Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oxid Med Cell Longev ; 2018: 4285694, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30228855

RESUMO

Cadmium (Cd) is a harmful heavy metal, which causes severe brain damage and neurotoxic effects. Polydeoxyribonucleotide (PDRN) stimulates adenosine A2A receptor, thus contrasting several deleterious mechanisms in course of tissue damages. We aimed to investigate the possible neuroprotective effect of PDRN in a murine model of Cd-induced brain toxicity. Male C57 BL/6J mice were treated as follows: vehicle (0.9% NaCl, 1 ml/kg/day), PDRN (8 mg/kg/day), CdCl2 (2 mg/kg/day), and CdCl2 + PDRN. Animals were tested with the Morris water maze test to assess spatial memory and learning. After 14 days of treatment, brains were processed to evaluate the presence of edema in the cerebral tissue, the expression of mammalian target of rapamycin kinase (mTOR) and brain-derived neurotrophic factor (BDNF), and the morphological behavior of the hippocampal structures. After CdCl2 administration, the escape latency was high, protein expression of BDNF was significantly decreased if compared to controls, mTOR levels were higher than normal controls, and brain edema and neuronal damages were evident. The coadministration of CdCl2 and PDRN significantly diminished the escape latency, increased BDNF levels, and decreased protein expression of mTOR. Furthermore, brain edema was reduced and the structural organization and the number of neurons, particularly in the CA1 and CA3 hippocampal areas, were improved. In conclusion, a functional, biochemical, and morphological protective effect of PDRN against Cd induced toxicity was demonstrated in mouse brain.


Assuntos
Cádmio/toxicidade , Fármacos Neuroprotetores/farmacologia , Polidesoxirribonucleotídeos/farmacologia , Animais , Edema Encefálico/patologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Glutationa/metabolismo , Hipocampo/patologia , Masculino , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Polidesoxirribonucleotídeos/administração & dosagem , Tempo de Reação/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
3.
Curr Med Chem ; 24(35): 3879-3893, 2017 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-28762312

RESUMO

BACKGROUND: Cadmium (Cd) is a heavy metal particularly hazardous for human health, as it is highly diffused and, therefore, a ubiquitous environmental toxicant. In fact, in the general population, the main sources of exposure are food, cigarette smoking, inhalation of ambient air, drinking water, contaminated soil or dust. Furthermore, an occupational exposure usually involves human during mining, fume inhalation or manufacturing nickel-cadmium battery, electroplating and paint pigments that utilize Cd. METHODS: We undertook a structured search in literature about Cd. This metal is noxious on the cells of many organs, among which the kidney, the testis and the brain will be considered in this review. RESULTS: The toxic effects induced by Cd include many specific mechanisms, such as the oxidative stress, cellular death and inflammation. As no specific therapy for the prevention or treatment of the morbidity and mortality associated with Cd exposure is available, the state of the art of the therapeutic approaches is illustrated. CONCLUSION: Nowadays, a therapy able to counteract Cd toxicity is still lacking and the development of new therapeutic agents is requested.


Assuntos
Encéfalo/efeitos dos fármacos , Cádmio/toxicidade , Poluentes Ambientais/toxicidade , Rim/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Encéfalo/metabolismo , Poluentes Ambientais/química , Humanos , Rim/metabolismo , Masculino , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Testículo/metabolismo
4.
Nutrients ; 9(6)2017 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-28590452

RESUMO

Genistein, a soy-derived isoflavone, may improve cardiovascular risk profile in postmenopausal women with metabolic syndrome (MetS), but few literature data on its cardiac effects in humans are available. The aim of this sub-study of a randomized double-blind case-control study was to analyze the effect on cardiac function of one-year genistein dietary supplementation in 22 post-menopausal patients with MetS. Participants received 54 mg/day of genistein (n = 11) or placebo (n = 11) in combination with a Mediterranean-style diet and regular exercise. Left ventricular (LV) systolic function was assessed as the primary endpoint, according to conventional and strain-echocardiography measurements. Also, left atrial (LA) morphofunctional indices were investigated at baseline and at the final visit. Results were expressed as median with interquartile range (IQ). A significant improvement of LV ejection fraction (20.3 (IQ 12.5) vs. -1.67 (IQ 24.8); p = 0.040)), and LA area fractional change (11.1 (IQ 22.6) vs. 2.8 (9.5); p = 0.034)) were observed in genistein patients compared to the controls, following 12 months of treatment. In addition, body surface area indexed LA systolic volume and peak LA longitudinal strain significantly changed from basal to the end of the study in genistein-treated patients. One-year supplementation with 54 mg/day of pure genistein improved both LV ejection fraction and LA remodeling and function in postmenopausal women with MetS.


Assuntos
Suplementos Nutricionais , Genisteína/farmacologia , Coração/efeitos dos fármacos , Síndrome Metabólica , Pós-Menopausa , Método Duplo-Cego , Feminino , Coração/fisiologia , Humanos , Pessoa de Meia-Idade
5.
Neuromolecular Med ; 17(2): 192-201, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25893744

RESUMO

Systemic administration of kainic acid causes inflammation and apoptosis in the brain, resulting in neuronal loss. Dual cyclooxygenase/5-lipoxygenase (COX/5-LOX) inhibitors could represent a possible neuroprotective approach in preventing glutamate excitotoxicity. Consequently, we investigated the effects of a dual inhibitor of COX/5-LOX following intraperitoneal administration of kainic acid (KA, 10 mg/kg) in rats. Animals were randomized to receive either the dual inhibitor of COX/5-LOX (flavocoxid, 20 mg/kg i.p.) or its vehicle (1 ml/kg i.p.) 30 min after KA administration. Sham brain injury rats were used as controls. We evaluated protein expression of phosphorylated extracellular signal-regulated kinase (p-ERK1/2) and tumor necrosis factor alpha (TNF-α) as well as levels of malondialdehyde (MDA), prostaglandin E2 (PGE2) and leukotriene B4 (LTB4) in the hippocampus. Animals were also observed for monitoring behavioral changes according to Racine Scale. Finally, histological analysis and brain edema evaluation were carried out. Treatment with the dual inhibitor of COX/5-LOX decreased protein expression of p-ERK1/2 and TNF-α in hippocampus, markedly reduced MDA, LTB4 and PGE2 hippocampal levels, and also ameliorated brain edema. Histological analysis showed a reduction in cell damage in rats treated with the dual inhibitor of COX/5-LOX, particularly in hippocampal subregion CA3c. Moreover, flavocoxid significantly improved behavioral signs following kainic acid administration. Our results suggest that dual inhibition of COX/5-LOX by flavocoxid has neuroprotective effects during kainic acid-induced excitotoxicity.


Assuntos
Edema Encefálico/prevenção & controle , Catequina/uso terapêutico , Inibidores de Ciclo-Oxigenase/uso terapêutico , Inibidores de Lipoxigenase/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Convulsões/prevenção & controle , Animais , Comportamento Animal/efeitos dos fármacos , Edema Encefálico/induzido quimicamente , Edema Encefálico/enzimologia , Edema Encefálico/patologia , Catequina/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/análise , Combinação de Medicamentos , Hipocampo/química , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Ácido Caínico/toxicidade , Leucotrieno B4/análise , Peroxidação de Lipídeos/efeitos dos fármacos , Inibidores de Lipoxigenase/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Malondialdeído/análise , Proteínas do Tecido Nervoso/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/toxicidade , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/enzimologia , Convulsões/patologia , Fator de Necrose Tumoral alfa/metabolismo
6.
Eur J Clin Invest ; 43(10): 1025-31, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23899172

RESUMO

BACKGROUND: Previous data have suggested that genistein could exert beneficial effects on endothelial function and on predictors of cardiovascular risk in healthy postmenopausal women. In a randomized clinical trial, we studied the effects of genistein on endothelial function in postmenopausal women with metabolic syndrome (MS). METHODS: Twenty postmenopausal women with MS, according to modified NCEP-ATP III criteria were randomly assigned to receive placebo or genistein (54 mg/day) for 6 months, along with a Mediterranean-style diet. Postmenopausal women without MS (n = 15), served as controls. The primary goal was the assessment of endothelial function by flow-mediated vasodilation (FMD) of brachial artery; moreover, time-to-peak dilation in the FMD response has been evaluated. Secondary outcomes were fasting glucose, fasting insulin, total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglycerides, visfatin, adiponectin and homocysteine blood levels. Data on adverse events were also recorded. RESULTS: After 6 months of treatment, FMD at 50s and peak FMD significantly increased in genistein recipients compared with placebo. Moreover, genistein significantly decreased the blood levels of total cholesterol, triglycerides, homocysteine and visfatin compared with placebo, while blood adiponectin levels were increased. Genistein recipients neither experienced more side-adverse effects than placebo nor discontinued the study. CONCLUSIONS: Six months of treatment with genistein effectively improves brachial artery flow-mediated vasodilation in postmenopausal women with metabolic syndrome.


Assuntos
Endotélio Vascular/fisiologia , Genisteína/uso terapêutico , Síndrome Metabólica/tratamento farmacológico , Fitoestrógenos/uso terapêutico , Pós-Menopausa/efeitos dos fármacos , Índice Tornozelo-Braço , Endotélio Vascular/efeitos dos fármacos , Feminino , Humanos , Síndrome Metabólica/fisiopatologia , Pessoa de Meia-Idade , Projetos Piloto , Resultado do Tratamento , Vasodilatação/efeitos dos fármacos
7.
J Clin Endocrinol Metab ; 98(8): 3366-74, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23824420

RESUMO

CONTEXT: This study was performed to evaluate the effects of genistein on metabolic and cardiovascular risk factors in Caucasian postmenopausal subjects with metabolic syndrome (MetS). OBJECTIVE: Our objective was to assess the effects of genistein on surrogate endpoints associated with diabetes and cardiovascular disease. DESIGN AND SETTING: This was a randomized, double-blind, placebo-controlled trial at 3 university medical centers in Italy. PATIENTS: Patients included 120 postmenopausal women with MetS according to modified Third Report of the National Cholesterol Education Program (NCEP), Expert Panel on Detection, Evaluation and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) criteria. INTERVENTION: After a 4-week stabilization period, postmenopausal women with MetS were randomly assigned to receive placebo (n = 60) or 54 mg genistein daily (n = 60) for 1 year. MAIN OUTCOME MEASURES: The primary outcome was homeostasis model assessment for insulin resistance (HOMA-IR) at 1 year. Secondary outcomes were fasting glucose, fasting insulin, total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides, visfatin, adiponectin, and homocysteine levels. Data on adverse events were also recorded. RESULTS: At 1 year in genistein recipients, fasting glucose, fasting insulin, and HOMA-IR (mean from 4.5 to 2.7; P < .001) decreased and were unchanged in placebo recipients. Genistein statistically increased HDL-C (mean from 46.4 to 56.8 mg/dL) and adiponectin and decreased total cholesterol, LDL-C (mean from 108.8 to 78.7 mg/dL), triglycerides, visfatin, and homocysteine (mean from 14.3 to 11.7 µmol/L) blood levels. Systolic and diastolic blood pressure was also reduced in genistein recipients. Genistein recipients neither experienced more side adverse effects than placebo nor discontinued the study. CONCLUSION: One year of treatment with genistein improves surrogate endpoints associated with risk for diabetes and cardiovascular disease in postmenopausal women with MetS.


Assuntos
Genisteína/uso terapêutico , Síndrome Metabólica/tratamento farmacológico , Idoso , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Método Duplo-Cego , Feminino , Genisteína/efeitos adversos , Humanos , Resistência à Insulina , Síndrome Metabólica/sangue , Pessoa de Meia-Idade
8.
Br J Pharmacol ; 165(4): 994-1005, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21827449

RESUMO

BACKGROUND AND PURPOSE: Ovariectomy accelerates age-related skin changes as adequate oestrogen levels are required to control structural integrity and functional capacity of skin. Genistein, a soy-derived isoflavone, has been tested in anti-ageing cosmetic preparations with interesting results on skin elasticity, photoaging and skin cancer prevention. We investigated the effects of genistein aglycone and compared them with systemic raloxifene hydrochloride and 17-α-ethinyloestradiol on skin changes in aged, ovariectomized (OVX) rats. EXPERIMENTAL APPROACH: Six months after ovariectomy, rats were randomly allocated to different groups and treated, daily, with genistein aglycone (1 and 10mg·kg(-1) s.c.), raloxifene hydrochloride (0.05 and 0.5mg·kg(-1) s.c.) or 17-α-ethinyloestradiol (0.003 and 0.03mg·kg(-1) s.c.) for 12 weeks. Controls were untreated OVX and sham OVX rats. At the end of the treatment period, a skin biopsy was carried out and skin samples were assessed for molecular, histological and functional changes. KEY RESULTS: Skin samples of untreated OVX rats showed a decrease in TGF-ß1, VEGF, MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 compared with sham OVX rats. All the treatments significantly restored this depressed molecular profile revealed in OVX rats. Genistein aglycone, 1mg·kg(-1) , also significantly increased the thickness of collagen and breaking strength of skin in the OVX rats. CONCLUSIONS AND IMPLICATIONS: Relatively long-term, systemic treatment with genistein aglycone shows comparable efficacy to oestrogen in reversing some molecular, histological and functional changes of the skin associated with ovariectomy in aged rats. This suggests that genistein aglycone might be an effective alternative therapy for the management of age-related skin changes in postmenopausal women.


Assuntos
Genisteína/farmacologia , Fitoestrógenos/farmacologia , Pós-Menopausa/metabolismo , Envelhecimento da Pele/efeitos dos fármacos , Animais , Etinilestradiol/farmacologia , Feminino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Modelos Animais , Ovariectomia , Cloridrato de Raloxifeno/farmacologia , Ratos , Ratos Sprague-Dawley , Envelhecimento da Pele/patologia , Envelhecimento da Pele/fisiologia , Glycine max , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Phytomedicine ; 17(11): 844-50, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20570122

RESUMO

OBJECTIVE: Endometrial hyperplasia without cytological atypia is commonly treated with progestins, but other treatment regimes may be available with equivalent efficacy and low side effects. DESIGN: A randomized double-blind, placebo and progesterone-controlled clinical trial to evaluate the effects of genistein aglycone in reducing endometrial hyperplasia. PATIENTS: A group of 56 premenopausal women with non-atypical endometrial hyperplasia were enrolled and received: genistein aglycone (n=19; 54 mg/day); norethisterone acetate (n=19; 10 mg/day on days 16-25 of the menstrual cycle) or placebo (n=18) for 6 months. MEASUREMENTS: Hysteroscopy was performed with biopsies and symptomology assessed at baseline, 3 and 6 months of administration. The effect on estrogen (ER) and progesterone receptors (PR) expression in uterine biopsies were assessed after 3 and 6 months. For each treatment follicle stimulating hormone (FSH), luteinizing hormone (LH), estradiol (E2), sex hormone-binding globulin (SHBG) and progesterone (PG) levels were also evaluated. RESULTS: After 6 months, 42% of genistein aglycone-administered subjects had a significant improvement of symptoms (histologically confirmed in the 29%) compared to 47% of norethisterone acetate subjects (histologically confirmed in the 31%), but only 12% in the placebo group with 19% exhibiting worsening symptoms and increased endometrial thickness. No significant differences were noted for hormone levels for any treatment, but immunohistochemical analysis revealed significantly reduced staining for ER-alpha and PR and enhanced ER-beta1 staining in genistein-administered subjects associated with a complete regression of bleeding. CONCLUSIONS: These results suggest that genistein aglycone might be useful for the management of endometrial hyperplasia without atypia in women that cannot be treated with progestin.


Assuntos
Anticoncepcionais Orais Sintéticos/uso terapêutico , Hiperplasia Endometrial/tratamento farmacológico , Genisteína/uso terapêutico , Noretindrona/análogos & derivados , Fitoestrógenos/uso terapêutico , Fitoterapia , Adulto , Anticoncepcionais Orais Sintéticos/farmacologia , Método Duplo-Cego , Hiperplasia Endometrial/patologia , Endométrio/efeitos dos fármacos , Endométrio/patologia , Feminino , Genisteína/análogos & derivados , Genisteína/farmacologia , Humanos , Pessoa de Meia-Idade , Noretindrona/farmacologia , Noretindrona/uso terapêutico , Acetato de Noretindrona , Fitoestrógenos/farmacologia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Pré-Menopausa , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Hemorragia Uterina/tratamento farmacológico
10.
J Clin Endocrinol Metab ; 95(6): 3067-72, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20357174

RESUMO

CONTEXT AND OBJECTIVE: Genistein aglycone positively affects postmenopausal symptoms. However, questions about its long-term safety on the thyroid gland still remain. DESIGN: The parent study was a randomized, double-blind, placebo-controlled trial involving 389 osteopenic, postmenopausal women for 24 months. A subcohort (138 patients) continued therapy for an additional year. SETTING: Patients received ambulatory care. PATIENTS AND INTERVENTIONS: Participants received 54 mg of genistein aglycone daily (n = 71) or placebo (n = 67), plus calcium and vitamin D(3) at therapeutic doses. Circulating thyroid hormones (TSH, free T(3), free T(4)) and autoantibodies (thyroid peroxidase, thyroglobulin, and thyroid microsomal antigen) were assessed in 40 genistein and 37 placebo subjects who completed 3 yr. Thyroid hormone receptor (THRalpha and THRbeta) and retinoid receptor (RARalpha, RARgamma, and RXRalpha) expression from peripheral blood monocytes was also evaluated at baseline, 12, 24, and 36 months in all 3-yr completers. RESULTS: Genistein administration over 3 yr did not affect serum thyroid hormones or autoantibodies. In addition, there were no differences in THRalpha, THRbeta, RARalpha, RARgamma, or RXRalpha mRNA expression between groups. CONCLUSION: These data suggest that genistein aglycone intake does not significantly increase the risk of clinical or subclinical hypothyroidism at the dose of 54 mg/d.


Assuntos
Genisteína/efeitos adversos , Pós-Menopausa/fisiologia , Glândula Tireoide/efeitos dos fármacos , Idoso , Índice de Massa Corporal , Dieta , Método Duplo-Cego , Feminino , Genisteína/sangue , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/sangue , Lipídeos/sangue , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores dos Hormônios Tireóideos/biossíntese , Receptores dos Hormônios Tireóideos/metabolismo , Receptores X de Retinoides/biossíntese , Receptores X de Retinoides/metabolismo , Testes de Função Tireóidea , Tireotropina/sangue , Tiroxina/sangue , Tri-Iodotironina/sangue
11.
Menopause ; 16(2): 301-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19034051

RESUMO

OBJECTIVE: To evaluate in a 24-month, prospective, randomized, double-blind, placebo-controlled study whether pure administration of the phytoestrogen genistein (54 mg/d) might reduce the number and severity of hot flushes in postmenopausal women, with no adverse effect on the endometrium and vagina. METHODS: A total of 389 participants met the parent study criteria and were randomly assigned to receive the phytoestrogen genistein (n = 198) or placebo (n = 191). About 40% of participants in both groups did not experience hot flushes, and the evaluation was performed in a subgroup of 236 participants (genistein, n = 119; placebo, n = 117). Reductions from the baseline in the frequency and severity of hot flushes were the principal criteria of efficacy. Endometrial thickness was evaluated by ultrasonography. The maturation value was also used to determine hormonal action on the vaginal cells. RESULTS: There were no significant differences in vasomotor symptoms between groups at the baseline (4.4 +/- 0.33 hot flushes per day in the genistein group and 4.2 +/- 0.35 hot flushes per day in the control group). After 12 months of genistein therapy, there was a significant reduction (-56.4%) in the mean number of hot flushes, with a significant difference compared with the control group. After 24 months, there was no further decrease in the number of hot flushes in both groups. No significant difference was found in mean endometrial thickness and the maturation value score between the two groups, either at the baseline or after 24 months. CONCLUSIONS: The phytoestrogen genistein has been shown to be effective on vasomotor symptoms without an adverse effect on the endometrium and vagina, but after the first year, there was no further improvement in the decrease in hot flushes.


Assuntos
Endométrio/efeitos dos fármacos , Fogachos/tratamento farmacológico , Fitoestrógenos/administração & dosagem , Pós-Menopausa , Vagina/efeitos dos fármacos , Método Duplo-Cego , Epitélio/efeitos dos fármacos , Feminino , Genisteína/administração & dosagem , Humanos , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Resultado do Tratamento
12.
J Clin Endocrinol Metab ; 93(12): 4787-96, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18796517

RESUMO

CONTEXT: Genistein aglycone improves bone metabolism in women. However, questions about the long-term safety of genistein on breast as well as its continued efficacy still remain. OBJECTIVE: We assessed the continued safety profile of genistein aglycone on breast and endometrium and its effects on bone after 3 yr of therapy. DESIGN: The parent study was a randomized, double-blind, placebo-controlled trial involving 389 osteopenic, postmenopausal women for 24-months. Subsequently, a subcohort (138 patients) continued therapy for an additional year. PATIENTS AND INTERVENTIONS: Participants received 54 mg of genistein aglycone daily (n = 71) or placebo (n = 67). Both treatment arms received calcium and vitamin D(3) in therapeutic doses. MAIN OUTCOMES: Mammographic density was assessed at baseline, 24 and 36 months by visual classification scale and digitized quantification. BRCA1 and BRCA2, sister chromatid exchange, and endometrial thickness were also evaluated. Lumbar spine and femoral neck bone mineral density were also assessed. Secondary outcomes were biochemical levels of bone markers. RESULTS: After 36 months, genistein did not significantly change mammographic breast density or endometrial thickness, BRCA1 and BRCA2 expression was preserved, whereas sister chromatid exchange was reduced compared with placebo. Bone mineral density increases were greater with genistein for both femoral neck and lumbar spine compared to placebo. Genistein also significantly reduced pyridinoline, as well as serum carboxy-terminal cross-linking telopeptide and soluble receptor activator of NF-kappaB ligand while increasing bone-specific alkaline phosphatase, IGF-I, and osteoprotegerin levels. There were no differences in discomfort or adverse events between groups. CONCLUSIONS: After 3 yr of treatment, genistein exhibited a promising safety profile with positive effects on bone formation in a cohort of osteopenic, postmenopausal women.


Assuntos
Neoplasias da Mama/epidemiologia , Genisteína/efeitos adversos , Genisteína/uso terapêutico , Osteoporose Pós-Menopausa/prevenção & controle , Fitoestrógenos/efeitos adversos , Fitoestrógenos/uso terapêutico , Idoso , Proteína BRCA1/sangue , Proteína BRCA2/sangue , Biomarcadores , Densidade Óssea , Doenças Ósseas Metabólicas/prevenção & controle , Método Duplo-Cego , Endométrio/patologia , Feminino , Humanos , Mamografia , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Troca de Cromátide Irmã
13.
J Bone Miner Res ; 23(5): 715-20, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18433304

RESUMO

INTRODUCTION: RANKL and its decoy receptor osteoprotegerin (OPG) constitute a complex physiological mediator system involved in the regulation of bone resorption and may be responsible for the homeostatic mechanism of normal bone remodeling. Genistein, an isoflavone representing 1-5% of total phytoestrogen content in soybean products, may positively regulate cellular bone metabolism, but its mechanism of action on bone is not yet fully understood. MATERIALS AND METHODS: We studied the serum levels of both soluble RANKL (sRANKL) and OPG and the sRANKL/OPG ratio in 389 postmenopausal women (age, 49-67 yr) with a femoral neck BMD <0.795 g/cm(2) and no significant comorbid conditions after 24-mo therapy with genistein, (n = 198; 54 mg/d) or placebo (n = 191). Both intervention and placebo contained calcium and vitamin D(3). All patients received dietary instruction in an isocaloric fat-reduced diet. RESULTS: In comparison with placebo, sRANKL level was lower (p < 0.001 versus placebo) and OPG higher in genistein recipients (p < 0.001 versus placebo) at 1 and 2 yr, respectively. Moreover, at the end of 24 mo, genistein produced a significant reduction in the sRANKL/OPG ratio compared with placebo (genistein = -0.021, 95% CI, -0.020 to -0.022; placebo = +0.004, 95% CI, 0.003-0.005; difference = -0.020, 95% CI, -0.015 to -0.025, p < 0.001). CONCLUSIONS: Our findings suggest that genistein plus calcium and vitamin D(3) as part of a healthy diet is able to positively modulate bone turnover in a cohort of osteopenic, postmenopausal women and improve sRANKL-OPG balance after 24 mo of treatment.


Assuntos
Genisteína/administração & dosagem , Osteoprotegerina/sangue , Pós-Menopausa , Ligante RANK/sangue , Absorciometria de Fóton , Idoso , Densidade Óssea , Feminino , Humanos , Pessoa de Meia-Idade , Placebos
14.
J Clin Endocrinol Metab ; 92(8): 3068-75, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17682090

RESUMO

CONTEXT: Genistein, a soy isoflavone, has received wide attention over the last few years because of its potential preventive role for cardiovascular disease. OBJECTIVE: Our objective was to assess the effects of genistein administration (54 mg/d) on some predictors of cardiovascular risk in osteopenic, postmenopausal women. DESIGN AND SETTING: We conducted a randomized, double-blind, placebo-controlled trial at three Italian university medical centers. INTERVENTION: After a 4-wk stabilization on a standard isocaloric, fat-reduced diet, participants were randomly assigned to receive genistein (n = 198) or placebo (n = 191) daily for 24 months. Both intervention and placebo contained calcium and vitamin D(3). OUTCOME MEASURES: Blood lipid profiles, fasting glucose and insulin, homeostasis model assessment for insulin resistance, fibrinogen, soluble intercellular adhesion molecule-1, soluble vascular cellular adhesion molecule-1, F2-isoprostanes, and osteoprotegerin at baseline and after 12 and 24 months of treatment were measured. RESULTS: Compared with placebo, genistein significantly reduced fasting glucose and insulin as well as homeostasis model assessment for insulin resistance after both 12 and 24 months of treatment. By contrast, genistein administration did not affect blood lipid levels although fibrinogen, F2-isoprostanes, soluble intercellular adhesion molecule-1, and soluble vascular cellular adhesion molecule-1 decreased significantly compared with placebo after 24 months. Serum osteoprotegerin was higher in the genistein group compared with placebo. At 24 months, the genistein group showed no change in endometrial thickness compared with placebo. Most treatment-related adverse events were moderate and composed of gastrointestinal side effects [genistein, n = 37 (19%); placebo, n = 15 (8%)]. CONCLUSIONS: These results suggest that 54 mg genistein plus calcium, vitamin D(3), and a healthy diet was associated with favorable effects on both glycemic control and some cardiovascular risk markers in a cohort of osteopenic, postmenopausal women.


Assuntos
Doenças Ósseas Metabólicas/complicações , Doenças Cardiovasculares/prevenção & controle , Genisteína/uso terapêutico , Pós-Menopausa/fisiologia , Idoso , Biomarcadores , Glicemia/metabolismo , Densidade Óssea , Doenças Ósseas Metabólicas/sangue , Doenças Cardiovasculares/sangue , Dieta , Método Duplo-Cego , Feminino , Fibrinogênio/metabolismo , Genisteína/efeitos adversos , Humanos , Insulina/sangue , Molécula 1 de Adesão Intercelular/sangue , Isoprostanos/sangue , Lipídeos/sangue , Pessoa de Meia-Idade , Osteoprotegerina/sangue , Fatores de Risco , Resultado do Tratamento , Ultrassonografia , Útero/diagnóstico por imagem , Molécula 1 de Adesão de Célula Vascular/sangue
15.
Ann Intern Med ; 146(12): 839-47, 2007 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-17577003

RESUMO

BACKGROUND: Observational studies and small trials of short duration suggest that the isoflavone phytoestrogen genistein reduces bone loss, but the evidence is not definitive. OBJECTIVE: To assess the effects of genistein on bone metabolism in osteopenic postmenopausal women. DESIGN: Randomized, double-blind, placebo-controlled trial. SETTING: 3 university medical centers in Italy. PATIENTS: 389 postmenopausal women with a bone mineral density (BMD) less than 0.795 g/cm2 at the femoral neck and no significant comorbid conditions. INTERVENTION: After a 4-week stabilization period during which participants received a low-soy, reduced-fat diet, participants were randomly assigned to receive placebo (n = 191) or 54 mg of genistein (n = 198) daily for 24 months. Both the genistein and placebo tablets contained calcium and vitamin D. MEASUREMENTS: The primary outcome was BMD at the anteroposterior lumbar spine and femoral neck at 24 months. Secondary outcomes were serum levels of bone-specific alkaline phosphatase and insulin-like growth factor I, urinary excretion of pyridinoline and deoxypyridinoline, and endometrial thickness. Data on adverse events were also collected. RESULTS: At 24 months, BMD had increased in genistein recipients and decreased in placebo recipients at the anteroposterior lumbar spine (change, 0.049 g/cm2 [95% CI, 0.035 to 0.059] vs. -0.053 g/cm2 [CI, -0.058 to -0.035]; difference, 0.10 g/cm2 [CI, 0.08 to 0.12]; P < 0.001) and the femoral neck (change, 0.035 g/cm2 [CI, 0.025 to 0.042] vs. -0.037 g/cm2 [CI, -0.044 to -0.027]; difference, 0.062 g/cm2 [CI, 0.049 to 0.073]; P < 0.001). Genistein statistically significantly decreased urinary excretion of pyridinoline and deoxypyridinoline, increased levels of bone-specific alkaline phosphatase and insulin-like growth factor I, and did not change endometrial thickness compared with placebo. More genistein recipients than placebo recipients experienced gastrointestinal side effects (19% vs. 8%; P = 0.002) and discontinued the study. LIMITATIONS: The study did not measure fractures and had limited power to evaluate adverse effects. CONCLUSION: Twenty-four months of treatment with genistein has positive effects on BMD in osteopenic postmenopausal women. ClinicalTrials.gov registration number: NCT00355953.


Assuntos
Densidade Óssea/efeitos dos fármacos , Doenças Ósseas Metabólicas/tratamento farmacológico , Osso e Ossos/metabolismo , Genisteína/farmacologia , Pós-Menopausa/metabolismo , Idoso , Doenças Ósseas Metabólicas/metabolismo , Método Duplo-Cego , Endométrio/efeitos dos fármacos , Feminino , Genisteína/efeitos adversos , Humanos , Pessoa de Meia-Idade
16.
Atherosclerosis ; 178(2): 295-302, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15694937

RESUMO

We investigated whether lipid peroxidation might influence activation of the mitogen activated protein kinase (MAPK) extracellular regulated kinase (ERK1/2) and c-Jun N-terminal kinase (JNK) in neointimal hyperplasia induced by flow interruption of carotid artery in mice. C57/BL6 mice were subjected to a complete ligation of the left common carotid artery or to a sham ligation. Animals were randomized to receive either IRFI-042, a Vitamin E-like inhibitor of lipid peroxidation (20 mg/kg/i.p., immediately after artery occlusion) or its vehicle (1 ml/kg of a NaCl-DMSO solution). The extent of lipid peroxidation (investigated by the means of conjugated dienes levels) and JNK and ERK activation were evaluated by Western blot analysis after blood flow interruption. ICAM-1 expression in injured arteries was investigated 4 days after artery ligation by the means of reverse transcriptase polymerase chain reaction (RT-PCR) and quantification of the ICAM-1 protein levels. Morphometric analysis of the structural alteration caused by the disruption of the arterial blood flow was performed 4 weeks after surgery. Flow interruption in the carotid artery resulted at 10 min, following occlusion in a marked increase in conjugated dienes tissue levels (5.8+/-0.44 DeltaABS/mg protein), caused at 30 min after occlusion peak increase in both ERK1/2 (45+/-8 integrated intensity) and JNK (38+/-6 integrated intensity) activities, enhanced ICAM-1 expression (1.5+/-0.45 relative amount of ICAM-1 mRNA) and ICAM-1 protein levels (55+/-12 pg/mg protein) and produced a marked neointimal hyperplasia (mean intimal area=101+/-14 microm2). Injured arteries harvested from IRFI-042-treated mice had reduced conjugated dienes tissue levels (2.9+/-0.5 DeltaABS/mg protein), attenuated ERK1/2 (19+/-6 integrated intensity) and JNK (2.9+/-0.5 integrated intensity) activities, blunted ICAM-1 expression (0.38+/-0.1 relative amount of ICAM-1 mRNA) and protein levels (26+/-8 pg/mg protein) and decreased neointimal hyperplasia (mean intimal area=4.5+/-1.5 microm2). Our data indicate that ERK1/2 and JNK kinases play a crucial role in neointimal hyperplasia induced by flow cessation in the mouse carotid artery. Furthermore, the present data suggest that lipid peroxidation triggers ERK and JNK activation.


Assuntos
Artérias Carótidas/patologia , Artérias Carótidas/fisiologia , Estenose das Carótidas/fisiopatologia , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/farmacologia , Peroxidação de Lipídeos , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Proteína Quinase 3 Ativada por Mitógeno/farmacologia , Animais , Benzofuranos/administração & dosagem , Benzofuranos/farmacologia , Western Blotting , Estenose das Carótidas/veterinária , Modelos Animais de Doenças , Hiperplasia , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Fluxo Sanguíneo Regional , Túnica Íntima/citologia
17.
Exp Neurol ; 188(1): 178-86, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15191814

RESUMO

Brain injury was induced by intraperitoneal administration of kainic acid (KA, 10 mg/kg). Animals were randomized to receive either IRFI 042 (20 mg/kg i.p.), a lipid peroxidation inhibitor, or its vehicle (NaCl 0.9% DMSO 10% 1 ml/kg i.p.) 30 min before KA administration. A first set of animals was sacrificed 6 h after KA injection to measure malondialdehyde (MDA) content, glutathione-reduced (GSH) levels and the mRNA for interleukin-1beta (IL-1beta) in the cortex and in the hippocampus. A second set of animals was sacrificed 48 h after KA administration for histological analysis. All animals were observed for monitoring the behavioral sequelae and for evaluating latency of convulsions. Sham brain injury rats were used as controls. Intraperitoneal administration of IRFI 042 significantly decreased brain MDA (cortex: KA + vehicle = 0.285 +/- 0.04 nmol/mg protein; KA + IRFI 042 = 0.156 +/- 0.02 nmol/mg protein, P < 0.005; hippocampus: KA + vehicle = 0.350 +/- 0.03 nmol/mg protein; KA + IRFI 042 = 0.17 +/- 0.04 nmol/mg protein, P < 0.005), prevented the brain loss of GSH in both cortex (KA + vehicle = 7.81 +/- 1 micromol/g protein; KA + IRFI 042 = 12.1 +/- 1 micromol/g protein; P < 0.005) and hippocampus (KA + vehicle = 5 +/- 0.8 micromol/g protein; KA + IRFI 042 = 9.4 +/- 1.8 micromol/g protein; P < 0.005), reduced both brain IL-1beta mRNA expression and oedema, and increased latency of convulsions. Histological analysis showed a reduction of cell damage in IRFI 042-treated samples. The present data indicate that lipid peroxidation inhibition reduces IL-1beta gene expression and protects against kainic acid-induced brain damage.


Assuntos
Lesões Encefálicas/fisiopatologia , Interleucina-1/genética , Peroxidação de Lipídeos/genética , Degeneração Neural/metabolismo , Estresse Oxidativo/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Benzofuranos/farmacologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatologia , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/genética , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Glutationa/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Degeneração Neural/induzido quimicamente , Degeneração Neural/tratamento farmacológico , Degeneração Neural/genética , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/tratamento farmacológico , Convulsões/metabolismo , Convulsões/fisiopatologia
18.
Life Sci ; 74(10): 1253-64, 2004 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-14697408

RESUMO

We investigated the Levetiracetam (LVT) ability to protect the brain against kainic acid (KA) induced neurotoxicity. Brain injury was induced by intraperitoneal administration of KA (10 mg/kg). Sham brain injury rats were used as controls. Animals were randomized to receive either LVT (50 mg/kg) or its vehicle (1 ml/kg) 30 min. before KA administration. Animals were sacrificed 6 hours after KA injection to measure brain malonildialdehyde (MDA), glutathione levels (GSH) and the mRNA for interleukin-1beta (IL-1beta) in the cortex and in the diencephalon. Behavioral changes were also monitored. Intraperitoneal administration of LVT decreased significantly MDA in the cortex (KA + vehicle = 0.25 +/- 0.03 nmol/mg protein; KA + LVT = 0.13 +/- 0.01 nmol/mg protein; P < 0.005), and in the diencephalons (KA + vehicle = 1,01 +/- 0.2 nmol/mg protein; KA + LVT = 0,33 +/- 0,08 nmol/mg protein; P < 0.005), prevented the brain loss of GSH in both cortex (KA + vehicle = 5 +/- 1 micromol/g protein; KA + LVT = 15 +/- 2 micromol/g protein; P < 0.005) and diencephalons (KA + vehicle = 9 +/- 0.8 micromol/g protein; KA + LVT = 13 +/- 0.3 micromol/g protein; P < 0.05), reduced brain IL-1beta mRNA and markedly controlled seizures. Histological analysis showed a reduction of cell damage in LVT treated samples. The present data indicate that LVT displays neuro-protective effects against KA induced brain toxicity and suggest that these effects are mediated, at least in part, by inhibition of lipid peroxidation.


Assuntos
Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/toxicidade , Síndromes Neurotóxicas/prevenção & controle , Nootrópicos/farmacologia , Piracetam/análogos & derivados , Piracetam/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/patologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Primers do DNA , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Glutationa/metabolismo , Inflamação/patologia , Interleucina-1/biossíntese , Levetiracetam , Peroxidação de Lipídeos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Neurônios/patologia , Síndromes Neurotóxicas/patologia , RNA/biossíntese , RNA/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Pharmacol Res ; 48(3): 309-17, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12860452

RESUMO

Angiogenesis induced by growth factors may represent a rational therapy for patients with stroke. Vascular endothelial growth factor (VEGF) plays a pivotal role in angiogenesis and VEGF expression is enhanced in the post-ischemic brain. VEGF induced by brain hypoxia can lead to the growth of new vessels and may represent a natural protective mechanism improving survival after stroke. In the light of these findings we investigated changes of VEGF expression in different brain regions after intracerebroventricular injection of adeno-associated virus transferring gene for VEGF (rAAV-VEGF) in the gerbil, and after transient brain ischemic injury, we studied the effects of rAAV-VEGF injection on survival, brain edema, delayed neuronal death in the CA1 area and learning ability. Treatment with rAAV-VEGF 6 days or 12 days before ischemia significantly improves survival, brain edema and CA1 delayed neuronal death and post-ischemic learning evaluated by passive avoidance test. Animals treated with rAAV-VEGF showed in the thalamus and the cortex, a significant positive immunostaining for VEGF similar to those subjected to brain ischemia and not treated with rAAV-VEGF. These data represent a further contribution to a possible employment of gene therapy by using rAAV-VEGF in brain ischemia and indicate that thalamus and cortex may be targets for neuroprotective effects of VEGF.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Ataque Isquêmico Transitório/terapia , Fator A de Crescimento do Endotélio Vascular/genética , Análise de Variância , Animais , Aprendizagem da Esquiva , Encéfalo/metabolismo , Encéfalo/patologia , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Técnicas de Transferência de Genes , Gerbillinae , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Masculino , Tálamo/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...