Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Oxid Med Cell Longev ; 2022: 1486157, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36046692

RESUMEN

Hyperhomocysteinemia (HHcy) is positively linked with several cardiovascular diseases; however, its role and underlying mechanisms in pathological cardiac hypertrophy are still unclear. Here, we focused on the effects and underlying mechanisms of HHcy in hypertensive cardiac hypertrophy, one of the most common and typical types of pathological cardiac hypertrophy. By a retrospective analysis of the association between HHcy and cardiac hypertrophy in a hypertensive cohort, we found that the prevalence of HHcy was higher in patients with hypertrophy and significantly associated with the presence of cardiac hypertrophy after adjusting for other conventional risk factors. In mice, HHcy induced by a methionine (2% wt/wt) diet feeding significantly promoted cardiac hypertrophy as well as cardiac inflammation and fibrosis induced by 3-week angiotensin ІІ (AngІІ) infusion (1000 ng/kg/min), while folic acid (0.006% wt/wt) supplement corrected HHcy and attenuated AngII-stimulated cardiac phenotypes. Mechanistic studies further showed that homocysteine (Hcy) exacerbated AngII-stimulated expression of Calcineurin and nuclear factor of activated T cells (NFAT), which could be attenuated by folic acid both in mice and in neonatal rat cardiomyocytes. Moreover, treatment with cyclosporin A, an inhibitor of Calcineurin, blocked Hcy-stimulated Calcineurin-NFAT signaling and hypertrophy in neonatal rat cardiomyocytes. In conclusion, our study indicates that HHcy promotes cardiac hypertrophy in hypertension, and Calcineurin-NFAT pathway might be involved in the pro-hypertrophic effect of Hcy.


Asunto(s)
Hiperhomocisteinemia , Hipertensión , Animales , Calcineurina/metabolismo , Cardiomegalia/complicaciones , Cardiomegalia/metabolismo , Ácido Fólico/farmacología , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Hipertensión/complicaciones , Hipertensión/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Factores de Transcripción NFATC/metabolismo , Ratas , Estudios Retrospectivos
2.
Neurosci Bull ; 38(8): 887-900, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35435568

RESUMEN

Chronic stress is generally accepted as the main risk factor in the development of cognitive decline; however, the underlying mechanisms remain unclear. Previous data have demonstrated that the levels of homocysteine (Hcy) are significantly elevated in the plasma of stressed animals, which suggests that Hcy is associated with stress and cognitive decline. To test this hypothesis, we analyzed the cognitive function, plasma concentrations of Hcy, and brain-derived neurotropic factor (BDNF) levels in rats undergoing chronic unpredicted mild stress (CUMS). The results showed that decreased cognitive behavioral performance and decreased BDNF transcription and protein expression were correlated with hyperhomocysteinemia (HHcy) levels in stressed rats. Diet-induced HHcy mimicked the cognitive decline and BDNF downregulation in the same manner as CUMS, while Hcy reduction (by means of vitamin B complex supplements) alleviated the cognitive deficits and BDNF reduction in CUMS rats. Furthermore, we also found that both stress and HHcy disturbed the DNA methylation process in the brain and induced DNA hypermethylation in the BDNF promoter. In contrast, control of Hcy blocked BDNF promoter methylation and upregulated BDNF levels in the brain. These results imply the possibility of a causal role of Hcy in stress-induced cognitive decline. We also used ten-eleven translocation (TET1), an enzyme that induces DNA demethylation, to verify the involvement of Hcy and DNA methylation in the regulation of BDNF expression and the development of stress-related cognitive decline. The data showed that TET1-expressing viral injection into the hippocampus inhibited BDNF promoter methylation and significantly mitigated the cognitive decline in HHcy rats. Taken together, novel evidence from the present study suggests that Hcy is likely involved in chronic stress-induced BDNF reduction and related cognitive deficits. In addition, the negative side-effects of HHcy may be associated with Hcy-induced DNA hypermethylation in the BDNF promoter. The results also suggest the possibility of Hcy as a target for therapy and the potential value of vitamin B intake in preventing stress-induced cognitive decline.


Asunto(s)
Disfunción Cognitiva , Homocisteína , Hiperhomocisteinemia , Estrés Psicológico , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Disfunción Cognitiva/complicaciones , Metilación de ADN , Homocisteína/efectos adversos , Homocisteína/metabolismo , Hiperhomocisteinemia/metabolismo , Ratas , Estrés Psicológico/fisiopatología
3.
Biomed Res Int ; 2021: 6652231, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34036101

RESUMEN

Homocysteine (Hct) is a substance produced in the metabolism of methionine. It is an essential type of amino acid gained from the daily diet. Methylenetetrahydrofolate reductase (MTHFR) gene mutation is related to elevated total homocysteine (tHct) expressions, in particular, among women with low folate intake. Hyperhomocysteinemia (HHct) is caused by numerous factors, such as genetic defects, lack of folic acid, vitamin B6 and B12 deficiency, hypothyroidism, drugs, aging, and renal dysfunction. Increased Hct in peripheral blood may lead to vascular illnesses, coronary artery dysfunction, atherosclerotic changes, and embolic diseases. Compared to nonpregnant women, the Hct level is lower in normal pregnancies. Recent studies have reported that HHct was associated with numerous pregnancy complications, including recurrent pregnancy loss (RPL), preeclampsia (PE), preterm delivery, placental abruption, fetal growth restriction (FGR), and gestational diabetes mellitus (GDM). Besides, it was discovered that neonatal birth weight and maternal Hct levels were negatively correlated. However, a number of these findings lack consistency. In this review, we summarized the metabolic process of Hct in the human body, the levels of Hct in different stages of normal pregnancy reported in previous studies, and the relationship between Hct and pregnancy complications. The work done is helpful for obstetricians to improve the likelihood of a positive outcome during pregnancy complications. Reducing the Hct level with a high dosage of folic acid supplements during the next pregnancy could be helpful for females who have suffered pregnancy complications due to HHct.


Asunto(s)
Homocisteína/sangre , Complicaciones del Embarazo , Aborto Habitual , Envejecimiento , Peso al Nacer , Diabetes Gestacional , Suplementos Dietéticos , Femenino , Retardo del Crecimiento Fetal , Ácido Fólico/sangre , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/metabolismo , Placenta , Preeclampsia , Embarazo , Vitamina B 12/sangre , Deficiencia de Vitamina B 12 , Vitamina B 6/sangre , Deficiencia de Vitamina B 6
4.
Phytomedicine ; 81: 153410, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33285470

RESUMEN

BACKGROUND: Homocysteine (Hcy) induced vascular endothelial dysfunction is known to be closely associated with oxidative stress and impaired NO system. 1,8-Dihydroxy-3-methoxy-6-methylanthracene-9,10-dione (physcion) has been known to has antioxidative and anti-inflammatory properties. PURPOSE: The purpose of the present study was to define the protective effect of physcion on Hcy-induced endothelial dysfunction and its mechanisms involved. STUDY DESIGN AND METHODS: Hyperhomocysteinemia (HHcy) rat model was induced by feeding 3% methionine. A rat thoracic aortic ring model was used to investigate the effects of physcion on Hcy-induced impairment of endothelium-dependent relaxation. Two doses, low (L, 30 mg/kg/day) and high (H, 50 mg/kg/day) of physcion were used in the present study. To construct Hcy-injured human umbilical vein endothelial cells (HUVECs) model, the cells treated with 3 mM Hcy. The effects of physcion on Hcy-induced HUVECs cytotoxicity and apoptosis were studied using MTT and flow cytometry. Confocal analysis was used to determine the levels of intracellular Ca2+. The levels of protein expression of the apoptosis-related markers Bcl-2, Bax, caspase-9/3, and Akt and endothelial nitric oxide synthase (eNOS) were evaluated by western blot. RESULTS: In the HHcy rat model, plasma levels of Hcy and malondialdehyde (MDA) were elevated (20.45 ± 2.42 vs. 4.67 ± 1.94 µM, 9.42 ± 0.48 vs. 3.47 ± 0.59 nM, p < 0.001 for both), whereas superoxide dismutase (SOD) and nitric oxide (NO) levels were decreased (77.11 ± 4.78 vs. 115.02 ± 5.63 U/ml, 44.51 ± 4.45 vs. 64.18 ± 5.34 µM, p < 0.001 and p < 0.01, respectively). However, treatment with physcion significantly reversed these changes (11.82 ± 2.02 vs. 20.45 ± 2.42 µM, 5.97 ± 0.72 vs. 9.42 ± 0.48 nM, 108.75 ± 5.65 vs. 77.11 ± 4.78 U/ml, 58.14 ± 6.02 vs. 44.51 ± 4.45 µM, p < 0.01 for all). Physcion also prevented Hcy-induced impairment of endothelium-dependent relaxation in HHcy rats (1.56 ± 0.06 vs. 15.44 ± 2.53 nM EC50 for ACh vasorelaxation, p < 0.05 vs. HHcy). In Hcy-injured HUVECs, physcion inhibited the impaired viability, apoptosis and reactive oxygen species. Hcy treatment significantly increased the protein phosphorylation levels of p38 (2.26 ± 0.20 vs. 1.00 ± 0.12, p <0.01), ERK (2.11 ± 0.21 vs. 1.00 ± 0.11, p <0.01) and JNK. Moreover, physcion reversed the Hcy-induced apoptosis related parameter changes such as decreased mitochondrial membrane potential (MMP) and Bcl-2/Bax protein ratio, and increased protein expression of caspase-9/3 in HUVECs. Furthermore, the downregulation of Ca2+, Akt, eNOS and NO caused by Hcy were recovered with physcion treatment in HUVECs. CONCLUSION: Physcion prevents Hcy-induced endothelial dysfunction by activating Ca2+- and Akt-eNOS-NO signaling pathways. This study provides the first evidence that physcion might be a candidate agent for the prevention of cardiovascular disease induced by Hcy.


Asunto(s)
Calcio/metabolismo , Emodina/análogos & derivados , Endotelio Vascular/efectos de los fármacos , Homocisteína/metabolismo , Hiperhomocisteinemia/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Caspasa 9/metabolismo , Emodina/farmacología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Hiperhomocisteinemia/metabolismo , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Sustancias Protectoras/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
5.
J Nutr ; 150(Suppl 1): 2524S-2531S, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33000164

RESUMEN

The metabolism of methionine and cysteine in the body tissues determines the concentrations of several metabolites with various biologic activities, including homocysteine, hydrogen sulfide (H2S), taurine, and glutathione. Hyperhomocysteinemia, which is correlated with lower HDL cholesterol in blood in volunteers and animal models, has been associated with an increased risk for cardiovascular diseases. In humans, the relation between methionine intake and hyperhomocysteinemia is dependent on vitamin status (vitamins B-6 and B-12 and folic acid) and on the supply of other amino acids. However, lowering homocysteinemia by itself is not sufficient for decreasing the risk of cardiovascular disease progression. Other compounds related to methionine metabolism have recently been identified as being involved in the risk of atherosclerosis and steatohepatitis. Indeed, the metabolism of sulfur amino acids has an impact on phosphatidylcholine (PC) metabolism, and anomalies in PC synthesis due to global hypomethylation have been associated with disturbances of lipid metabolism. In addition, impairment of H2S synthesis from cysteine favors atherosclerosis and steatosis in animal models. The effects of taurine on lipid metabolism appear heterogeneous depending on the populations of volunteers studied. A decrease in the concentration of intracellular glutathione, a tripeptide involved in redox homeostasis, is implicated in the etiology of cardiovascular diseases and steatosis. Last, supplementation with betaine, a compound that allows remethylation of homocysteine to methionine, decreases basal and methionine-stimulated homocysteinemia; however, it adversely increases plasma total and LDL cholesterol. The study of these metabolites may help determine the range of optimal and safe intakes of methionine and cysteine in dietary proteins and supplements. The amino acid requirement for protein synthesis in different situations and for optimal production of intracellular compounds involved in the regulation of lipid metabolism also needs to be considered for dietary attenuation of atherosclerosis and steatosis risk.


Asunto(s)
Aterosclerosis/etiología , Cisteína/metabolismo , Hígado Graso/etiología , Metabolismo de los Lípidos , Metionina/metabolismo , Estado Nutricional , Azufre/metabolismo , Aminoácidos Sulfúricos/metabolismo , Animales , Aterosclerosis/metabolismo , Betaína/metabolismo , Betaína/farmacología , Colesterol/sangre , Proteínas en la Dieta/química , Suplementos Dietéticos , Hígado Graso/metabolismo , Glutatión/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/etiología , Hiperhomocisteinemia/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Necesidades Nutricionales , Fosfatidilcolinas/metabolismo , Compuestos de Azufre/metabolismo , Taurina/metabolismo , Taurina/farmacología
6.
J Nutr ; 150(Suppl 1): 2538S-2547S, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33000166

RESUMEN

The central position of methionine (Met) in protein metabolism indicates the importance of this essential amino acid for growth and maintenance of lean body mass. Therefore, Met might be a tempting candidate for supplementation. However, because Met is also the precursor of homocysteine (Hcy), a deficient intake of B vitamins or excessive intake of Met may result in hyperhomocysteinemia (HHcy), which is a risk factor for cardiovascular disease. This review discusses the evidence generated in preclinical and clinical studies on the importance and potentially harmful effects of Met supplementation and elaborates on potential clinical applications of supplemental Met with reference to clinical studies performed over the past 20 y. Recently acquired knowledge about the NOAEL (no observed adverse effect level) of 46.3 mg · kg-1 · d-1 and the LOAEL (lowest observed adverse effect level) of 91 mg · kg-1 · d-1 of supplemented Met will guide the design of future studies to further establish the role of Met as a potential (safe) candidate for nutritional supplementation in clinical applications.


Asunto(s)
Compartimentos de Líquidos Corporales/metabolismo , Enfermedades Cardiovasculares/etiología , Suplementos Dietéticos , Homocisteína/metabolismo , Hiperhomocisteinemia/etiología , Metionina , Deficiencia de Vitamina B/complicaciones , Animales , Enfermedades Cardiovasculares/metabolismo , Femenino , Humanos , Hiperhomocisteinemia/metabolismo , Masculino , Metionina/efectos adversos , Metionina/metabolismo , Metionina/farmacología , Metionina/uso terapéutico , Proteínas/metabolismo , Complejo Vitamínico B/sangre , Deficiencia de Vitamina B/sangre
7.
J Neuroinflammation ; 17(1): 274, 2020 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943069

RESUMEN

BACKGROUND: Elevated blood homocysteine levels, termed hyperhomocysteinemia (HHcy), is a prevalent risk factor for Alzheimer's disease (AD) in elderly populations. While dietary supplementation of B-vitamins is a generally effective method to lower homocysteine levels, there is little if any benefit to cognition. In the context of amyloid pathology, dietary-induced HHcy is known to enhance amyloid deposition and certain inflammatory responses. Little is known, however, about whether there is a more specific effect on microglia resulting from combined amyloid and HHcy pathologies. METHODS: The present study used a knock-in mouse model of amyloidosis, aged to 12 months, given 8 weeks of B-vitamin deficiency-induced HHcy to better understand how microglia are affected in this comorbidity context. RESULTS: We found that HHcy-inducing diet increased amyloid plaque burden, altered the neuroinflammatory milieu, and upregulated the expression of multiple damage-associated and "homeostatic" microglial genes. CONCLUSIONS: Taken together, these data indicate complex effects of comorbid pathologies on microglial function that are not driven solely by increased amyloid burden. Given the highly dynamic nature of microglia, their central role in AD pathology, and the frequent occurrence of various comorbidities in AD patients, it is increasingly important to understand how microglia respond to mixed pathological processes.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Técnicas de Sustitución del Gen/métodos , Hiperhomocisteinemia/metabolismo , Microglía/metabolismo , Placa Amiloide/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/patología , Ratones , Ratones Transgénicos , Microglía/patología , Placa Amiloide/genética , Placa Amiloide/patología
8.
Appl Biochem Biotechnol ; 191(3): 1127-1139, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31960366

RESUMEN

Hyperglycemia, the hallmark of diabetes mellitus, is considered one of the endothelial dysfunction risk factors, the main reason of vascular complication. In this study, we aimed to evaluate homocysteine (Hcy) and asymmetrical dimethylarginine (ADMA) levels in diabetic rats and the possibility to attenuate the elevation of these two parameters by supplementation of docosahexaenoic acid (DHA) alone or loaded zinc oxide nanoparticles (ZnONPs) to improve endothelial dysfunction in streptozotocin (STZ)-induced diabetic rats. Forty male albino rats weighing 180-200 g were classified as control, diabetic, diabetic treated with DHA, and diabetic treated with DHA-loaded zinc oxide nanoparticles (DHA/ZnONPs) groups. Fasting blood glucose, insulin, ADMA, Hcy, and nitric oxide (NO) were estimated. Fatty acids (linoleic acid (LA), arachidonic acid (AA), DHA, α-linolenic acid (ALA), and oleic acid (OA)) were also evaluated by reversed phase HPLC using a UV detector. The results showed that fasting blood sugar, insulin resistance, LA, AA, OA, ADMA, and Hcy increased significantly in diabetic rats compared with control while fasting insulin, DHA, ALA, and NO decreased significantly in diabetic rats. In both treated groups, fasting blood sugar, insulin resistance, LA, AA, OA, ADMA, and Hcy significantly decreased as compared with the diabetic group while fasting insulin, DHA, ALA, and NO were significantly increased. In conclusion, DHA and DHA/ZnONP supplementation protect against diabetic complications and improve endothelial dysfunction as well as hyperhomocysteinemia in diabetes. DHA/ZnONP-treated group appeared more efficient than DHA alone.


Asunto(s)
Arginina/análogos & derivados , Diabetes Mellitus Experimental/terapia , Ácidos Docosahexaenoicos/química , Homocisteína/química , Óxido de Zinc/química , Animales , Arginina/química , Celulosa/química , Hiperhomocisteinemia/metabolismo , Insulina/metabolismo , Resistencia a la Insulina , Masculino , Nanopartículas del Metal/química , Microscopía Electrónica de Transmisión , Óxido Nítrico/metabolismo , Ratas , Factores de Riesgo
9.
Br J Pharmacol ; 176(17): 3180-3192, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31140595

RESUMEN

BACKGROUND AND PURPOSE: Hydrogen sulfide donors can block the cardiovascular injury of hyperhomocysteinemia. H2 S also lowers serum homocysteine in rats with mild hyperhomocysteinemia, but the pharmacological mechanism is unknown. The present study investigated the mechanism(s) involved. EXPERIMENTAL APPROACH: ApoE-knockout mice were fed a Paigen diet and L-methionine in drinking water for 16 weeks to create a mouse model of atherosclerosis with hyperhomocysteinemia. H2 S donors (NaHS and GYY4137) were administered by intraperitoneal injection. We also assayed the H2 S produced (by methylene blue assay and mito-HS [H2 S fluorescence probe]), cystathionine γ lyase (CSE) mRNA and protein expression, and CSE sulfhydration and nitrosylation and its activity. KEY RESULTS: H2 S donor treatment significantly lowered atherosclerotic plaque area, macrophage infiltration, and serum homocysteine level in the mouse model of atherosclerosis with co-existing hyperhomocysteinemia. mRNA and protein levels of CSE, a key enzyme catalyzing homocysteine trans-sulfuration, were down-regulated with hyperhomocysteinemia, and CSE catalytic activity was inhibited. All these effects were reversed with H2 S donor treatment. Hyperhomocysteinemia induced CSE nitrosylation, whereas H2 S sulfhydrated CSE at the same cysteine residues. Nitrosylated CSE decreased and sulfhydrated CSE increased its catalytic and binding activities towards L-homocysteine. Mutation of C252, C255, C307, and C310 residues in CSE abolished CSE nitrosylation or sulfhydration and prevented its binding to L-homocysteine. CONCLUSIONS AND IMPLICATIONS: Sulfhydration or nitrosylation of CSE represents a yin/yang regulation of catalysis or binding to L-homocysteine. H2 S donor treatment enhanced CSE sulfhydration, thus lowering serum L-homocysteine, which contributed in part to the anti-atherosclerosis effects in ApoE-knockout mice with hyperhomocysteinemia.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Cistationina gamma-Liasa/metabolismo , Sulfuro de Hidrógeno/farmacología , Hiperhomocisteinemia/tratamiento farmacológico , Animales , Aterosclerosis/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células HEK293 , Células Hep G2 , Humanos , Sulfuro de Hidrógeno/análisis , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/metabolismo , Masculino , Ratones , Ratones Noqueados para ApoE , Relación Estructura-Actividad
11.
Neurosci Bull ; 35(2): 325-335, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30264378

RESUMEN

Hyperhomocysteinemia (Hhcy) is an independent risk factor for Alzheimer's disease (AD). Visual dysfunction is commonly found and is positively correlated with the severity of cognitive defects in AD patients. Our previous study demonstrated that Hhcy induces memory deficits with AD-like tau and amyloid-ß (Aß) pathologies in the hippocampus, and supplementation with folate and vitamin B12 (FB) prevents the Hhcy-induced AD-like pathologies in the hippocampus. Here, we investigated whether Hhcy also induces AD-like pathologies in the retina and the effects of FB. An Hhcy rat model was produced by vena caudalis injection of homocysteine for 14 days, and the effects of FB were assessed by simultaneous supplementation with FB in drinking water. We found that Hhcy induced vessel damage with Aß and tau pathologies in the retina, while simultaneous supplementation with FB remarkably attenuated the Hhcy-induced tau hyperphosphorylation at multiple AD-related sites and Aß accumulation in the retina. The mechanisms involved downregulation of amyloid precursor protein (APP), presenilin-1, beta-site APP-cleaving enzyme 1, and protein phosphatase-2A. Our data suggest that the retina may serve as a window for evaluating the effects of FB on hyperhomocysteinemia-induced Alzheimer-like pathologies.


Asunto(s)
Enfermedad de Alzheimer/terapia , Ácido Fólico/uso terapéutico , Hiperhomocisteinemia/terapia , Retina/patología , Vitamina B 12/uso terapéutico , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Homocisteína , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/patología , Masculino , Ratas Sprague-Dawley , Retina/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Proteínas tau/metabolismo
12.
Neuromolecular Med ; 20(4): 475-490, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30105650

RESUMEN

Homocysteine (Hcy) is a sulfur-containing amino acid derived from methionine metabolism. Elevated plasma Hcy levels (> 15 µM) result in a condition called hyperhomocysteinemia (HHcy), which is an independent risk factor in the development of various neurodegenerative disorders. Reactive oxygen species (ROS) produced by auto-oxidation of Hcy have been implicated in HHcy-associated neurological conditions. Hydrogen sulfide (H2S) is emerging as a potent neuroprotective and neuromodulator molecule. The present study was aimed to evaluate the ability of NaHS (a source of H2S) to attenuate Hcy-induced oxidative stress and altered antioxidant status in animals subjected to HHcy. Impaired cognitive functions assessed by Y-maze and elevated plus maze in Hcy-treated animals were reversed on NaHS administration. Increased levels of ROS, lipid peroxidation, protein carbonyls, and 4-hydroxynonenal (4-HNE)-modified proteins were observed in the cortex and hippocampus of Hcy-treated animals suggesting accentuated oxidative stress. This increase in Hcy-induced oxidative stress was reversed following NaHS supplementation. GSH/GSSG ratio, activity of antioxidant enzymes viz; superoxide dismutase, glutathione peroxidase, glutathione reductase, and glutathione-S-transferase were decreased in Hcy-treated animals. NaHS supplementation, on the otherhand, restored redox ratio and activity of antioxidant enzymes in the brains of animals with HHcy. Further, NaHS administration normalized nuclear factor erythroid 2-related factor 2 expression and acetylcholinesterase (AChE) activity in the brain of Hcy-treated animals. Histopathological studies using cresyl violet indicated higher number of pyknotic neurons in the cortex and hippocampus of HHcy animals, which were reversed by NaHS administration. The results clearly demonstrate that NaHS treatment significantly ameliorates Hcy-induced cognitive impairment by attenuating oxidative stress, improving antioxidant status, and modulating AChE activity thereby suggesting potential of H2S as a therapeutic molecule.


Asunto(s)
Antioxidantes/farmacología , Corteza Cerebral/efectos de los fármacos , Trastornos del Conocimiento/prevención & control , Hipocampo/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Hiperhomocisteinemia/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/fisiología , Fármacos Neuroprotectores/farmacología , Acetilcolinesterasa/metabolismo , Animales , Corteza Cerebral/metabolismo , Trastornos del Conocimiento/etiología , Hipocampo/metabolismo , Homocisteína/toxicidad , Sulfuro de Hidrógeno/uso terapéutico , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/psicología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Oxidación-Reducción , Estrés Oxidativo , Oxidorreductasas/metabolismo , Carbonilación Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
13.
Dig Dis Sci ; 63(12): 3339-3347, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29974377

RESUMEN

BACKGROUND: The hyperhomocysteinaemia (Hhcy) is a common phenomenon observed in patients with inflammatory bowel disease (IBD). Our previous study showed that Hhcy aggravated intestinal inflammation in an animal model of colitis. Increased levels of IL-17 and RORγt were also observed in this animal model of colitis with Hhcy. However, the direct effect of homocysteine on the differentiation of Th17 cells has never been studied. The aim of this study was to investigate the direct effect of Hhcy on the differentiation of CD4+ T cells into Th17 cells. METHOD: Lamina propria lymphocytes (LPLs) in colonic mucosa of Wistar rats were isolated and cultured under Th17-inducing (iTH17) environments. Different concentrations of the Hcy (0-100 µmol/ml) were added alone or combined with IL-23 (100 ng/ml) or folate (5 µmol/ml). The LPLs were divided into eight groups as follows: (1) Control group; (2) 10 µmol/ml Hcy group; (3) 25 µmol/ml Hcy group; (4) 50 µmol/ml Hcy group; (5) 100 µmol/ml Hcy group; (6) 100 ng/ml IL-23 group; (7) 50 µmol/ml Hcy + 100 ng/ml IL-23 group and (8) 50 µmol/ml Hcy + 100 ng/ml IL-23 + 5 µmol/ml folate group. The protein expression levels of IL-17, retinoid-related orphan nuclear receptor-γt (RORγt), p38 MAPK, phosphorylated p38 MAPK, cytosolic phospholipase A2 (cPLA2), phosphorylated-cPLA2 and cyclooxygenase 2 (COX2) were detected by immunoblot analysis. The protein level of prostaglandin E2 (PGE2) and IL-17 was detected by ELISA, and IL-17 and RORγt-positive CD4+ T cells were stained and analyzed by flow cytometry. RESULTS: Hcy increased the protein levels of IL-17, RORγt, the ratio of phosphorylated p38 MAPK to p38 MAPK (p-p38/p38), the ratio of phosphorylated cPLA2 to cPLA2 (p-cPLA2/cPLA2) and COX2. The effect was concentration dependent to a certain degree; Hcy of 50 µmol/ml was the optimal concentration to increase the protein levels of those molecules. The level of IL-17 and PGE2 in the cell culture supernatants and the expression of IL-17 and RORγt in positive CD4+ T cells were also increased in the group of Hhcy. IL-23 showed a cooperative effect with Hcy on the differentiation of CD4+ Th cells into Th17 cells, whereas folate supplementation showed an inhibition action. CONCLUSIONS: Homocysteine promoted the differentiation of CD4+ T cells into Th17 cells in a dose-dependant manner. This effect could be inhibited by folate.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Diferenciación Celular , Ácido Fólico/farmacología , Homocisteína/metabolismo , Hiperhomocisteinemia , Enfermedades Inflamatorias del Intestino , Células Th17/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Colon/metabolismo , Colon/patología , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Fosfolipasas A2 Grupo IV/metabolismo , Hiperhomocisteinemia/tratamiento farmacológico , Hiperhomocisteinemia/etiología , Hiperhomocisteinemia/metabolismo , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/metabolismo , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Ratas , Ratas Wistar , Resultado del Tratamiento , Complejo Vitamínico B/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
J Biol Chem ; 293(15): 5544-5555, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29414770

RESUMEN

S-Adenosyl-l-homocysteine hydrolase (AdoHcy hydrolase; Sah1 in yeast/AHCY in mammals) degrades AdoHcy, a by-product and strong product inhibitor of S-adenosyl-l-methionine (AdoMet)-dependent methylation reactions, to adenosine and homocysteine (Hcy). This reaction is reversible, so any elevation of Hcy levels, such as in hyperhomocysteinemia (HHcy), drives the formation of AdoHcy, with detrimental consequences for cellular methylation reactions. HHcy, a pathological condition linked to cardiovascular and neurological disorders, as well as fatty liver among others, is associated with a deregulation of lipid metabolism. Here, we developed a yeast model of HHcy to identify mechanisms that dysregulate lipid metabolism. Hcy supplementation to wildtype cells up-regulated cellular fatty acid and triacylglycerol content and induced a shift in fatty acid composition, similar to changes observed in mutants lacking Sah1. Expression of the irreversible bacterial pathway for AdoHcy degradation in yeast allowed us to dissect the impact of AdoHcy accumulation on lipid metabolism from the impact of elevated Hcy. Expression of this pathway fully suppressed the growth deficit of sah1 mutants as well as the deregulation of lipid metabolism in both the sah1 mutant and Hcy-exposed wildtype, showing that AdoHcy accumulation mediates the deregulation of lipid metabolism in response to elevated Hcy in yeast. Furthermore, Hcy supplementation in yeast led to increased resistance to cerulenin, an inhibitor of fatty acid synthase, as well as to a concomitant decline of condensing enzymes involved in very long-chain fatty acid synthesis, in line with the observed shift in fatty acid content and composition.


Asunto(s)
Adenosilhomocisteinasa/metabolismo , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos , S-Adenosilhomocisteína/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Adenosilhomocisteinasa/genética , Ácidos Grasos/genética , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Modelos Biológicos , Mutación , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
15.
Curr Alzheimer Res ; 15(1): 89-99, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28847282

RESUMEN

BACKGROUND: Ginkgo biloba extract EGb761 has shown the neuroprotective effects on Alzheimer's disease (AD) through the protection against the Aß-induced neurotoxicity. However, it is not completedly clear whether EGb761 attenuates tau hyperphosphorylation, another of the most prominent mechanisms underlying the pathology of AD. METHODS: we employed hyperhomocysteinemia (HHcy) to mimic AD like pathological alterations and memory deficits in rats as model, and injected EGb761 with or after HHcy injection as prevention and treatment, injected saline as control. We measured the status of oxidative damage and spatial and learning memory in rats. Then we detected the level of memory-related proteins, tau phosphorylation and the level and activity of tau kinase (GSK-3ß) and phosphatase (PP2A) by Western blotting and Immunohistochemistry. RESULTS: We found that EGb761 could significantly antagonize HHcy-induced oxidative damage, recover PP2Ac and GSK3ß activities deregulated by HHcy. Furthermore, tau was hyperphosphorylated at Thr231, Ser262, Ser396, and Ser404, most common PP2Ac and GSK3ß targeted sites in the hippocampus and prefrontal cortex of HHcy rats, whereas EGb761 recovered the tau phosphorylation at those sites. Behavioral tests revealed that EGb761 rescued HHcy-induced spatial reference memory deficit and upregulated the expression of synapse-associated protein PSD95 and synapsin-1. CONCLUSION: EGb761 might be a promising drug to treat AD through its anti-oxidative activity and decreasing tau hyperphosphorylation besides the protection against the Aß-induced neurotoxicity.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Hiperhomocisteinemia/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Extractos Vegetales/farmacología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Ginkgo biloba , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/patología , Hiperhomocisteinemia/psicología , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Sinapsinas/metabolismo
16.
Amino Acids ; 50(1): 3-9, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29018979

RESUMEN

Hyperhomocysteinemia is an independent risk factor for cardiovascular disease and is associated with primary causes of mortality and morbidity throughout the world. Several studies have been carried out to evaluate the effects of a diet inducing cystathionine-ß-synthase, methyltetrafolate, folic acid, and vitamin B supplemented with methionine on the homocysteine metabolism and in lowering the plasma total homocysteine levels. A large number of molecular and biomedical studies in numerous animals, such as mice, rabbits, and pigs, have sought to elevate the plasma total homocysteine levels and to identify a disease model for human hyperhomocysteinemia. However, a specific animal model is not suitable for hyperhomocysteinemia in terms of all aspects of cardiovascular disease. In this review article, the experimental progress of animal models with plasma total homocysteine levels is examined to identify a feasible animal model of hyperhomocysteinemia for different aspects.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Modelos Animales de Enfermedad , Hiperhomocisteinemia/metabolismo , Animales , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Homocisteína/biosíntesis , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/enzimología , Hiperhomocisteinemia/fisiopatología , Metionina/metabolismo , Factores de Riesgo
17.
Br J Pharmacol ; 175(8): 1173-1189, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28836260

RESUMEN

Homocysteine is a sulphur-containing non-proteinogenic amino acid. Hyperhomocysteinaemia (HHcy), the pathogenic elevation of plasma homocysteine as a result of an imbalance of its metabolism, is an independent risk factor for various vascular diseases, such as atherosclerosis, hypertension, vascular calcification and aneurysm. Treatments aimed at lowering plasma homocysteine via dietary supplementation with folic acids and vitamin B are more effective in preventing vascular disease where the population has a normally low folate consumption than in areas with higher dietary folate. To date, the mechanisms of HHcy-induced vascular injury are not fully understood. HHcy increases oxidative stress and its downstream signalling pathways, resulting in vascular inflammation. HHcy also causes vascular injury via endoplasmic reticulum stress. Moreover, HHcy up-regulates pathogenic genes and down-regulates protective genes via DNA demethylation and methylation respectively. Homocysteinylation of proteins induced by homocysteine also contributes to vascular injury by modulating intracellular redox state and altering protein function. Furthermore, HHcy-induced vascular injury leads to neuronal damage and disease. Also, an HHcy-activated sympathetic system and HHcy-injured adipose tissue also cause vascular injury, thus demonstrating the interactions between the organs injured by HHcy. Here, we have summarized the recent developments in the mechanisms of HHcy-induced vascular injury, which are further considered as potential therapeutic targets in this condition. LINKED ARTICLES: This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.


Asunto(s)
Hiperhomocisteinemia/complicaciones , Lesiones del Sistema Vascular/etiología , Animales , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/metabolismo , Lesiones del Sistema Vascular/metabolismo
18.
Int J Immunopathol Pharmacol ; 30(4): 371-382, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29171783

RESUMEN

It has been reported that hyperhomocysteinemia (HHcy) is associated with neurodegenerative and cardiovascular diseases. However, little is known about brain histomorphology, neuronal organelles, and hairy enhancer of split ( hes) expression under HHcy. In this study, non-HHcy and HHcy induced by high-methionine diet in apolipoprotein E-deficient (Apo E-/-) mice were comparatively investigated. The histomorphology, ultrastructure, autophagosomes, apoptosis, and expression of proteins, HES1, HES5 and P62, were designed to assess the effects of HHcy on brain. The results showed that compared to the non-HHcy mice, the HHcy group had an increase in autophagosomes, vacuolization in mitochondria, and neuron apoptosis; treatment with folate and vitamin B12 reduced the extent of these lesions. However, the elementary histomorphology, the numbers of cortical neurons, and Nissl bodies had no significant difference between the HHcy and the non-HHcy groups or the group treated with folate and vitamin B12. Immunohistochemistry and immunofluorescence demonstrated a decrease in HES1- or HES5-positive neurons in the HHcy group when compared to the non-HHcy groups, wild-type, and Apo E-/- controls, or the HHcy mice with folate and vitamin B12 supplement. Western blots showed that HHcy induced a decreased expression of HES1 and HES5, or P62, in which the expression of HES1 and P62 was elevated by treating with folate and vitamin B12 supplement. These results suggest that HHcy-enhanced brain damage is associated with increased autophagy and neuronal apoptosis in Apo E-/- mice, in which downregulation of hes1 and hes5 is involved.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hiperhomocisteinemia/metabolismo , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Factor de Transcripción HES-1/metabolismo , Animales , Apoptosis , Autofagia , Regulación hacia Abajo , Lóbulo Frontal/metabolismo , Lóbulo Frontal/ultraestructura , Homocisteína/sangre , Hiperhomocisteinemia/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Microscopía Electrónica de Transmisión , Neuronas/ultraestructura
19.
J Med Case Rep ; 11(1): 250, 2017 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-28863787

RESUMEN

BACKGROUND: Hyperhomocysteinemia has been suspected of favoring thrombosis. Several case-control studies and even a meta-analysis have confirmed a link between venous thrombosis and hyperhomocysteinemia. Homocysteine is due to genetic and acquired factors (poor diet in folate and vitamin B12, older age, renal impairment, thyroid diseases, and malignancies) induced by the intake and the concentrations of vitamin B9 or B12 in the majority of cases. CASES PRESENTATION: We report the cases of four Moroccan patients who presented with acute vein thrombosis of different sites: a 34-year-old man, a 60-year-old man, a 58-year-old man, and a 47-year-old woman. All patients had a low level of cobalamin with marked hyperhomocysteinemia with normal serum and red cell folic acid. Venous thrombosis revealed pernicious anemia in all patients. Their low levels of cobalamin, atrophic gastritis, and positive results for gastric parietal cell antibodies confirmed the diagnosis of pernicious anemia. There was no evidence of immobilization, recent surgery, malignancy, antiphospholipid antibody, myeloproliferative disorder, or hormone replacement therapy. No deficiencies in protein C and protein S were detected; they had normal antithrombin III function and factor V Leiden; no prothrombin gene mutations were detected. Treatment included orally administered anticoagulation therapy and cobalamin supplementation. The outcome was favorable in all cases. CONCLUSIONS: These reports demonstrate that pernicious anemia, on its own, can lead to hyperhomocysteinemia that is significant enough to lead to thrombosis. Understanding the molecular pathogenesis of the development of thrombosis in patients with hyperhomocysteinemia related to Biermer disease would help us to identify patients at risk and to treat them accordingly. The literature concerning the relationship between homocysteine and venous thrombosis is briefly reviewed.


Asunto(s)
Anemia Perniciosa , Anticoagulantes/administración & dosificación , Células Parietales Gástricas/inmunología , Tromboembolia Venosa , Vitamina B 12 , Adulto , Anemia Perniciosa/sangre , Anemia Perniciosa/complicaciones , Anemia Perniciosa/diagnóstico , Anemia Perniciosa/terapia , Anticuerpos/sangre , Femenino , Ácido Fólico/sangre , Humanos , Hiperhomocisteinemia/diagnóstico , Hiperhomocisteinemia/etiología , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/terapia , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/etiología , Tromboembolia Venosa/metabolismo , Tromboembolia Venosa/terapia , Vitamina B 12/administración & dosificación , Vitamina B 12/sangre , Deficiencia de Vitamina B 12/diagnóstico , Deficiencia de Vitamina B 12/etiología , Vitaminas/administración & dosificación
20.
Congenit Anom (Kyoto) ; 57(5): 142-149, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28598562

RESUMEN

Moderate hyperhomocysteinemia-induced low folate status is an independent risk factor for cardiovascular disease, dementia, and depression. Folate is an essential cofactor in the one-carbon metabolism pathway and is necessary in amino acid metabolism, purine and thymidylate synthesis, and DNA methylation. In the folate cycle and homocysteine metabolism, folate, vitamin B12, vitamin B6, and vitamin B2 are important cofactors. Many enzymes are involved in folate transport and uptake, the folate pathway, and homocysteine (Hcy) metabolism, and various polymorphisms have been documented in these enzymes. Serum folate and total Hcy (tHcy) levels are influenced by folate intake and genetic polymorphisms in 5,10-methylenetertahydrofolate reductase (MTHFR) such as C677T. The prevalence of the MTHFR 677TT genotype varies across ethnic groups and regions, with a frequency of approximately 15% in Japanese populations. Individuals with the TT genotype have significantly higher tHcy levels and lower folate levels in serum than those with the CT and TT genotypes. However, administration of folic acid has been shown to eliminate these differences. Moreover, data have suggested that interventions based on genotype may be effective for motivating individuals to change their lifestyle and improve their nutrition status. Accordingly, in this review, we discuss the effects of MTHFR C677T polymorphisms on serum tHcy and folate levels with folic acid intervention and evaluate approaches for overcoming folic acid deficiency and related symptoms.


Asunto(s)
Suplementos Dietéticos , Deficiencia de Ácido Fólico/prevención & control , Ácido Fólico/sangre , Hiperhomocisteinemia/prevención & control , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo Genético , Vitamina B 12/sangre , Femenino , Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/fisiopatología , Expresión Génica , Genotipo , Humanos , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/fisiopatología , Masculino , Redes y Vías Metabólicas , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA