Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cancer Cell ; 38(4): 567-583.e11, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32976774

RESUMO

Oncogenic transformation alters lipid metabolism to sustain tumor growth. We define a mechanism by which cholesterol metabolism controls the development and differentiation of pancreatic ductal adenocarcinoma (PDAC). Disruption of distal cholesterol biosynthesis by conditional inactivation of the rate-limiting enzyme Nsdhl or treatment with cholesterol-lowering statins switches glandular pancreatic carcinomas to a basal (mesenchymal) phenotype in mouse models driven by KrasG12D expression and homozygous Trp53 loss. Consistently, PDACs in patients receiving statins show enhanced mesenchymal features. Mechanistically, statins and NSDHL loss induce SREBP1 activation, which promotes the expression of Tgfb1, enabling epithelial-mesenchymal transition. Evidence from patient samples in this study suggests that activation of transforming growth factor ß signaling and epithelial-mesenchymal transition by cholesterol-lowering statins may promote the basal type of PDAC, conferring poor outcomes in patients.


Assuntos
Vias Biossintéticas/genética , Carcinoma Ductal Pancreático/genética , LDL-Colesterol/biossíntese , Neoplasias Pancreáticas/genética , Fator de Crescimento Transformador beta/genética , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Atorvastatina/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Estimativa de Kaplan-Meier , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
N Engl J Med ; 356(2): 148-56, 2007 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-17215532

RESUMO

BACKGROUND: Patients with homozygous familial hypercholesterolemia have markedly elevated cholesterol levels, which respond poorly to drug therapy, and a very high risk of premature cardiovascular disease. Inhibition of the microsomal triglyceride transfer protein may be effective in reducing cholesterol levels in these patients. METHODS: We conducted a dose-escalation study to examine the safety, tolerability, and effects on lipid levels of BMS-201038, an inhibitor of the microsomal triglyceride transfer protein, in six patients with homozygous familial hypercholesterolemia. All lipid-lowering therapies were suspended 4 weeks before treatment. The patients received BMS-201038 at four different doses (0.03, 0.1, 0.3, and 1.0 mg per kilogram of body weight per day), each for 4 weeks, and returned for a final visit after a 4-week drug washout period. Analysis of lipid levels, safety laboratory analyses, and magnetic resonance imaging of the liver for fat content were performed throughout the study. RESULTS: All patients tolerated titration to the highest dose, 1.0 mg per kilogram per day. Treatment at this dose decreased low-density lipoprotein (LDL) cholesterol levels by 50.9% and apolipoprotein B levels by 55.6% from baseline (P<0.001 for both comparisons). Kinetic studies showed a marked reduction in the production of apolipoprotein B. The most serious adverse events were elevation of liver aminotransferase levels and accumulation of hepatic fat, which at the highest dose ranged from less than 10% to more than 40%. CONCLUSIONS: Inhibition of the microsomal triglyceride transfer protein by BMS-201038 resulted in the reduction of LDL cholesterol levels in patients with homozygous familial hypercholesterolemia, owing to reduced production of apolipoprotein B. However, the therapy was associated with elevated liver aminotransferase levels and hepatic fat accumulation.


Assuntos
Benzimidazóis/administração & dosagem , Proteínas de Transporte/antagonistas & inibidores , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Adolescente , Adulto , Alanina Transaminase/sangue , Apolipoproteínas B/sangue , Benzimidazóis/efeitos adversos , LDL-Colesterol/biossíntese , LDL-Colesterol/sangue , Terapia Combinada , Feminino , Homozigoto , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/dietoterapia , Fígado/efeitos dos fármacos , Masculino
4.
Blood ; 104(6): 1816-24, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15161671

RESUMO

Cholesterol levels are abnormally increased in many acute myeloid leukemia (AML) samples exposed in vitro to chemotherapy. Blocking these acute cholesterol responses selectively sensitizes AML cells to therapeutics. Thus, defining the molecular mechanisms by which AML cells accomplish these protective cholesterol increments might elucidate novel therapeutic targets. We now report that the levels of mRNAs encoding the cholesterol synthesis-regulating enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase, and the cholesterol-importing low-density lipoprotein (LDL) receptor were both increased by daunorubicin (DNR) or cytarabine (ARA-C) treatments in almost three fourths of cultured AML samples. However, less than one third of AML samples significantly increased LDL accumulation during drug treatments, suggesting that de novo synthesis is the primary mechanism by which most AML cells increase cholesterol levels during drug exposures. LDL increments were not correlated with cholesterol increments in ARA-C-treated AML samples. However, LDL and cholesterol increments did correlate in DNR-treated AML samples where they were measured, suggesting that a subset of AMLs may rely on increased LDL accumulation during treatment with particular drugs. Our data suggest that cholesterol synthesis inhibitors may improve the efficacy of standard antileukemia regimens, but that for maximum benefit, therapy may need to be tailored for individual patients with leukemia.


Assuntos
LDL-Colesterol/biossíntese , LDL-Colesterol/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Lovastatina/análogos & derivados , Transporte Biológico , Coenzima A Ligases/genética , Citarabina/farmacologia , Citometria de Fluxo , Humanos , Hidroximetilglutaril-CoA Sintase , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Lovastatina/farmacologia , Lovastatina/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de LDL/genética , Células Tumorais Cultivadas
5.
Int J Vitam Nutr Res ; 73(1): 39-47, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12690910

RESUMO

Amaranth was an important ancient grain and has current nutritional potential, being high in protein, fiber, lysine, magnesium, calcium, and squalene. Limited, inconsistent evidence demonstrates amaranth grain or oil can lower cholesterol in animal models. In the present study, hamsters received hypercholesterolemic diets consisting of a control, 10 or 20% Amaranthus cruentus grain, or 2.5 or 5% crude amaranth oil for four weeks. Amaranth oil (5%) decreased total and non-high-density lipoprotein (HDL) cholesterol by 15 and 22%, respectively, compared to control. Amaranth grain (20%; providing 1.4% amaranth oil) lowered non-HDL cholesterol and raised HDL cholesterol. Amaranth grain and oil decreased very low-density lipoprotein (VLDL) cholesterol by 21-50%; and increased fecal excretion of particular neutral sterols and the bile acid ursodeoxycholate. Amaranth oil (5%) additionally increased the cholesterol synthesis rate, possibly due to compensatory mechanisms; and decreased hepatic cholesterol ester, indicating reduced cholesterol ester availability for VLDL secretion and consistency with reduced VLDL cholesterol. Amaranth thus affected absorption of cholesterol and bile acids, cholesterol lipoprotein distribution, hepatic cholesterol content, and cholesterol biosynthesis. Amaranth grain and oil did not affect these pathways identically.


Assuntos
Amaranthus , Anticolesterolemiantes/farmacologia , Colesterol/metabolismo , Hipercolesterolemia/tratamento farmacológico , Fitoterapia , Óleos de Plantas/farmacologia , Animais , Colesterol/sangue , Colesterol/farmacocinética , HDL-Colesterol/biossíntese , HDL-Colesterol/sangue , LDL-Colesterol/biossíntese , LDL-Colesterol/sangue , Cricetinae , Modelos Animais de Doenças , Fezes/química , Hipercolesterolemia/metabolismo , Absorção Intestinal , Fígado/metabolismo , Masculino , Mesocricetus , Distribuição Aleatória
6.
Atherosclerosis ; 165(1): 137-44, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12208479

RESUMO

Animal and human studies have shown that the biosynthesis of cholesterol exhibits diurnal periodicity with nocturnal increases in the level of cholesterol precursors. Dietary cholesterol, which increases the intracellular pool of cholesterol and plasma cholesterol levels, has been shown to blunt the nocturnal increases in cholesterol biosynthesis. Patients with heterozygous familial hypercholesterolemia (FH) have very high levels of plasma low-density lipoprotein cholesterol (LDL) due to their reduced ability to metabolize LDL particles. The present studies were carried out to determine whether diurnal variations in cholesterol synthesis occur in FH patients and to test the effects of 3-hydroxy-3-methyl glutaryl CoA (HMG CoA) reductase inhibitors on the diurnal cycle of cholesterol biosynthesis in these patients. Diurnal rates of cholesterol synthesis were assessed by measuring the plasma concentrations of mevalonate, an intermediate in the pathway of cholesterol biosynthesis. Female FH patients exhibited a diurnal pattern in plasma mevalonate levels similar to that previously reported in controls with peak values occurring at night. Treatment with lovastatin and simvastatin (40 mg b.i.d.) significantly reduced 24-h mean plasma mevalonate levels from baseline values. Administration of lovastatin in the evening reduced the nocturnal increases in mevalonate levels, and the administration of simvastatin completely abolished the nighttime rise. These results demonstrate that inhibition of cholesterol biosynthesis by lovastatin and simvastatin modifies the normal diurnal rhythm of cholesterol biosynthesis in female FH patients.


Assuntos
HDL-Colesterol/biossíntese , LDL-Colesterol/biossíntese , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Lovastatina/administração & dosagem , Ácido Mevalônico/sangue , Sinvastatina/administração & dosagem , Adulto , Análise de Variância , Biomarcadores/sangue , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Ritmo Circadiano , Feminino , Heterozigoto , Humanos , Hiperlipoproteinemia Tipo II/genética , Pessoa de Meia-Idade , Periodicidade , Probabilidade , Estudos Prospectivos , Estudos de Amostragem , Urinálise
7.
Am J Clin Nutr ; 71(1 Suppl): 228S-31S, 2000 01.
Artigo em Inglês | MEDLINE | ID: mdl-10617976

RESUMO

n-3 Fatty acids have been shown to modify several key risk factors for cardiovascular disease. However, it is not clear whether the apparent protection against cardiovascular disease is directly related to antiatherogenic functions of these fatty acids or is mediated through their modification of the risk factors through mechanisms not directly related to lipids. A major question concerns the importance of lipid modification, which is a potent outcome of fish-oil supplementation. On balance, lipid modification is likely to represent a significant antiatherogenic factor. The benefits include increased HDL(2)-cholesterol concentrations, reduced triacylglycerol-rich lipoprotein concentrations, reduced postprandial lipemia, and reduced remnant concentrations. In contrast, LDL-cholesterol concentrations have often been noted to rise and the potential of increased oxidizability of LDLs is potentially adverse with lipid modification, but this potential can be overcome with vitamin E supplementation. The characteristic lipid changes and the underlying mechanisms are reviewed. Additional benefits of fish oils include improved endothelial function and better arterial compliance (elasticity). Future trials will be needed to determine minimum effective dosages of eicosapentaenoic and docosahexaenoic acids over lengthy periods and to show cardiovascular disease reduction through intervention.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Óleos de Peixe/metabolismo , Lipoproteínas/metabolismo , Animais , Artérias/fisiologia , HDL-Colesterol/biossíntese , HDL-Colesterol/metabolismo , LDL-Colesterol/biossíntese , LDL-Colesterol/metabolismo , Quilomícrons/metabolismo , Quilomícrons/farmacologia , Ácidos Graxos Ômega-3/metabolismo , Peixes , Humanos , Peróxidos Lipídicos/efeitos adversos , Lipoproteínas/biossíntese , Triglicerídeos/biossíntese , Triglicerídeos/metabolismo
8.
Scand J Clin Lab Invest ; 59(3): 159-66, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10400160

RESUMO

In order to test whether a 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor has an anti-atherogenic activity, the effects of carvastatin, a newly developed potent inhibitor, and pravastatin were examined on the intimal thickening of the artery after the endothelial denudation induced by balloon catheter injury. Rabbits were divided into four groups; control, pravastatin-treated (20 mg kg(-1) day(-1)) and two of carvastatin-treated groups (10 or 20 mg kg(-1) day(-1)). Two weeks after balloon catheter injury, the areas of intima and media of the injured carotid arteries were determined, and the ratios of intima to media (I/M) were calculated as an index of intimal thickening. Average I/M ratios of the injured artery were 0.42+/-0.05 for control, 0.49+/-0.07 for pravastatin, 0.19+/-0.03 (10 mg kg(-1) day(-1)) and 0.20+/-0.04 (20 mg kg(-1) day(-1)) for carvastatin-treated rabbits, respectively. Thus, carvastatin reduced I/M ratio of the injured artery to approximately half versus control, but pravastatin failed to suppress the intimal thickening. For in vitro study, vascular smooth muscle cells (SMC) from rabbit aorta were explanted, then cultured, and the effects of carvastatin on SMC migration and SMC proliferation were also examined. Carvastatin inhibited dose-dependently SMC migration and SMC proliferation with IC50 values of 0.5 microM and 1 microM, respectively. These inhibitory effects of carvastatin were cancelled by the coexistence of mevalonate, a metabolite of cholesterol synthesis. Our results suggest that carvastatin may be useful in rabbits as an anti-atherogenic drug by means of the inhibition of SMC migaration or SMC proliferation.


Assuntos
Artérias Carótidas/efeitos dos fármacos , Cateterismo/efeitos adversos , Movimento Celular/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Músculo Liso Vascular/citologia , Naftalenos/farmacologia , Piranos/farmacologia , Animais , Anticoagulantes/farmacologia , Arteriosclerose/tratamento farmacológico , Arteriosclerose/patologia , Becaplermina , Carcinoma Hepatocelular , Artérias Carótidas/patologia , Divisão Celular/efeitos dos fármacos , LDL-Colesterol/biossíntese , Humanos , Masculino , Ácido Mevalônico/farmacologia , Músculo Liso Vascular/química , Músculo Liso Vascular/metabolismo , Naftalenos/química , Fator de Crescimento Derivado de Plaquetas/farmacologia , Pravastatina/farmacologia , Proteínas Proto-Oncogênicas c-sis , Piranos/química , Coelhos , Receptores de LDL/biossíntese , Células Tumorais Cultivadas , Túnica Íntima/efeitos dos fármacos
9.
J Lipid Res ; 38(10): 2071-8, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9374129

RESUMO

Apheresis only partially controls raised low density lipoprotein cholesterol levels in patients with homozygous familial hypercholesterolemia, who usually respond poorly to lipid-lowering drugs. The efficacy and mechanism of action of a new 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, atorvastatin, was therefore investigated in seven homozygotes undergoing apheresis. One receptor-negative and six receptor-defective homozygotes undergoing plasma exchange or LDL apheresis every 2 weeks were studied during 2 months each on placebo and on atorvastatin 80 mg daily. Changes in plasma lipids and mevalonic acid, an index of cholesterol synthesis, were measured and the kinetics of the rebound of low density lipoprotein cholesterol and apolipoprotein B after apheresis were analyzed. All subjects had significant improvements on atorvastatin. Mean decreases in low density lipoprotein cholesterol were 31% greater both pre- and post-apheresis on atorvastatin compared with placebo, accompanied by a 63% decrease in mevalonic acid. Percentage changes in low density lipoprotein cholesterol and mevalonic acid were closely correlated (r = 0.89, P = 0.007). The mean production rates of low density lipoprotein cholesterol and apolipoprotein B were 21% and 25% lower, respectively, on atorvastatin than on placebo (P < 0.005 and <0.02) but changes in mean fractional clearance rates were not statistically significant. We conclude that atorvastatin enhances the efficacy of plasma exchange and low density lipoprotein apheresis in patients who lack low density lipoprotein receptors. This effect appears to be due to marked inhibition of cholesterol synthesis which results in a decreased rate of production of low density lipoprotein.


Assuntos
Anticolesterolemiantes/uso terapêutico , Remoção de Componentes Sanguíneos , LDL-Colesterol/sangue , Ácidos Heptanoicos/uso terapêutico , Hiperlipoproteinemia Tipo II/terapia , Lipoproteínas LDL/sangue , Pirróis/uso terapêutico , Adolescente , Adulto , Apolipoproteínas B/sangue , Atorvastatina , LDL-Colesterol/biossíntese , Terapia Combinada , Inibidores Enzimáticos/uso terapêutico , Feminino , Homozigoto , Humanos , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/metabolismo , Cinética , Lipídeos/sangue , Masculino , Ácido Mevalônico/sangue
10.
Atherosclerosis ; 135(2): 249-56, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9430375

RESUMO

Patients with homozygous familial hypercholesterolaemia (HFH) have abnormalities in both low-density lipoprotein (LDL) receptor alleles, resulting in severe hypercholesterolaemia and premature coronary heart disease. Limited treatment options are available and the response to drug therapy has been poor. In the present paper, we have evaluated the efficacy and safety of simvastatin at doses beyond the current maximal dose of 40 mg/day in patients with HFH. After a 4 week placebo diet run-in period, 12 patients with well-characterized HFH were randomized to simvastatin 80 mg/day administered in three divided doses (n = 8; group 1) or 40 mg once daily (n = 4; group 2). After 9 weeks, the dose in group 1 was increased to 160 mg/day while the dose in group 2 was kept at 40 mg/day, but with the drug given in three divided doses and treatment continued for an additional 9 weeks. All 12 patients completed the study and there were no serious or unexpected adverse effects. LDL-cholesterol concentrations fell by 14% at the 40 mg/day dose, but were reduced further at the higher doses (25% at the 80 mg/day and by 31% at the 160 mg/day dosage, P < 0.0001). Excretion of urinary mevalonic acid, as an index of in vivo cholesterol biosynthesis, was reduced but did not correlate with reduction in LDL-cholesterol in the individual patients. The magnitude of response to therapy was not predicted by the LDL-receptor gene defect as patients with the same LDL-receptor mutations responded differently to the same dose of simvastatin therapy. The ability of expanded doses of simvastatin (80 or 160 mg/day) to reduce LDL-cholesterol levels in patients with HFH, even if receptor negative, suggests that at these doses, the drug reduces LDL production. Simvastatin therapy, at doses of 80 or 160 mg/day, should therefore be considered in all patients with HFH, either as an adjunct to apheresis, or as monotherapy for those patients who do not have access to apheresis or other such treatment modalities.


Assuntos
Homozigoto , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/genética , Sinvastatina/administração & dosagem , Adolescente , Adulto , Anticolesterolemiantes/administração & dosagem , Apolipoproteínas B/biossíntese , LDL-Colesterol/antagonistas & inibidores , LDL-Colesterol/biossíntese , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Feminino , Humanos , Hiperlipoproteinemia Tipo II/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino
11.
J Nutr ; 126(8): 1972-8, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8759369

RESUMO

All amaranth varieties contain tocotrienols and squalene compounds which are known to affect cholesterol biosynthesis. Therefore, in the present study, the influence of dietary supplementation of whole seed, popped, and milled amaranth and amaranth oil on cholesterogenesis was studied in 6-wk-old female chickens. Serum total cholesterol and LDL-cholesterol were lowered 10-30% and 7-70% (P < 0.01), respectively, in birds fed amaranth-containing diets. HDL-cholesterol was not affected by amaranth supplementation. Activities of liver cholesterol 7alpha-hydroxylase (the enzyme responsible for cholesterol breakdown into bile acids) were 10-18% higher (P < 0.01) than those of controls for birds fed most forms of amaranth and its oil, whereas activities of liver 3-hydroxy-3-methylglutaryl coenzyme A reductase (the rate-limiting enzyme for cholesterol biosynthesis) were lowered by about only 9% (P < 0.01) by popped, milled amaranth and its oil. This lack of marked inhibition of this enzyme suggests the presence of some other potent cholesterol inhibitor(s) apart from tocotrienols and squalene in amaranth.


Assuntos
Colesterol/biossíntese , Magnoliopsida , Óleos de Plantas/farmacologia , Amaranthus , Animais , Apolipoproteína A-I/sangue , Apolipoproteínas B/sangue , Peso Corporal/fisiologia , Galinhas , Colesterol/sangue , HDL-Colesterol/biossíntese , HDL-Colesterol/sangue , LDL-Colesterol/biossíntese , LDL-Colesterol/sangue , Fibras na Dieta/farmacologia , Feminino , Manipulação de Alimentos , Hidroximetilglutaril-CoA Redutases/análise , Fígado/enzimologia , Magnoliopsida/química , Sementes/química , Glycine max/normas , Zea mays/normas
12.
Acta bioquím. clín. latinoam ; 24(2): 147-58, jun. 1990. ilus
Artigo em Espanhol | LILACS | ID: lil-95763

RESUMO

En este trabajo se determinaron los valores promedio, desviación estándar y percentilos para el cociente Apo B/C-HDL y, por otra parte, se analizó el efecto de las lipoproteínas ricas en triglicéridos sobre la relación entre C-LDL y Apo B, determinada por electroinmunodifusión en suero total. Se estudiaron 74 individuos de 20 y más años, aparentemente sanos. Para Apo B/C-HDL se obtuvo un valor medio de 2,16+0,78. El percentilo 50 fue 2,00, el percentilo 75 fue 2,60 y el percentilo 95 fue 3,60. Los individuos con Apo B/C-HDL>3,60 estarían en riesgo respecto de la aterosclerosis coronaria. La relación entre C-LDL y Apo B, es importante para la detección de sujetos con hiperapo B, la cual está fuertemente relacionada con la aterosclerosis coronaria. Utilizando los triglicéridos como estimadores de masa de las lipoproteínas ricas en triglicéridos, se halló que mientras TG<160 mg/dl la correlación entre Apo B y C-LDL fue r= +0,65,P <0,001. Un 86% de los pacientes con C-LDL entre 80 y 165 mg/dl tenían Apo B entre 55 y 120 mg/dl, mientras que un 12% tenían Apo B por encima de 120 mg/dl. En estos pacientes se puede suponer la presencia de hiperapo B en estas condiciones experimentales. Cuando TG>160 mg/dl, la correlación entre Apo B y C-LDL disminuye a r=+0,39,P<0,1. Un 21% de los pacientes con C-LDL entre 80 y 165 mg/dl tenían Apo B entre 55 y 120 mg/dl, pero un 79% tenían Apo B por encima de 120 mg/dl. En estas condiciones experimentales no se podrían discriminar los pacientes con hiperapo B.


Assuntos
Humanos , Masculino , Feminino , Apolipoproteínas B/sangue , Apolipoproteínas C/sangue , LDL-Colesterol , Doença das Coronárias/fisiopatologia , Imunodifusão , Lipoproteínas HDL/sangue , Fatores de Risco , Apolipoproteínas B/biossíntese , Apolipoproteínas B/metabolismo , Apolipoproteínas C/biossíntese , Apolipoproteínas C/metabolismo , LDL-Colesterol/biossíntese , LDL-Colesterol/metabolismo , Hipertrigliceridemia/diagnóstico , Hipertrigliceridemia/metabolismo , Lipoproteínas HDL/biossíntese , Lipoproteínas HDL/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA