Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Sci Rep ; 12(1): 21707, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36522378

RESUMEN

Diabetic nephropathy is characterized by excessive accumulation of extracellular matrix (ECM) leading to renal fibrosis, progressive deterioration of renal function, and eventually to end stage renal disease. Matrix metalloproteinases (MMPs) are known to regulate synthesis and degradation of the ECM. Earlier, we demonstrated that imbalanced MMPs promote adverse ECM remodeling leading to renal fibrosis in type-1 diabetes. Moreover, elevated macrophage infiltration, pro-inflammatory cytokines and epithelial‒mesenchymal transition (EMT) are known to contribute to the renal fibrosis. Various bioactive compounds derived from the medicinal plant, Azadirachta indica (neem) are shown to regulate inflammation and ECM proteins in different diseases. Nimbidiol is a neem-derived diterpenoid that is considered as a potential anti-diabetic compound due to its glucosidase inhibitory properties. We investigated whether Nimbidiol mitigates adverse ECM accumulation and renal fibrosis to improve kidney function in type-1 diabetes and the underlying mechanism. Wild-type (C57BL/6J) and type-1 diabetic (C57BL/6-Ins2Akita/J) mice were treated either with saline or with Nimbidiol (0.40 mg kg-1 d-1) for eight weeks. Diabetic kidney showed increased accumulation of M1 macrophages, elevated pro-inflammatory cytokines and EMT. In addition, upregulated MMP-9 and MMP-13, excessive collagen deposition in the glomerular and tubulointerstitial regions, and degradation of vascular elastin resulted to renal fibrosis in the Akita mice. These pathological changes in the diabetic mice were associated with functional impairments that include elevated resistive index and reduced blood flow in the renal cortex, and decreased glomerular filtration rate. Furthermore, TGF-ß1, p-Smad2/3, p-P38, p-ERK1/2 and p-JNK were upregulated in diabetic kidney compared to WT mice. Treatment with Nimbidiol reversed the changes to alleviate inflammation, ECM accumulation and fibrosis and thus, improved renal function in Akita mice. Together, our results suggest that Nimbidiol attenuates inflammation and ECM accumulation and thereby, protects kidney from fibrosis and dysfunction possibly by inhibiting TGF-ß/Smad and MAPK signaling pathways in type-1 diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Nefropatías Diabéticas , Diterpenos , Ratones , Animales , Nefropatías Diabéticas/patología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Ratones Endogámicos C57BL , Fibrosis , Factor de Crecimiento Transformador beta1/metabolismo , Riñón/metabolismo , Diterpenos/metabolismo , Inflamación/patología , Glucosidasas
2.
Medicina (Kaunas) ; 58(1)2021 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-35056324

RESUMEN

Impaired folate-mediated one-carbon metabolism (FOCM) is associated with many pathologies and developmental abnormalities. FOCM is a metabolic network of interdependent biosynthetic pathways that is known to be compartmentalized in the cytoplasm, mitochondria and nucleus. Currently, the biochemical mechanisms and causal metabolic pathways responsible for the initiation and/or progression of folate-associated pathologies have yet to be fully established. This review specifically examines the role of impaired FOCM in type 2 diabetes mellitus, Alzheimer's disease and the emerging Long COVID/post-acute sequelae of SARS-CoV-2 (PASC). Importantly, elevated homocysteine may be considered a biomarker for impaired FOCM, which is known to result in increased oxidative-redox stress. Therefore, the incorporation of hyperhomocysteinemia will be discussed in relation to impaired FOCM in each of the previously listed clinical diseases. This review is intended to fill gaps in knowledge associated with these clinical diseases and impaired FOCM. Additionally, some of the therapeutics will be discussed at this early time point in studying impaired FOCM in each of the above clinical disease states. It is hoped that this review will allow the reader to better understand the role of FOCM in the development and treatment of clinical disease states that may be associated with impaired FOCM and how to restore a more normal functional role for FOCM through improved nutrition and/or restoring the essential water-soluble B vitamins through oral supplementation.


Asunto(s)
Enfermedad de Alzheimer , COVID-19 , Diabetes Mellitus Tipo 2 , COVID-19/complicaciones , Carbono , Ácido Fólico , Humanos , SARS-CoV-2 , Síndrome Post Agudo de COVID-19
3.
Int J Mol Sci ; 21(23)2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33265949

RESUMEN

This study aimed to examine the effects of diallyl trisulfide (DATS), the most potent polysulfide derived from garlic, on metabolic syndrome and myocardial function in rats with metabolic syndrome (MetS). For that purpose, we used 36 male Wistar albino rats divided into control rats, rats with MetS and MetS rats treated with 40 mg/kg of DATS every second day for 3 weeks. In the first part, we studied the impact of DATS on MetS control and found that DATS significantly raised H2S, decreased homocysteine and glucose levels and enhanced lipid and antioxidative, while reducing prooxidative parameters. Additionally, this polysulfide improved cardiac function. In the second part, we investigated the impact of DATS on ex vivo induced ischemia/reperfusion (I/R) heart injury and found that DATS consumption significantly improved cardiodynamic parameters and prevented oxidative and histo-architectural variation in the heart. In addition, DATS significantly increased relative gene expression of eNOS, SOD-1 and -2, Bcl-2 and decreased relative gene expression of NF-κB, IL-17A, Bax, and caspases-3 and -9. Taken together, the data show that DATS can effectively mitigate MetS and have protective effects against ex vivo induced myocardial I/R injury in MetS rat.


Asunto(s)
Compuestos Alílicos/uso terapéutico , Cardiotónicos/uso terapéutico , Ajo/química , Síndrome Metabólico/tratamiento farmacológico , Sulfuros/uso terapéutico , Compuestos Alílicos/farmacología , Animales , Glucemia/metabolismo , Cardiotónicos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Pruebas de Función Cardíaca/efectos de los fármacos , Insulina/sangre , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Síndrome Metabólico/sangre , Síndrome Metabólico/fisiopatología , Miocardio/patología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Sulfuros/farmacología
4.
Mol Cell Biochem ; 460(1-2): 151-164, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31280436

RESUMEN

Diallyl trisulfide (DATS) is distinguished as the most potent polysulfide isolated from garlic. The aim of our study was to investigate effects of oral administration of DATS on healthy and diabetic rats, with special attention on heart function. Rats were randomly divided into four groups: CTRL (healthy rats), DATS (healthy rats treated with DATS), DM (diabetic rats), DM + DATS (diabetic rats treated with DATS). DATS (40 mg/kg of body weight) was administered every other day for 3 weeks, at the end of which rats underwent echocardiography, glycemic measurement and redox status assessment. Isolated rat hearts were subjected to 30 min global ischemia and 60 min reperfusion, after which heart tissue was counterstain with hematoxylin and eosin and cardiac Troponin T staining (cTnT), while expression of Bax, B cell lymphoma 2 (Bcl-2), caspase-3, caspase-9 and superoxide dismutase-2 were examined in the left ventricle. DATS treatment significantly reduced blood glucose levels of diabetic rats, and improved cardiac function recovery, diminished oxidation status, attenuated cardiac remodeling and inhibited myocardial apoptosis in healthy and diabetic rats. DATS treatment causes promising cardioprotective effects on ex vivo-induced ischemia/reperfusion (I/R) injury in diabetic and healthy rat heart probably mediated by inhibited myocardial apoptosis. Moreover, appropriate DATS consumption may provide potential co-therapy or prevention of hyperglycemia and various cardiac complications in rats with DM.


Asunto(s)
Compuestos Alílicos/uso terapéutico , Cardiotónicos/uso terapéutico , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Daño por Reperfusión/complicaciones , Daño por Reperfusión/tratamiento farmacológico , Sulfuros/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Cardiotónicos/farmacología , Diabetes Mellitus Experimental/fisiopatología , Masculino , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Daño por Reperfusión/fisiopatología
5.
J Cell Biochem ; 118(8): 2108-2117, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27982479

RESUMEN

A detrimental consequence of hypermethylation is hyperhomocysteinemia (HHcy), that causes oxidative stress, inflammation, and matrix degradation, which leads to multi-pathology in different organs. Although, it is well known that hypermethylation leads to overall gene silencing and hypomethylation leads to overall gene activation, the role of such process in skeletal muscle dysfunction during HHcy condition is unclear. In this study, we emphasized the multiple mechanisms including epigenetic alteration by which HHcy causes skeletal muscle myopathy. This review also highlights possible role of methylation, histone modification, and RNA interference in skeletal muscle dysfunction during HHcy condition and potential therapeutic molecules, putative challenges, and methodologies to deal with HHcy mediated skeletal muscle dysfunction. We also highlighted that B vitamins (mainly B12 and B6), with folic acid supplementation, could be useful as an adjuvant therapy to reverse these consequences associated with this HHcy conditions in skeletal muscle. However, we would recommend to further study involving long-term trials could help to assess efficacy of the use of these therapeutic agents. J. Cell. Biochem. 118: 2108-2117, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Epigénesis Genética , Hiperhomocisteinemia/diagnóstico , Músculo Esquelético/metabolismo , Enfermedades Musculares/diagnóstico , Vitamina B 12/administración & dosificación , Vitamina B 6/administración & dosificación , Animales , Metilación de ADN/efectos de los fármacos , Ácido Fólico/administración & dosificación , Histonas/genética , Histonas/metabolismo , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/tratamiento farmacológico , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Metformina/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Estrés Oxidativo , Interferencia de ARN , Tiazolidinedionas/uso terapéutico
6.
J Mol Neurosci ; 52(2): 202-15, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24122186

RESUMEN

Epigenetic mechanisms underlying nutrition (nutrition epigenetics) are important in understanding human health. Nutritional supplements, for example folic acid, a cofactor in one-carbon metabolism, regulate epigenetic alterations and may play an important role in the maintenance of neuronal integrity. Folic acid also ameliorates hyperhomocysteinemia, which is a consequence of elevated levels of homocysteine. Hyperhomocysteinemia induces oxidative stress that may epigenetically mediate cerebrovascular remodeling and leads to neurodegeneration; however, the mechanisms behind such alterations remain unclear. Therefore, the present study was designed to observe the protective effects of folic acid against hyperhomocysteinemia-induced epigenetic and molecular alterations leading to neurotoxic cascades. To test this hypothesis, we employed 8-weeks-old male wild-type (WT) cystathionine-beta-synthase heterozygote knockout methionine-fed (CBS+/− + Met), WT, and CBS+/− + Met mice supplemented with folic acid (FA) [WT + FA and CBS+/− + Met + FA, respectively, 0.0057-µg g−1 day−1 dose in drinking water/4 weeks]. Hyperhomocysteinemia in CBS+/− + Met mouse brain was accompanied by a decrease in methylenetetrahydrofolate reductase and an increase in S-adenosylhomocysteine hydrolase expression, symptoms of oxidative stress, upregulation of DNA methyltransferases, rise in matrix metalloproteinases, a drop in the tissue inhibitors of metalloproteinases, decreased expression of tight junction proteins, increased permeability of the blood-brain barrier, neurodegeneration, and synaptotoxicity. Supplementation of folic acid to CBS+/− + Met mouse brain led to a decrease in the homocysteine level and rescued pathogenic and epigenetic alterations, showing its protective efficacy against homocysteine-induced neurotoxicity.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Epigénesis Genética , Ácido Fólico/uso terapéutico , Hiperhomocisteinemia/dietoterapia , Fármacos Neuroprotectores/uso terapéutico , Adenosilhomocisteinasa/genética , Adenosilhomocisteinasa/metabolismo , Animales , Barrera Hematoencefálica/patología , Cistationina betasintasa/genética , Dieta , Ácido Fólico/administración & dosificación , Ácido Fólico/farmacología , Heterocigoto , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Metionina/administración & dosificación , Metionina/farmacología , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo
7.
Am J Physiol Cell Physiol ; 303(1): C41-51, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22517358

RESUMEN

Hydrogen sulfide (H(2)S) has recently been identified as a regulator of various physiological events, including vasodilation, angiogenesis, antiapoptotic, and cellular signaling. Endogenously, H(2)S is produced as a metabolite of homocysteine (Hcy) by cystathionine ß-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3MST). Although Hcy is recognized as vascular risk factor at an elevated level [hyperhomocysteinemia (HHcy)] and contributes to vascular injury leading to renovascular dysfunction, the exact mechanism is unclear. The goal of the current study was to investigate whether conversion of Hcy to H(2)S improves renovascular function. Ex vivo renal artery culture with CBS, CSE, and 3MST triple gene therapy generated more H(2)S in the presence of Hcy, and these arteries were more responsive to endothelial-dependent vasodilation compared with nontransfected arteries treated with high Hcy. Cross section of triple gene-delivered renal arteries immunostaining suggested increased expression of CD31 and VEGF and diminished expression of the antiangiogenic factor endostatin. In vitro endothelial cell culture demonstrated increased mitophagy during high levels of Hcy and was mitigated by triple gene delivery. Also, dephosphorylated Akt and phosphorylated FoxO3 in HHcy were reversed by H(2)S or triple gene delivery. Upregulated matrix metalloproteinases-13 and downregulated tissue inhibitor of metalloproteinase-1 in HHcy were normalized by overexpression of triple genes. Together, these results suggest that H(2)S plays a key role in renovasculopathy during HHcy and is mediated through Akt/FoxO3 pathways. We conclude that conversion of Hcy to H(2)S by CBS, CSE, or 3MST triple gene therapy improves renovascular function in HHcy.


Asunto(s)
Cistationina betasintasa/genética , Cistationina gamma-Liasa/genética , Terapia Genética , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/terapia , Sulfurtransferasas/genética , Animales , Células Cultivadas , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Endostatinas/biosíntesis , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Homocisteína/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Hipertensión Renovascular/genética , Hipertensión Renovascular/terapia , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Arteria Renal/metabolismo , Sulfurtransferasas/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Lesiones del Sistema Vascular
8.
Am J Physiol Heart Circ Physiol ; 302(3): H688-96, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22101525

RESUMEN

Although right ventricular failure (RVF) is the hallmark of pulmonary arterial hypertension (PAH), the mechanism of RVF is unclear. Development of PAH-induced RVF is associated with an increased reactive oxygen species (ROS) production. Increases in oxidative stress lead to generation of nitro-tyrosine residues in tissue inhibitor of metalloproteinase (TIMPs) and liberate active matrix metalloproteinase (MMPs). To test the hypothesis that an imbalance in MMP-to-TIMP ratio leads to interstitial fibrosis and RVF and whether the treatment with folic acid (FA) alleviates ROS generation, maintains MMP/TIMP balance, and regresses interstitial fibrosis, we used a mouse model of pulmonary artery constriction (PAC). After surgery mice were given FA in their drinking water (0.03 g/l) for 4 wk. Production of ROS in the right ventricle (RV) was measured using oxidative fluorescent dye. The level of MMP-2, -9, and -13 and TIMP-4, autophagy marker (p62), mitophagy marker (LC3A/B), collagen interstitial fibrosis, and ROS in the RV wall was measured. RV function was measured by Millar catheter. Treatment with FA decreased the pressure to 35 mmHg from 50 mmHg in PAC mice. Similarly, RV volume in PAC mice was increased compared with the Sham group. A robust increase of ROS was observed in RV of PAC mice, which was decreased by treatment with FA. The protein level of MMP-2, -9, and -13 was increased in RV of PAC mice in comparison with that in the sham-operated mice, whereas supplementation with FA abolished this effect and mitigated MMPs levels. The protein level of TIMP-4 was decreased in RV of PAC mice compared with the Sham group. Treatment with FA helped PAC mice to improve the level of TIMP-4. To further support the claim of mitophagy occurrence during RVF, the levels of LC3A/B and p62 were measured by Western blot and immunohistochemistry. LC3A/B was increased in RV of PAC mice. Similarly, increased p62 protein level was observed in RV of PAC mice. Treatment with FA abolished this effect in PAC mice. These results suggest that FA treatment improves MMP/TIMP balance and ameliorates mitochondrial dysfunction that results in protection of RV failure during pulmonary hypertension.


Asunto(s)
Autofagia/fisiología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/fisiopatología , Arteria Pulmonar/fisiopatología , Remodelación Ventricular/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Autofagia/efectos de los fármacos , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Ácido Fólico/farmacología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/tratamiento farmacológico , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/patología , Masculino , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Remodelación Ventricular/efectos de los fármacos , Complejo Vitamínico B/farmacología , Inhibidor Tisular de Metaloproteinasa-4
9.
Artículo en Inglés | MEDLINE | ID: mdl-22162783

RESUMEN

Accumulating evidences suggest that homocysteine, a non-protein amino acid, is involved in vessel remodeling and blood flow at elevated level, although the exact mechanism is unclear. Here we hypothesized that homocysteine affects vein in such a way that vein develops arterial phenotype. We tested our hypothesis employing wild type (WT, C57BL/6J) and CBS+/- (cystathionine ß-synthase heterozygote, a genetic model of hyperhomocysteinemia) supplemented with or without folic acid (FA, a homocysteine lowering agent). Vena cava blood flow was measured by ultrasound transonic flow probe. Tissue collagen and elastin were detected by histochemistry. Super oxide was detected by dihydroethidium (DHE) staining. Expressions of MMP-2, -9, -12, TIMP -2,-4, were measured by Western blot. MMP-13, TIMP-1, -3, and vein and aortic markers, EphB4 and EphrinB2, respectively were measured by RT-PCR. The results indicated relatively low blood flow and significant increase of collagen/elastin ratio in the CBS+/- mice compared to WT. Although FA treatment did not alter blood flow in CBS+/- mice, the collagen/elastin ratio was normalized. A relatively increased content of super oxide and gelatinase activity was observed in CBS+/- vena cava vs WT and normalized by FA treatment. Western blot analyses showed significant increase in MMP-9,-12 and decrease in TIMP-2, -4 expressions. Expressions of MMP-13, TIMP-1 and -3, Ephrin B2 were increased, whereas EphB4 was decreased with reverse change in FA treatment, with no change in MMP-13 and TIMP-1. We conclude that chronic HHcy causes vascular remodeling that expresses arterial phenotype in vein.

10.
Artículo en Inglés | MEDLINE | ID: mdl-21760968

RESUMEN

We tested the hypothesis that exercise ameliorates contractile dysfunction by interfering with homocysteine - ß2-adrenergic receptor (AR) interactions, inducing ß2-adrenergic response and Gs (stimulatory G adenylyl cyclase dependent protein kinase), and lowering homocysteine level in diabetes. The effect of homocysteine on ß2-AR was determined by (a) scoring the ß2-AR in the cardiomyocytes treated with high dose of homocysteine using flow cytometry, and (b) co-localizing homocysteine with Gs (an inducer of ß2-AR) in the cardiomyocytes obtained from C57BL/ 6J (WT) and db/ db mice using confocal microscopy. The effect of exercise on the protein-protein interactions of homocysteine and ß2-AR in diabetes was evaluated by co-immunoprecipitation in the four groups of db/db mice: (1) sedentary, (2) treated with salbutamol (a ß2-AR agonist), (3) swimming exercise, and (4) swimming + salbutamol treatment. The effect of exercise on ß2-AR was determined by RT-PCR and Western blotting while cardiac dysfunction was assessed by echocardiography, and contractility and calcium transient of cardiomyocytes from the above four groups. The results revealed that elevated level of homocysteine decreases the number of ß2-AR and inhibits Gs in diabetes. However, exercise mitigates the interactions of homocysteine with ß2-AR and induces ß2-AR. Exercise also ameliorates cardiac dysfunction by enhancing the calcium transient of cardiomyocytes. To our knowledge, this is the first report showing mechanism of homocysteine mediated attenuation of ß2-AR response in diabetes and effect of exercise on homocysteine - ß2-AR interactions.

11.
J Cell Physiol ; 226(10): 2712-20, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21792928

RESUMEN

Human atherosclerotic coronary vessels elicited vasoconstriction to acetylcholine (Ach) and revealed a phenomenon of vasospasm. Homocysteine (Hcy) levels are elevated in the atherosclerotic plaque tissue, suggesting its pathological role in endothelial damage in atherosclerotic diseases. Accordingly, we examined the role hyperhomocysteinemia in coronary endothelial dysfunction, vessel wall thickness, lumen narrowing, leading to acute/chronic coronary vasospasm. The therapeutic potential and mechanisms of folic acid (FA) using hyperhomocysteinemic cystathionine beta synthase heterozygote (CBS-/+) and wild type (CBS+/+) mice were addressed. The CBS-/+ and CBS+/+ mice were treated with or without a Hcy lowering agent FA in drinking water (0.03 g/L) for 4 weeks. The isolated mouse septum coronary artery was cannulated and pressurized at 60 mmHg. The wall thickness and lumen diameters were measured by Ion-Optic. The vessels were treated with Ach (10(-8) -10(-5) M) and, for comparison, with non-endothelial vasodilator sodium nitroprusside (10(-5) M). The endothelium-impaired arteries from CBC-/+ mice constricted in response to Ach and this vasoconstriction was mitigated with FA supplementation. The level of endothelial nitric oxide synthase (eNOS) was lower in coronary artery in CBS-/+ than of CBS+/+ mice. Treatment with FA increased the levels of Ach-induced NO generation in the coronary artery of CBS-/+ mice. The results suggest that Ach induced coronary vasoconstriction in CBS-/+ mice and this vasoconstriction was ameliorated by FA treatment. The mechanisms for the impairment of vascular function and therapeutic effects of FA may be related to the regulation of eNOS expression, NO availability and tissue homocysteine.


Asunto(s)
Acetilcolina/farmacología , Vasoespasmo Coronario/metabolismo , Vasos Coronarios/efectos de los fármacos , Ácido Fólico/farmacología , Hiperhomocisteinemia/metabolismo , Vasoconstricción/efectos de los fármacos , Animales , Agonistas Colinérgicos/farmacología , Vasoespasmo Coronario/tratamiento farmacológico , Vasoespasmo Coronario/genética , Vasos Coronarios/fisiología , Sinergismo Farmacológico , Femenino , Hiperhomocisteinemia/tratamiento farmacológico , Hiperhomocisteinemia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Vasoconstricción/fisiología , Complejo Vitamínico B/farmacología
12.
J Orthop Res ; 29(10): 1511-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21469179

RESUMEN

Deficiencies in folate lead to increased serum concentrations of homocysteine (Hcy), which is known as hyperhomocysteinemia (HHcy), is associated with bone disorders. Although, Hcy accumulates collagen in bone and contribute to decrease in bone strength. The mechanism of Hcy induced bone loss and remodeling is unclear. Therefore, the present study was aimed to determine the role of folic acid (FA) in genetically HHcy-associated decrease in bone blood flow and remodeling. Wild type (WT) and cystathionine-ß-synthase heterozygous (CBS+/-) mice were used in this study and supplemented with or without FA (300 mg/kg, Hcy reducing agent) in drinking water for 6 weeks. The tibial bone blood flow was measured by laser Doppler and ultrasonic flow probe method. The tibial bone density (BD) was assessed by dual energy X-ray absorptiometry. The bone homogenates were analyzed for oxidative stress, NOX-4 as oxidative marker and thioredoxin-1 (Trx-1) as anti-oxidant marker, bone remodeling (MMP-9) and bio-availability of nitric oxide (eNOS/iNOS/NO) by Western blot method. The results suggested that there was decrease in tibial blood flow in CBS+/- mice. The BD was also reduced in CBS+/- mice. There was an increase in NOX-4, iNOS, MMP-9 protein as well as MMP-9 activity in CBS+/- mice and decrease in Trx-1, eNOS protein levels, in part by decreasing NO bio-availability in CBS+/- mice. Interestingly, these effects were ameliorated by FA and suggested that FA supplementation may have therapeutic potential against genetically HHcy induced bone loss.


Asunto(s)
Remodelación Ósea , Ácido Fólico/fisiología , Homocisteína/fisiología , Hiperhomocisteinemia/complicaciones , Osteoporosis/etiología , Animales , Densidad Ósea , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Óxido Nítrico/metabolismo , Estrés Oxidativo , Flujo Sanguíneo Regional , Tibia/irrigación sanguínea , Arterias Tibiales/fisiología
13.
Am J Physiol Heart Circ Physiol ; 299(5): H1484-93, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20802128

RESUMEN

Myocardial infarction (MI) results in significant metabolic derangement, causing accumulation of metabolic by product, such as homocysteine (Hcy). Hcy is a nonprotein amino acid generated during nucleic acid methylation and demethylation of methionine. Folic acid (FA) decreases Hcy levels by remethylating the Hcy to methionine, by 5-methylene tetrahydrofolate reductase (5-MTHFR). Although clinical trials were inconclusive regarding the role of Hcy in MI, in animal models, the levels of 5-MTHFR were decreased, and FA mitigated the MI injury. We hypothesized that FA mitigated MI-induced injury, in part, by mitigating cardiac remodeling during chronic heart failure. Thus, MI was induced in 12-wk-old male C57BL/J mice by ligating the left anterior descending artery, and FA (0.03 g/l in drinking water) was administered for 4 wk after the surgery. Cardiac function was assessed by echocardiography and by a Millar pressure-volume catheter. The levels of Hcy-metabolizing enzymes, cystathionine ß-synthase (CBS), cystathionine γ-lyase (CSE), and 5-MTHFR, were estimated by Western blot analyses. The results suggest that FA administered post-MI significantly improved cardiac ejection fraction and induced tissue inhibitor of metalloproteinase, CBS, CSE, and 5-MTHFR. We showed that FA supplementation resulted in significant improvement of myocardial function after MI. The study eluted the importance of homocysteine (Hcy) metabolism and FA supplementation in cardiovascular disease.


Asunto(s)
Ácido Fólico/farmacología , Corazón/efectos de los fármacos , Hematínicos/farmacología , Homocisteína/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Infarto del Miocardio/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Animales , Suplementos Dietéticos , Ácido Fólico/administración & dosificación , Ácido Fólico/uso terapéutico , Corazón/fisiopatología , Hematínicos/administración & dosificación , Hematínicos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Resultado del Tratamiento , Función Ventricular Izquierda/efectos de los fármacos , Función Ventricular Izquierda/fisiología
14.
Curr Vasc Pharmacol ; 8(1): 64-74, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19485933

RESUMEN

Elevated levels of serum homocysteine (Hcy) resulting in hyperhomocysteinemia (HHcy) have been implicated in cardiac pathological conditions including: coronary heart disease (CHD), acute myocardial infarction, arrhythmogenesis and sudden cardiac death (SCD). The mechanisms by which HHcy leads to arrhythmogenesis and SCD are unknown. Novel findings indicate that Hcy is an agonist of the N-methyl-D-aspartate receptor (NMDA-R), known to be present in cardiac tissue, and when activated, increases intracellular calcium leading to increased cell excitability. Also, HHcy induces oxidative stress in cardiac cells and activates matrix metalloproteinases (MMPs) that degrade cell membranes and proteins. Here we review the literature relevant to HHcy-induced oxidative stress leading to cardiac tissue remodelling that may adversely affect cell-to-cell impulse conduction, in particular on the heart's specialized conduction system, and may provide substrate for arrhythmogenesis and SCD. Efficacy of B vitamin supplementation in patient populations with HHcy and CHD is also reviewed.


Asunto(s)
Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Muerte Súbita Cardíaca/etiología , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/fisiopatología , Animales , Arritmias Cardíacas/prevención & control , Señalización del Calcio/fisiología , Conexinas/fisiología , Enfermedad Coronaria/terapia , Muerte Súbita Cardíaca/prevención & control , Suplementos Dietéticos , Corazón/fisiopatología , Sistema de Conducción Cardíaco/fisiopatología , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/terapia , Metaloproteinasas de la Matriz/metabolismo , Miocardio/enzimología , Miocardio/metabolismo , Estrés Oxidativo/fisiología , Receptores de N-Metil-D-Aspartato/agonistas , Factores de Riesgo , Complejo Vitamínico B/uso terapéutico
15.
Pharmacology ; 82(3): 201-13, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18810244

RESUMEN

BACKGROUND/AIMS: Sodium thiosulfate (STS) has been shown to be an antioxidant and calcium solubilizer, but the possible role of STS in dysfunctional ventricles remains unknown. Here, we assessed the effects of STS in the failing heart. METHODS: Heart failure was created by an arteriovenous fistula (AVF). Mice were divided into 4 groups: sham, AVF, sham + STS, and AVF + STS. STS (3 mg/ml) was supplemented with drinking water for 6 weeks in the appropriate surgery groups after surgery. RESULTS: M-mode echocardiograms showed ventricular contractile dysfunction with reduced aortic blood flow in AVF mice, whereas STS treatment prevented the decline in cardiac function. Ventricular collagen, MMP-2 and -9, and TIMP-1 were robustly increased with a decreasing trend in adenylate cyclase VI expression; however, STS supplementation reversed these effects in AVF mice. Among 2 enzymes that produce endogenous hydrogen sulfide (H(2)S), cystathionine-gamma-lyase (CSE) expression was attenuated in AVF mice with no changes in cystathionine-beta-synthase (CBS) expression. In addition, reduced production of H(2)S in AVF ventricular tissue was normalized with STS supplementation. Moreover, cardiac tissues were more responsive to H(2)S when AVF mice were supplemented with STS compared to AVF alone. CONCLUSIONS: These results suggested that STS modulated cardiac dysfunction and the extracellular matrix, in part, by increasing ventricular H(2)S generation.


Asunto(s)
Cardiotónicos/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Sulfuro de Hidrógeno/metabolismo , Tiosulfatos/farmacología , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Aorta/fisiopatología , Fístula Arteriovenosa , Enfermedad Crónica , Colágeno/efectos de los fármacos , Colágeno/metabolismo , Cistationina gamma-Liasa/efectos de los fármacos , Cistationina gamma-Liasa/metabolismo , Ecocardiografía , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/fisiopatología , Masculino , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica/efectos de los fármacos , Inhibidor Tisular de Metaloproteinasa-1/efectos de los fármacos , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
16.
Cardiovasc Toxicol ; 8(3): 137-44, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18679830

RESUMEN

Elevated levels of homocysteine (Hcy) (known as hyperhomocysteinemia HHcy) are involved in dilated cardiomyopathy. Hcy chelates copper and impairs copper-dependent enzymes. Copper deficiency has been linked to cardiovascular disease. We tested the hypothesis that copper supplement regresses left ventricular hypertrophy (LVH), fibrosis and endothelial dysfunction in pressure overload DCM mice hearts. The mice were grouped as sham, sham + Cu, aortic constriction (AC), and AC + Cu. Aortic constriction was performed by transverse aortic constriction. The mice were treated with or without 20 mg/kg copper supplement in the diet for 12 weeks. The cardiac function was assessed by echocardiography and electrocardiography. The matrix remodeling was assessed by measuring matrix metalloproteinase (MMP), tissue inhibitor of metalloproteinases (TIMPs), and lysyl oxidase (LOX) by Western blot analyses. The results suggest that in AC mice, cardiac function was improved with copper supplement. TIMP-1 levels decreased in AC and were normalized in AC + Cu. Although MMP-9, TIMP-3, and LOX activity increased in AC and returned to baseline value in AC + Cu, copper supplement showed no significant effect on TIMP-4 activity after pressure overload. In conclusion, our data suggest that copper supplement helps improve cardiac function in a pressure overload dilated cardiomyopathic heart.


Asunto(s)
Cardiomiopatía Dilatada/tratamiento farmacológico , Cobre/administración & dosificación , Suplementos Dietéticos , Insuficiencia Cardíaca/tratamiento farmacológico , Hemodinámica/efectos de los fármacos , Homocisteína/metabolismo , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Miocardio/metabolismo , Animales , Aorta/cirugía , Presión Sanguínea , Western Blotting , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Constricción , Cobre/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Electrocardiografía , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Femenino , Fibrosis , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Homocisteína/sangre , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocardio/enzimología , Miocardio/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Remodelación Ventricular/efectos de los fármacos
17.
J Cell Physiol ; 215(3): 771-81, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18181170

RESUMEN

Hyperhomocysteinemia (HHcy) is associated with atherosclerotic events involving the modulation of arachidonic acid (AA) metabolism and the activation of matrix metalloproteinase-9 (MMP-9). Cytochrome P450 (CYP) epoxygenase-2J2 (CYP2J2) is abundant in the heart endothelium, and its AA metabolites epoxyeicosatrienoic acids (EETs) mitigates inflammation through NF-kappabeta. However, the underlying molecular mechanisms for MMP-9 regulation by CYP2J2 in HHcy remain obscure. We sought to determine the molecular mechanisms by which P450 epoxygenase gene transfection or EETs supplementation attenuate homocysteine (Hcy)-induced MMP-9 activation. CYP2J2 was over-expressed in mouse aortic endothelial cells (MAECs) by transfection with the pcDNA3.1/CYP2J2 vector. The effects of P450 epoxygenase transfection or exogenous supplementation of EETs on NF-kappabeta-mediated MMP-9 regulation were evaluated using Western blot, in-gel gelatin zymography, electromobility shift assay, immunocytochemistry. The result suggested that Hcy downregulated CYP2J2 protein expression and dephosphorylated PI3K-dependent AKT signal. Hcy induced the nuclear translocation of NF-kappabeta via downregulation of IKbetaalpha (endogenous cytoplasmic inhibitor of NF-kappabeta). Hcy induced MMP-9 activation by increasing NF-kappabeta-DNA binding. Moreover, P450 epoxygenase transfection or exogenous addition of 8,9-EET phosphorylated the AKT and attenuated Hcy-induced MMP-9 activation. This occurred, in part, by the inhibition of NF-kappabeta nuclear translocation, NF-kappabeta-DNA binding and activation of IKbetaalpha. The study unequivocally suggested the pivotal role of EETs in the modulation of Hcy/MMP-9 signal.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Hiperhomocisteinemia/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Oxigenasas/genética , Oxigenasas/metabolismo , Factor de Transcripción ReIA/antagonistas & inhibidores , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacología , Animales , Células Cultivadas , Citocromo P-450 CYP2J2 , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Homocisteína/farmacología , Hiperhomocisteinemia/enzimología , Proteínas I-kappa B/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Inhibidor NF-kappaB alfa , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción ReIA/metabolismo , Transfección
18.
Clin Chem Lab Med ; 45(12): 1633-44, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18067448

RESUMEN

Chronic heart failure (CHF) is a major public health problem causing considerable morbidity and mortality. Recently, plasma homocysteine (HCY) has been suggested to be significantly increased in CHF patients. This article reviews the relation between hyperhomocysteinemia (HHCY) and CHF. Clinical data indicate that HHCY is associated with an increased incidence, as well as severity, of CHF. In addition, HCY correlates with brain natriuretic peptide (BNP), a modern biochemical marker of CHF, which is used for diagnosis, treatment guidance and risk assessment. Animal studies showed that experimental HHCY induces systolic and diastolic dysfunction, as well as an increased BNP expression. Moreover, hyperhomocysteinemic animals exhibit an adverse cardiac remodeling characterized by accumulation of interstitial and perivascular collagen. In vitro superfusion experiments with increasing concentrations of HCY in the superfusion medium stimulated myocardial BNP release independent from myocardial wall stress. Thus, clinical and experimental data underline a correlation between HHCY and BNP supporting the role of HHCY as a causal factor for CHF. The mechanisms leading from an elevated HCY level to reduced pump function and adverse cardiac remodeling are a matter of speculation. Existing data indicate that direct effects of HCY on the myocardium, as well as nitric oxide independent vascular effects, are involved. Preliminary data from small intervention trials have initiated the speculation that HCY lowering therapy by micronutrients may improve clinical as well as laboratory markers of CHF. In conclusion, HHCY might be a potential etiological factor in CHF. Future studies need to explore the pathomechanisms of HHCY in CHF. Moreover, larger intervention trials are needed to clarify whether modification of plasma HCY by B-vitamin supplementation improves the clinical outcome in CHF patients.


Asunto(s)
Insuficiencia Cardíaca/etiología , Homocisteína/sangre , Péptido Natriurético Encefálico/sangre , Animales , Enfermedad Crónica , Humanos
19.
J Exp Med ; 204(3): 657-66, 2007 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-17339407

RESUMEN

Sustained pressure overload causes cardiac hypertrophy and the transition to heart failure. We show here that dietary supplementation with physiologically relevant levels of copper (Cu) reverses preestablished hypertrophic cardiomyopathy caused by pressure overload induced by ascending aortic constriction in a mouse model. The reversal occurs in the continued presence of pressure overload. Sustained pressure overload leads to decreases in cardiac Cu and vascular endothelial growth factor (VEGF) levels along with suppression of myocardial angiogenesis. Cu supplementation replenishes cardiac Cu, increases VEGF, and promotes angiogenesis. Systemic administration of anti-VEGF antibody blunts Cu regression of hypertrophic cardiomyopathy. In cultured human cardiomyocytes, Cu chelation blocks insulin-like growth factor (IGF)-1- or Cu-stimulated VEGF expression, which is relieved by addition of excess Cu. Both IGF-1 and Cu activate hypoxia-inducible factor (HIF)-1alpha and HIF-1alpha gene silencing blocks IGF-1- or Cu-stimulated VEGF expression. HIF-1alpha coimmunoprecipitates with a Cu chaperone for superoxide dismutase-1 (CCS), and gene silencing of CCS, but not superoxide dismutase-1, prevents IGF-1- or Cu-induced HIF-1alpha activation and VEGF expression. Therefore, dietary Cu supplementation improves the condition of hypertrophic cardiomyopathy at least in part through CCS-mediated HIF-1alpha activation of VEGF expression and angiogenesis.


Asunto(s)
Cardiomiopatía Hipertrófica/dietoterapia , Cardiomiopatía Hipertrófica/etiología , Cobre/uso terapéutico , Suplementos Dietéticos , Hipertensión/complicaciones , Animales , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL
20.
Nutr J ; 3: 4, 2004 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15134582

RESUMEN

Homocysteine has emerged as a novel independent marker of risk for the development of cardiovascular disease over the past three decades. Additionally, there is a graded mortality risk associated with an elevated fasting plasma total homocysteine (tHcy). Metabolic syndrome (MS) and type 2 diabetes mellitus (T2DM) are now considered to be a strong coronary heart disease (CHD) risk enhancer and a CHD risk equivalent respectively. Hyperhomocysteinemia (HHcy) in patients with MS and T2DM would be expected to share a similar prevalence to the general population of five to seven percent and of even greater importance is: Declining glomerular filtration and overt diabetic nephropathy is a major determinant of tHcy elevation in MS and T2DM. There are multiple metabolic toxicities resulting in an excess of reactive oxygen species associated with MS, T2DM, and the accelerated atherosclerosis (atheroscleropathy). HHcy is associated with an increased risk of cardiovascular disease, and its individual role and how it interacts with the other multiple toxicities are presented.The water-soluble B vitamins (especially folate and cobalamin-vitamin B12) have been shown to lower HHcy. The absence of the cystathionine beta synthase enzyme in human vascular cells contributes to the importance of a dual role of folic acid in lowering tHcy through remethylation, as well as, its action of being an electron and hydrogen donor to the essential cofactor tetrahydrobiopterin. This folate shuttle facilitates the important recoupling of the uncoupled endothelial nitric oxide synthase enzyme reaction and may restore the synthesis of the omnipotent endothelial nitric oxide to the vasculature.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA