Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 19438, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37945738

RESUMO

To provide a theoretical basis for the prevention and treatment of atherosclerosis (As), the current study aimed to investigate the mechanism underlying the effect of homocysteine (Hcy) on inducing the lipid deposition and foam cell formation of the vascular smooth muscle cell (VSMC) via C1q/Tumor necrosis factor-related protein9 (CTRP9) promoter region Hypermethylation negative regulating endoplasmic reticulum stress (ERs). Therefore, apolipoprotein E deficient (ApoE-/-) mice were randomly divided into the control [ApoE-/- + normal diet (NC)] and high methionine [ApoE-/- + (normal diet supplemented with 1.7% methionine (HMD)] groups (n = 6 mice/group). Following feeding for 15 weeks, the serum levels of Homocysteine (Hcy), total cholesterol (TC), and triglyceride (TG) were measured using an automatic biochemical analyzer. HE and oil red O staining were performed on the aorta roots to observe the pathological changes. Additionally, immunofluorescence staining was performed to detect the protein expression levels of CTRP9, glucose-regulated protein 78 kD (GRP78), phosphorylated protein kinase RNA-like ER kinase (p-PERK), activating transcription factor 6a (ATF6a), phosphorylated inositol-requiring enzyme-1α (p-IRE1α), sterol regulatory element binding proteins-1c (SREBP1c) and sterol regulatory element binding proteins-2 (SREBP2) in VSMC derived from murine aortic roots. In vitro, VSMC was stimulated with 100 µmol/l Hcy. After transfection of plasmids with overexpression and interference of CTRP9, ERs agonist (TM) and inhibitor (4-PBA) were given to stimulate VSMC cells. HE staining and oil red O staining were used to observe the effect of Hcy stimulation on lipid deposition in VSMC. Additionally, The mRNA and protein expression levels of CTRP9, GRP78, PERK, ATF6a, IRE1α, SREBP1c, and SREBP2 in VSMC were detected by RT-qPCR and western blot analysis, respectively. Finally, The methylation modification of the CTRP9 promoter region has been studied. The NCBI database was used to search the promoter region of the CTRP9 gene, and CpG Island was used to predict the methylation site. After Hcy stimulation of VSMC, overexpression of DNMT1, and intervention with 5-Azc, assess the methylation level of the CTRP9 promoter through bisulfite sequencing PCR (BSP). The results showed that the serum levels of Hcy, TC, and TG in the ApoE-/- + HMD group were significantly increased compared with the ApoE-/- + NC group. In addition, HE staining and oil red O staining showed obvious AS plaque formation in the vessel wall, and a large amount of fat deposition in VSMC, thus indicating that the hyperhomocysteinemia As an animal model was successfully established. Furthermore, CTRP9 were downregulated, while GRP78, p-PERK, ATF6a, p-IRE1α, SREBP1c, SREBP2 was upregulated in aortic VSMC in the ApoE-/- + HMD group. Consistent with the in vivo results, Hcy can inhibit the expression of CTRP9 in VSMC and induce ERs and lipid deposition in VSMC. Meanwhile, the increased expression of CTRP9 can reduce ERs and protect the lipid deposition in Hcy induced VSMC. Furthermore, ERs can promote Hcy induced VSMC lipid deposition, inhibition of ERs can reduce Hcy induced VSMC lipid deposition, and CTRP9 may play a protective role in Hcy induced VSMC lipid deposition and foam cell transformation through negative regulation of ERs. In addition, The CTRP9 promoter in the Hcy group showed hypermethylation. At the same time as Hcy intervention, overexpression of DNMT1 increases the methylation level of the CTRP9 promoter, while 5-Azc can reduce the methylation level of the CTRP9 promoter. Finally, Hcy can up-regulate the expression of DNMT1 and down-regulate the expression of CTRP9. After overexpression of DNMT1, the expression of CTRP9 is further decreased. After 5-Azc inhibition of DNMT1, the expression of DNMT1 decreases, while the expression of CTRP9 increases. It is suggested that the molecular mechanism of Hcy inhibiting the expression of CTRP9 is related to the hypermethylation of the CTRP9 promoter induced by Hcy and regulated by DNMT1. 5-Azc can inhibit the expression of DNMT1 and reverse the regulatory effect of DNMT1 on CTRP9. Overall, the results of the present study suggested that Hcy induces DNA hypermethylation in the CTRP9 promoter region by up-regulating DNMT1 expression, and negatively regulates ERs mediated VSMC lipid deposition and foam cell formation. CTRP9 may potentially be a therapeutic target in the treatment of hyperhomocysteinemia and As.


Assuntos
Aterosclerose , Hiper-Homocisteinemia , Camundongos , Animais , Endorribonucleases/metabolismo , Chaperona BiP do Retículo Endoplasmático , Músculo Liso Vascular/metabolismo , Células Espumosas/metabolismo , Hiper-Homocisteinemia/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Aterosclerose/metabolismo , Regiões Promotoras Genéticas , Metionina/metabolismo , Apolipoproteínas E/metabolismo , Lipídeos/farmacologia , Homocisteína/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Estresse do Retículo Endoplasmático
2.
Rev Port Cardiol ; 41(10): 813-819, 2022 10.
Artigo em Inglês, Português | MEDLINE | ID: mdl-36210587

RESUMO

OBJECTIVES: Hyperhomocysteinemia (HHcy) can induce vascular inflammatory and oxidative damage and accelerate intimal hyperplasia. This study investigated the protective effect of pirfenidone (PFD) on the recovery process of injured endothelial arteries during HHcy. MATERIALS AND METHODS: Thirty rabbits were randomly separated into three groups: A control group (n=10, standard rabbit chow), a model group (n=10, control diet plus 30 g methionine/kg food), and a PFD group (n=10, model diet plus oral administration of 90 mg/day of PFD). After 14 weeks of arterial injury, histopathological changes were determined. Plasma homocysteine (Hcy) concentrations, lipid profiles and oxidant and antioxidant status were evaluated. Macrophage infiltration was assessed using immunohistochemical staining. RESULTS: PFD supplementation decreased macrophage infiltration of iliac artery significantly without changes in blood lipids and Hcy concentrations. Compared with the model group, PFD restored superoxide dismutase and glutathione peroxidase activities and reduced malondialdehyde and reactive oxygen species levels. A high-methionine diet significantly increased neointimal area and the ratio between neointimal and media area. Systemic administration of PFD inhibited neointimal formation. CONCLUSIONS: PFD can partly alleviate intimal hyperplasia by inhibiting inflammatory and oxidative stress response induced by HHcy during endothelial injury. It may be a potential therapeutic agent for the prevention and treatment of endothelial injury-associated diseases such as atherosclerosis.


Assuntos
Hiper-Homocisteinemia , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Glutationa Peroxidase/farmacologia , Glutationa Peroxidase/uso terapêutico , Homocisteína/farmacologia , Homocisteína/uso terapêutico , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/tratamento farmacológico , Hiper-Homocisteinemia/patologia , Hiperplasia/patologia , Lipídeos , Malondialdeído/farmacologia , Metionina/farmacologia , Metionina/uso terapêutico , Oxidantes/farmacologia , Oxidantes/uso terapêutico , Piridonas , Coelhos , Espécies Reativas de Oxigênio/farmacologia , Espécies Reativas de Oxigênio/uso terapêutico , Superóxido Dismutase/farmacologia , Superóxido Dismutase/uso terapêutico , Túnica Íntima/patologia
3.
Curr Med Sci ; 41(3): 548-554, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34169425

RESUMO

Ligustrazine, an alkaloid extracted from the traditional Chinese herbal medicine Ligusticum Chuanxiong Hort, has been clinically applied to treat the cerebrovascular diseases. Hyperhomocysteinemia (Hhcy) is an independent risk factor for Alzheimer's disease (AD). Memory deficits can be caused by Hhcy via pathologies of AD-like tau and amyloid-ß (Aß) in the hippocampus. Here, we investigated whether homocysteine (Hcy) can induce AD-like pathologies and the effects of ligustrazine on these pathologies. The Hcy rat model was constructed by 14-day Hcy injection via vena caudalis, and rats were treated with daily intragastric administration of ligustrazine at the same time. We found that the pathologies of tau and Aß were induced by Hcy in the hippocampus, while the Hcy-induced tau hyperphosphorylation and Aß accumulation could be markedly attenuated by simultaneous ligustrazine treatment. Our data demonstrate that ligustrazine may be used as a promising neuroprotective agent to treat the Hcy-induced AD-like pathologies.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Hiper-Homocisteinemia/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Pirazinas/farmacologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/patologia , Transtornos da Memória/etiologia , Transtornos da Memória/genética , Transtornos da Memória/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
4.
J Neuroinflammation ; 17(1): 274, 2020 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-32943069

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Técnicas de Introdução de Genes/métodos , Hiper-Homocisteinemia/metabolismo , Microglia/metabolismo , Placa Amiloide/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/patologia , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/patologia , Camundongos , Camundongos Transgênicos , Microglia/patologia , Placa Amiloide/genética , Placa Amiloide/patologia
5.
J Pathol ; 248(3): 291-303, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30734924

RESUMO

The pathomechanisms that associate a deficit in folate and/or vitamin B12 and the subsequent hyperhomocysteinemia with pathological brain ageing are unclear. We investigated the homocysteinylation of microtubule-associated proteins (MAPs) in brains of patients with Alzheimer's disease or vascular dementia, and in rats depleted in folate and vitamin B12, Cd320 KO mice with selective B12 brain deficiency and H19-7 neuroprogenitors lacking folate. Compared with controls, N-homocysteinylated tau and MAP1 were increased and accumulated in protein aggregates and tangles in the cortex, hippocampus and cerebellum of patients and animals. N-homocysteinylation dissociated tau and MAPs from ß-tubulin, and MS analysis showed that it targets lysine residues critical for their binding to ß-tubulin. N-homocysteinylation increased in rats exposed to vitamin B12 and folate deficit during gestation and lactation and remained significantly higher when they became 450 days-old, despite returning to normal diet at weaning, compared with controls. It was correlated with plasma homocysteine (Hcy) and brain expression of methionine tRNAsynthetase (MARS), the enzyme required for the synthesis of Hcy-thiolactone, the substrate of N-homocysteinylation. Experimental inactivation of MARS prevented the N-homocysteinylation of tau and MAP1, and the dissociation of tau and MAP1 from ß-tubulin and PSD95 in cultured neuroprogenitors. In conclusion, increased N-homocysteinylation of tau and MAP1 is a mechanism of brain ageing that depends on Hcy concentration and expression of MARS enzyme. Its irreversibility and cumulative occurrence throughout life may explain why B12 and folate supplementation of the elderly has limited effects, if any, to prevent pathological brain ageing and cognitive decline. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Doença de Alzheimer/patologia , Demência Vascular/patologia , Hiper-Homocisteinemia/patologia , Proteínas tau/metabolismo , Envelhecimento/fisiologia , Doença de Alzheimer/metabolismo , Animais , Autopsia/métodos , Encéfalo/metabolismo , Encéfalo/patologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Demência Vascular/metabolismo , Feminino , Humanos , Camundongos Knockout , Ratos
6.
Neurosci Bull ; 35(2): 325-335, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30264378

RESUMO

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.


Assuntos
Doença de Alzheimer/terapia , Ácido Fólico/uso terapêutico , Hiper-Homocisteinemia/terapia , Retina/patologia , Vitamina B 12/uso terapêutico , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Suplementos Nutricionais , Modelos Animais de Doenças , Homocisteína , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Masculino , Ratos Sprague-Dawley , Retina/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Proteínas tau/metabolismo
7.
Curr Alzheimer Res ; 15(1): 89-99, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28847282

RESUMO

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.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Hiper-Homocisteinemia/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Nootrópicos/farmacologia , Extratos Vegetais/farmacologia , Proteínas tau/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large/metabolismo , Ginkgo biloba , Glicogênio Sintase Quinase 3 beta/metabolismo , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/psicologia , Masculino , Memória/efeitos dos fármacos , Memória/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos Sprague-Dawley , Sinapsinas/metabolismo
8.
J Cell Biochem ; 118(9): 2921-2932, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28230279

RESUMO

Folate deficiency is a known risk factor for liver injury; however, the underlying mechanism remains unclear. In this study, we employed a high homocysteine-induced liver injury model of Apolipoprotein E-deficient (ApoE-/- ) mice fed high-methionine diet and found that high homocysteine induced endoplasmic reticulum (ER) stress and liver cell apoptosis by downregulation of cystic fibrosis transmembrane conductance regulator (CFTR) expression; observations that were attenuated with supplementation of dietary folate. The regulation on CFTR expression was mediated by CFTR promoter methylation and trimethylation of lysine 27 on histone H3 (H3K27me3). Mechanistically, folate inhibited homocysteine-induced CFTR promoter methylation and H3K27me3, which resulted in upregulation of CFTR expression, and reduced ER stress and liver cell apoptosis. Further study showed that folate inhibited the expression of DNA methyltransferase 1 and enhancer of zeste homolog 2, downregulated the cellular concentrations of S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) and upregulated the SAM/SAH ratio, leading to the inhibition of Hcy-induced DNA hypermethylation and H3K27me3 in CFTR promoter. In conclusion, our results provide insight into the protective role of folate in homocysteine-induced ER stress and liver cell apoptosis through the regulation of CFTR expression. J. Cell. Biochem. 118: 2921-2932, 2017. © 2017 Wiley Periodicals, Inc. HIGHLIGHTS: Folate protects hepatocytes of hyperhomocysteinemia mice from apoptosis. Folate alleviates Hcy-induced hepatocyte apoptosis. Folate inhibits Hcy-induced ER stress via upregulation of CFTR expression in hepatocytes. Folate inhibits Hcy-induced methylation of CFTR promotor and H3K27me3.


Assuntos
Apoptose/efeitos dos fármacos , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Metilação de DNA/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ácido Fólico/farmacologia , Hepatócitos/metabolismo , Hiper-Homocisteinemia/metabolismo , Regiões Promotoras Genéticas , Animais , Apoptose/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Metilação de DNA/genética , Estresse do Retículo Endoplasmático/genética , Hepatócitos/patologia , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/patologia , Camundongos , Camundongos Knockout
9.
Cell Death Dis ; 7(12): e2513, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929536

RESUMO

Hyperhomocysteinemia (HHcy) is a well-known risk factor for stroke; however, its underlying molecular mechanism remains unclear. Using both mouse and cell culture models, we have provided evidence that impairment of autophagy has a central role in HHcy-induced cellular injury in the mouse brain. We observed accumulation of LC3B-II and p62 that was associated with increased MTOR signaling in human and mouse primary astrocyte cell cultures as well as a diet-induced mouse model of HHcy, HHcy decreased lysosomal membrane protein LAMP2, vacuolar ATPase (ATP6V0A2), and protease cathepsin D, suggesting that lysosomal dysfunction also contributed to the autophagic defect. Moreover, HHcy increased unfolded protein response. Interestingly, Vitamin B supplementation restored autophagic flux, alleviated ER stress, and reversed lysosomal dysfunction due to HHCy. Furthermore, the autophagy inducer, rapamycin was able to relieve ER stress and reverse lysosomal dysfunction caused by HHcy in vitro. Inhibition of autophagy by HHcy exacerbated cellular injury during oxygen and glucose deprivation and reperfusion (OGD/R), and oxidative stress. These effects were prevented by Vitamin B co-treatment, suggesting that it may be helpful in relieving detrimental effects of HHcy in ischemia/reperfusion or oxidative stress. Collectively, these findings show that Vitamin B therapy can reverse defects in cellular autophagy and ER stress due to HHcy; and thus may be a potential treatment to reduce ischemic damage caused by stroke in patients with HHcy.


Assuntos
Autofagia/efeitos dos fármacos , Suplementos Nutricionais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hiper-Homocisteinemia/patologia , Vitamina B 12/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Dieta , Ácido Fólico/farmacologia , Glucose/deficiência , Humanos , Hiper-Homocisteinemia/tratamento farmacológico , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Oxigênio , Traumatismo por Reperfusão/patologia , Proteína Sequestossoma-1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Vitamina B 12/uso terapêutico
10.
Int J Biochem Cell Biol ; 67: 158-66, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26117455

RESUMO

Transmethylation reactions utilize S-adenosylmethionine (SAM) as a methyl donor and are central to the regulation of many biological processes: more than fifty SAM-dependent methyltransferases methylate a broad spectrum of cellular compounds including DNA, histones, phospholipids and other small molecules. Common to all SAM-dependent transmethylation reactions is the release of the potent inhibitor S-adenosylhomocysteine (SAH) as a by-product. SAH is reversibly hydrolyzed to adenosine and homocysteine by SAH hydrolase. Hyperhomocysteinemia is an independent risk factor for cardiovascular disease. However, a major unanswered question is if homocysteine is causally involved in disease pathogenesis or simply a passive and indirect indicator of a more complex mechanism. A chronic elevation in homocysteine levels results in a parallel increase in intracellular or plasma SAH, which is a more sensitive biomarker of cardiovascular disease than homocysteine and suggests that SAH is a critical pathological factor in homocysteine-associated disorders. Previous reports indicate that supplementation with folate and B vitamins efficiently lowers homocysteine levels but not plasma SAH levels, which possibly explains the failure of homocysteine-lowering vitamins to reduce vascular events in several recent clinical intervention studies. Furthermore, more studies are focusing on the role and mechanisms of SAH in different chronic diseases related to hyperhomocysteinemia, such as cardiovascular disease, kidney disease, diabetes, and obesity. This review summarizes the current role of SAH in cardiovascular disease and its effect on several related risk factors. It also explores possible the mechanisms, such as epigenetics and oxidative stress, of SAH. This article is part of a Directed Issue entitled: Epigenetic dynamics in development and disease.


Assuntos
Aterosclerose/metabolismo , Epigênese Genética , S-Adenosil-Homocisteína/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Estresse Oxidativo , S-Adenosilmetionina/metabolismo
11.
J Inherit Metab Dis ; 38(5): 863-72, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25633902

RESUMO

In the folate cycle MTHFD1, encoded by MTHFD1, is a trifunctional enzyme containing 5,10-methylenetetrahydrofolate dehydrogenase, 5,10-methenyltetrahydrofolate cyclohydrolase and 10-formyltetrahydrofolate synthetase activity. To date, only one patient with MTHFD1 deficiency, presenting with hyperhomocysteinemia, megaloblastic anaemia, hemolytic uremic syndrome (HUS) and severe combined immunodeficiency, has been identified (Watkins et al J Med Genet 48:590-2, 2011). We now describe four additional patients from two different families. The second patient presented with hyperhomocysteinemia, megaloblastic anaemia, HUS, microangiopathy and retinopathy; all except the retinopathy resolved after treatment with hydroxocobalamin, betaine and folinic acid. The third patient developed megaloblastic anaemia, infection, autoimmune disease and moderate liver fibrosis but not hyperhomocysteinemia, and was successfully treated with a regime that included and was eventually reduced to folic acid. The other two, elder siblings of the third patient, died at 9 weeks of age with megaloblastic anaemia, infection and severe acidosis and had MTFHD1 deficiency diagnosed retrospectively. We identified a missense mutation (c.806C > T, p.Thr296Ile) and a splice site mutation (c.1674G > A) leading to exon skipping in the second patient, while the other three harboured a missense mutation (c.146C > T, p.Ser49Phe) and a premature stop mutation (c.673G > T, p.Glu225*), all of which were novel. Patient fibroblast studies revealed severely reduced methionine formation from [(14)C]-formate, which did not increase in cobalamin supplemented culture medium but was responsive to folic and folinic acid. These additional cases increase the clinical spectrum of this intriguing defect, provide in vitro evidence of disturbed methionine synthesis and substantiate the effectiveness of folic or folinic acid treatment.


Assuntos
Ácido Fólico/uso terapêutico , Leucovorina/uso terapêutico , Metilenotetra-Hidrofolato Desidrogenase (NADP)/deficiência , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Anemia Megaloblástica/tratamento farmacológico , Anemia Megaloblástica/genética , Anemia Megaloblástica/patologia , Células Cultivadas , Evolução Fatal , Feminino , Deficiência de Ácido Fólico/tratamento farmacológico , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/patologia , Humanos , Hiper-Homocisteinemia/tratamento farmacológico , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/patologia , Lactente , Recém-Nascido , Masculino , Antígenos de Histocompatibilidade Menor , Imunodeficiência Combinada Severa/tratamento farmacológico , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/patologia , Adulto Jovem
12.
Eur J Pharmacol ; 714(1-3): 163-9, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23792140

RESUMO

This study was designed to investigate the effect and mechanism of allicin on hyperhomocysteinemia-induced experimental vascular endothelial dysfunction in rats. Fifty male Wistar rats were randomly divided into five groups: the normal control rats (NC), the high-methionine-diet rats (Met), the high-methionine-diet rats treated with folic acid, vitaminB6 and vitaminB12 (Met+F), or with low-dose allicin (Met+L), or with high-dose allicin (Met+H). After 6 weeks, we collected blood samples of all groups to determine plasma endothelin (ET), serum homocysteine (Hcy), nitric oxide (NO), superoxide dismutase (SOD), malondialdehyde (MDA), and detected the expression of basic fibroblast growth factors (bFGF), transforming growth factor beta (TGF-ß), tumor necrosis factor-alpha (TNF-α), and intercellular adhesion molecule-1 (ICAM-1) in the aorta. The Hcy and the expression of TGF-ß in both the Met+L and Met+H groups were significantly lower than the Met and Met+F groups. The ET, ET/NO ratio and the MDA levels of the Met+L and Met+H groups were significantly lower than the Met group. The SOD and NO levels and the expression of bFGF, TNF-α and ICAM-1 of the Met+L and Met+H groups were significantly higher than the Met group. Our data indicate that allicin inhibits lipid peroxidation induced by hyperhomocysteinemia and regulates the excretion and equilibrium of ET and NO, and suggest that allicin might be useful in the prevention of endothelial dysfunction caused by hyperhomocysteinemia.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Ácidos Sulfínicos/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Dissulfetos , Endotelinas/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Homocisteína/sangue , Hiper-Homocisteinemia/sangue , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Malondialdeído/sangue , Óxido Nítrico/metabolismo , Ratos , Ratos Wistar , Superóxido Dismutase/sangue , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
13.
Nutrition ; 29(4): 661-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23410631

RESUMO

OBJECTIVE: The aim of this study was to determine the effects of quercetin on homocysteine (Hcy) metabolism and hepatic antioxidant status in high methionine (Met)-fed rats. METHODS: Rats were fed for 6 wk the following diets: control, 1.0% Met, 1.0% Met and 0.1% quercetin, 1.0% Met and 0.5% quercetin, 1.0% Met and 2.5% quercetin-supplemented diets. Serum Hcy, Met, cysteine, serine, taurine, glutathione (GSH), quercetin and its metabolites, and activities of alanine transaminase (ALT) and aspartate transaminase (AST) were assayed. Hepatic malondialdehyde, GSH and carbonyls, and activity of superoxide dismutase and ferric-reducing antioxidant power also were measured. RESULTS: Serum Hcy was increased significantly after Met treatment and decreased after quercetin supplementation. Meanwhile, serum taurine was increased and serine decreased. However, the content of GSH in serum and liver was decreased in the quercetin-supplemented groups and activities of serum ALT and AST were enhanced in the 1.0% Met and 2.5% quercetin-supplemented groups. CONCLUSIONS: Quercetin is effective in decreasing serum Hcy level in high Met-fed rats and one of possible mechanisms is associated with increased transsulfuration of Hcy. Quercetin can acts as a prooxidant at high intake levels.


Assuntos
Antioxidantes/uso terapêutico , Suplementos Nutricionais , Modelos Animais de Doenças , Hiper-Homocisteinemia/prevenção & controle , Fígado/metabolismo , Estresse Oxidativo , Quercetina/uso terapêutico , Animais , Antioxidantes/administração & dosagem , Antioxidantes/efeitos adversos , Antioxidantes/metabolismo , Suplementos Nutricionais/efeitos adversos , Glutationa/sangue , Glutationa/metabolismo , Homocisteína/sangue , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/fisiopatologia , Peroxidação de Lipídeos , Fígado/patologia , Fígado/fisiopatologia , Masculino , Malondialdeído/metabolismo , Metionina , Tamanho do Órgão , Carbonilação Proteica , Quercetina/administração & dosagem , Quercetina/efeitos adversos , Quercetina/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Aumento de Peso
14.
Acta Biochim Pol ; 59(4): 495-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251907

RESUMO

Stroke is one of the most devastating neurological conditions, with an approximate worldwide mortality of 5.5 million annually and loss of 44 million disability-adjusted life-years. The etiology of stroke is often unknown; it has been estimated that the etiology and pathophysiology remains unexplained in more than 40% of stroke cases. The conventional stroke risk factors, including hypertension, diabetes mellitus, smoking, and cardiac diseases, do not fully account for the risk of stroke, and stroke victims, especially young subjects, often do not have any of these factors. It is very likely that inflammation, specific genetic predispositions and traditional risk factors interact with each other and may together increase the risk of stroke. Inflammatory and immune responses play important roles in the course of ischemic stroke. Hyperhomocysteinemia (hcy) is considered a modifiable risk factor for stroke, possibly through an atherogenic and prothrombotic mechanism. Both genetic and environmental factors (e.g., dietary intake of folic acid and B vitamins) affect homocysteine level. Identification of the role of hcy as a modifiable risk factor for stroke and of HSPs as regulators of the immune response may lead to more effective prevention and treatment of stroke through dietary and pharmacological intervention. Dietary modification may also include supplementation with novel preventive compounds, such as the antioxidative isoflavones--genistein or daidzein.


Assuntos
Proteínas de Choque Térmico/metabolismo , Homocisteína/metabolismo , Acidente Vascular Cerebral , Vitaminas , Suplementos Nutricionais , Genisteína/uso terapêutico , Humanos , Hiper-Homocisteinemia/dietoterapia , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Fatores de Risco , Acidente Vascular Cerebral/dietoterapia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Vitaminas/metabolismo , Vitaminas/uso terapêutico
15.
Nutrition ; 27(10): 1034-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21353475

RESUMO

OBJECTIVE: The purpose of this study was to investigate the protective effects of combined treatment of folate and vitamin B12 against alcoholic liver disease. METHODS: Male Wistar rats weighing about 160 g were divided into four groups: an ethanol group fed an ethanol liquid diet; a control group pair-fed an isoenergetic diet without ethanol; an ethanol and vitamin group fed an ethanol-containing diet that was supplemented with folate (10 mg/kg of body weight per day) and vitamin B12 (0.5 mg/kg of body weight per day); and a control and vitamin group fed an isoenergetic diet without ethanol, which was supplemented with folate (10 mg/kg of body weight per day) and vitamin B12 (0.5 mg/kg of body weight per day). RESULTS: After 16 wk, the plasma folate concentration in the ethanol group was significantly lower than in the other three groups. The plasma homocysteine concentration in the ethanol group was significantly higher than in the other three groups. The hepatic matrix metalloproteinase-2 concentration in the ethanol group was significantly higher than in the control and ethanol/vitamin groups. Furthermore, the plasma homocysteine concentration at the 16th week and the hepatic matrix metalloproteinase-2 concentration showed a significant positive correlation in rats of each group. In addition, pathologic evidence of liver fibrosis was observed only in the ethanol group. Furthermore, hepatic cytochrome 2E1 protein expression in group E increased significantly. CONCLUSION: These results suggest that combined treatment of folate and vitamin B12 can alleviate alcoholic liver injury that may be related to normalization of plasma homocysteine levels.


Assuntos
Ácido Fólico/uso terapêutico , Homocisteína/sangue , Hiper-Homocisteinemia/tratamento farmacológico , Cirrose Hepática Alcoólica/tratamento farmacológico , Fígado/efeitos dos fármacos , Vitamina B 12/uso terapêutico , Complexo Vitamínico B/uso terapêutico , Animais , Citocromos/metabolismo , Ácido Fólico/sangue , Ácido Fólico/farmacologia , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática Alcoólica/patologia , Testes de Função Hepática , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Ratos , Ratos Wistar , Vitamina B 12/farmacologia , Complexo Vitamínico B/farmacologia
16.
Am J Clin Nutr ; 93(5): 941-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21430116

RESUMO

BACKGROUND: Observational studies have shown that low folate and elevated homocysteine concentrations are risk factors for vascular disease in the general population. Randomized controlled trials in vascular patients have failed to show that folic acid reduces the risk of recurrent vascular disease, whereas such trials are lacking in the general population. OBJECTIVE: The objective was to determine whether folic acid supplementation reduces the progression of atherosclerosis as measured by common carotid intima-media thickness (CIMT)-a validated marker of atherosclerosis and predictor of vascular disease risk. DESIGN: A randomized, double-blind, placebo-controlled study in 819 men and postmenopausal women aged 50-70 y, free-living in the Netherlands, and with a total homocysteine concentration ≥13 µmol/L at screening was conducted. Participants received either 800 µg folic acid or placebo daily for 3 y. Rate of change in CIMT and arterial distensibility were the primary and secondary outcomes, respectively. RESULTS: Compared with placebo, serum folate increased by 577% and plasma total homocysteine concentrations decreased by 26% after 3 y of folic acid supplementation. The mean (±SE) rate of change in CIMT was 1.9 ± 0.9 µm/y in the folic acid arm and 1.3 ± 0.8 µm/y in the placebo arm (mean difference: 0.7 µm/y; 95% CI: -1.8, 3.1 µm/y; P = 0.59). No difference was observed (P = 0.23) between the rates of change in distensibility in the folic acid arm (-0.53 ± 0.06 × 10(-3) kPa(-1)) and in the placebo arm (-0.62 ± 0.06 × 10(-3) kPa(-1)). CONCLUSION: Despite a considerable increase in folate concentrations and a reduction in total homocysteine concentrations, 3-y folic acid supplementation did not slow down atherosclerotic progression or arterial stiffening. This trial was registered at clinicaltrials.gov as NCT00110604.


Assuntos
Aterosclerose/prevenção & controle , Artéria Carótida Primitiva/patologia , Suplementos Nutricionais , Deficiência de Ácido Fólico/tratamento farmacológico , Ácido Fólico/uso terapêutico , Túnica Íntima/patologia , Túnica Média/patologia , Idoso , Aterosclerose/epidemiologia , Aterosclerose/etiologia , Doenças das Artérias Carótidas/epidemiologia , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/prevenção & controle , Artéria Carótida Primitiva/diagnóstico por imagem , Progressão da Doença , Método Duplo-Cego , Elasticidade , Feminino , Ácido Fólico/sangue , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/patologia , Deficiência de Ácido Fólico/fisiopatologia , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/tratamento farmacológico , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Fatores de Risco , Túnica Íntima/diagnóstico por imagem , Túnica Média/diagnóstico por imagem , Ultrassonografia
17.
J Thromb Thrombolysis ; 32(1): 82-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21234645

RESUMO

Increased homocysteine levels can be responsible for arterial ischemic events, such as MI, stroke or peripheral vascular disease. Homocysteine is metabolized by two pathways: re-methylation and trans-sulfuration. Both involve folic acid, and vitamins B(6-12.) Several studies assumed that the folates and vitamins B supplementation or dietary source to normalize plasma homocysteine. But, even if tends to normalize homocysteine levels, lowering homocysteine by B-group vitamins and/or folates does not reduce cardiovascular risk. In fact, recent reports confirmed that hyper-homocysteinemia is not directly responsible for cardiovascular disease, but is merely present in individuals suffering for acute and/or chronic cardiovascular events, as a collateral finding. Reduced methylation potential (MP) [due to decreased S-adenosyl-methionine (AdoMet)/S-adenosyl-homocysteine (AdoHcy) ratio] induced by the elevated plasma homocysteine levels seems to be the true responsible for cardiovascular diseases (CVD). The pathogenic mechanisms responsible for CVD appear to be dependent of DNA hypomethylation inducing an inhibition of cyclin A transcription and a reduction of endothelial cells growth. But, other human studies performed in a wide range are requested.


Assuntos
Doenças Cardiovasculares/metabolismo , Homocisteína/metabolismo , Hiper-Homocisteinemia/metabolismo , Doença Aguda , Biomarcadores/metabolismo , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/patologia , Doença Crônica , Humanos , Hiper-Homocisteinemia/tratamento farmacológico , Hiper-Homocisteinemia/patologia , Fatores de Risco , Complexo Vitamínico B/uso terapêutico
18.
J Inherit Metab Dis ; 34(1): 93-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20567905

RESUMO

Mild to moderate hyperhomocysteinemia has been identified as a strong predictor of cardiovascular disease, independent from classical atherothrombotic risk factors. In the last decade, a number of large intervention trials using B vitamins have been performed and have shown no benefit of homocysteine-lowering therapy in high-risk patients. In addition, Mendelian randomization studies failed to convincingly demonstrate that a genetic polymorphism commonly associated with higher homocysteine levels (methylenetetrahydrofolate reductase 677 C>T) is a risk factor for cardiovascular disease. Together, these findings have cast doubt on the role of homocysteine in cardiovascular disease pathogenesis, and the homocysteine hypothesis has turned into a homocysteine controversy. In this review, we attempt to find solutions to this controversy. First, we explain that the Mendelian randomization analyses have limitations that preclude final conclusions. Second, several characteristics of intervention trials limit interpretation and generalizability of their results. Finally, the possibility that homocysteine lowering is in itself beneficial but is offset by adverse side effects of B vitamins on atherosclerosis deserves serious attention. As we explain, such side effects may relate to direct adverse effects of the B-vitamin regimen (in particular, the use of high-dose folic acid) or to proinflammatory and proproliferative effects of B vitamins on advanced atherosclerotic lesions.


Assuntos
Homocisteína/fisiologia , Animais , Dissidências e Disputas , Projetos de Pesquisa Epidemiológica , Ácido Fólico/efeitos adversos , Ácido Fólico/uso terapêutico , Homocisteína/efeitos adversos , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/patologia , Análise da Randomização Mendeliana/métodos , Análise da Randomização Mendeliana/normas , Modelos Biológicos , Índice de Gravidade de Doença , Complexo Vitamínico B/efeitos adversos , Complexo Vitamínico B/uso terapêutico
19.
Rev Neurol (Paris) ; 165(12): 1099-102, 2009 Dec.
Artigo em Francês | MEDLINE | ID: mdl-19446857

RESUMO

INTRODUCTION: Biermer's disease is an autoimmune disorder characterized by vitamin B12 deficiency. Ischemic stroke is an uncommon complication of Biermer's disease, possibly though hyperhomocysteinemia. CASE REPORT: A 58-year-old male presented with recurrent ischemic stroke. Extensive investigations were normal, except for a high plasma level of homocysteine in the context of pernicious anemia which was otherwise asymptomatic. DISCUSSION: Hyperhomocysteinemia is a known marker, and probably a risk factor, for stroke, fostering atherosclerosis and thrombosis. It can be found among individuals suffering from homocysteinuria, but also when there is deficiency of vitamin B12 or folic acid. Vitamin B12 supplementation would reduce homocysteine concentration which in turn would reduce the risk of ischemic stroke.


Assuntos
Anemia Perniciosa/diagnóstico , Hiper-Homocisteinemia/diagnóstico , Ataque Isquêmico Transitório/etiologia , Anemia Perniciosa/sangue , Anemia Perniciosa/patologia , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/patologia , Ataque Isquêmico Transitório/sangue , Ataque Isquêmico Transitório/patologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Recidiva , Vitamina B 12/uso terapêutico
20.
Indian J Exp Biol ; 46(7): 534-40, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18807758

RESUMO

The present study was designed to investigate the antioxidant effect of curcumin on methionine-induced hyperlipidemia and hyperhomocysteinemia in Wistar rats (200-250 g) of either sex. The vehicle control rats were treated with 1% Tween 80 in normal saline (2 ml/kg, po) for 30 days. Hyperlipidemia and hyperhomocysteinemia was induced by methionine administration (1 g/kg, po) for 30 days. A significant increase in total cholesterol, triglycerides, low density lipoprotein cholesterol (LDL-C) and homocysteine levels in serum and thiobarbituric acid reactive substances (TBARS) levels in heart homogenates were observed with a concomitant decrease in serum high density lipoprotein (HDL-C) levels in pathogenic control (i.e. group II) rats, as compared to vehicle control (i.e. group I) rats. Further, curcumin (200 mg/kg, p.o.) treatment in methionine treated rats for 30 days significantly decreased the total cholesterol, triglycerides, LDL-C and homocysteine levels in serum and TBARS levels in heart homogenates and increased serum HDL-C levels, as compared to pathogenic control (i.e. group II) rats. The results of biochemical observations were supplemented by histopathological examination of rat's aortic section. The results of test drug were comparable to that obtained with folic acid (100 mg/kg, p.o.). The results suggest that curcumin has significant antihyperlipidemic and antihyperhomocysteinemic effect against methionine-induced hyperlipidemia and hyperhomocysteinemia in rats.


Assuntos
Curcumina/farmacologia , Curcumina/uso terapêutico , Hiper-Homocisteinemia/tratamento farmacológico , Hiperlipidemias/tratamento farmacológico , Metionina/farmacologia , Animais , Colesterol/sangue , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/patologia , Hiperlipidemias/sangue , Hiperlipidemias/induzido quimicamente , Hiperlipidemias/patologia , Masculino , Fitoterapia , Ratos , Ratos Wistar , Triglicerídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA