Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell Biochem ; 465(1-2): 175-185, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31853800

RESUMO

Cutaneous changes like rash and hair loss, as well as other neurogenic inflammation side effects, occur frequently during anticancer treatment with the epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI), erlotinib. These adverse events may be so severe that they impair the patient's compliance with the treatment or even cause its discontinuation. In the current preclinical study, rats (9.2 weeks) were treated with erlotinib (10 mg/kg/day) ± aprepitant (2 mg/kg/day) for 12 weeks. Visual changes in the development of facial skin lesions/hair loss and SP-receptor expression (immunohistochemically) in facial skin tissue were assessed; also changes in plasma magnesium, 8-isoprostane, substance P (SP), neutrophil superoxide production, and cardiac function (echocardiography) were measured. Erlotinib lowered plasma magnesium 14%, elevated SP 65%, caused 3.7-fold higher basal superoxide production, 2.5-fold higher 8-isoprostane levels, 11.6% lower cardiac systolic, and 10.9% lower diastolic function. Facial dermatological changes (alopecia, skin reddening, scabbing, nose crusting) occurred by 4 weeks (± + to ++) in erlotinib-treated rats, and progressively worsened (±++ to +++) by week 12. Facial skin SP-receptor upregulation (78% higher) occurred in epidermal and hair follicle cells. All adverse effects were substantially and significantly mitigated by aprepitant, including a 62% lowering of skin SP-receptors (p < 0.05). Elevated SP levels mediated the side effects of erlotinib treatment, but aprepitant's significant prevention of the systemic and cutaneous adverse events indicates a novel potential therapy against the side effects of this anticancer treatment.


Assuntos
Aprepitanto/farmacologia , Toxidermias , Cloridrato de Erlotinib/efeitos adversos , Doenças do Sistema Nervoso , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Animais , Toxidermias/tratamento farmacológico , Toxidermias/metabolismo , Toxidermias/patologia , Cloridrato de Erlotinib/farmacologia , Masculino , Doenças do Sistema Nervoso/induzido quimicamente , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Ratos , Ratos Sprague-Dawley
2.
PLoS One ; 14(1): e0210107, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30668566

RESUMO

We determined if HIV-1 expression in transgenic (HIV-1-Tg) rats enhanced hepatic genomic changes related to oxidative/nitrosative stress and lipogenesis during cART-treatment, and assessed effects of Mg-supplementation. A clinically used cART (atazanavir-ritonavir+Truvada) was given orally to control and HIV-1-Tg rats (18 weeks) with normal or 6-fold dietary-Mg. Oxidative/nitrosative and lipogenic genes were determined by real-time RT-PCR. cART induced a 4-fold upregulation of sterol regulatory element-binding protein-1 (SREBP-1) in HIV-1-Tg-rats, but not in controls; Tg rats displayed a 2.5-fold higher expression. Both were completely prevented by Mg-supplementation. Nrf2 (Nuclear erythroid-derived factor 2), a master transcription factor controlling redox homeostasis, was down-regulated 50% in HIV-Tg rats, and reduced further to 25% in Tg+cART-rats. Two downstream antioxidant genes, heme oxygenase-1(HmOX1) and Glutathione-S-transferase(GST), were elevated in HIV-Tg alone but were suppressed by cART treatment. Decreased Nrf2 in Tg±cART were normalized by Mg-supplementation along with the reversal of altered HmOX1 and GST expression. Concomitantly, iNOS (inducible nitric oxide synthase) was upregulated 2-fold in Tg+cART rats, which was reversed by Mg-supplementation. In parallel, cART-treatment led to substantial increases in plasma 8-isoprostane, nitrotyrosine, and RBC-GSSG (oxidized glutathione) levels in HIV-1-Tg rats; all indices of oxidative/nitrosative stress were suppressed by Mg-supplementation. Both plasma triglyceride and cholesterol levels were elevated in Tg+cART rats, but were lowered by Mg-supplementation. Thus, the synergistic effects of cART and HIV-1 expression on lipogenic and oxidative/nitrosative effects were revealed at the genomic and biochemical levels. Down-regulation of Nrf2 in the Tg+cART rats suggested their antioxidant response was severely compromised; these abnormal metabolic and oxidative stress effects were effectively attenuated by Mg-supplementation at the genomic level.


Assuntos
Antirretrovirais/efeitos adversos , Suplementos Nutricionais , Regulação da Expressão Gênica/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Magnésio/administração & dosagem , Animais , Sulfato de Atazanavir/efeitos adversos , Modelos Animais de Doenças , Combinação Emtricitabina e Fumarato de Tenofovir Desoproxila/efeitos adversos , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/patogenicidade , Humanos , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos , Ritonavir/efeitos adversos
3.
Diabetes Metab Syndr Obes ; 11: 389-400, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30122966

RESUMO

PURPOSE: In this clinical trial, we assessed the efficacy of magnesium (Mg) supplementation in hypomagnesemic type 2 diabetes patients in restoring serum and intracellular Mg levels. The study had two coprimary end points: the change in serum and intracellular Mg level between baseline and after 3 months of supplementation. We compared the efficacy with regard to lowering hemoglobin A1c (HbA1c), C-reactive protein (CRP), tumor necrosis factor-α (TNF-α), and 8-isoprostane as secondary end points. PATIENTS AND METHODS: In an open-label trial, 47 hypomagnesemic type 2 diabetes patients were administered 336 mg Mg daily. At baseline and after 3 months, serum, cellular Mg, and inflammation biomarkers were measured. For intracellular Mg levels, sublingual epithelial cells were analyzed by analytical scanning electron microscopy using computerized elemental X-ray analysis. Blood samples were analyzed for Mg, creatinine, HbA1c, and CRP. Systemic inflammatory markers including TNF-α and the oxidative stress marker 8-isoprostane were determined using enzyme-linked immunosorbent assay. RESULTS: Mg supplementation significantly increased the intracellular and serum levels. Statistically clinical improvement in HbA1c and CRP levels was not observed, but significant decreases in TNF-α as well as in 8-isoprostane were found. CONCLUSION: A feasible clinical method for the assessment of intracellular Mg was demonstrated in tissue samples obtained noninvasively, providing evidence for potential clinical translation of this method to routinely determine intracellular Mg concentration.

4.
Int J Mol Sci ; 19(8)2018 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-30111743

RESUMO

Chronic effects of a combination antiretroviral therapy (cART = tenofovir/emtricitatine + atazanavir/ritonavir) on systemic and cardiac oxidative stress/injury in HIV-1 transgenic (Tg) rats and protection by Mg-supplementation were assessed. cART (low doses) elicited no significant effects in normal rats, but induced time-dependent oxidative/nitrosative stresses: 2.64-fold increased plasma 8-isoprostane, 2.0-fold higher RBC oxidized glutathione (GSSG), 3.2-fold increased plasma 3-nitrotyrosine (NT), and 3-fold elevated basal neutrophil superoxide activity in Tg rats. Increased NT staining occurred within cART-treated HIV-Tg hearts, and significant decreases in cardiac systolic and diastolic contractile function occurred at 12 and 18 weeks. HIV-1 expression alone caused modest levels of oxidative stress and cardiac dysfunction. Significantly, cART caused up to 24% decreases in circulating Mg in HIV-1-Tg rats, associated with elevated renal NT staining, increased creatinine and urea levels, and elevated plasma substance P levels. Strikingly, Mg-supplementation (6-fold) suppressed all oxidative/nitrosative stress indices in the blood, heart and kidney and substantially attenuated contractile dysfunction (>75%) of cART-treated Tg rats. In conclusion, cART caused significant renal and cardiac oxidative/nitrosative stress/injury in Tg-rats, leading to renal Mg wasting and hypomagnesemia, triggering substance P-dependent neurogenic inflammation and cardiac dysfunction. These events were effectively attenuated by Mg-supplementation likely due to its substance P-suppressing and Mg's intrinsic anti-peroxidative/anti-calcium properties.


Assuntos
Antirretrovirais/efeitos adversos , Coração/efeitos dos fármacos , Magnésio/uso terapêutico , Inflamação Neurogênica/induzido quimicamente , Inflamação Neurogênica/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/uso terapêutico , Animais , Terapia Antirretroviral de Alta Atividade/efeitos adversos , Cardiotoxinas/efeitos adversos , Expressão Gênica , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , HIV-1/genética , Coração/fisiopatologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Masculino , Inflamação Neurogênica/fisiopatologia , Ativação de Neutrófilo/efeitos dos fármacos , Estresse Nitrosativo/efeitos dos fármacos , Ratos Endogâmicos F344 , Ratos Transgênicos
5.
J Cardiovasc Pharmacol ; 65(1): 54-61, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25343568

RESUMO

To determine whether the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib may cause hypomagnesemia, inflammation, and cardiac stress, erlotinib was administered to rats (10 mg · kg(-1)· d(-1)) for 9 weeks. Plasma magnesium decreased progressively between 3 and 9 weeks (-9% to -26%). Modest increases in plasma substance P (SP) occurred at 3 (27%) and 9 (25%) weeks. Neutrophil superoxide-generating activity increased 3-fold, and plasma 8-isoprostane rose 210%, along with noticeable appearance of cardiac perivascular nitrotyrosine. The neurokinin-1 (NK-1) receptor antagonist, aprepitant (2 mg · kg(-1) · d(-1)), attenuated erlotinib-induced hypomagnesemia up to 42%, reduced circulating SP, suppressed neutrophil superoxide activity and 8-isoprostane elevations; cardiac nitrotyrosine was diminished. Echocardiography revealed mild to moderately decreased left ventricular ejection fraction (-11%) and % fractional shortening (-17%) by 7 weeks of erlotinib treatment and significant reduction (-17.5%) in mitral valve E/A ratio at week 9 indicative of systolic and early diastolic dysfunction. Mild thinning of the left ventricular posterior wall suggested early dilated cardiomyopathy. Aprepitant completely prevented the erlotinib-induced systolic and diastolic dysfunction and partially attenuated the anatomical changes. Thus, chronic erlotinib treatment does induce moderate hypomagnesemia, triggering SP-mediated oxidative/inflammation stress and mild-to-moderate cardiac dysfunction, which can largely be corrected by the administration of the SP receptor blocker.


Assuntos
Receptores ErbB/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/toxicidade , Quinazolinas/toxicidade , Animais , Aprepitanto , Ecocardiografia , Cloridrato de Erlotinib , Magnésio/sangue , Masculino , Morfolinas/farmacologia , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-1/efeitos dos fármacos , Receptores da Neurocinina-1/metabolismo , Substância P/sangue , Função Ventricular Esquerda/efeitos dos fármacos
6.
Am J Physiol Regul Integr Comp Physiol ; 305(10): R1102-11, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24049113

RESUMO

Use of protease inhibitors (PI) in HIV patients is associated with hyperlipidemia and increased risk of coronary heart disease. Chronic systemic and cardiac effects of ritonavir (RTV), a universal PI booster, and Mg supplementation were examined. RTV was administered (75 mg·kg(-1)·day(-1) po) to Lewis × Brown-Norway hybrid (LBNF1) rats for up to 8 wk; significant increases in plasma triglyceride and cholesterol occurred from 8 days to 8 wk. At 5 wk, the expression of selected hepatic genes (CYP7A1, CITED2, G6PC, and ME-1), which are key to lipid catabolism/synthesis, were altered toward lipogenesis. Dietary Mg supplementation (six-fold higher) completely reversed the altered expression of these genes and attenuated both hypertriglyceridemia and hypercholesterolemia. Neutrophils isolated from the RTV-treated rats displayed a three-fold higher basal and a twofold higher stimulated superoxide production; plasma isoprostane and red blood cell (RBC) GSSG levels were elevated two- to three-fold. All oxidative indices were normalized by Mg supplementation. After 5 wk, RTV caused significant decreases in cardiac left ventricular (LV) shortening fraction and LV ejection fraction; mitral valve early/late atrial ventricular filling (E/A) ratio was reduced accompanied by LV posterior wall thinning. Immunohistochemical staining revealed significant white blood cell (WBC) infiltration (5 wk) and prominent fibrosis (8 wk) in the RTV hearts. Mg supplementation attenuated RTV-induced declines in systolic and diastolic (improved mitral valve E/A ratio) function (>70%), lessened LV posterior wall thinning (by 75%), and substantially decreased the pathological markers. The known clinical hyperlipidemia effects of RTV can be mimicked in the LBNF1 rats; in association, systemic oxidative stress and progressive cardiac dysfunction occurred. Remarkably, Mg supplementation alone suppressed RTV-mediated hyperlipidemia, oxidative stress, and cardiac dysfunction.


Assuntos
Cardiopatias/induzido quimicamente , Hiperlipidemias/induzido quimicamente , Magnésio/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Ritonavir/toxicidade , Ração Animal , Animais , Dieta , Suplementos Nutricionais , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores da Protease de HIV/toxicidade , Cardiopatias/tratamento farmacológico , Masculino , Ratos , Aumento de Peso
7.
Can J Physiol Pharmacol ; 90(10): 1413-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23067376

RESUMO

Angiotensin may promote endothelial dysfunction through iron accumulation. To research this, bovine endothelial cells (ECs) were incubated with iron (30 µmol·L⁻¹) with or without angiotensin II (100 nmol·L⁻¹). After incubation for 6 h, it was observed that the addition of angiotensin enhanced EC iron accumulation by 5.1-fold compared with a 1.8-fold increase for cells incubated with iron only. This enhanced iron uptake was attenuated by losartan (100 nmol·L⁻¹), d-propranolol (10 µmol·L⁻¹), 4-HO-propranolol (5 µmol·L⁻¹), and methylamine, but not by vitamin E or atenolol. After 6 h of incubation, angiotensin plus iron provoked intracellular oxidant formation (2'7'-dichlorofluorescein diacetate (DCF-DA) fluorescence) and elevated oxidized glutathione; significant loss of cell viability occurred at 48 h. Stimulated prostacyclin release decreased by 38% (6 h) and NO synthesis was reduced by 41% (24 h). Both oxidative events and functional impairment were substantially attenuated by losartan or d-propranolol. It is concluded that angiotensin promoted non-transferrin-bound iron uptake via AT-1 receptor activation, leading to EC oxidative functional impairment. The protective effects of d-propranolol and 4-HO-propranolol may be related to their lysosomotropic properties.


Assuntos
Angiotensina II/metabolismo , Endotélio Vascular/metabolismo , Epoprostenol/metabolismo , Ferro/metabolismo , Losartan/farmacologia , Óxido Nítrico/metabolismo , Propranolol/análogos & derivados , Antagonistas Adrenérgicos beta/química , Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Angiotensina II/química , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/metabolismo , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Epoprostenol/agonistas , Epoprostenol/antagonistas & inibidores , Glutationa/metabolismo , Ferro/efeitos adversos , Sobrecarga de Ferro/prevenção & controle , Losartan/uso terapêutico , Óxido Nítrico/agonistas , Óxido Nítrico/antagonistas & inibidores , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Propranolol/farmacologia , Propranolol/uso terapêutico , Receptor Tipo 1 de Angiotensina/metabolismo
8.
Can J Physiol Pharmacol ; 90(9): 1257-68, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22913465

RESUMO

d-Propranolol (d-Pro: 2-8 mg·(kg body mass)(-1)·day(-1)) protected against cardiac dysfunction and oxidative stress during 3-5 weeks of iron overload (2 mg Fe-dextran·(g body mass)(-1)·week(-1)) in Sprague-Dawley rats. At 3 weeks, hearts were perfused in working mode to obtain baseline function; red blood cell glutathione, plasma 8-isoprostane, neutrophil basal superoxide production, lysosomal-derived plasma N-acetyl-ß-galactosaminidase (NAGA) activity, ventricular iron content, and cardiac iron deposition were assessed. Hearts from the Fe-treated group of rats exhibited lower cardiac work (26%) and output (CO, 24%); end-diastolic pressure rose 1.8-fold. Further, glutathione levels increased 2-fold, isoprostane levels increased 2.5-fold, neutrophil superoxide increased 3-fold, NAGA increased 4-fold, ventricular Fe increased 4.9-fold; and substantial atrial and ventricular Fe-deposition occurred. d-Pro (8 mg) restored heart function to the control levels, protected against oxidative stress, and decreased cardiac Fe levels. After 5 weeks of Fe treatment, echocardiography revealed that the following were depressed: percent fractional shortening (%FS, 31% lower); left ventricular (LV) ejection fraction (LVEF, 17%), CO (25%); and aortic pressure maximum (P(max), 24%). Mitral valve E/A declined by 18%, indicating diastolic dysfunction. Cardiac CD11b+ infiltrates were elevated. Low d-Pro (2 mg) provided modest protection, whereas 4-8 mg greatly improved LVEF (54%-75%), %FS (51%-81%), CO (43%-78%), P(max) (56%-100%), and E/A >100%; 8 mg decreased cardiac inflammation. Since d-Pro is an antioxidant and reduces cardiac Fe uptake as well as inflammation, these properties may preserve cardiac function during Fe overload.


Assuntos
Antagonistas Adrenérgicos beta/uso terapêutico , Cardiopatias/prevenção & controle , Sobrecarga de Ferro/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Propranolol/uso terapêutico , Acetilglucosaminidase/sangue , Antagonistas Adrenérgicos beta/administração & dosagem , Antagonistas Adrenérgicos beta/química , Animais , Débito Cardíaco/efeitos dos fármacos , Progressão da Doença , Relação Dose-Resposta a Droga , Ecocardiografia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Glutationa , Cardiopatias/sangue , Cardiopatias/etiologia , Cardiopatias/metabolismo , Sobrecarga de Ferro/sangue , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/metabolismo , Masculino , Miocárdio/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Perfusão , Propranolol/administração & dosagem , Propranolol/química , Ratos , Ratos Sprague-Dawley , Estereoisomerismo , Superóxido Dismutase/metabolismo , Resultado do Tratamento
9.
Can J Physiol Pharmacol ; 90(8): 1145-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22646904

RESUMO

We determined whether the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) N-​(3-​chlorophenyl)-​6,​7-​dimethoxy-​4-​quinazolinamine (tyrphostin AG-1478) causes hypomagnesemia and cardiac dysfunction in rats. Tyrphostin was administered (3 times per week, intraperitoneal injection, to achieve 21.4 mg·(kg body mass)(-1)·day(-1)) to normomagnesemic rats for 5 weeks. Levels of magnesium in the plasma of the tyrphostin-treated rats decreased significantly by the following amount: 17% at week 1, 27% at week 2, and 26%-35% between weeks 3 to 5. Levels of the plasma lipid peroxidation marker 8-isoprostane rose significantly: by 58% at week 1, 168% at week 3, and 113% at week 5. At week 5, blood neutrophils from the tyrphostin-treated group displayed a 2.26-fold higher basal level of O(2)(·-) generation; the ratio of oxidized glutathione (glutathione disulfide; GSSG) to reduced glutathione (GSH) in the red blood cells increased 2.5-fold. At week 5, echocardiography revealed that TKI treatment resulted in significant cardiac systolic dysfunction, with impaired diastolic function and dilated cardiomyopathy. Since hypomagnesemia alone can trigger oxidative stress and cardiac injury, we suggest that inhibition of EGFR-TK caused magnesium wasting, which partly contributed to decreased cardiac contractility.


Assuntos
Ecocardiografia/efeitos dos fármacos , Inibidores Enzimáticos/efeitos adversos , Receptores ErbB/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/efeitos adversos , Erros Inatos do Transporte Tubular Renal/induzido quimicamente , Tirfostinas/efeitos adversos , Animais , Biomarcadores/sangue , Cálcio/sangue , Dinoprosta/análogos & derivados , Dinoprosta/sangue , Ecocardiografia/métodos , Ecocardiografia/estatística & dados numéricos , Glutationa/sangue , Masculino , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Erros Inatos do Transporte Tubular Renal/sangue , Superóxidos/sangue
10.
Annu Rev Nutr ; 32: 55-71, 2012 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-22404119

RESUMO

In recent years, increasing awareness of hypomagnesemia has resulted in clinical trials that associate this mineral deficiency with diabetes, metabolic syndrome, and drug therapies for cancer and cardiovascular diseases. However, diagnostic testing for tissue deficiency of magnesium still presents a challenge. Investigations of animal and cellular responses to magnesium deficiency have found evidence of complex proinflammatory pathways that may lead to greater understanding of mediators of the pathobiology in neuronal, cardiovascular, intestinal, renal, and hematological tissues. The roles of free radicals, cytokines, neuropeptides, endotoxin, endogenous antioxidants, and vascular permeability, and interventions to limit the inflammatory response associated with these parameters, are outlined in basic studies of magnesium deficiency. It is hoped that this limited review of inflammation associated with some diseases complicated by magnesium deficiency will prompt greater awareness by clinicians and other health providers and in turn increase efforts to prevent and treat this disorder.


Assuntos
Deficiência de Magnésio/etiologia , Deficiência de Magnésio/imunologia , Alcoolismo/imunologia , Alcoolismo/fisiopatologia , Animais , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/fisiopatologia , Citocinas/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/imunologia , Humanos , Deficiência de Magnésio/sangue , Deficiência de Magnésio/prevenção & controle , Síndrome Metabólica/imunologia , Síndrome Metabólica/fisiopatologia , Neuropeptídeos/sangue , Osteoporose/imunologia , Osteoporose/fisiopatologia
12.
Exp Clin Cardiol ; 16(4): 121-4, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22131854

RESUMO

BACKGROUND/OBJECTIVE: Hypomagnesemia (Hypo-Mg) in rodents leads to neurogenic inflammation associated with substance P (SP) elevations; neutral endopeptidase (NEP) is a principle cell surface proteolytic enzyme, which degrades SP. The effects of chronic Hypo-Mg on neutrophil NEP activity, cell activation and the associated cardiac dysfunction were examined. METHODS/RESULTS: Male Sprague-Dawley rats (180 g) were fed Mg-sufficient or Mg-deficient (Hypo-Mg) diets for five weeks. Enriched blood neutrophils were isolated at the end of one, three and five weeks by step gradient centrifugation. NEP enzymatic activity decreased by 20% (P value was nonsignificant), 50% (P<0.025) and 57% (P<0.01), respectively, for week 1, 3 and 5 Hypo-Mg rats. In association, neutrophil basal superoxide (•O(2) (-))-generating activities were elevated: 30% at week 1 (P value was nonsignificant), and fourfold to sevenfold for weeks 3 to 5 (P<0.01). Maximal phorbol myristate acetate-stimulated •O(2) (-) production by Hypo-Mg neutrophils increased twofold at week 5. Also, plasma 8-isoprostane levels were elevated twofold to threefold, and red blood cell glutathione decreased by 50% (P<0.01) after three to five weeks of chronic Hypo-Mg. When Hypo-Mg rats were treated with the SP receptor blocker (L-703,606), neutrophil NEP activities were retained at 75% (week 3) and 77% (week 5) (P<0.05); activation of neutrophil •O(2) (-) and other oxidative indexes were also significantly (P<0.05) attenuated. After five weeks, histochemical (hematoxylin and eosin) staining of Hypo-Mg-treated rat ventricles revealed significant white blood cell infiltration, which was substantially reduced by L-703,606. Echocardiography after three weeks of Hypo-Mg only showed modest diastolic impairment, but five weeks resulted in significant (P<0.05) depression in both left ventricular systolic and diastolic functions; changes in these functional parameters were attenuated by L-703,606. CONCLUSION: NEP activity regulates neutrophil •O(2) (-) formation by controlling SP bioavailability. When oxidative inactivation of NEP is prevented by SP receptor blockade, partial protection is afforded against cardiac contractile dysfunction.

13.
Am J Med Sci ; 342(2): 125-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21747282

RESUMO

In rodents with dietary magnesium deficiency (Mg deficiency), hypomagnesemia, occurs leading to a rise in circulating substance P from neuronal tissues to trigger systemic inflammatory stress in cardiac and intestinal tissues. Sustained elevations of substance P may result from impaired neutral endopeptidase (NEP) activity due to reactive oxygen and reactive nitrogen species. Associated increase in intestinal permeability includes infiltration of WBC and endotoxemia, which can further amplify the systemic inflammatory response that leads to impaired contractile function associated with up-regulation of the cardiac CD14 endotoxin receptor. The neurogenic signal transduction pathways that we have identified in the pro-oxidant/pro-inflammatory processes found with prolonged hypomagnesemia are described in this report.


Assuntos
Sistema Cardiovascular/metabolismo , Mucosa Intestinal/metabolismo , Deficiência de Magnésio , Estresse Oxidativo/fisiologia , Animais , Sistema Cardiovascular/fisiopatologia , Humanos , Inflamação/metabolismo , Inflamação/fisiopatologia , Intestinos/fisiopatologia , Deficiência de Magnésio/metabolismo , Deficiência de Magnésio/fisiopatologia , Neprilisina/metabolismo , Ratos , Espécies Reativas de Nitrogênio/metabolismo , Receptores Imunológicos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Substância P/fisiologia
14.
Magnes Res ; 23(4): S199-206, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20971697

RESUMO

Hypomagnesemia continues to cause difficult clinical problems, such as significant cardiac arrhythmias where intravenous magnesium therapy can be lifesaving. Nutritional deficiency of magnesium may present with some subtle symptoms such as leg cramps and occasional palpitation. We have investigated dietary-induced magnesium deficiency in rodent models to assess the pathobiology associated with prolonged hypomagnesemia. We found that neuronal sources of the neuropeptide, substance P (SP), contributed to very early prooxidant/proinflammatory changes during Mg deficiency. This neurogenic inflammation is systemic in nature, affecting blood cells, cardiovascular, intestinal, and other tissues, leading to impaired cardiac contractility similar to that seen in patients with heart failure. We have used drugs that block the release of SP from neurons and SP-receptor blockers to prevent some of these pathobiological changes; whereas, blocking SP catabolism enhances inflammation. Our findings emphasize the essential role of this cation in preventing cardiomyopathic changes and intestinal inflammation in a well-studied animal model, and also implicate the need for more appreciation of the potential clinical relevance of optimal magnesium nutrition and therapy.


Assuntos
Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Deficiência de Magnésio/metabolismo , Deficiência de Magnésio/fisiopatologia , Animais , Humanos , Deficiência de Magnésio/imunologia , Receptores da Neurocinina-1/metabolismo , Substância P/metabolismo
15.
Magnes Res ; 22(3): 167S-173S, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19780404

RESUMO

During dietary deficiency of magnesium neurogenic inflammation is mediated, primarily, by elevated levels of substance P (SP). The enzyme most specific for degrading this neuropeptide is neutral endopeptidase (NEP). In recent studies we found that pharmacological inhibition of NEP by phosphoramidon resulted in elevated plasma levels of SP and greater oxidative stress. We also observed that hypomagnesemia reduced cardiac and intestinal expression of NEP. In these magnesium-deficient rats increased intestinal permeability and impaired cardiac contractility occurred. In our colony of genetically-engineered NEP knockout mice that have reduced ability to degrade SP, we found increased oxidative stress that was prevented by SP (neurokinin-1) receptor blockade. Thus, we submit that inhibition of NEP by pharmacological, genetic and dietary approaches (magnesium restriction), causes greater neurogenic inflammation that may result in increased intestinal and cardiac dysfunction.


Assuntos
Deficiência de Magnésio/complicações , Neprilisina/antagonistas & inibidores , Inflamação Neurogênica/etiologia , Substância P/metabolismo , Animais , Glicopeptídeos/farmacologia , Coração/efeitos dos fármacos , Imuno-Histoquímica , Intestinos/efeitos dos fármacos , Inflamação Neurogênica/metabolismo , Inibidores de Proteases/farmacologia , Ratos , Regulação para Cima
16.
Am J Med Sci ; 338(1): 22-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19593099

RESUMO

Hypomagnesemia continues to be a significant clinical disorder that is present in patients with diabetes mellitus, alcoholism, and treatment with magnesuric drugs (diuretics, cancer chemotherapy agents, etc.). To determine the role of magnesium in cardiovascular pathophysiology, we have used dietary restriction of this cation in animal models. This review highlights some key observations that helped formulate the hypothesis that release of substance P (SP) during experimental dietary Mg deficiency (MgD) may initiate a cascade of deleterious inflammatory, oxidative, and nitrosative events, which ultimately promote cardiomyopathy, in situ cardiac dysfunction, and myocardial intolerance to secondary stresses. SP acts primarily through neurokinin-1 receptors of inflammatory and endothelial cells, and may induce production of reactive oxygen and nitrogen species (superoxide anion, NO*, peroxynitrite, hydroxyl radical), leading to enhanced consumption of tissue antioxidants; stimulate release of inflammatory mediators; promote tissue adhesion molecule expression; and enhance inflammatory cell tissue infiltration and cardiovascular lesion formation. These SP-mediated events may predispose the heart to injury if faced with subsequent oxidative stressors (ischemia/reperfusion, certain drugs) or facilitate development of in situ cardiac dysfunction, especially with prolonged dietary Mg restriction. Significant protection against most of these MgD-mediated events has been observed with interventions that modulate neuronal SP release or its bioactivity, and with several antioxidants (vitamin E, probucol, epicaptopril, d-propranolol). In view of the clinical prevalence of hypomagnesemia, new treatments, beyond magnesium repletion, may be needed to diminish deleterious neurogenic and prooxidative components described in this article.


Assuntos
Cardiomiopatias , Deficiência de Magnésio/complicações , Inflamação Neurogênica , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/fisiopatologia , Dieta , Endotoxemia/metabolismo , Humanos , Traumatismo por Reperfusão Miocárdica/metabolismo , Inflamação Neurogênica/etiologia , Inflamação Neurogênica/fisiopatologia , Neuropeptídeos/metabolismo , Estresse Oxidativo , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores da Neurocinina-1/metabolismo
17.
Cardiovasc Toxicol ; 9(2): 78-85, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19484392

RESUMO

Cardiovascular effects of chronic AZT treatment on SD male rats (185 g) fed either a normal Mg diet (0.1% MgO) or a high Mg diet (0.6% MgO) were examined. AZT treatment (1 mg/ml drinking water) for 3 weeks led to a 5.5-fold (0.88 +/- 0.11 nmol/min/10(6) cells, P < 0.05) elevation in neutrophil basal activity of O2(-) production versus controls (0.16 +/- 0.03 nmol/min, assayed ex vivo as SOD-inhibitable cytochrome c reduction). Concomitantly, plasma 8-isoprostane and PGE(2) levels rose 2.1-fold and 3-fold (both P < 0.05), respectively, compared to control; however, RBC GSH decreased 28% (P < 0.02) with GSSG content increased 3-fold, indicative of systemic oxidative stress. High Mg diet substantially attenuated the AZT-induced neutrophil activation by 70% (0.26 +/- 0.05 nmol/min, P < 0.05); reduced plasma 8-isoprostane and PGE(2) to levels comparable to normal; and RBC GSH was restored back to 92% of control. AZT alone caused moderate, but significant vascular inflammatory lesions in the heart (assessed by H&E staining). Immunohistochemical staining revealed significantly higher (about 4-fold) infiltration of CD11b positive cells (WBC surface marker) in the atria and ventricles of AZT-treated rats. However, these inflammatory pathological markers were minimal in samples of rats treated with AZT plus high Mg diet. Moreover, AZT alone significantly (P < 0.02) decreased rat weight gain by 21% at 3 weeks; Mg-supplementation completely prevented (P < 0.05) the weight gain loss due to AZT intake. It is concluded that high dietary Mg may provide beneficial effects against AZT toxicity due to its systemic antioxidative/anti-inflammatory properties.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Suplementos Nutricionais , Magnésio/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Zidovudina/toxicidade , Animais , Doenças Cardiovasculares/induzido quimicamente , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Masculino , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley
18.
Cardiovasc Res ; 81(3): 500-8, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18835843

RESUMO

Hypertension and heart failure are worldwide health problems of ever-increasing proportions. A failure of the heart, during either systolic and/or diastolic phases of the cardiac cycle, has its origins rooted in an adverse structural, biochemical, and molecular remodelling of myocardium that involves its cellular constituents, extracellular matrix, and intramural coronary vasculature. Herein we focus on the pathogenic role of a dyshomeostasis of several macro- (i.e. Ca(2+) and Mg(2+)) and micronutrients (i.e. Zn(2+), Se(2+), and vitamin D) in contributing to adverse remodelling of the myocardium and its failure as a pulsatile muscular pump. An improved understanding of how these macro- and micronutrients account for the causes and consequences of adverse myocardial remodelling carries with it the potential of identifying new biomarkers predictive of risk, onset and progression, and response to intervention(s), which could be monitored non-invasively and serially over time. Moreover, such incremental knowledge will serve as the underpinning to the development of novel strategies aimed at preventing and/or regressing the ongoing adverse remodelling of myocardium. The time is at hand to recognize the importance of macro- and micronutrient dyshomeostasis in the evaluation and management of hypertension and heart failure.


Assuntos
Insuficiência Cardíaca/metabolismo , Hipertensão/metabolismo , Micronutrientes/metabolismo , Miocárdio/metabolismo , Remodelação Ventricular , Animais , Biomarcadores/metabolismo , Cálcio/metabolismo , Cobre/metabolismo , Progressão da Doença , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Homeostase , Humanos , Hipertensão/complicações , Hipertensão/fisiopatologia , Hipertensão/terapia , Magnésio/metabolismo , Miocárdio/enzimologia , Prognóstico , Selênio/metabolismo , Vitamina D/metabolismo , Zinco/metabolismo
19.
Magnes Res ; 20(3): 208-12, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17972464

RESUMO

Severe Mg2+ deficiency provokes pro-oxidative and pro-inflammatory changes, and also has been shown to be pro-apoptotic in thymus and certain cell cultures. In this study we examined the extent that chronic severe dietary Mg2+ deficiency induces apoptosis in the heart. Sprague-Dawley rats were fed during three weeks with normal (25 mM, Mg-control) or magnesium deficient (2.25 mM, Mg-deficient) diets, after which, hearts were harvested and frozen. DNA fragmentation was examined in heart tissue sections, and while < 1% of nuclei were positive for apoptosis in Mg-control rat tissue, over 32% of nuclei gave positive for Klenow fragments in hearts from Mg-deficient rats. Caspase 3 activity measurements in heart homogenates showed a 3.9-fold increase in enzyme activity in Mg-deficient rat hearts compared to Mg-controls (p < 0.002); and furthermore, western blot analysis of cleaved PARP (caspase 3 substrate), showed a 4.6-fold increase of cleaved PARP in Mg-deficient rat hearts (p < 0.002). In summary, our data indicate that chronic Mg2+ deficiency induces apoptosis of myocardium in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Coração/efeitos dos fármacos , Magnésio/administração & dosagem , Miocárdio/metabolismo , Animais , Western Blotting , Caspase 3/metabolismo , Doença Crônica , Fragmentação do DNA/efeitos dos fármacos , Suplementos Nutricionais , Magnésio/metabolismo , Deficiência de Magnésio/fisiopatologia , Masculino , Miocárdio/citologia , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Ratos Sprague-Dawley
20.
Mol Cell Biochem ; 306(1-2): 59-69, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17657590

RESUMO

The aim of this study was to determine the effect of magnesium deficiency on small intestinal morphology and function. Rats were assigned to 4 groups and placed on magnesium sufficient or deficient diet for 1 or 3 weeks. Infiltration of neutrophils and mucosal injury were assessed in stained sections of small intestine. Magnesium deficiency alone induced a significant increase in neutrophil infiltration and increased vascular ICAM-1 expression, in the absence of changes in mucosal injury or expression of proinflammatory mediators. Magnesium deficiency was associated with hyposecretory epithelial cell responses and vascular macromolecular leak in the small intestine and lung, which was attributed partly to reduced expression of NOS-3. To determine the effect of hypomagnesmia on the intestinal responses to a known oxidative stress, groups of rats were randomized to either sham operation or superior mesenteric artery occlusion for 10 (non-injurious) or 30 (injurious) minutes followed by a 1- or 4-hour reperfusion period. In response to mesenteric ischemia/reperfusion, deficient rats showed exaggerated PMN influx, but similar mucosal injury. Intestinal ischemia in sufficient animals induced vascular macromolecular leak in the small intestine and lung at 4 hours of reperfusion, with levels similar to those observed in untreated deficient rats. Acute magnesium repletion of deficient rats 24 h before surgery attenuated the exaggerated inflammation in deficient rats. These data show that magnesium deficiency induced a subclinical inflammation in the small intestine in the absence of mucosal injury, but with significant functional changes in local and remote organs and increased sensitivity to oxidative stress.


Assuntos
Enterite/etiologia , Doenças do Jejuno/etiologia , Deficiência de Magnésio/complicações , Estresse Oxidativo , Animais , Modelos Animais de Doenças , Enterite/metabolismo , Enterite/patologia , Membrana Eritrocítica/metabolismo , Glutationa/metabolismo , Técnicas Imunoenzimáticas , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Doenças do Jejuno/metabolismo , Doenças do Jejuno/patologia , Magnésio/sangue , Masculino , Neutrófilos/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...