Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 32(10): 5506-5519, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29741927

RESUMEN

Cobalamin [Cbl (or B12)] deficiency causes megaloblastic anemia and a variety of neuropathies. However, homeostatic mechanisms of cyanocobalamin (CNCbl) and other Cbls by vascular endothelial cells are poorly understood. Herein, we describe our investigation into whether cultured bovine aortic endothelial cells (BAECs) perform transcytosis of B12, namely, the complex formed between serum transcobalamin and B12, designated as holo-transcobalamin (holo-TC). We show that cultured BAECs endocytose [57Co]-CNCbl-TC (source material) via the CD320 receptor. The bound Cbl is transported across the cell both via exocytosis in its free form, [57Co]-CNCbl, and via transcytosis as [57Co]-CNCbl-TC. Transcellular mobilization of Cbl occurred in a bidirectional manner. A portion of the endocytosed [57Co]-CNCbl was enzymatically processed by methylmalonic aciduria combined with homocystinuria type C (cblC) with subsequent formation of hydroxocobalamin, methylcobalamin, and adenosylcobalamin, which were also transported across the cell in a bidirectional manner. This demonstrates that transport mechanisms for Cbl in vascular endothelial cells do not discriminate between various ß-axial ligands of the vitamin. Competition studies with apoprotein- and holo-TC and holo-intrinsic factor showed that only holo-TC was effective at inhibiting transcellular transport of Cbl. Incubation of BAECs with a blocking antibody against the extracellular domain of the CD320 receptor inhibited uptake and transcytosis by ∼40%. This study reveals that endothelial cells recycle uncommitted intracellular Cbl for downstream usage by other cell types and suggests that the endothelium is self-sufficient for the specific acquisition and subsequent distribution of circulating B12 via the CD320 receptor. We posit that the endothelial lining of the vasculature is an essential component for the maintenance of serum-tissue homeostasis of B12.-Hannibal, L., Bolisetty, K., Axhemi, A., DiBello, P. M., Quadros, E. V., Fedosov, S., Jacobsen, D. W. Transcellular transport of cobalamin in aortic endothelial cells.


Asunto(s)
Aorta/metabolismo , Células Endoteliales/metabolismo , Transcitosis/fisiología , Vitamina B 12/farmacocinética , Animales , Aorta/citología , Bovinos , Células Endoteliales/citología , Vitamina B 12/farmacología
2.
J Lipid Res ; 56(3): 653-664, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25561462

RESUMEN

HDL functions are impaired by myeloperoxidase (MPO), which selectively targets and oxidizes human apoA1. We previously found that the 4WF isoform of human apoA1, in which the four tryptophan residues are substituted with phenylalanine, is resistant to MPO-mediated loss of function. The purpose of this study was to generate 4WF apoA1 transgenic mice and compare functional properties of the 4WF and wild-type human apoA1 isoforms in vivo. Male mice had significantly higher plasma apoA1 levels than females for both isoforms of human apoA1, attributed to different production rates. With matched plasma apoA1 levels, 4WF transgenics had a trend for slightly less HDL-cholesterol versus human apoA1 transgenics. While 4WF transgenics had 31% less reverse cholesterol transport (RCT) to the plasma compartment, equivalent RCT to the liver and feces was observed. Plasma from both strains had similar ability to accept cholesterol and facilitate ex vivo cholesterol efflux from macrophages. Furthermore, we observed that 4WF transgenic HDL was partially (∼50%) protected from MPO-mediated loss of function while human apoA1 transgenic HDL lost all ABCA1-dependent cholesterol acceptor activity. In conclusion, the structure and function of HDL from 4WF transgenic mice was not different than HDL derived from human apoA1 transgenic mice.


Asunto(s)
Apolipoproteína A-I/metabolismo , HDL-Colesterol/metabolismo , Macrófagos/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Apolipoproteína A-I/genética , HDL-Colesterol/genética , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Oxidación-Reducción , Peroxidasa/genética , Peroxidasa/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Relación Estructura-Actividad
3.
Clin Chem Lab Med ; 51(3): 477-88, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23241609

RESUMEN

The causes of cobalamin (B12, Cbl) deficiency are multifactorial. Whether nutritional due to poor dietary intake, or functional due to impairments in absorption or intracellular processing and trafficking events, the major symptoms of Cbl deficiency include megaloblastic anemia, neurological deterioration and in extreme cases, failure to thrive and death. The common biomarkers of Cbl deficiency (hyperhomocysteinemia and methylmalonic acidemia) are extremely valuable diagnostic indicators of the condition, but little is known about the changes that occur at the protein level. A mechanistic explanation bridging the physiological changes associated with functional B12 deficiency with its intracellular processers and carriers is lacking. In this article, we will cover the effects of B12 deficiency in a cblC-disrupted background (also referred to as MMACHC) as a model of functional Cbl deficiency. As will be shown, major protein changes involve the cytoskeleton, the neurological system as well as signaling and detoxification pathways. Supplementation of cultured MMACHC-mutant cells with hydroxocobalamin (HOCbl) failed to restore these variants to the normal phenotype, suggesting that a defective Cbl processing pathway produces irreversible changes at the protein level.


Asunto(s)
Proteómica , Vitamina B 12/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Regulación de la Expresión Génica , Humanos , Sistema Nervioso/metabolismo , Oxidorreductasas , Biosíntesis de Proteínas , Transducción de Señal , Vitamina B 12/química , Deficiencia de Vitamina B 12/diagnóstico , Deficiencia de Vitamina B 12/metabolismo , Deficiencia de Vitamina B 12/patología
4.
Mol Nutr Food Res ; 56(12): 1812-24, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23097236

RESUMEN

SCOPE: Selenium has complex effects in vivo on multiple homeostatic mechanisms such as redox balance, methylation balance, and epigenesis, via its interaction with the methionine-homocysteine cycle. In this study, we examined the hypothesis that selenium status would modulate both redox and methylation balance and thereby modulate myocardial structure and function. METHODS AND RESULTS: We examined the effects of selenium-deficient (<0.025 mg/kg), control (0.15 mg/kg), and selenium-supplemented (0.5 mg/kg) diets on myocardial histology, biochemistry and function in adult C57/BL6 mice. Selenium deficiency led to reactive myocardial fibrosis and systolic dysfunction accompanied by increased myocardial oxidant stress. Selenium supplementation significantly reduced methylation potential, DNA methyltransferase activity and DNA methylation. In mice fed the supplemented diet, inspite of lower oxidant stress, myocardial matrix gene expression was significantly altered resulting in reactive myocardial fibrosis and diastolic dysfunction in the absence of myocardial hypertrophy. CONCLUSION: Our results indicate that both selenium deficiency and modest selenium supplementation leads to a similar phenotype of abnormal myocardial matrix remodeling and dysfunction in the normal heart. The crucial role selenium plays in maintaining the balance between redox and methylation pathways needs to be taken into account while optimizing selenium status for prevention and treatment of heart failure.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Metilación de ADN/efectos de los fármacos , Suplementos Dietéticos , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Selenio/deficiencia , Selenio/farmacología , Animales , Cardiomiopatías/fisiopatología , Cisteína/sangre , Dieta , Epigenómica , Fibrosis , Glutatión/sangre , Homocisteína/sangre , Isoprostanos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Selenio/sangre , Selenoproteínas/genética , Selenoproteínas/metabolismo
5.
Mol Genet Metab ; 103(3): 226-39, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21497120

RESUMEN

Cobalamin (Cbl, B(12)) is an essential micronutrient required to fulfill the enzymatic reactions of cytosolic methylcobalamin-dependent methionine synthase and mitochondrial adenosylcobalamin-dependent methylmalonyl-CoA mutase. Mutations in the MMACHC gene (cblC complementation group) disrupt processing of the upper-axial ligand of newly internalized cobalamins, leading to functional deficiency of the vitamin. Patients with cblC disease present with both hyperhomocysteinemia and methylmalonic acidemia, cognitive dysfunction, and megaloblastic anemia. In the present study we show that cultured skin fibroblasts from cblC patients export increased levels of both homocysteine and methylmalonic acid compared to control skin fibroblasts, and that they also have decreased levels of total intracellular folates. This is consistent with the clinical phenotype of functional cobalamin deficiency in vivo. The protein changes that accompany human functional Cbl deficiency are unknown. The proteome of control and cblC fibroblasts was quantitatively examined by two dimensional difference in-gel electrophoresis (2D-DIGE) and liquid chromatography-electrospray ionization-mass spectrometry (LC/ESI/MS). Major changes were observed in the expression levels of proteins involved in cytoskeleton organization and assembly, the neurological system and cell signaling. Pathway analysis of the differentially expressed proteins demonstrated strong associations with neurological disorders, muscular and skeletal disorders, and cardiovascular diseases in the cblC mutant cell lines. Supplementation of the cell cultures with hydroxocobalamin did not restore the cblC proteome to the patterns of expression observed in control cells. These results concur with the observed phenotype of patients with the cblC disorder and their sometimes poor response to treatment with hydroxocobalamin. Our findings could be valuable for designing alternative therapies to alleviate the clinical manifestation of the cblC disorder, as some of the protein changes detected in our study are common hallmarks of known pathologies such as Alzheimer's and Parkinson's diseases as well as muscular dystrophies.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteoma , Deficiencia de Vitamina B 12/fisiopatología , Errores Innatos del Metabolismo de los Aminoácidos , Proteínas Portadoras/genética , Línea Celular , Células Cultivadas , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Ácido Fólico/metabolismo , Homocisteína/metabolismo , Humanos , Hidroxocobalamina/farmacología , Espacio Intracelular/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Ácido Metilmalónico/metabolismo , Mutación/genética , Oxidorreductasas , Fenotipo , Vimentina/metabolismo , Vitamina B 12/metabolismo , Deficiencia de Vitamina B 12/genética , Complejo Vitamínico B/farmacología
6.
Mol Cell Proteomics ; 9(3): 471-85, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20008833

RESUMEN

Hyperhomocysteinemia has long been associated with atherosclerosis and thrombosis and is an independent risk factor for cardiovascular disease. Its causes include both genetic and environmental factors. Although homocysteine is produced in every cell as an intermediate of the methionine cycle, the liver contributes the major portion found in circulation, and fatty liver is a common finding in homocystinuric patients. To understand the spectrum of proteins and associated pathways affected by hyperhomocysteinemia, we analyzed the mouse liver proteome of gene-induced (cystathionine beta-synthase (CBS)) and diet-induced (high methionine) hyperhomocysteinemic mice using two-dimensional difference gel electrophoresis and Ingenuity Pathway Analysis. Nine proteins were identified whose expression was significantly changed by 2-fold (p < or = 0.05) as a result of genotype, 27 proteins were changed as a result of diet, and 14 proteins were changed in response to genotype and diet. Importantly, three enzymes of the methionine cycle were up-regulated. S-Adenosylhomocysteine hydrolase increased in response to genotype and/or diet, whereas glycine N-methyltransferase and betaine-homocysteine methyltransferase only increased in response to diet. The antioxidant proteins peroxiredoxins 1 and 2 increased in wild-type mice fed the high methionine diet but not in the CBS mutants, suggesting a dysregulation in the antioxidant capacity of those animals. Furthermore, thioredoxin 1 decreased in both wild-type and CBS mutants on the diet but not in the mutants fed a control diet. Several urea cycle proteins increased in both diet groups; however, arginase 1 decreased in the CBS(+/-) mice fed the control diet. Pathway analysis identified the retinoid X receptor signaling pathway as the top ranked network associated with the CBS(+/-) genotype, whereas xenobiotic metabolism and the NRF2-mediated oxidative stress response were associated with the high methionine diet. Our results show that hyperhomocysteinemia, whether caused by a genetic mutation or diet, alters the abundance of several liver proteins involved in homocysteine/methionine metabolism, the urea cycle, and antioxidant defense.


Asunto(s)
Antioxidantes/metabolismo , Cistationina betasintasa/genética , Dieta/efectos adversos , Homocisteína/metabolismo , Hiperhomocisteinemia/enzimología , Hígado/enzimología , Metionina/metabolismo , Nutrigenómica , Urea/metabolismo , Adenosilhomocisteinasa/metabolismo , Animales , Betaína-Homocisteína S-Metiltransferasa/metabolismo , Glicina N-Metiltransferasa/metabolismo , Hiperhomocisteinemia/inducido químicamente , Hiperhomocisteinemia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Peroxirredoxinas/metabolismo , Proteómica
7.
Hepatology ; 49(5): 1709-17, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19205032

RESUMEN

UNLABELLED: Ethanol-induced liver injury is characterized by increased formation of reactive oxygen species (ROS) and inflammatory cytokines, resulting in the development of hepatic steatosis, injury, and cell death by necrosis and apoptosis. Thioredoxin (Trx), a potent antioxidant and antiinflammatory molecule with antiapoptotic properties, protects animals from a number of inflammatory diseases. However, the effects of ethanol on Trx or its role in ethanol-induced liver injury are not known. Female C57BL/6 mice were allowed ad libitum access to a Lieber-deCarli ethanol diet with 5.4% of calories as ethanol for 2 days to acclimate them to the diet, followed by 2 days with 32.4% of calories as ethanol or pair-fed control diet. Hepatic Trx-1 was decreased by ethanol feeding; daily supplementation with recombinant human Trx (rhTrx) prevented this ethanol-induced decrease. Therefore, we tested the hypothesis that administration of rhTrx during ethanol exposure would attenuate ethanol-induced oxidative stress, inflammatory cytokine production, and apoptosis. Mice were treated with a daily intraperitoneal injection of either 5 g/kg of rhTrx or phosphate-buffered saline (PBS). CONCLUSION: Ethanol feeding increased accumulation of hepatic 4-hydroxynonenal protein adducts, expression of hepatic tumor necrosis factor alpha, and resulted in hepatic steatosis and increased plasma aspartate aminotransferase and alanine aminotransferase. In ethanol-fed mice, treatment with rhTrx reduced 4-hydroxynonenal adduct accumulation, inflammatory cytokine expression, decreased hepatic triglyceride, and improved liver enzyme profiles. Ethanol feeding also increased transferase-mediated dUTP-biotin nick-end labeling-positive cells, caspase-3 activity, and cytokeratin-18 staining in the liver. rhTrx treatment prevented these increases. In summary, rhTrx attenuated ethanol-induced increases in markers of oxidative stress, inflammatory cytokine expression, and apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Etanol/farmacología , Hepatopatías Alcohólicas/prevención & control , Estrés Oxidativo/efectos de los fármacos , Tiorredoxinas/uso terapéutico , Animales , Citocinas/metabolismo , Femenino , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Tiorredoxinas/farmacología
8.
FASEB J ; 22(7): 2569-78, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18364397

RESUMEN

A causal relationship between diet-induced hyperhomocysteinemia (HHcy) and accelerated atherosclerosis has been established in apolipoprotein E-deficient (apoE(-/-)) mice. However, it is not known whether the proatherogenic effect of HHcy in apoE(-/-) mice is independent of hyperlipidemia and/or deficiency of apoE. In this study, a comprehensive dietary approach using C57BL/6J mice was used to investigate whether HHcy is an independent risk factor for accelerated atherosclerosis or dependent on additional dietary factors that increase plasma lipids and/or inflammation. C57BL/6J mice at 4 wk of age were divided into 6 dietary groups: chow diet (C), chow diet + methionine (C+M), western-type diet (W), western-type diet + methionine (W+M), atherogenic diet (A), or atherogenic diet + methionine (A+M). After 2, 10, 20, or 40 wk on the diets, mice were sacrificed, and the levels of total plasma homocysteine, cysteine, and glutathione, as well as total plasma cholesterol and triglycerides were analyzed. Aortic root sections were examined for atherosclerotic lesions. HHcy was induced in all groups supplemented with methionine, compared to diet-matched control groups. Plasma total cholesterol was significantly increased in mice fed the W or A diet. However, the W diet increased LDL/IDL and HDL levels, while the A diet significantly elevated plasma VLDL and LDL/IDL levels without increasing HDL. No differences in plasma total cholesterol levels or lipid profiles were observed between methionine-supplemented groups and the diet-matched control groups. Early atherosclerotic lesions containing macrophage foam cells were only observed in mice fed the A or A + M diet. Furthermore, lesion size was significantly larger in the A + M group compared to the A group at 10 and 20 wk; however, mature lesions were never observed even after 40 wk on these diets. The presence of lymphocytes, increased hyaluronan staining, and the expression of endoplasmic reticulum (ER) stress markers were also increased in atherosclerotic lesions from the A + M group. Taken together, these results suggest that HHcy does not independently cause atherosclerosis in C57BL/6J mice even in the presence of increased total plasma lipids induced by the W diet. However, HHcy can accelerate atherosclerotic lesion development under dietary conditions that increase plasma VLDL levels and/or inflammation.


Asunto(s)
Aterosclerosis/fisiopatología , Hiperhomocisteinemia/inducido químicamente , Hiperhomocisteinemia/fisiopatología , Metionina/farmacología , Animales , Aterosclerosis/sangre , Aterosclerosis/inducido químicamente , Aterosclerosis/patología , Dieta Aterogénica , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Homocisteína/sangre , Ácido Hialurónico/metabolismo , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/patología , Inmunohistoquímica , Lípidos/sangre , Metionina/administración & dosificación , Ratones , Ratones Endogámicos C57BL
9.
Haematologica ; 93(2): 279-82, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18223282

RESUMEN

In Chuvash polycythemia, homozygous von Hippel-Lindau (VHL) 598C>T leads to increased hypoxia inducible factor-1alpha and 2alpha, thromboses and lower systemic blood pressures. Circulating homocysteine, glutathione, gamma-glutamyltransferase and cysteinylglycine concentrations were higher in 34 VHL598C>T homozygotes than in 37 normal controls and cysteine was lower. Multivariate analysis showed elevated homocysteine independently associated with higher mean systemic blood pressures and elevated glutathione was associated with lower pressures to a similar degree. Among VHL598C>T homozygotes, homocysteine was elevated with low and normal folate concentrations, consistent with a possible defect in the remethylation pathway. The elevated glutathione and gamma-glutamyltransferase levels correlated positively with cysteinylglycine, consistent with possible upregulation of a glutathione synthetic enzyme and gamma-glutamyltransferase. Cysteinylglycine correlated inversely with cysteine, consistent with possible reduced cysteinyldipeptidase activity. We conclude that up-regulated hypoxia-sensing may influence multiple steps in thiol metabolism. The effects of the resultant elevated levels of homocysteine and glutathione on systemic blood pressure may largely balance each other out.


Asunto(s)
Dipéptidos/sangre , Enfermedades Genéticas Congénitas/sangre , Glutatión/sangre , Homocisteína/sangre , Homocigoto , Mutación Puntual , Policitemia/sangre , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Presión Sanguínea/genética , Femenino , Enfermedades Genéticas Congénitas/metabolismo , Humanos , Masculino , Policitemia/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , gamma-Glutamiltransferasa/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 27(1): 49-54, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17082481

RESUMEN

OBJECTIVE: L-homocysteine and/or L-homocystine interact in vivo with albumin and other extracellular proteins by forming mixed-disulfide conjugates. Because of its extremely rich cysteine content, we hypothesized that metallothionein, a ubiquitous intracellular zinc-chaperone and superoxide anion radical scavenger, reacts with L-homocysteine and that homocysteinylated-metallothionein suffers loss of function. METHODS AND RESULTS: 35S-homocysteinylated-metallothionein was resolved in lysates of cultured human aortic endothelial cells in the absence and presence of reduced glutathione by SDS-PAGE and identified by Western blotting and phosphorimaging. Using zinc-Sepharose chromatography, L-homocysteine was shown to impair the zinc-binding capacity of metallothionein even in the presence of reduced glutathione. L-Homocysteine induced a dose-dependent increase in intracellular free zinc in zinquin-loaded human aortic endothelial cells within 30 minutes, followed by the appearance of early growth response protein-1 within 60 minutes. In addition, intracellular reactive oxygen species dramatically increased 6 hours after L-homocysteine treatment. In vitro studies demonstrated that L-homocysteine is a potent inhibitor of the superoxide anion radical scavenging ability of metallothionein. CONCLUSIONS: These studies provide the first evidence that L-homocysteine targets intracellular metallothionein by forming a mixed-disulfide conjugate and that loss of function occurs after homocysteinylation. The data support a novel mechanism for disruption of zinc and redox homeostasis.


Asunto(s)
Endotelio Vascular/metabolismo , Homeostasis/fisiología , Homocisteína/farmacología , Metalotioneína/metabolismo , Zinc/metabolismo , Aorta Torácica/citología , Células Cultivadas , Cromatografía en Agarosa , Relación Dosis-Respuesta a Droga , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Regulación de la Expresión Génica , Homocisteína/metabolismo , Humanos , Oxidación-Reducción , Superóxidos/metabolismo , Factores de Tiempo
11.
Arch Biochem Biophys ; 446(2): 119-30, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16455044

RESUMEN

Hyperhomocysteinemia is an independent risk factor for cardiovascular disease. Transport of L-homocysteine into and out of the human vascular endothelium is poorly understood. We hypothesized that cultured human aortic endothelial cells (HAEC) would import L-homocysteine on one or more of the L-cysteine transport systems. Inhibitors of the transporters were used to characterize the uptake of [35S]L-homocysteine, [35S]L-homocystine, and [35S]L-cysteine. We found that L-homocysteine uptake is mediated by the sodium-dependent cysteine transport systems X(AG), ASC, and A, and the sodium-independent transport system L. Thus, HAEC utilize multiple cysteine transporters (X(AG) > or = L > ASC > A) to import L-homocysteine. Kinetic analysis supported the uptake results. Michaelis-Menten constants (Km) for the four systems yielded values of 19.0, 27.1, 112, and 1000 microM for systems L, X(AG), ASC, and A, respectively. The binding and uptake of [35S]L-homocystine, the disulfide homodimer of L-homocysteine, was mediated by systems X(AG), L, and ASC but not by system A. In contrast to [35S]L-homocysteine, system x(c) was active for [35S]L-homocystine uptake. A similar pattern was observed for [35S]L-cysteine. Thus, L-homocysteine and L-homocystine found in hyperhomocysteinemic subjects can gain entry into the vascular endothelium by way of multiple L-cysteine transporters.


Asunto(s)
Células Endoteliales/metabolismo , Homocisteína/metabolismo , Sistema de Transporte de Aminoácidos A/fisiología , Sistema de Transporte de Aminoácidos ASC/fisiología , Sistema de Transporte de Aminoácidos L/fisiología , Sistema de Transporte de Aminoácidos X-AG/fisiología , Aorta Torácica/citología , Transporte Biológico Activo , Células Cultivadas , Cisteína/metabolismo , Cisteína/farmacocinética , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Homocisteína/farmacocinética , Humanos , Unión Proteica , Radioisótopos de Azufre
12.
Clin Chem Lab Med ; 43(10): 1076-83, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16197301

RESUMEN

Hyperhomocysteinemia is an independent risk factor for cardiovascular disease. Although there is a growing body of evidence that homocysteine plays a causal role in atherogenesis, specific mechanisms to explain the underlying pathology have remained elusive. This review focuses on chemistry unique to the homocysteine molecule to explain its inherent cytotoxicity. Thus, the high pKa of the sulfhydryl group (pKa=10.0) of homocysteine underlies its ability to form stable disulfide bonds with protein cysteine residues, and in the process, alters or impairs the function of the protein. Albumin, fibronectin, transthyretin, annexin II, and factor V have now been identified as molecular targets for homocysteine, and in the case of albumin, the mechanism of targeting has been elucidated.


Asunto(s)
Homocisteína/metabolismo , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología , Animales , Proteínas Sanguíneas/metabolismo , Células Endoteliales/metabolismo , Homocisteína/química , Humanos , Proteínas de la Membrana/metabolismo
13.
Circ Res ; 94(10): 1318-24, 2004 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-15105297

RESUMEN

Elevated total plasma homocysteine is an independent risk factor in the development of vascular disease in humans. Cystathionine beta-synthase (CBS) is an enzyme that condenses homocysteine with serine to form cystathionine. In this article, we describe the effects of modulating CBS activity using a transgenic mouse that contains the human CBS cDNA under control of the zinc-inducible metallothionein promoter (Tg-CBS). In the presence of zinc, Tg-CBS mice have a 2- to 4-fold increase in liver and kidney CBS activity compared with nontransgenic littermates. Transgenic mice on standard mouse chow had a 45% decrease in their serum homocysteine (12.1 to 7.2 micromol/L; P<0.0001) when zinc was added to drinking water, although zinc had minimal effect on their nontransgenic siblings (13.2 micromol/L versus 13.0 micromol/L; P=NS). Tg-CBS mice maintained on a high-methionine, low-folate diet also had significantly lower serum homocysteine compared with control animals (179 micromol/L versus 242 micromol/L; P<0.02). CBS overexpression also significantly lowered serum cysteinylglycine (3.6 versus 2.8 micromol/L; P<0.003) levels and reduced the levels of many amino acids in the liver. We also found that expression of Tg-CBS rescued the severe hyperhomocysteinemia and neonatal lethality of Cbs deletion animals. Our results show that elevating CBS activity is an effective method to lower plasma homocysteine levels. In addition, the creation of an inducible mouse system to modulate plasma homocysteine will also be useful in the study of homocysteine-related vascular disease.


Asunto(s)
Cistationina betasintasa/metabolismo , Homocisteína/sangre , Aminoácidos/metabolismo , Animales , Cistationina betasintasa/genética , Humanos , Hígado/metabolismo , Metionina/metabolismo , Ratones , Ratones Transgénicos
15.
Thromb Haemost ; 89(1): 48-52, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12540953

RESUMEN

Previous studies showed that alpha-fibrin monomer (lacking both A-fibrinopeptides, FPA) is normally cleared from the circulation before it assembles into a clot. Recent studies indicate that substantial quantities of an intermediate, alpha-profibrin lacking only one of the two FPA are produced in the course of conversion of human fibrinogen to fibrin. Since clearance of the alpha-fibrin monomer is saturable and receptor mediated, the extent to which alpha-profibrin or other fibrin(ogen) derivatives might compete for monomer uptake was deemed important. We compared plasma decay of injected human alpha-fibrin, fibrinogen, and alpha-profibrin in rabbits using rabbit anti-human fibrinogen for assays. The circulatory half-life of human alpha-fibrin monomer was short (t(1/2) = 2.3 h) and followed a simple exponential decay curve, as anticipated from clearance of rabbit alpha-fibrin. It was absorbed as fast as it permeated the extravascular space with no redistribution. Human fibrinogen had a long half-life (t(1/2) = 39.5 h), calculated from the double exponential plasma decay curves (redistribution + catabolism) observed over 28 h. The alpha-profibrin had an intermediary half-life (t(1/2) = 11 h) determined from double exponential decay curves. Since redistribution accompanied the slow clearance of alpha-profibrin, its binding by the fibrin receptor(s) must be weak, probably too weak to compete with the clearance of alpha-fibrin monomer. The initial production of alpha-fibrin monomer is only partially dependent on prior formation of alpha-profibrin, as recently shown. Thus, it is the slow clearance and the weak competition from alpha-profibrin that underlie the occurrence of substantial levels of alpha-profibrin unaccompanied by detectable levels of alpha-fibrin monomer in many subjects with vascular disease.


Asunto(s)
Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Fibrina/metabolismo , Fibrinógeno/metabolismo , Precursores de Proteínas/metabolismo , Animales , Unión Competitiva , Circulación Sanguínea , Productos de Degradación de Fibrina-Fibrinógeno/administración & dosificación , Productos de Degradación de Fibrina-Fibrinógeno/farmacocinética , Fibrinógeno/administración & dosificación , Fibrinógeno/farmacocinética , Semivida , Humanos , Cinética , Tasa de Depuración Metabólica , Precursores de Proteínas/administración & dosificación , Precursores de Proteínas/farmacocinética , Conejos , Receptores de Péptidos/metabolismo
16.
J Biol Chem ; 277(22): 19367-73, 2002 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-11891217

RESUMEN

Fibrin formation depends on the release of the two N-terminal fibrinopeptides A (FPA) from fibrinogen, and its formation is accompanied by an intermediate, alpha-profibrin, which lacks only one of the FPA. In this study, we confirm that the maximal levels of alpha-profibrin found over the course of thrombin reactions with human fibrinogen are only half of what would be expected if the first and second FPA were being released independently with equal rate constants. The rapidity of release of the fibrinopeptides by thrombin had been shown to depend on an allosteric transformation that is induced when Na(+) binds to a site defined by the 215-227 residues of thrombin, a transformation that results in the exposure of its fibrinogen-binding exosites transforming the thrombin from a slow to a fast acting form toward fibrinogen. When choline was substituted for sodium to transform thrombin to its slow form, the maximal levels of alpha-profibrin rose to those expected for independent release of the two FPA. Thus, it is only the fast thrombin that releases the second FPA fast, and that fast release only occurs when both FPA are present because of a partial coupling of its release with that of the first FPA. The release of the FPA from purified alpha-profibrin with the first FPA already missing is no faster than the release of any FPA. Surprisingly, we also found that slow thrombin became increasingly transformed to a fast form in the absence of sodium when the fibrinogen was elevated to high concentrations. This potentiation by concentrated fibrinogen also occurs with the recombinant mutant thrombin (Y225P), which is otherwise slow in both the presence and absence of Na(+). The potentiation of thrombin by fibrinogen must be short-lived so that the thrombin reverts to its slow acting form in the interim among encounters with other fibrinogen molecules in dilute fibrinogen solutions lacking Na(+), whereas at high fibrinogen concentrations the thrombin encounters other molecules before it reverts back to the slow form.


Asunto(s)
Fibrinógeno/química , Trombina/química , Sitio Alostérico , Sitios de Unión , Colina/química , Colina/metabolismo , Fibrinógeno/metabolismo , Humanos , Cinética , Mutación , Unión Proteica , Sodio/química , Sodio/metabolismo , Temperatura , Trombina/genética , Trombina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...