Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Chinese Medical Journal ; (24): 671-679, 2019.
Article in English | WPRIM | ID: wpr-774771

ABSTRACT

BACKGROUND@#Fatty acid oxidation (FAO) disorder is involved in the pathogenesis of some cases of preeclampsia (PE). Several studies show that mammalian target of rapamycin (mTOR) signaling pathway is related to FAO. Pravastatin (Pra) can promote FAO in Nω-nitro-L-arginine methyl ester (L-NAME) PE-like mouse model in our previous study. This study aimed to investigate the effect of mTOR signaling pathway in PE-like model treated with Pra.@*METHODS@#Pregnant mice were randomly injected with L-NAME as PE-like model group or saline as control group respectively, from gestational 7th to 18th day. Giving Pra (L-NAME + Pra, Control + Pra, n = 8) or normal saline (NS; L-NAME + NS, Control + NS, n = 8) from gestational 8th to 18th day, the mice were sacrificed on day 18 and their liver and placental tissues were collected. Then the activation of mTOR and its substrates in the liver and placenta were detected. And the association between mTOR activation and serum free fatty acid (FFA) levels and the expression of long-chain 3-hydroxyacyl-coenzyme A dehydrogenase (LCHAD) were evaluated using Pearson correlation test. Differences between groups were analyzed using independent t-test or one-way analysis of variance (ANOVA).@*RESULTS@#Both in the maternal liver and placenta, the activation of mTOR protein and its effect on substrates increased significantly in the L-NAME + NS group and decreased significantly in the L-NAME + Pra group. The p-mTOR/mTOR protein ratio decreased in the L-NAME + Pra group significantly than that in the L-NAME + NS group both in liver and placenta (liver: 0.74 ± 0.08 vs. 0.85 ± 0.06, t = 2.95, P < 0.05; placenta: 0.63 ± 0.06 vs. 0.77 ± 0.06, t = 4.64, P < 0.05). The activation of mTOR protein in the liver and placenta negatively correlated with the expression of LCHAD in the L-NAME + NS group (liver: r = -0.745, P < 0.05; placenta: r = -0.833, P < 0.05) and that in the maternal liver negatively correlated with the expression of LCHAD (r = -0.733, P < 0.05) and positively with the serum FFA levels (r = 0.841, P < 0.05) in the L-NAME + Pra group.@*CONCLUSION@#The inhibition of mTOR signaling pathway might be involved in the regulation of FAO in mouse model treated with Pra.


Subject(s)
Animals , Female , Mice , Pregnancy , Blotting, Western , Fatty Acids , Metabolism , Immunohistochemistry , Liver , Metabolism , Mice, Inbred C57BL , Oxidation-Reduction , Placenta , Metabolism , Pravastatin , Therapeutic Uses , Pre-Eclampsia , Drug Therapy , Signal Transduction , TOR Serine-Threonine Kinases , Metabolism
2.
Rev. bras. neurol ; 54(3): 35-38, jul.-ago. 2018. graf
Article in English | LILACS | ID: biblio-948100

ABSTRACT

Statins are frequently prescribed in clinical practice for their proven efficacy in prevention of cardiovascular and cerebrovascular diseases. Despite the recognized beneficial effects of this class of drugs, in recent years, many studies published in medical literature have shown a wide range of adverse effects as a consequence of this therapy, including the risk of peripheral neuropathy. The purpose of this article is to report a case in which clinical features consistent with multiple mononeuropathy probably secondary to use of pravastatin were observed. The case report is followed by a review of the relevant literature.


As estatinas são frequentemente prescritas na prática clínica por sua comprovada eficácia na prevenção de doenças cardiovasculares e cérebrovasculares. Apesar dos reconhecidos efeitos benéficos dessa classe medicamentosa, nos últimos anos, diversos estudos publicados na literatura médica vem evidenciando uma ampla variedade de efeitos colaterais como consequência desta terapia, incluindo o risco de neuropatias periféricas. O objetivo deste artigo é relatar um caso no qual foram observadas manifestações clínicas compatíveis com o diagnóstico de mononeuropatia múltipla sensitiva, provavelmente secundária ao uso de pravastatina. O relato de caso é acompanhando de uma revisão de dados pertinentes da literatura.


Subject(s)
Humans , Male , Middle Aged , Pravastatin/adverse effects , Pravastatin/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Mononeuropathies/diagnosis , Mononeuropathies/chemically induced , Paresthesia/etiology , Review Literature as Topic , Hyperesthesia
3.
The Korean Journal of Internal Medicine ; : 1210-1223, 2018.
Article in English | WPRIM | ID: wpr-718010

ABSTRACT

BACKGROUND/AIMS: The co-occurrence of obesity aggravates asthma symptoms. Diet-induced obesity increases helper T cell (TH) 17 cell differentiation in adipose tissue and the spleen. The 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor pravastatin can potentially be used to treat asthma in obese patients by inhibiting interleukin 17 (IL-17) expression. This study investigated the combined effects of pravastatin and anti-IL-17 antibody treatment on allergic inflammation in a mouse model of obesity-related asthma. METHODS: High-fat diet (HFD)-induced obesity was induced in C57BL/6 mice with or without ovalbumin (OVA) sensitization and challenge. Mice were administered the anti-IL-17 antibody, pravastatin, or both, and pathophysiological and immunological responses were analyzed. RESULTS: HFD exacerbated allergic airway inflammation in the bronchoalveolar lavage fluid of HFD-OVA mice as compared to OVA mice. Blockading of the IL-17 in the HFD-OVA mice decreased airway hyper-responsiveness (AHR) and airway inflammation compared to the HFD-OVA mice. Moreover, the administration of the anti-IL-17 antibody decreased the leptin/adiponectin ratio in the HFD-OVA but not the OVA mice. Co-administration of pravastatin and anti-IL-17 inhibited airway inflammation and AHR, decreased goblet cell numbers, and increased adipokine levels in obese asthmatic mice. CONCLUSIONS: These results suggest that the IL-17–leptin/adiponectin axis plays a key role in airway inflammation in obesity-related asthma. Our findings suggest a potential new treatment for IL-17 as a target that may benefit obesity-related asthma patients who respond poorly to typical asthma medications.


Subject(s)
Animals , Humans , Mice , Adipokines , Adipose Tissue , Asthma , Bronchoalveolar Lavage Fluid , Cell Differentiation , Diet, High-Fat , Goblet Cells , Inflammation , Interleukin-17 , Obesity , Ovalbumin , Ovum , Oxidoreductases , Pravastatin , Respiratory Hypersensitivity , Spleen
4.
Journal of Korean Diabetes ; : 53-61, 2017.
Article in Korean | WPRIM | ID: wpr-726828

ABSTRACT

BACKGROUND: The effect of pravastatin on insulin resistance (IR) is controversial and poorly studied in prediabetes. METHODS: This study was performed in hyperglycemic patients at Saint Carollo Hospital from January 1, 2013 to December 31, 2015. Among them, we selected 40 patients (24 prediabetes and 16 new onset diabetes [NOD]) who had been treated with pravastatin 20 mg daily for 2 or 4 months and in whom fasting insulin and fasting glucose had been measured before and after administration of pravastatin. IR was defined as a fasting insulin level ≥ 12.94 µU/mL, homeostasis model for IR (HOMA-IR) ≥ 3.04 or quantitative insulin sensitivity check index (QUICKI) ≤ 0.32. RESULTS: Pravastatin treatment decreased total cholesterol and low-density lipoprotein cholesterol levels by 25.2% and 32.3% respectively (P = 0.000 for all), but did not affect fasting insulin level, HOMA-IR, or QUICKI in total, prediabetes, and NOD groups. Prevalence of IR was significantly different between prediabetes and NOD groups both before and after pravastatin treatment (0% versus 37.5%, P = 0.001), but pravastatin treatment did not affect the prevalence of IR in the prediabetes or NOD group. Fasting glucose level was not significantly different before and after pravastatin treatment in prediabetes (106.8 ± 6.4 mg/dL versus 103.8 ± 8.4 mg/dL, P = 0.223) but was significantly different in the NOD group (171.5 ± 70.1 mg/dL versus 124.4 ± 26.7 mg/dL, P = 0.017). CONCLUSION: Pravastatin treatment did not affect IR or fasting glucose level in hyperglycemic patients. Therefore, we suggest pravastatin can be prescribed to hypercholesterolemic patients with hyperglycemia.


Subject(s)
Humans , Cholesterol , Fasting , Glucose , Homeostasis , Hyperglycemia , Insulin Resistance , Insulin , Lipoproteins , Pravastatin , Prediabetic State , Prevalence , Saints
6.
Diabetes & Metabolism Journal ; : 23-30, 2017.
Article in English | WPRIM | ID: wpr-222885

ABSTRACT

BACKGROUND: There has been evidences of ethnic differences in the low density lipoprotein cholesterol (LDL-C) lowering effect of statin. We aimed to evaluate the efficacy of moderate-intensity statins in the treatment of dyslipidemia among Korean patients with type 2 diabetes mellitus (T2DM). METHODS: We analyzed a retrospective cohort that consisted of Korean patients with T2DM aged 40 to 75 years who had been prescribed any of the moderate-intensity statins (atorvastatin 10 or 20 mg, rosuvastatin 5 or 10 mg, pitavastatin 2 mg, or pravastatin 40 mg). Among them, only patients with baseline lipid profiles before starting statin treatment were selected, and changes in their lipid profiles before and 6 months after statin therapy were analyzed. RESULTS: Following the first 6 months of therapy, the overall LDL-C reduction was −47.4% (interquartile range, −56.6% to −34.1%). In total, 92.1% of the participants achieved an LDL-C level of <100 mg/dL, 38.3% had a 30% to 50% reduction in their LDL-C levels, and 42.3% had a reduction in their LDL-C levels greater than 50%. The response rates of each drug for achieving a LDL-C level <100 mg/dL were 81.7%, 93.1%, 95.0%, 95.0%, 96.5%, and 91.7% for treatment with atorvastatin doses of 10 or 20 mg, rosuvastatin 5 or 10 mg, pitavastatin 2 mg, and pravastatin 40 mg, respectively. CONCLUSION: In conclusion, the use of moderate-intensity statins reduced LDL-C levels less than 100 mg/dL in most of the Korean patients studied with T2DM. The efficacies of those statins were higher than expected in about 42% of Korean patients with T2DM.


Subject(s)
Humans , Atorvastatin , Cholesterol, LDL , Cohort Studies , Diabetes Mellitus, Type 2 , Dyslipidemias , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Pravastatin , Retrospective Studies , Rosuvastatin Calcium
7.
Korean Circulation Journal ; : 432-439, 2017.
Article in English | WPRIM | ID: wpr-195067

ABSTRACT

Hypercholesterolemia and hypertension are among the most important risk factors for cardiovascular (CV) disease. They are also important contributors to metabolic diseases including diabetes that further increase CV risk. Updated guidelines emphasize targeted reduction of overall CV risks but do not explicitly incorporate potential adverse metabolic outcomes that also influence CV health. Hypercholesterolemia and hypertension have synergistic deleterious effects on interrelated insulin resistance and endothelial dysfunction. Dysregulation of the renin-angiotensin system is an important pathophysiological mechanism linking insulin resistance and endothelial dysfunction to atherogenesis. Statins are the reference standard treatment to prevent CV disease in patients with hypercholesterolemia. Statins work best for secondary CV prevention. Unfortunately, most statin therapies dose-dependently cause insulin resistance, increase new onset diabetes risk and exacerbate existing type 2 diabetes mellitus. Pravastatin is often too weak to achieve target low-density lipoprotein cholesterol levels despite having beneficial metabolic actions. Renin-angiotensin system inhibitors improve both endothelial dysfunction and insulin resistance in addition to controlling blood pressure. In this regard, combined statin-based and renin-angiotensin system (RAS) inhibitor therapies demonstrate additive/synergistic beneficial effects on endothelial dysfunction, insulin resistance, and other metabolic parameters in addition to lowering both cholesterol levels and blood pressure. This combined therapy simultaneously reduces CV events when compared to either drug type used as monotherapy. This is mediated by both separate and interrelated mechanisms. Therefore, statin-based therapy combined with RAS inhibitors is important for developing optimal management strategies in patients with hypertension, hypercholesterolemia, diabetes, metabolic syndrome, or obesity. This combined therapy can help prevent or treat CV disease while minimizing adverse metabolic consequences.


Subject(s)
Humans , Atherosclerosis , Blood Pressure , Cardiovascular Diseases , Cholesterol , Diabetes Mellitus, Type 2 , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hypercholesterolemia , Hypertension , Insulin Resistance , Lipoproteins , Metabolic Diseases , Obesity , Pravastatin , Renin-Angiotensin System , Risk Factors
8.
Braz. j. biol ; 76(1): 59-65, Feb. 2016. graf
Article in English | LILACS | ID: lil-774508

ABSTRACT

Abstract Multiple myeloma (MM) is a B cell bone marrow neoplasia characterized by inflammation with an intense secretion of growth factors that promote tumor growth, cell survival, migration and invasion. The aim of this study was to evaluate the effects of pravastatin, a drug used to reduce cholesterol, in a MM cell line.Cell cycle and viability were determinate by Trypan Blue and Propidium Iodide. IL6, VEGF, bFGF and TGFβ were quantified by ELISA and qRT-PCR including here de HMG CoA reductase. It was observed reduction of cell viability, increase of cells in G0/G1 phase of the cell cycle and reducing the factors VEGF and bFGF without influence on 3-Methyl-Glutaryl Coenzyme A reductase expression.The results demonstrated that pravastatin induces cell cycle arrest in G0/G1 and decreased production of growth factors in Multiple Myeloma cell line.


Resumo O Mieloma Múltiplo é uma neoplasia de linfócitos B da medula óssea, caracterizada por inflamação com uma intensa secreção de fatores de crescimento que promovem o aumento do volume do tumor, sobrevivência celular, migração e invasão. O objetivo deste estudo foi avaliar os efeitos da pravastatina, uma droga usada para reduzir o colesterol, em um linhagem de MM. O ciclo celular e viabilidade foram determinadas por Trypan Blue e iodeto de propídio. IL6, VEGF, bFGF e TGF foram quantificadas por ELISA e qRT-PCR, incluindo aqui de HMG CoA redutase. Observou-se a redução da viabilidade das células, aumento de células na fase G0/G1 do ciclo celular e redução no VEGF e bFGF, sem influência na expressão da enzima 3-Metil-Glutaril Coenzima A redutase. Os resultados demonstraram que a pravastatina induz parada no ciclo celular em G0/G1 e diminuição da produção de fatores de crescimento em várias linhas de células de Mieloma.


Subject(s)
Humans , Fibroblast Growth Factors/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Multiple Myeloma/metabolism , Pravastatin/pharmacology , Vascular Endothelial Growth Factor A/genetics , Anticholesteremic Agents/pharmacology , Cell Line , Cell Cycle Checkpoints/drug effects , Cholesterol/metabolism , Fibroblast Growth Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Diabetes & Metabolism Journal ; : 376-385, 2016.
Article in English | WPRIM | ID: wpr-84891

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease is the most common form of chronic liver disease in industrialized countries. Recent studies have highlighted the association between peroxisomal dysfunction and hepatic steatosis. Peroxisomes are intracellular organelles that contribute to several crucial metabolic processes, such as facilitation of mitochondrial fatty acid oxidation (FAO) and removal of reactive oxygen species through catalase or plasmalogen synthesis. Statins are known to prevent hepatic steatosis and non-alcoholic steatohepatitis (NASH), but underlying mechanisms of this prevention are largely unknown. METHODS: Seven-week-old C57BL/6J mice were given normal chow or a methionine- and choline-deficient diet (MCDD) with or without various statins, fluvastatin, pravastatin, simvastatin, atorvastatin, and rosuvastatin (15 mg/kg/day), for 6 weeks. Histological lesions were analyzed by grading and staging systems of NASH. We also measured mitochondrial and peroxisomal FAO in the liver. RESULTS: Statin treatment prevented the development of MCDD-induced NASH. Both steatosis and inflammation or fibrosis grades were significantly improved by statins compared with MCDD-fed mice. Gene expression levels of peroxisomal proliferator-activated receptor α (PPARα) were decreased by MCDD and recovered by statin treatment. MCDD-induced suppression of mitochondrial and peroxisomal FAO was restored by statins. Each statin's effect on increasing FAO and improving NASH was independent on its effect of decreasing cholesterol levels. CONCLUSION: Statins prevented NASH and increased mitochondrial and peroxisomal FAO via induction of PPARα. The ability to increase hepatic FAO is likely the major determinant of NASH prevention by statins. Improvement of peroxisomal function by statins may contribute to the prevention of NASH.


Subject(s)
Animals , Mice , Atorvastatin , Catalase , Cholesterol , Developed Countries , Diet , Fatty Liver , Fibrosis , Gene Expression , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Inflammation , Liver Diseases , Liver , Metabolism , Non-alcoholic Fatty Liver Disease , Organelles , Peroxisomes , Pravastatin , Reactive Oxygen Species , Rosuvastatin Calcium , Simvastatin
11.
Yonsei Medical Journal ; : 1087-1094, 2016.
Article in English | WPRIM | ID: wpr-34057

ABSTRACT

PURPOSE: The effects of short-term intensive lipid-lowering treatment on coronary plaque composition have not yet been sufficiently evaluated. We investigated the influence of short-term intensive lipid-lowering treatment on quantitative and qualitative changes in plaque components of non-culprit lesions in patients with acute coronary syndrome. MATERIALS AND METHODS: This was a prospective, randomized, open-label, single-center trial. Seventy patients who underwent both baseline and three-month follow-up virtual histology intravascular ultrasound were randomly assigned to either an intensive lipid-lowering treatment group (ezetimibe/simvastatin 10/40 mg, n=34) or a control statin treatment group (pravastatin 20 mg, n=36). Using virtual histology intravascular ultrasound, plaque was characterized as fibrous, fibro-fatty, dense calcium, or necrotic core. Changes in plaque components during the three-month lipid-lowering treatment were compared between the two groups. RESULTS: Compared with the control statin treatment group, there was a significant reduction in low-density lipoprotein cholesterol in the intensive lipid-lowering treatment group (-20.4±17.1 mg/dL vs. -36.8±17.4 mg/dL, respectively; p<0.001). There were no statistically significant differences in baseline, three-month follow-up, or serial changes of gray-scale intravascular ultrasound parameters between the two groups. The absolute volume of fibro-fatty plaque was significantly reduced in the intensive lipid-lowering treatment group compared with the control group (-1.5±3.4 mm3 vs. 0.8±4.7 mm3, respectively; p=0.024). A linear correlation was found between changes in low-density lipoprotein cholesterol levels and changes in the absolute volumes of fibro-fatty plaque (p<0.001, R2=0.209). CONCLUSION: Modification of coronary plaque may be attainable after only three months of intensive lipid-lowering treatment.


Subject(s)
Aged , Female , Humans , Male , Middle Aged , Cholesterol, LDL/blood , Coronary Artery Disease/diagnostic imaging , Drug Administration Schedule , Ezetimibe, Simvastatin Drug Combination/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Plaque, Atherosclerotic/diagnostic imaging , Pravastatin/administration & dosage , Prospective Studies , Time Factors , Treatment Outcome , Ultrasonography, Interventional
12.
Rev. cuba. farm ; 49(1)ene.-mar. 2015. ilus, tab
Article in Spanish | LILACS, CUMED | ID: lil-770992

ABSTRACT

Introducción: el desarrollo de medicamentos transdérmicos manifiesta gran interés en los últimos años debido a las ventajas que ofrece; sin embargo, muchos de los sistemas desarrollados utilizan componentes solubles lo cual podría llevar a una ineficacia terapéutica si la matriz polimérica del sistema se solubiliza muy rápido, por ello se ensayan polímeros insolubles que permitan modular la liberación de un ingrediente farmacéuticamente activo. Objetivo: evaluar la liberación de pravastatina sódica en matrices poliméricas insolubles de quitosan/PF-127 con el método de paleta sobre disco para obtener su perfil cinético de liberación, con la finalidad de proponerse como matrices viables para la elaboración de parches transdérmicos. Métodos: se realizaron estudios de contenido químico, diámetro y espesor de las películas, calorimetrías de barrido diferencial y estudios de liberación. La cuantificación del principio activo se realizó mediante espectrofotometría UV-Vis a 238 nm. Resultados: se obtuvieron parches transdérmicos con buena uniformidad de contenido, espesor, diámetro, con una buena estabilidad en base a los estudios de calorimetría. El uso de PF-127 incrementó o retardó la liberación de pravastatina de la matriz polimérica dependiendo de su concentración y al realizarse los perfiles cinéticos de liberación las formulaciones se ajustaron a una cinética de orden 0 que describe el comportamiento de algunos sistemas transdérmicos. Conclusiones: los resultados manifiestan la posibilidad de usar esta matriz polimérica insoluble de quitosana con PF-127 para modular la liberación de pravastatina sódica y de fármacos con estructura similar a la misma por vía transdérmica, lo que generará de esta manera nuevas alternativas a las formas farmacéuticas orales para el tratamiento de padecimientos y enfermedades(AU)


Introduction: the development of transdermal drugs has aroused great interest in recent years due to their advantages, however many of the drug delivery systems use soluble matrix components which could trigger therapeutic problems due to a rapid release of the active ingredient. Therefore, insoluble polymers are being tested that can modulate the release of a pharmaceutically active ingredient. Objective: to evaluate the release of pravastatin sodium in insoluble polymer chitosan/PF-127 matrices by VER to obtain kinetic profile of release in order to submit them as viable systems for the manufacture of transdermal patches. Methods: studies on the chemical content, diameter and thickness of films, differential scanning calorimetry and release studies were performed. The UV-Vis spectrophotometry at 238 nm allowed quantitating the active principle. Results: transdermal patches with adequate uniform drug content, suitable thickness and diameter with good stability, based on calorimetric studies, were obtained. The use of PF-127 increased or delayed the release of pravastatin sodium from the polymeric matrix depending on concentration. When performing the kinetic profiles of release, the formulations were regulated to zero kinetic that describes the behavior of some transdernal systems. Conclusions: the results demonstrated the possibility of using these insoluble polymer chitosan/PF-127 matrices to modulate the release of pravastin sodium and of other structurally similar drugs, thus creating new alternatives to existing pharmaceutical oral forms for treatment of diseases(AU)


Subject(s)
Humans , Male , Female , Pravastatin/therapeutic use , Drug Delivery Systems/methods , Chitosan , Chitosan/therapeutic use , Transdermal Patch , Calorimetry, Differential Scanning/methods , Mexico
13.
Journal of Southern Medical University ; (12): 879-882, 2015.
Article in Chinese | WPRIM | ID: wpr-355265

ABSTRACT

<p><b>OBJECTIVE</b>To prepare pravastatin sodium-loaded chitosan microspheres to allow sustained drug release.</p><p><b>METHODS</b>The drug-loaded chitosan microspheres were prepared by using genipin as the cross-linker. The influences of molecular weight of chitosan, volume ratio of oil and water, reaction temperature, and stirring speed on the formation of chitosan microspheres were investigated. The morphology of the microspheres was observed using scanning electron microscopy. The encapsulation efficiency, swelling ratio under different pH conditions, and in vitro drug release were measured.</p><p><b>RESULTS</b>The in vitro release of pravastatin sodium could last for at least 31 days. The drug release rate varied with the reaction condition. The drug entrapment efficiency of the microsphere was 54.7%. The optimal processing conditions were as follows: chitosan viscosity of 200-400 mPa·s, oil-water proportion of 10:1, stirring speed of 850 r/min, and reaction temperature at 40 degrees celsius;.</p><p><b>CONCLUSION</b>The pravastatin sodium-loaded microspheres show good sustained drug release property, and the drug release rate can be modified by controlling the cross-linking time.</p>


Subject(s)
Chitosan , Cross-Linking Reagents , Delayed-Action Preparations , Iridoids , Microscopy, Electron, Scanning , Microspheres , Pravastatin , Chemistry
14.
Yonsei Medical Journal ; : 45-52, 2015.
Article in English | WPRIM | ID: wpr-201311

ABSTRACT

PURPOSE: To evaluate the effect of statin treatment on strut coverage after drug-eluting stent (DES) implantation. MATERIALS AND METHODS: In this study, 60 patients were randomly assigned to undergo sirolimus-eluting stent (SES) or biolimus-eluting stent (BES) implantation, after which patients were randomly treated with pitavastatin 2 mg or pravastatin 20 mg for 6 months. The degree of strut coverage was assessed by 6-month follow-up optical coherence tomography, which was performed in 52 DES-implanted patients. RESULTS: The percentages of uncovered struts were 19.4+/-14.7% in pitavastatin-treated patients (n=25) and 19.1+/-15.2% in pravastatin-treated patients (n=27; p=0.927). A lower percentage of uncovered struts was significantly correlated with a lower follow-up low-density lipoprotein (LDL) cholesterol level (r=0.486; p=0.009) and a greater decline of the LDL cholesterol level (r=-0.456; p=0.015) in SES-implanted patients, but not in BES-implanted patients. In SES-implanted patients, the percentage of uncovered struts was significantly lower among those with LDL cholesterol levels of less than 70 mg/dL after 6 months of follow-up (p=0.025), but no significant difference in this variable according to the follow-up LDL cholesterol level was noted among BES-implanted patients (p=0.971). CONCLUSION: Lower follow-up LDL cholesterol levels, especially those less than 70 mg/dL, might have a protective effect against delayed strut coverage after DES implantation. This vascular healing effect of lower LDL cholesterol levels could differ according to the DES type.


Subject(s)
Adult , Female , Humans , Male , Middle Aged , Coronary Angiography , Drug-Eluting Stents , Follow-Up Studies , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipoproteins, LDL/blood , Pravastatin/therapeutic use , Prosthesis Implantation , Quinolines/therapeutic use , Tomography, Optical Coherence , Treatment Outcome
15.
Allergy, Asthma & Respiratory Disease ; : 446-448, 2015.
Article in Korean | WPRIM | ID: wpr-56774

ABSTRACT

Stevens-Johnson syndrome (SJS) manifests with severe cutaneous reactions, most commonly triggered by medications, which are characterized by fever and mucocutaneous lesions leading to necrosis and sloughing of the epidermis. To our knowledge, pravastatin-induced SJS has not yet been reported. Here, we describe a case of SJS due to pravastatin, which was diagnosed by a patch test. A 70-year-old woman presented with maculopapular skin rashes, which developed 2 weeks after medication of bisoprolol, amlodipine, pravastatin, spironolactone, and indobufene for cardiac problems. Various bullous-erosive mucocutaneous lesions occupied less than 10% of the total body surface area. Painful oropharyngeal mucous membrane lesions were observed. The vermilion border of the lips became denuded and developed serosanguinous crusts. With the drug withdrawal and the use of systemic corticosteroids, her manifestations resolved. Drug patch tests with bisoprolol, amlodipine, pravastatin, spironolactone, and indobufene were performed, resulting in a positive reaction to pravastatin, but not to the other drugs. To the best of our knowledge, this is the first case of pravastatin-induced SJS.


Subject(s)
Aged , Female , Humans , Adrenal Cortex Hormones , Amlodipine , Bisoprolol , Body Surface Area , Epidermis , Exanthema , Fever , Lip , Mucous Membrane , Necrosis , Patch Tests , Pravastatin , Spironolactone , Stevens-Johnson Syndrome
16.
Saudi Medical Journal. 2014; 35 (11): 1339-1347
in English | IMEMR | ID: emr-153959

ABSTRACT

To investigate the combined effects of rosiglitazone and pravastatin on renal functions in early streptozotocin induced diabetic nephropathy [DN]. This study was carried out at King Khalid University Hospital Animal House, Riyadh, Saudi Arabia from August 2013 to February 2014. Fifty male Wistar rats were assigned to normal control rats and diabetic rats that received saline, rosiglitazone, pravastatin, or rosiglitazone+pravastatin for 2 months. Their weight range was 230-250 gm, and age range was from 18-20 weeks. At the end of experiment, creatinine clearance, and urinary albumin to creatinine ratio [ACR] were measured. Blood samples were analyzed for transferrin, glycosylated hemoglobin [HbA1c], lipid profile, tumor necrosis factor-alpha [TNF-alpha], intercellular adhesion molecule-1 [ICAM-1], and lipid peroxide. Rosiglitazone treatment increased creatinine clearance and plasma transferrin, and decreased urinary ACR, HbA1c, plasma TNF- alpha, ICAM-1, and serum lipid peroxide levels without affecting the altered lipid profile. Pravastatin treatment produced similar results and normalized the lipid alteration. The combination of rosiglitazone and pravastatin was more effective in attenuating the diabetes-induced nephropathy compared with treatment with either drug alone. The combination strategy of rosiglitazone and pravastatin may provide a potential synergistic renoprotective effect against DN by improving renal functions and reducing indices of DN


Subject(s)
Animals, Laboratory , Diabetic Nephropathies/veterinary , Pravastatin , Rats, Wistar , Diabetes Mellitus, Experimental/complications , Peroxisome Proliferators , Drug Therapy, Combination
17.
Acta Pharmaceutica Sinica ; (12): 72-77, 2014.
Article in Chinese | WPRIM | ID: wpr-297968

ABSTRACT

The study aims to establish a method for simultaneous determination of repaglinide and pravastatin sodium in rat plasma by LC-MS/MS and to study its pharmacokinetic interactions. Eighteen male SD rats were divided into repaglinide group, pravastatin sodium group and co-administration group. Blood samples were collected at different times after oral administration. Repaglinide and pravastatin sodium in rat plasma were separated by Agilent HC-C18 with the mobile phase consisting of methanol-0.1% formic acid (80 : 20). Detection and quantification were performed by using ESI-MS. The detector was operated in selected Reaction-monitoring mode at m/z 453.3-->230.1 for repaglinide, m/z 447.2-->327.4 for pravastatin sodium and m/z 285.1-->192.9 for diazepam as the internal standard. The calibration curve obtained was linear (R2>0.99) over the concentration range of 9.77-10,000 ng.mL-1 for repaglinide and 4.88-625 ng.mL-1 for pravastatin sodium. Compared with the single administration group, Cmax and AUC0-6h of repaglinide increased significantly (P<0.05) and tmax of pravastatin sodium prolonged (P<0.05) in co-administration group. The method is found to be simple, sensitive and accurate for determining the concentration of repaglinide and pravastatin sodium in rat plasma. There exists pharmacokinetic interactions in the co-administration of repaglinide and pravastatin sodium.


Subject(s)
Animals , Male , Rats , Administration, Oral , Carbamates , Blood , Pharmacokinetics , Chromatography, High Pressure Liquid , Drug Interactions , Piperidines , Blood , Pharmacokinetics , Pravastatin , Blood , Pharmacokinetics , Random Allocation , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
18.
Journal of Lipid and Atherosclerosis ; : 21-28, 2014.
Article in Korean | WPRIM | ID: wpr-65823

ABSTRACT

OBJECTIVE: The aim of this study is to compare cholesterol lowering effects of low dose 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) in Korean patients. METHODS: A total of 909 consecutive patients were enrolled prospectively according to the criteria of National Cholesterol Education Program guidelines. Lipid profiles were obtained before and 2 months after statin therapy. RESULTS: Atorvastatin 10 mg (n=260), lovastatin 20 mg (n=145), pitavastatin 2 mg (n=80), pravastatin 20 mg (n=28), rosuvastatin 5 mg (n=145), and simvastatin 20 mg (n=208) reduced low density lipoprotein (LDL) cholesterol by -41.8+/-11.0%, -33.8+/-12.8%, -39.3+/-10.8%, -31.5+/-8.9%, -48.8+/-12.3%, and -42.8+/-13.5%, respectively. LDL cholesterol less than 130 mg/dL was achieved in 90.3%, 76.9%, 88.5%, 85.2%, 97.2%, and 94.2%, respectively. The reduction of LDL cholesterol by 30% or more was obtained in 84.4%, 60.7%, 81.6%, 63.0%, 93.0%, and 83.5%, respectively. LDL cholesterol less than 70 mg/dL or the reduction by 50% or more was observed in a small portion of patients and was variable according to the different types of statins. CONCLUSION: A low dose statin was enough to manage dyslipidemia in most Korean patients with low to moderate risks and was even effective in a subpopulation of high risk patients.


Subject(s)
Humans , Cholesterol , Cholesterol, LDL , Coenzyme A , Dyslipidemias , Education , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hypercholesterolemia , Lipoproteins , Lovastatin , Oxidoreductases , Pravastatin , Prospective Studies , Simvastatin , Atorvastatin , Rosuvastatin Calcium
20.
The Korean Journal of Internal Medicine ; : 754-763, 2014.
Article in English | WPRIM | ID: wpr-126100

ABSTRACT

BACKGROUND/AIMS: This study was designed to evaluate the dose-effect relationship of statins in patients with ischemic congestive heart failure (CHF), since the role of statins in CHF remains unclear. METHODS: The South koreAn Pitavastatin Heart FaIluRE (SAPHIRE) study was designed to randomize patients with ischemic CHF into daily treatments of 10 mg pravastatin or 4 mg pitavastatin. RESULTS: The low density lipoprotein cholesterol level decreased by 30% in the pitavastatin group compared with 12% in the pravastatin (p < 0.05) group. Left ventricular systolic dimensions decreased significantly by 9% in the pitavastatin group and by 5% in the pravastatin group. Left ventricular ejection fraction (EF) improved significantly from 37% to 42% in the pitavastatin group and from 35% to 39% in the pravastatin group. Although the extent of the EF change was greater in the pitavastatin group (16% vs. 11%) than that in the pravastatin group, no significant difference was observed between the groups (p = 0.386). Exercise capacity, evaluated by the 6-min walking test, improved significantly in the pravastatin group (p < 0.001), but no change was observed in the pitavastatin group (p = 0.371). CONCLUSIONS: Very low dose/low potency pravastatin and high dose/high potency pitavastatin had a beneficial effect on cardiac reverse remodeling and improved systolic function in patients with ischemic CHF. However, only pravastatin significantly improved exercise capacity. These findings suggest that lowering cholesterol too much may not be beneficial for patients with CHF.


Subject(s)
Aged , Female , Humans , Male , Middle Aged , Biomarkers/blood , Cholesterol, LDL/blood , Down-Regulation , Dyslipidemias/blood , Exercise Tolerance/drug effects , Heart Failure/diagnosis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Myocardial Ischemia/diagnosis , Pravastatin/administration & dosage , Prospective Studies , Quinolines/administration & dosage , Recovery of Function , Republic of Korea , Stroke Volume/drug effects , Time Factors , Treatment Outcome , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL