Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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.

10.
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
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
Mol Cell Biochem ; 288(1-2): 213-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16718379

RESUMO

In this study we have tested the effects of d-propranolol (D-Pro) on the iron uptake, iron release and oxidative response of iron-loaded cells in a cellular model of iron-overload using isolated rat peritoneal macrophages incubated with iron-dextran (Fe-D). Pretreatment of macrophages with D-Pro (5-200 microM) prior to Fe-D exposure decreased the cellular iron content and partially prevented iron release from latex-activated macrophages. Release of reactive oxygen species from activated cells was detected by dichlorodihydrofluorescein (DCDHF, 5 microM) oxidation. We found that loading cells with Fe-D increased their response to latex, which was prevented by the lysosomotropic antioxidant agent D-Pro (10 microM).


Assuntos
Ferro/metabolismo , Macrófagos Peritoneais/metabolismo , Propranolol/farmacologia , Animais , Relação Dose-Resposta a Droga , Complexo Ferro-Dextran/metabolismo , Ativação de Macrófagos , Macrófagos Peritoneais/efeitos dos fármacos , Masculino , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
18.
J Pharmacol Exp Ther ; 317(2): 522-8, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16456084

RESUMO

The influence of selected beta-receptor blockers on iron overload and oxidative stress in endothelial cells (ECs) was assessed. Confluent bovine ECs were loaded with iron dextran (15 muM) for 24 h and then exposed to dihydroxyfumarate (DHF), a source of reactive oxygen species, for up to 2 h. Intracellular oxidant formation, monitored by fluorescence of 2',7'-dichlorofluorescin (DCF; 30 microM), increased and peaked at 30 min; total glutathione decreased by 52 +/- 5% (p < 0.01) at 60 min. When the ECs were pretreated 30 min before iron loading with 1.25 to 10 microM d-propranolol, glutathione losses were attenuated 15 to 80%, with EC(50) = 3.1 microM. d-Propranolol partially inhibited the DCF intensity increase, but atenolol up to 10 microM was ineffective. At 2 h, caspase 3 activity was elevated 3.2 +/- 0.3-fold (p < 0.01) in the iron-loaded and DHF-treated ECs, and cell survival, determined 24 h later, decreased 47 +/- 6% (p < 0.01). Ten micromoles of d-propranolol suppressed the caspase 3 activation by 63% (p < 0.05) and preserved cell survival back to 88% of control (p < 0.01). In separate experiments, 24-h iron loading resulted in a 3.6 +/- 0.8-fold increase in total EC iron determined by atomic absorption spectroscopy; d-propranolol at 5 microM reduced this increase to 1.5 +/- 0.4-fold (p < 0.01) of controls. Microscopic observation by Perls' staining revealed that the excessive iron accumulated in vesicular endosomal/lysosomal structures, which were substantially diminished by d-propranolol. We previously showed that propranolol could readily concentrate into the lysosomes and raise the intralysosomal pH; it is suggested that the lysosomotropic properties of d-propranolol retarded the EC iron accumulation and thereby conferred the protective effects against iron load-mediated cytotoxicity.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Células Endoteliais/efeitos dos fármacos , Ferro/metabolismo , Lisossomos/efeitos dos fármacos , Estresse Oxidativo , Propranolol/farmacologia , Animais , Caspase 3 , Caspases/metabolismo , Bovinos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Glutationa/metabolismo , Lisossomos/enzimologia , Lisossomos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
19.
Mol Cell Biochem ; 278(1-2): 53-7, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16180088

RESUMO

Substance P is elevated in plasma and in other tissues during Mg-deficiency, and was found localised to neuronal C-fibres of cardiac and intestinal tissues, where it could promote neurogenic inflammation. Plasma prostaglandin E2 (PGE2), indicative of systemic inflammation, rose significantly (>or=4 fold, p<0.01) after 1 week and remained elevated through week 2 and 3 in rat on the Mg-deficient (MgD) diet. Concomitantly, total blood glutathione decreased by 50%. Immunohistochemical staining for endotoxin (lipopolysaccaride, LPS) receptor, CD14 was prominent in macrophage-type cells in intestinal tissue; more importantly, cardiac tissue revealed both CD11b (monocyte/macrophage surface protein) and CD14 positive cells after 3 weeks in rats on MgD diet. Western blot analysis indicated a significant increase in the endotoxin receptor protein level in the 3 week MgD hearts. Since CD14 is known to be up-regulated in cells exposed to LPS, these observations suggest that prolonged Mg-deficiency results in increased intestinal permeability to bacterial products that induce the endotoxin receptor in cells localized to myocardial and intestinal tissues. These CD14 positive cells may amplify the cardiomyopathic inflammatory process by stimulating TNF-alpha and other pro-inflammatory cytokines.


Assuntos
Inflamação/metabolismo , Deficiência de Magnésio/complicações , Receptores Imunológicos/metabolismo , Animais , Imunofluorescência , Mucosa Intestinal/metabolismo , Intestinos/citologia , Masculino , Miocárdio/citologia , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Imunológicos/sangue , Especificidade da Espécie , Fatores de Tempo
20.
J Pharmacol Exp Ther ; 311(1): 8-13, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15169828

RESUMO

Elevated plasma levels of the neuropeptide substance P (SP) precede the perivascular inflammatory infiltrate seen in hearts of Mg(2+)-deficient (MgD) animals. The N-methyl-d-aspartate (NMDA) receptor is found in neurons, and activation of this receptor participates in SP release; under normal circumstances, this release can be blocked by Mg(2+). Therefore, we reasoned that blockade of the NMDA receptor with dizolcipine maleate (a noncompetitive NMDA receptor antagonist) would prevent SP release from C-fibers due to MgD. In this study, animals were implanted with slow-release pellets containing dizolcipine or placebo and were fed with diet sufficient in Mg(2+) or deficient with only 9% of USDA-recommended Mg(2+). SP immunostaining of dorsal root ganglia showed a time-dependent depletion of SP in the MgD animals, with a dramatic decrease of SP by week 2; this depletion was prevented by pretreatment with dizolcipine maleate. The significant increase in plasma prostaglandin E(2) levels during MgD was prevented by dizolcipine, and the loss of total red blood cell glutathione content was significantly attenuated by NMDA blockade after 3 weeks of MgD (p < 0.01 versus controls). Immunohistochemical and Western blot analyses of ventricular tissue demonstrated that NMDA receptor blockade abolished MgD-related increase of endothelium adhesion molecule CD54 (weeks 1 and 2; p < 0.05), and of monocyte/macrophage surface protein CD11b expression (week 3; p < 0.05). We conclude that NMDA receptor blockade with dizolcipine maleate prevented SP depletion and reduced perivascular inflammatory infiltrates, thus decreasing cardiac injury due to Mg(2+) deficiency.


Assuntos
Maleato de Dizocilpina/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Cardiopatias/prevenção & controle , Deficiência de Magnésio/complicações , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Imunofluorescência , Cardiopatias/etiologia , Cardiopatias/patologia , Inflamação/prevenção & controle , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Estresse Fisiológico/etiologia , Substância P/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...