Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 12(4): e0174264, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28388629

RESUMO

Tubulin alpha 8 (Tuba8) is the most divergent member of the highly conserved alpha tubulin family, and uniquely lacks two key post-translational modification sites. It is abundantly expressed in testis and muscle, with lower levels in the brain. We previously identified homozygous hypomorphic TUBA8 mutations in human subjects with a polymicrogyria (PMG) syndrome, suggesting its involvement in development of the cerebral cortex. We have now generated and characterized a Tuba8 knockout mouse model. Homozygous mice were confirmed to lack Tuba8 protein in the testis, but did not display PMG and appeared to be neurologically normal. In response to this finding, we re-analyzed the human PMG subjects using whole exome sequencing. This resulted in identification of an additional homozygous loss-of-function mutation in SNAP29, suggesting that SNAP29 deficiency, rather than TUBA8 deficiency, may underlie most or all of the neurodevelopmental anomalies in these subjects. Nonetheless, in the mouse brain, Tuba8 specifically localised to the cerebellar Purkinje cells, suggesting that the human mutations may affect or modify motor control. In the testis, Tuba8 localisation was cell-type specific. It was restricted to spermiogenesis with a strong acrosomal localization that was gradually replaced by cytoplasmic distribution and was absent from spermatozoa. Although the knockout mice were fertile, the localisation pattern indicated that Tuba8 may have a role in spermatid development during spermatogenesis, rather than as a component of the mature microtubule-rich flagellum itself.


Assuntos
Encéfalo/embriologia , Espermatogênese/genética , Tubulina (Proteína)/genética , Animais , Exoma , Homozigoto , Camundongos , Camundongos Knockout
2.
Biochem J ; 466(3): 537-46, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25562606

RESUMO

Reactive oxygen species (ROS) can cause pancreatic ß-cell death by activating transient receptor potential (melastatin) 2 (TRPM2) channels. Cell death has been attributed to the ability of these channels to raise cytosolic Ca2+. Recent studies however revealed that TRPM2 channels can also conduct Zn2+, but the physiological relevance of this property is enigmatic. Given that Zn2+ is cytotoxic, we asked whether TRPM2 channels can permeate sufficient Zn2+ to affect cell viability. To address this, we used the insulin secreting (INS1) ß-cell line, human embryonic kidney (HEK)-293 cells transfected with TRPM2 and pancreatic islets. H2O2 activation of TRPM2 channels increases the cytosolic levels of both Ca2+ and Zn2+ and causes apoptotic cell death. Interestingly, chelation of Zn2+ alone was sufficient to prevent ß-cell death. The source of the cytotoxic Zn2+ is intracellular, found largely sequestered in lysosomes. Lysosomes express TRPM2 channels, providing a potential route for Zn2+ release. Zn2+ release is potentiated by extracellular Ca2+ entry, indicating that Ca2+-induced Zn2+ release leads to apoptosis. Knockout of TRPM2 channels protects mice from ß-cell death and hyperglycaemia induced by multiple low-dose streptozotocin (STZ; MLDS) administration. These results argue that TRPM2-mediated, Ca2+-potentiated Zn2+ release underlies ROS-induced ß-cell death and Zn2+, rather than Ca2+, plays a primary role in apoptosis.


Assuntos
Células Secretoras de Insulina/metabolismo , Líquido Intracelular/metabolismo , Canais de Cátion TRPM/fisiologia , Zinco/metabolismo , Animais , Morte Celular/fisiologia , Células HEK293 , Humanos , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo
3.
J Am Soc Nephrol ; 25(11): 2526-38, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24876114

RESUMO

Diabetes is associated with activation of the polyol pathway, in which glucose is converted to sorbitol by aldose reductase. Previous studies focused on the role of sorbitol in mediating diabetic complications. However, in the proximal tubule, sorbitol can be converted to fructose, which is then metabolized largely by fructokinase, also known as ketohexokinase, leading to ATP depletion, proinflammatory cytokine expression, and oxidative stress. We and others recently identified a potential deleterious role of dietary fructose in the generation of tubulointerstitial injury and the acceleration of CKD. In this study, we investigated the potential role of endogenous fructose production, as opposed to dietary fructose, and its metabolism through fructokinase in the development of diabetic nephropathy. Wild-type mice with streptozotocin-induced diabetes developed proteinuria, reduced GFR, and renal glomerular and proximal tubular injury. Increased renal expression of aldose reductase; elevated levels of renal sorbitol, fructose, and uric acid; and low levels of ATP confirmed activation of the fructokinase pathway. Furthermore, renal expression of inflammatory cytokines with macrophage infiltration was prominent. In contrast, diabetic fructokinase-deficient mice demonstrated significantly less proteinuria, renal dysfunction, renal injury, and inflammation. These studies identify fructokinase as a novel mediator of diabetic nephropathy and document a novel role for endogenous fructose production, or fructoneogenesis, in driving renal disease.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Frutoquinases/metabolismo , Frutose/biossíntese , Frutose/metabolismo , Túbulos Renais Proximais/enzimologia , Animais , Glicemia/metabolismo , Peso Corporal , Linhagem Celular Transformada , Quimiocinas/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/patologia , Humanos , Córtex Renal/enzimologia , Córtex Renal/patologia , Glomérulos Renais/citologia , Glomérulos Renais/patologia , Túbulos Renais Proximais/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polímeros/metabolismo
4.
Nat Commun ; 4: 2434, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24022321

RESUMO

Carbohydrates with high glycaemic index are proposed to promote the development of obesity, insulin resistance and fatty liver, but the mechanism by which this occurs remains unknown. High serum glucose concentrations are known to induce the polyol pathway and increase fructose generation in the liver. Here we show that this hepatic, endogenously produced fructose causes systemic metabolic changes. We demonstrate that mice unable to metabolize fructose are protected from an increase in energy intake and body weight, visceral obesity, fatty liver, elevated insulin levels and hyperleptinaemia after exposure to 10% glucose for 14 weeks. In normal mice, glucose consumption is accompanied by aldose reductase and polyol pathway activation in steatotic areas. In this regard, we show that aldose reductase-deficient mice are protected against glucose-induced fatty liver. We conclude that endogenous fructose generation and metabolism in the liver represents an important mechanism by which glucose promotes the development of metabolic syndrome.


Assuntos
Frutose/biossíntese , Frutose/metabolismo , Fígado/metabolismo , Fígado/patologia , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Aldeído Redutase/metabolismo , Animais , Metabolismo Energético , Fígado Gorduroso/metabolismo , Comportamento Alimentar , Frutoquinases/deficiência , Frutoquinases/metabolismo , Glucose/metabolismo , Células Hep G2 , Humanos , Fígado/enzimologia , Fígado/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polímeros/metabolismo
5.
Hepatology ; 58(5): 1632-43, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23813872

RESUMO

UNLABELLED: Fructose intake from added sugars has been implicated as a cause of nonalcoholic fatty liver disease. Here we tested the hypothesis that fructose may interact with a high-fat diet to induce fatty liver, and to determine if this was dependent on a key enzyme in fructose metabolism, fructokinase. Wild-type or fructokinase knockout mice were fed a low-fat (11%), high-fat (36%), or high-fat (36%) and high-sucrose (30%) diet for 15 weeks. Both wild-type and fructokinase knockout mice developed obesity with mild hepatic steatosis and no evidence of hepatic inflammation on a high-fat diet compared to a low-fat diet. In contrast, wild-type mice fed a high-fat and high-sucrose diet developed more severe hepatic steatosis with low-grade inflammation and fibrosis, as noted by increased CD68, tumor necrosis factor alpha, monocyte chemoattractant protein-1, alpha-smooth muscle actin, and collagen I and TIMP1 expression. These changes were prevented in the fructokinase knockout mice. CONCLUSION: An additive effect of high-fat and high-sucrose diet on the development of hepatic steatosis exists. Further, the combination of sucrose with high-fat diet may induce steatohepatitis. The protection in fructokinase knockout mice suggests a key role for fructose (from sucrose) in this development of steatohepatitis. These studies emphasize the important role of fructose in the development of fatty liver and nonalcoholic steatohepatitis.


Assuntos
Dieta Hiperlipídica , Fígado Gorduroso/etiologia , Frutoquinases/fisiologia , Sacarose/administração & dosagem , Animais , Ingestão de Energia , Frutose/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Aumento de Peso
6.
Proc Natl Acad Sci U S A ; 109(11): 4320-5, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22371574

RESUMO

Fructose intake from added sugars correlates with the epidemic rise in obesity, metabolic syndrome, and nonalcoholic fatty liver disease. Fructose intake also causes features of metabolic syndrome in laboratory animals and humans. The first enzyme in fructose metabolism is fructokinase, which exists as two isoforms, A and C. Here we show that fructose-induced metabolic syndrome is prevented in mice lacking both isoforms but is exacerbated in mice lacking fructokinase A. Fructokinase C is expressed primarily in liver, intestine, and kidney and has high affinity for fructose, resulting in rapid metabolism and marked ATP depletion. In contrast, fructokinase A is widely distributed, has low affinity for fructose, and has less dramatic effects on ATP levels. By reducing the amount of fructose for metabolism in the liver, fructokinase A protects against fructokinase C-mediated metabolic syndrome. These studies provide insights into the mechanisms by which fructose causes obesity and metabolic syndrome.


Assuntos
Frutoquinases/metabolismo , Síndrome Metabólica/enzimologia , Animais , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Frutose/administração & dosagem , Frutose/metabolismo , Frutose/farmacologia , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Nat Genet ; 41(7): 829-32, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19525956

RESUMO

Aicardi-Goutières syndrome is a mendelian mimic of congenital infection and also shows overlap with systemic lupus erythematosus at both a clinical and biochemical level. The recent identification of mutations in TREX1 and genes encoding the RNASEH2 complex and studies of the function of TREX1 in DNA metabolism have defined a previously unknown mechanism for the initiation of autoimmunity by interferon-stimulatory nucleic acid. Here we describe mutations in SAMHD1 as the cause of AGS at the AGS5 locus and present data to show that SAMHD1 may act as a negative regulator of the cell-intrinsic antiviral response.


Assuntos
Encefalopatias Metabólicas Congênitas/genética , Imunidade Inata , Proteínas Monoméricas de Ligação ao GTP/genética , Substituição de Aminoácidos , Encefalopatias Metabólicas Congênitas/imunologia , Humanos , Proteínas Monoméricas de Ligação ao GTP/imunologia , Proteína 1 com Domínio SAM e Domínio HD
8.
J Histochem Cytochem ; 57(8): 763-74, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19365088

RESUMO

Ketohexokinase (KHK, also known as fructokinase) initiates the pathway through which most dietary fructose is metabolized. Very little is known about the cellular localization of this enzyme. Alternatively spliced KHK-C and KHK-A mRNAs are known, but the existence of the KHK-A protein isoform has not been demonstrated in vivo. Using antibodies to KHK for immunohistochemistry and Western blotting of rodent tissues, including those from mouse knockouts, coupled with RT-PCR assays, we determined the distribution of the splice variants. The highly expressed KHK-C isoform localized to hepatocytes in the liver and to the straight segment of the proximal renal tubule. In both tissues, cytoplasmic and nuclear staining was observed. The KHK-A mRNA isoform was observed exclusively in a range of other tissues, and by Western blotting, the presence of endogenous immunoreactive KHK-A protein was shown for the first time, proving that the KHK-A mRNA is translated into KHK-A protein in vivo, and supporting the suggestion that this evolutionarily conserved isoform is physiologically functional. However, the low levels of KHK-A expression prevented its immunohistochemical localization within these tissues. Our results highlight that the use of in vivo biological controls (tissues from knockout animals) is required to distinguish genuine KHK immunoreactivity from experimental artifact.


Assuntos
Frutoquinases/metabolismo , Frutose/metabolismo , Processamento Alternativo , Animais , Western Blotting , Linhagem Celular Tumoral , Escherichia coli/metabolismo , Feminino , Frutoquinases/genética , Humanos , Imuno-Histoquímica , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie
9.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 3): 201-11, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19237742

RESUMO

A molecular understanding of the unique aspects of dietary fructose metabolism may be the key to understanding and controlling the current epidemic of fructose-related obesity, diabetes and related adverse metabolic states in Western populations. Fructose catabolism is initiated by its phosphorylation to fructose 1-phosphate, which is performed by ketohexokinase (KHK). Here, the crystal structures of the two alternatively spliced isoforms of human ketohexokinase, hepatic KHK-C and the peripheral isoform KHK-A, and of the ternary complex of KHK-A with the substrate fructose and AMP-PNP are reported. The structure of the KHK-A ternary complex revealed an active site with both the substrate fructose and the ATP analogue in positions ready for phosphorylation following a reaction mechanism similar to that of the pfkB family of carbohydrate kinases. Hepatic KHK deficiency causes the benign disorder essential fructosuria. The effects of the disease-causing mutations (Gly40Arg and Ala43Thr) have been modelled in the context of the KHK structure.


Assuntos
Processamento Alternativo , Frutoquinases/química , Trifosfato de Adenosina/metabolismo , Adenilil Imidodifosfato/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Frutoquinases/genética , Frutoquinases/metabolismo , Frutose/metabolismo , Humanos , Magnésio/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Fosforilação , Mutação Puntual , Conformação Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
10.
Biochem Biophys Res Commun ; 348(3): 1123-31, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16904639

RESUMO

ATP-sensitive potassium (KATP) channels play a central role in glucose-stimulated insulin secretion (GSIS) by pancreatic beta-cells. Activity of these channels is determined by their open probability (Po) and the number of channels present in a cell. Glucose is known to reduce Po, but whether it also affects the channel density is unknown. Using INS-1 model beta-cell line, we show that the expression of K(ATP) channel subunits, Kir6.2 and SUR1, is high at low glucose, but declines sharply when the ambient glucose concentration exceeds 5mM. In response to glucose deprivation, channel synthesis increases rapidly by up-regulating translation of existing mRNAs. The effects of glucose deprivation could be mimicked by pharmacological activation of 5'-AMP-activated protein kinase with 5-aminoimidazole-4-carboxamide ribonucleotide and metformin. Pancreatic beta-cells which have lost their ability for GSIS do not show such changes implicating a possible (patho-)physiological link between glucose-regulated KATP channel expression and the capacity for normal GSIS.


Assuntos
Trifosfato de Adenosina/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/biossíntese , Glucose/deficiência , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Proteínas Quinases Ativadas por AMP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Meios de Cultivo Condicionados , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Glucose/fisiologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Camundongos , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Complexos Multienzimáticos/fisiologia , Canais de Potássio/biossíntese , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Ratos , Receptores de Droga/biossíntese , Receptores de Sulfonilureias
11.
Brain Res Mol Brain Res ; 121(1-2): 37-49, 2004 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-14969735

RESUMO

The expression of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor subunits GluR1-4 in the nucleus of the solitary tract (NTS) of adult Wistar rats was examined by polymerase chain reaction (PCR), and the neuronal localisation of these receptor subunits in the NTS were confirmed by immunohistochemistry using subunit-specific antibodies. Semi-quantitative PCR was used to investigate differences in AMPA receptor subunit expression between spontaneously hypertensive rats (SH) and age-matched normotensive Wistar Kyoto rats (WKY). All four receptor subunits were expressed in both strains, but compared to WKY, total AMPA receptor and the GluR3 mRNA expressions were significantly higher in SH. No differences were detected in cDNA form the cerebral cortex or cerebellum. Immunolabelling for GluRs 1, 2 and 2/3 in the neuropil relative to neuronal somata in the cardioregulatory areas of the NTS appeared to be increased in SH, with an overall increase in the density of GluR2/3 labelling in the medial and commissural NTS of SH. These results indicate a possible role for changes in AMPA receptor subunit expression in NTS neurones, involving an increase in GluR3 associated with development of hypertension in SH.


Assuntos
Hipertensão/metabolismo , Subunidades Proteicas/metabolismo , Receptores de AMPA/metabolismo , Núcleo Solitário/metabolismo , Animais , Contagem de Células/métodos , Cerebelo/metabolismo , Cerebelo/patologia , Expressão Gênica , Hipertensão/genética , Imuno-Histoquímica/métodos , Masculino , Subunidades Proteicas/genética , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Wistar , Receptores de AMPA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Núcleo Solitário/patologia
12.
Diabetes ; 52(9): 2426-32, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12941785

RESUMO

Alternative splicing of the ketohexokinase (fructokinase) gene generates a "central" predominantly hepatic isoform (ketohexokinase-C) and a more widely distributed ketohexokinase-A. Only the abundant hepatic isoform is known to possess activity, and no function is defined for the lower levels of ketohexokinase-A in peripheral tissues. Hepatic ketohexokinase deficiency causes the benign disorder essential fructosuria. The molecular basis of this has been defined in one family (compound heterozygosity for mutations Gly40Arg and Ala43Thr). Here we show that both ketohexokinase isoforms are indeed active. Ketohexokinase-A has much poorer substrate affinity than ketohexokinase-C for fructose but is considerably more thermostable. The Gly40Arg mutation seems null, rendering both ketohexokinase-A and ketohexokinase-C inactive and largely insoluble. The Ala43Thr mutant retains activity, but this mutation decreases the thermal stability of both ketohexokinase-A and ketohexokinase-C. At physiologic temperature, this results in significant loss of ketohexokinase-C activity but not of ketohexokinase-A. Affected individuals who carry both mutations therefore probably have a selective deficiency of hepatic ketohexokinase, with peripheral ketohexokinase-A being preserved. These findings raise the possibility that ketohexokinase-A serves an unknown physiologic function that remains intact in essential fructosuria. Further mutation analysis in this rare disorder could illuminate the question of whether ketohexokinase-A activity is, unlike that of ketohexokinase-C, physiologically indispensable.


Assuntos
Frutoquinases/genética , Frutoquinases/metabolismo , Erros Inatos do Metabolismo da Frutose/enzimologia , Erros Inatos do Metabolismo da Frutose/genética , Frutose/urina , Processamento Alternativo , Escherichia coli , Erros Inatos do Metabolismo da Frutose/urina , Regulação Enzimológica da Expressão Gênica , Temperatura Alta , Humanos , Cinética , Proteínas Recombinantes/genética , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...