatments (10 weeks for the mice, and 7 days for the rats) isoflurane (3%) was administered in an anesthesia chamber, and the time until anesthetic induction (time to immobility) was measured. Blood ketone levels were measured after anesthetic induction and correlation was calculated for blood beta-hydroxybutyrate (ßHB) and anesthesia latency. RESULTS: Both KD and exogenous ketone supplementation increased blood ketone levels and delayed the onset of isoflurane-induced immobility in all investigated rodent models, showing positive correlation between the two measurements. These results demonstrate that elevated blood ketone levels by either KD or exogenous ketones delayed the onset of isoflurane-induced anesthesia in these animal models. CONCLUSIONS: These findings suggest that ketone levels might affect surgical anesthetic needs, or could potentially decrease or delay effects of other narcotic gases.


Asunto(s)
Anestesia/estadística & datos numéricos , Interacciones Alimento-Droga , Isoflurano/farmacología , Cetosis/inducido químicamente , Ácido 3-Hidroxibutírico/sangre , Administración por Inhalación , Animales , Dieta Cetogénica/efectos adversos , Transportador de Glucosa de Tipo 1/deficiencia , Isoflurano/administración & dosificación , Cetonas/sangre , Cetonas/farmacología , Cetosis/sangre , Masculino , Ratones , Ratas , Especificidad de la Especie , Factores de Tiempo
19.
Nat Med ; 24(5): 617-627, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29662201

RESUMEN

Proliferating cells, compared with quiescent cells, are more dependent on glucose for their growth. Although glucose transport in keratinocytes is mediated largely by the Glut1 facilitative transporter, we found that keratinocyte-specific ablation of Glut1 did not compromise mouse skin development and homeostasis. Ex vivo metabolic profiling revealed altered sphingolipid, hexose, amino acid, and nucleotide metabolism in Glut1-deficient keratinocytes, thus suggesting metabolic adaptation. However, cultured Glut1-deficient keratinocytes displayed metabolic and oxidative stress and impaired proliferation. Similarly, Glut1 deficiency impaired in vivo keratinocyte proliferation and migration within wounded or UV-damaged mouse skin. Notably, both genetic and pharmacological Glut1 inactivation decreased hyperplasia in mouse models of psoriasis-like disease. Topical application of a Glut1 inhibitor also decreased inflammation in these models. Glut1 inhibition decreased the expression of pathology-associated genes in human psoriatic skin organoids. Thus, Glut1 is selectively required for injury- and inflammation-associated keratinocyte proliferation, and its inhibition offers a novel treatment strategy for psoriasis.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Psoriasis/terapia , Piel/lesiones , Piel/metabolismo , Animales , Transporte Biológico/efectos de la radiación , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Cultivadas , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Eliminación de Gen , Transportador de Glucosa de Tipo 1/deficiencia , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Ratones Endogámicos C57BL , Oxidación-Reducción , Psoriasis/patología , Piel/patología , Estrés Fisiológico , Rayos Ultravioleta
20.
Int J Mol Sci ; 19(1)2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29303961

RESUMEN

Monogenic and polygenic mutations are important contributors in patients suffering from epilepsy, including metabolic epilepsies which are inborn errors of metabolism with a good respond to specific dietetic treatments. Heterozygous variation in solute carrier family 2, facilitated glucose transporter member 1 (SLC2A1) and mutations of the GLUT1/SLC2A2 gene results in the failure of glucose transport, which is related with a glucose type-1 transporter (GLUT1) deficiency syndrome (GLUT1DS). GLUT1 deficiency syndrome is a treatable disorder of glucose transport into the brain caused by a variety of mutations in the SLC2A1 gene which are the cause of different neurological disorders also with different types of epilepsy and related clinical phenotypes. Since patients continue to experience seizures due to a pharmacoresistance, an early clinical diagnosis associated with specific genetic testing in SLC2A1 pathogenic variants in clinical phenotypes could predict pure drug response and might improve safety and efficacy of treatment with the initiation of an alternative energy source including ketogenic or analog diets in such patients providing individualized strategy approaches.


Asunto(s)
Epilepsia/tratamiento farmacológico , Pruebas Genéticas/métodos , Transportador de Glucosa de Tipo 1/genética , Medicina de Precisión/métodos , Anticonvulsivantes/uso terapéutico , Epilepsia/diagnóstico , Epilepsia/genética , Transportador de Glucosa de Tipo 1/deficiencia , Humanos , Farmacogenética/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA