Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 372
Filtrar
1.
JAMA ; 327(17): 1679-1687, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35368052

RESUMO

Importance: Lipoprotein(a) (Lp[a]) is an important risk factor for atherothrombotic cardiovascular disease and aortic stenosis, for which there are no treatments approved by regulatory authorities. Objectives: To assess adverse events and tolerability of a short interfering RNA (siRNA) designed to reduce hepatic production of apolipoprotein(a) and to assess associated changes in plasma concentrations of Lp(a) at different doses. Design, Setting, and Participants: A single ascending dose study of SLN360, an siRNA targeting apolipoprotein(a) synthesis conducted at 5 clinical research unit sites located in the US, United Kingdom, and Australia. The study enrolled adults with Lp(a) plasma concentrations of 150 nmol/L or greater at screening and no known clinically overt cardiovascular disease. Participants were enrolled between November 18, 2020, and July 21, 2021, with last follow-up on December 29, 2021. Interventions: Participants were randomized to receive placebo (n = 8) or single doses of SLN360 at 30 mg (n = 6), 100 mg (n = 6), 300 mg (n = 6), or 600 mg (n = 6), administered subcutaneously. Main Outcomes and Measures: The primary outcome was evaluation of safety and tolerability. Secondary outcomes included change in plasma concentrations of Lp(a) to a maximum follow-up of 150 days. Results: Among 32 participants who were randomized and received the study intervention (mean age, 50 [SD, 13.5] years; 17 women [53%]), 32 (100%) completed the trial. One participant experienced 2 serious adverse event episodes: admission to the hospital for headache following SARS-CoV-2 vaccination and later for complications of cholecystitis, both of which were judged to be unrelated to study drug. Median baseline Lp(a) concentrations were as follows: placebo, 238 (IQR, 203-308) nmol/L; 30-mg SLN360, 171 (IQR, 142-219) nmol/L; 100-mg SLN360, 217 (IQR, 202-274) nmol/L; 300-mg SLN360, 285 (IQR, 195-338) nmol/L; and 600-mg SLN360, 231 (IQR, 179-276) nmol/L. Maximal median changes in Lp(a) were -20 (IQR, -61 to 3) nmol/L, -89 (IQR, -119 to -61) nmol/L, -185 (IQR, -226 to -163) nmol/L, -268 (IQR, -292 to -189) nmol/L, and -227 (IQR, -270 to -174) nmol/L, with maximal median percentage changes of -10% (IQR, -16% to 1%), -46% (IQR, -64% to -40%), -86% (IQR, -92% to -82%), -96% (IQR, -98% to -89%), and -98% (IQR, -98% to -97%), for the placebo group and the 30-mg, 100-mg, 300-mg, and 600-mg SLN360 groups, respectively. The duration of Lp(a) lowering was dose dependent, persisting for at least 150 days after administration. Conclusions and Relevance: In this phase 1 study of 32 participants with elevated Lp(a) levels and no known cardiovascular disease, the siRNA SLN360 was well tolerated, and a dose-dependent lowering of plasma Lp(a) concentrations was observed. The findings support further study to determine the safety and efficacy of this siRNA. Trial Registration: ClinicalTrials.gov Identifier: NCT04606602; EudraCT Identifier: 2020-002471-35.


Assuntos
Apoproteína(a) , Hiperlipoproteinemias , RNA Interferente Pequeno , Adulto , Apoproteína(a)/efeitos adversos , Apoproteína(a)/biossíntese , Apoproteína(a)/sangue , Doenças Cardiovasculares/etiologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Hiperlipoproteinemias/sangue , Hiperlipoproteinemias/genética , Hiperlipoproteinemias/metabolismo , Hiperlipoproteinemias/terapia , Injeções Subcutâneas , Lipoproteína(a)/efeitos adversos , Lipoproteína(a)/biossíntese , Lipoproteína(a)/sangue , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/efeitos adversos , RNA Interferente Pequeno/uso terapêutico , Resultado do Tratamento
4.
Rev. Hosp. Clin. Univ. Chile ; 33(2): 97-107, 2022. tab, ilus
Artigo em Espanhol | LILACS | ID: biblio-1401167

RESUMO

Chylomicronemia syndrome is a metabolic condition characterized by severe hypertriglyceridemia and fasting chylomicronemia, secondary to an alteration in the ability to metabolize triglycerides. It can respond to different etiologies, the most frequent being multifactorial. Familial chylomicronemia syndrome, on the other hand, represents an infrequent cause of chylomicronemia syndrome, showing an autosomal recessive inheritance pattern. It's caused by pathogenic variants in genes related to chylomicron's metabolism, mainly LPL1 gene. One of the main associated risks is the occurrence of acute pancreatitis, which can also have a recurrent course. The primary therapy goal in patients with this condition is prevention of pancreatitis and related comorbidities. The treatment basis consists in reduce chylomicron formation by restriction of dietary fat, in association with physical activity and pharmacologic therapy. It is important to distinguish the etiology of chylomicronemia syndrome since it has repercussions in terms of response to treatment, complications, and recurrence risk. (AU)


Assuntos
Humanos , Animais , Masculino , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Hiperlipoproteinemias/genética , Hiperlipoproteinemias/diagnóstico , Hiperlipoproteinemias/tratamento farmacológico , Hiperlipoproteinemias/terapia , Hiperlipoproteinemia Tipo I/genética
5.
J Hum Genet ; 66(5): 465-473, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33100326

RESUMO

Observational studies have identified gout patients are often comorbid with dyslipidemia. However, the relationship between dyslipidemia and gout is still unclear. We first performed Mendelian randomization (MR) to evaluate the causal effect of four lipid traits on gout and serum urate based on publicly available GWAS summary statistics (n ~100,000 for lipid, 69,374 for gout and 110,347 for serum urate). MR showed each standard deviation (SD) (~12.26 mg/dL) increase in HDL resulted in about 25% (95% CI 9.0%-38%, p = 3.31E-3) reduction of gout risk, with 0.09 mg/dL (95% CI: -0.12 to -0.05, p = 7.00E-04) decrease in serum urate, and each SD (~112.33 mg/dL) increase of TG was associated with 0.10 mg/dL (95% CI: 0.06-0.14, p = 9.87E-05) increase in serum urate. Those results were robust against various sensitive analyses. Additionally, independent effects of HDL and TG on gout/serum urate were confirmed with multivariable MR. Finally, mediation analysis demonstrated HDL or TG could also indirectly affect gout via the pathway of serum urate. In conclusion, our study confirmed the causal associations between HDL (and TG) and gout, and further revealed the effect of HDL or TG on gout could also be mediated via serum urate.


Assuntos
Dislipidemias/complicações , Estudo de Associação Genômica Ampla , Gota/sangue , Lipídeos/sangue , Análise de Mediação , Análise da Randomização Mendeliana/métodos , Polimorfismo de Nucleotídeo Único , Fatores Etários , Causalidade , Colesterol/sangue , Dislipidemias/genética , Gota/etiologia , Gota/genética , Humanos , Hiperlipoproteinemias/complicações , Hiperlipoproteinemias/genética , Hipertrigliceridemia/complicações , Hipertrigliceridemia/genética , Funções Verossimilhança , Modelos Lineares , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Modelos Biológicos , Sensibilidade e Especificidade , Fatores Sexuais , Triglicerídeos/sangue , Ácido Úrico/sangue , População Branca
6.
J Tradit Chin Med ; 40(4): 640-645, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32744031

RESUMO

OBJECTIVE: To study the effects of rutin on serum glucose and lipid levels in hyperglycemic rats. METHODS: Male Wistar rats were subjected to intraperitoneal streptozotocin injections and a high-sugar, high-fat diet to establish a hyperglycemic and hyperlipidemic model. The model was considered to be successfully established in rats with fasting blood sugar (FBS) ≥ 11.1 mmol/L. The study included 6 groups with 10 rats each: a blank control group, a model group, a metformin group, and groups on large, medium and small doses of rutin. The groups received intraperitoneal streptozotocin or normal saline for 21 d. FBS, serum lipids, serum insulin, insulin sensitivity index (ISI), and levels of catalase (CAT), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and malondialdehyde (MDA) levels were evaluated in all rats. Pancreatic tissue samples were harvested to observe structural changes in islet cells. RESULTS: Large, medium, and small doses of rutin were associated with significantly reduced FBS (P < 0.05), and increased levels of ISI, CAT, GSH-Px and SOD, as well as decreased MDA (P < 0.05). Rutin administration was also related with reduced total cholesterol, triglycerides and low density lipoprotein chesterol, as well as increased high density lipoprotein chesterol (P < 0.05). Histologic evaluation revealed rutin induced repair of damaged islet cells. CONCLUSION: In diabetic rat models, rutin can significantly reduce FBS and blood lipids, improve anti-oxidant activity, increase insulin sensitivity, and induce repair of damaged islet cells.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hiperlipoproteinemias/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Hipolipemiantes/administração & dosagem , Rutina/administração & dosagem , Animais , Glicemia/metabolismo , LDL-Colesterol/sangue , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Glutationa Peroxidase/metabolismo , Humanos , Hiperlipoproteinemias/genética , Hiperlipoproteinemias/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Triglicerídeos/sangue
7.
Arterioscler Thromb Vasc Biol ; 39(12): 2468-2479, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597442

RESUMO

OBJECTIVE: A high level of LDL-C (low-density lipoprotein cholesterol) is a major risk factor for cardiovascular disease. The E3 ubiquitin ligase named IDOL (inducible degrader of the LDLR [LDL receptor]; also known as MYLIP [myosin regulatory light chain interacting protein]) mediates degradation of LDLR through ubiquitinating its C-terminal tail. But the expression profile of IDOL differs greatly in the livers of mice and humans. Whether IDOL is able to regulate LDL-C levels in humans remains to be determined. Approach and Results: By using whole-exome sequencing, we identified a nonsynonymous variant rs149696224 in the IDOL gene that causes a G51S (Gly-to-Ser substitution at the amino acid site 51) from a Chinese Uygur family. Large cohort analysis revealed IDOL G51S carriers (+/G51S) displayed significantly higher LDL-C levels. Mechanistically, the G51S mutation stabilized IDOL protein by inhibiting its dimerization and preventing self-ubiquitination and subsequent proteasomal degradation. IDOL(G51S) exhibited a stronger ability to promote ubiquitination and degradation of LDLR. Adeno-associated virus-mediated expression of IDOL(G51S) in mouse liver decreased hepatic LDLR and increased serum levels of LDL-C, total cholesterol, and triglyceride. CONCLUSIONS: Our study demonstrates that IDOL(G51S) is a gain-of-function variant responsible for high LDL-C in both humans and mice. These results suggest that IDOL is a key player regulating cholesterol level in humans.


Assuntos
LDL-Colesterol/sangue , Regulação da Expressão Gênica , Hiperlipoproteinemias/genética , RNA/genética , Ubiquitina-Proteína Ligases/genética , Adulto , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Hiperlipoproteinemias/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Receptores de LDL/sangue , Ubiquitina-Proteína Ligases/biossíntese , Sequenciamento Completo do Genoma/métodos
8.
PLoS One ; 13(12): e0209229, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30592719

RESUMO

The aim of this study was a comparison of aortic valve calcium score (AVCS) between patients with hypercholesterolemia and genetic diagnosis of familial hypercholesterolemia with low-density lipoprotein receptor gene mutation (LDLR-M group), versus patients with hypercholesterolemia without LDLR gene mutation (LDLR-WT group). A total of 72 LDLR-M patients and 50 LDLR-WT patients were enrolled in the study and underwent CT as a part of an assessment of coronary calcium scoring. AVCS was determined and compared between the two patient groups. AVCS was significantly higher in the LDLR-M group in comparison to the LDLR-WT group (13.8 ± 37.9 vs. 0.94 ± 3.1, p = 0.03). The Yates' chi-squared test for independence revealed that LDLR mutation and AVCS were significantly dependable (Chi^2 = 6.106, p = 0.013). The LDLR mutation was a strong predictor of a high AVCS (OR 7.83, 95% CI 2.08-29.50, p = 0.002) on multivariate regression analysis. Among the traditional risk factors, age (odds ratio 1.12, 95% CI 1.05-1.18, p<0.001) and SBP (OR 1.04, 95% CI 1.00-1.07, p = 0.045) were also significant for high result of AVCS. An assessment of computed tomography calcium scores showed that LDLR-M patients have increased AVCS in comparison to those with LDLR-WT. In addition, LDLR mutation can be considered as an independent risk factor of having high AVSC even after adjustment for risk factors including cholesterol levels. This may result from the associated process connected with the regulatory role of LDLR in evolution of aortic valve calcifications.


Assuntos
Estenose da Valva Aórtica/diagnóstico por imagem , Estenose da Valva Aórtica/genética , Valva Aórtica/patologia , Calcinose/diagnóstico por imagem , Calcinose/genética , Hiperlipoproteinemias/diagnóstico por imagem , Hiperlipoproteinemias/genética , Receptores de LDL/genética , Valva Aórtica/diagnóstico por imagem , Valva Aórtica/fisiopatologia , Estenose da Valva Aórtica/fisiopatologia , Calcinose/fisiopatologia , Feminino , Humanos , Hiperlipoproteinemias/fisiopatologia , Masculino , Pessoa de Meia-Idade , Mutação , Fatores de Risco , Tomografia Computadorizada por Raios X
9.
J Clin Lipidol ; 12(1): 110-115, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29198934

RESUMO

BACKGROUND: Hyperalphalipoproteinemia (HALP) is inversely correlated with coronary heart disease (CHD) although genetic variants associated with high serum levels of high-density lipoprotein cholesterol (HDL-C) have not been shown to be cardioprotective. OBJECTIVE: The objective of the study was to uncover novel genetic variants associated with HALP and possibly with reduced risk of CHD. METHODS: Exome sequencing data, HDL-C, and triglyceride levels were analyzed in 1645 subjects. They included the University of Maryland outpatients with high HDL-C (n = 12), Cardiovascular Health Study (n = 210), Jackson Heart Study (n = 402), Multi-Ethnic Study of Atherosclerosis (n = 404), Framingham Heart Study (n = 463), and Old Order Amish (n = 154). RESULTS: Novel nonsynonymous single-nucleotide polymorphisms (nsSNPs) were identified in men and women with primary HALP (mean HDL-C, 145 ± 30 mg/dL). Using PolyPhen-2 and Combined Annotation Dependent Depletion to estimate the predictive effect of each nsSNP on the gene product, rare, deleterious polymorphisms in UGT1A3, PLLP, PLEKHH1, ANK2, DIS3L, ACACB, and LRP4 were identified in 16 subjects with HALP but not in any tested subject with low HDL-C (<40 mg/dL). In addition, a single novel polymorphism, rs376849274, was found in OSBPL1A. The majority of these candidate genes have been implicated in fat and lipid metabolism, and none of these subjects has a history of CHD despite 75% of subjects having risk factors for CHD. Overall, the probability of finding these nsSNPs in a non-high HDL-C population ranges from 1 × 10-17 to 1 × 10-25. CONCLUSION: Novel functional polymorphisms in 8 candidate genes are associated with HALP in the absence of CHD. Future study is required to examine the extent to which these genes may affect HDL function and serve as potential therapeutic targets for CHD risk reduction.


Assuntos
Hiperlipoproteinemias/patologia , Polimorfismo de Nucleotídeo Único , Idoso , Idoso de 80 Anos ou mais , HDL-Colesterol/sangue , Feminino , Frequência do Gene , Glucuronosiltransferase/genética , Humanos , Hiperlipoproteinemias/epidemiologia , Hiperlipoproteinemias/genética , Masculino , Pessoa de Meia-Idade , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/genética , Linhagem , Fenótipo , Receptores de Esteroides/genética , Triglicerídeos/sangue , Sequenciamento do Exoma
10.
Atheroscler Suppl ; 30: 200-208, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29096839

RESUMO

BACKGROUND AND AIMS: Dyslipidaemias are associated with cardiovascular mortality and morbidity, driven by unstable atherosclerotic plaques with inflammatory infiltrates. Levels of messenger RNA (mRNA) for pro-inflammatory cytokines have been positively correlated with atherosclerotic disease progression. Therapeutic lipoprotein apheresis (LA) reduces plasma lipid levels and reduces inflammation. We evaluated the effects of LA on expression of mRNA coding for key pro-inflammatory cytokines in patients with dyslipidaemia, homo-/hetero-zygous familial hypercholesterolaemia (HoFH, HeFH) or hyperlipoprotein(a)aemia [hyperLp(a)] and associated coronary artery disease (CAD). APPROACH: Ten patients (five males and five females, mean age 47 ± 9.2 years) were enrolled, all with HyperLp(a) or confirmed genetic diagnoses of dyslipidaemia, HoFH, or HeFH; all had associated CAD. mRNA determinations were via reverse transcriptase polymer chain reaction (RT-qPCR). RESULTS: LA was associated with downregulation of mRNA expression for IL-1α, IL-6 and TNF-α, starting after the first LA session. The observed reduction was progressively enhanced during the interval between the first and second LA sessions to achieve a maximum decrease by the end of the second session (IL-1α: -49%, p < 0.001; IL-6: -35%, p < 0.001; TNF-α: -56%, p < 0.001). CONCLUSIONS: LA suppresses the expression of IL-1α, IL-6 and TNF-α mRNA in patients with dyslipidaemias. This may contribute to the arterial anti-inflammatory effect of LA.


Assuntos
Remoção de Componentes Sanguíneos/métodos , Hiperlipoproteinemias/terapia , Mediadores da Inflamação , Interleucina-1alfa/genética , Interleucina-6/genética , Lipoproteínas/sangue , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/genética , Adulto , Biomarcadores/sangue , Regulação para Baixo , Feminino , Heterozigoto , Homozigoto , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/terapia , Hiperlipoproteinemias/sangue , Hiperlipoproteinemias/diagnóstico , Hiperlipoproteinemias/genética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento
11.
J Clin Lipidol ; 11(2): 357-361, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28502491

RESUMO

BACKGROUND: Type I hyperlipoproteinemia, manifesting as chylomicronemia and severe hypertriglyceridemia, is a rare autosomal recessive disorder usually caused by mutations in the lipoprotein lipase gene (LPL). OBJECTIVE: We sought to determine whether mutations in LPL could explain the clinical indications of a patient presenting with pancreatitis and hypertriglyceridemia. METHODS: Coding regions of LPL were amplified by polymerase chain reaction and analyzed by nucleotide sequencing. The LPL messenger RNA transcript was also analyzed to investigate whether alternative splicing was occurring. RESULTS: The patient was homozygous for the mutation c.767_768insTAAATATT in exon 5 of the LPL gene. This mutation is predicted to result in either a truncated nonfunctional LPL, or alternatively a new 5' donor splice site may be used, resulting in a full-length LPL with an in-frame deletion of 3 amino acids. Analysis of messenger RNA from the patient showed that the new splice site is used in vivo. CONCLUSION: Homozygosity for a mutation in the LPL gene was consistent with the clinical findings. Use of the new splice site created by the insertion mutation rescues an otherwise damaging frameshift mutation, resulting in expression of an almost full-length LPL that is predicted to be partially functional. The patient therefore has a less severe form of type I hyperlipoproteinemia than would be expected if she lacked any functional LPL.


Assuntos
Mutação da Fase de Leitura , Lipase Lipoproteica/genética , Splicing de RNA , Adulto , Sequência de Bases , Éxons/genética , Feminino , Homozigoto , Humanos , Hiperlipoproteinemias/enzimologia , Hiperlipoproteinemias/genética , Mutagênese Insercional , RNA Mensageiro/genética
12.
Clin Res Cardiol Suppl ; 12(Suppl 1): 12-17, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28185213

RESUMO

A high level of lipoprotein(a) (Lp(a)) is recognized as an independent and additional cardiovascular risk factor contributing to the risk of early onset and progressive course of cardiovascular disease (CVD). All lipid lowering medications in use mainly lower low density lipoprotein-cholesterol (LDL-c) with no or limited effect on levels of Lp(a). Niacin, the only component lowering Lp(a), is firstly often poorly tolerated and secondly not available anymore in many countries. A level of <50 mg/dl was recommended recently as the cut off level for clinical use and decision making. Since lipoprotein apheresis (LA) lowers not only LDL-c but also Lp(a) significantly, its use is recommended in some countries in very high-risk patients with early or progressive CVD. Retrospective analyses show that regular LA improves the course of CVD. This is supported by a recent prospective observational trial and data of the German Lipoprotein Apheresis Registry. Despite many treatment options, all too often it is not possible to reduce LDL-c levels to target and to reduce Lp(a) levels sustainably at all. Therefore, new drug therapies are awaited. Some of the lipid modifying drugs in development lower Lp(a) to some extent in addition to LDL-c; the only specific approach is the apoprotein(a) antisense oligonucleotide. Currently LA is the standard of care as a last resort treatment in high-risk patients with elevated Lp(a) and severe CVD despite optimal control of all other cardiovascular risk factors.


Assuntos
Remoção de Componentes Sanguíneos , Doenças Cardiovasculares/prevenção & controle , Terapia Genética/métodos , Hiperlipoproteinemias/terapia , Hipolipemiantes/uso terapêutico , Lipoproteína(a)/sangue , Oligonucleotídeos Antissenso/uso terapêutico , Animais , Biomarcadores/sangue , LDL-Colesterol/sangue , Humanos , Hiperlipoproteinemias/sangue , Hiperlipoproteinemias/diagnóstico , Hiperlipoproteinemias/genética , Terapia de Alvo Molecular , Inibidores de PCSK9 , Pró-Proteína Convertase 9/metabolismo , Fatores de Risco , Inibidores de Serina Proteinase/uso terapêutico , Resultado do Tratamento
13.
Clin Res Cardiol Suppl ; 12(Suppl 1): 38-43, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28185214

RESUMO

Lipoprotein(a) (Lp(a)) is an independent cardiovascular risk factor playing a causal role for atherosclerotic cardiovascular disease (CVD). Lipoprotein apheresis (LA) is a safe well-tolerated outpatient treatment to lower LDL-C and Lp(a) by 60-70%, and is the ultimate escalating therapeutic option in patients with hyperlipoproteinemias (HLP) involving LDL particles. Major therapeutic effect of LA is preventing cardiovascular events. Lp(a)-HLP associated with progressive CVD has been approved as indication for regular LA in Germany since 2008. The Pro(a)LiFe-study investigated with a prospective multicenter design the long-term preventive effect of LA on incidence rates of cardiovascular events prospectively over a period of 5 years in 170 consecutive patients who commenced regular LA. During a median period of 4.7 years of the pre-LA period, Lp(a) associated progressive CVD became apparent. Apolipoprotein(a) (apo(a)) isoforms and polymorphisms at the apo(a) gene (LPA) were analyzed to assess hypothetical clinical correlations. 154 patients (90.6%) completed 5­years follow-up. Significant decline of the mean annual major adverse cardiac event (MACE) rate was observed from 0.41 ± 0.45 two years prior to regular LA to 0.06 ± 0.11 during 5 years with regular LA (p < 0.0001). 95.3% of patients expressed at least one small apo(a) isoform. Calculation of isoform specific concentrations allowed to confirm the equivalence of 60 mg/dl or 120 nmol/l as Lp(a) thresholds of the German LA guideline. Results of 5 years prospective follow-up confirmed that LA has a lasting effect on prevention of cardiovascular events in patients with Lp(a)-HLP and afore progressive CVD.


Assuntos
Remoção de Componentes Sanguíneos/normas , Doenças Cardiovasculares/prevenção & controle , Fidelidade a Diretrizes/normas , Hiperlipoproteinemias/terapia , Lipoproteína(a)/sangue , Guias de Prática Clínica como Assunto/normas , Biomarcadores/sangue , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/epidemiologia , Alemanha/epidemiologia , Humanos , Hiperlipoproteinemias/sangue , Hiperlipoproteinemias/epidemiologia , Hiperlipoproteinemias/genética , Incidência , Lipoproteína(a)/genética , Polimorfismo Genético , Estudos Prospectivos , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
14.
Arterioscler Thromb Vasc Biol ; 36(9): 2019-27, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27417585

RESUMO

OBJECTIVE: Lipoprotein(a)-hyperlipoproteinemia (Lp(a)-HLP) along with progressive cardiovascular disease has been approved as indication for regular lipoprotein apheresis (LA) in Germany since 2008. We aimed to study the long-term preventive effect of LA and to assess hypothetical clinical correlations of apolipoprotein(a) (apo(a)) by analyzing genotypes and phenotypes. APPROACH AND RESULTS: This prospective observational multicenter study included 170 patients with Lp(a)-HLP and progressive cardiovascular disease (48.9 years median age at diagnosis) despite other cardiovascular risk factors, including low-density lipoprotein cholesterol had maximally been treated (mean baseline low-density lipoprotein cholesterol: measured, 2.56 mmol/L [98.9 mg/dL] and corrected, 1.72 mmol/L [66.3 mg/dL]). Patients were prospectively investigated during a 5-year period about annual incidence rates of cardiovascular events. In addition, apo(a) isoforms and polymorphisms at the apo(a) gene (LPA) were characterized. One hundred fifty-four patients (90.6%) completed 5 years of follow-up. Mean Lp(a) concentration before commencing regular LA was 108.1 mg/dL. This was reduced by a single LA treatment by 68.1% on average. Significant decline of the mean annual cardiovascular event rate was observed from 0.58±0.53 2 years before regular LA to 0.11±0.15 thereafter (P<0.0001); 95.3% of patients expressed at least 1 small apo(a) isoform. Small apo(a) isoform (35.2%) carrying phenotypes were not tagged by single-nucleotide polymorphisms rs10455872 or rs3798220. CONCLUSIONS: Results of 5 years of prospective follow-up confirm that LA has a lasting effect on prevention of cardiovascular events in patients with Lp(a)-HLP. Patients clinically selected by progressive cardiovascular disease were characterized by a highly frequent expression of small apo(a) isoforms. Only Lp(a) concentration seemed to comprehensively reflect Lp(a)-associated cardiovascular risk, however.


Assuntos
Apoproteína(a)/sangue , Remoção de Componentes Sanguíneos/métodos , Doenças Cardiovasculares/prevenção & controle , Hiperlipoproteinemias/terapia , Lipoproteína(a)/sangue , Idoso , Biomarcadores/sangue , Remoção de Componentes Sanguíneos/efeitos adversos , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/epidemiologia , Feminino , Seguimentos , Predisposição Genética para Doença , Alemanha , Humanos , Hiperlipoproteinemias/sangue , Hiperlipoproteinemias/epidemiologia , Hiperlipoproteinemias/genética , Incidência , Lipoproteína(a)/genética , Masculino , Pessoa de Meia-Idade , Fenótipo , Polimorfismo de Nucleotídeo Único , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
15.
FEBS Lett ; 590(16): 2650-60, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27339627

RESUMO

We previously showed that feeding a Western-type diet (WTD) to Ldlr(-/-) mice lacking serum amyloid A (SAA) (Saa(-/-) Ldlr(-/-) mice), the level of total blood monocytes was higher than in Ldlr(-/-) mice. In this investigation we demonstrate that higher levels of bone marrow monocytes and macrophage-dendritic cell progenitor (MDP) cells were found in WTD-fed Saa(-/-) Ldlr(-/-) mice compared to Ldlr(-/-) mice and lower levels of GMP cells and CMP cells in Ldlr(-/-) mice. These data indicate that SAA regulates the level of bone marrow monocytes and their myeloid progenitors in hyperlipidemic Ldlr(-/-) mice.


Assuntos
Hiperlipoproteinemias/genética , Receptores de LDL/genética , Proteína Amiloide A Sérica/metabolismo , Animais , Aterosclerose/sangue , Aterosclerose/genética , Medula Óssea/metabolismo , Transplante de Medula Óssea , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Humanos , Hiperlipoproteinemias/sangue , Hiperlipoproteinemias/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Células Mieloides/metabolismo , Proteína Amiloide A Sérica/genética
17.
Nat Commun ; 7: 10353, 2016 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-26790392

RESUMO

Apolipoprotein C-III deficiency provides cardiovascular protection, but apolipoprotein C-III is not known to be associated with human amyloidosis. Here we report a form of amyloidosis characterized by renal insufficiency caused by a new apolipoprotein C-III variant, D25V. Despite their uremic state, the D25V-carriers exhibit low triglyceride (TG) and apolipoprotein C-III levels, and low very-low-density lipoprotein (VLDL)/high high-density lipoprotein (HDL) profile. Amyloid fibrils comprise the D25V-variant only, showing that wild-type apolipoprotein C-III does not contribute to amyloid deposition in vivo. The mutation profoundly impacts helical structure stability of D25V-variant, which is remarkably fibrillogenic under physiological conditions in vitro producing typical amyloid fibrils in its lipid-free form. D25V apolipoprotein C-III is a new human amyloidogenic protein and the first conferring cardioprotection even in the unfavourable context of renal failure, extending the evidence for an important cardiovascular protective role of apolipoprotein C-III deficiency. Thus, fibrate therapy, which reduces hepatic APOC3 transcription, may delay amyloid deposition in affected patients.


Assuntos
Amiloidose/genética , Amiloidose/metabolismo , Apolipoproteína C-III/metabolismo , Doenças Cardiovasculares/prevenção & controle , Lipoproteínas HDL/metabolismo , Lipoproteínas VLDL/metabolismo , Mutação de Sentido Incorreto , Adulto , Idoso , Idoso de 80 Anos ou mais , Apolipoproteína C-III/química , Apolipoproteína C-III/genética , Sequência de Bases , Feminino , França , Humanos , Hiperlipoproteinemias/genética , Hiperlipoproteinemias/metabolismo , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem
18.
Vnitr Lek ; 62(11): 887-894, 2016.
Artigo em Tcheco | MEDLINE | ID: mdl-28128575

RESUMO

Hyperlipoproteinemia (HLP) and dyslipidemia (DLP) are of course mainly perceived as diseases of common incidence and are typically seen as the greatest risk factors (RF) in the context of the pandemic of cardiovascular diseases. This is certainly true and HLP or DLP overall affect tens of percents of adults. However we cannot overlook the fact that disorders (mostly congenital) of lipid metabolism exist which, though not formally defined as such, amply satisfy the conditions for classification as rare diseases. Our account only includes a brief overview of the rare HLPs based on the dominant disorder of lipid metabolism, i.e. we shall mention the rare primary forms of hypercholesterolemia, primary forms of hypertriglyceridemia and the rare primary combined forms of HLP. In recent years an amazing progress has been reached relating to these diseases, in particular in the area of exact identification of the genetic defect and the mechanism of defect formation, however each of these diseases would require a separate article, though outside the field of clinical internal medicine. Therefore we shall discuss homozygous familial hypercholesterolemia (FH) in greater depth, partially also the "severe" form of heterozygous FH and in the following part the lipoprotein lipase deficiency; that means, diseases which present an extreme and even fatal risk for their carriers at a young age, but on the other hand, new therapeutic possibilities are offered within their treatment. An internist then should be alert to the suspicion that the described diseases may be involved, know about their main symptoms, where to refer the patient and how to treat them. Also dysbetalipoproteinemia (or type III HLP) will be briefly mentioned. Homozygous FH occurs with the frequency of 1 : 1 000 000 (maybe even more frequently, 1 : 160 000), it is characterized by severe isolated hypercholesterolemia (overall cholesterol typically equal to 15 mmol/l or more), xanthomatosis and first of all by a very early manifestation of a cardiovascular disease. Myocardial infarction is not an exception even in childhood. The therapy is based on high-dose statins, statins in combination with ezetimib and now also newly on PCSK9 inhibitors. Lomitapid and partly also mipomersen hold great promise for patients. LDL apheresis then represents an aggressive form of treatment. Lipoprotein lipase deficiency (type I HLP) is mainly characterized by severe hypertriglyceridemia, serum milky in colour, and xanthomatosis. A fatal complication is acute recurrent pancreatitis. A critical part of the treatment is diet, however it alone is not enough to control a genetic disorder. The only approved treatment is gene therapy. Experimentally, as an "off label" therapy, it is used in case studies with a lomitapid effect. We have our own experience with this experimental therapy. Dysbetalipoproteinemia is a congenital disorder of lipoprotein metabolism, characterized by high cholesterol (CH) and triglyceride (TG) levels. The underlying cause of this disease is the defect of the gene providing for apolipoprotein E. It is clinically manifested by xanthomatosis, however primarily by an early manifestation of atherosclerosis (rather peripheral than coronary).Key words: Lipoprotein lipase deficiency - dysbetalipoproteinemia - familial hypercholesterolemia - gene therapy - homozygous FH - LDL apheresis - lomitapid - mipomersen - PCSK9 inhibitors - rare diseases.


Assuntos
Anticolesterolemiantes/uso terapêutico , Remoção de Componentes Sanguíneos , Dislipidemias/terapia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hiperlipoproteinemias/terapia , Doenças Raras , Adulto , Aterosclerose , Benzimidazóis/uso terapêutico , Doenças Cardiovasculares , Colesterol , Dislipidemias/genética , Ezetimiba/uso terapêutico , Heterozigoto , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/terapia , Hiperlipoproteinemia Tipo I/complicações , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/terapia , Hiperlipoproteinemia Tipo II/complicações , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/terapia , Hiperlipoproteinemia Tipo III/genética , Hiperlipoproteinemia Tipo III/terapia , Hiperlipoproteinemias/genética , Masculino , Oligonucleotídeos/uso terapêutico , Inibidores de PCSK9 , Fatores de Risco , Xantomatose/etiologia
19.
Circulation ; 132(17): 1648-66, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26503748

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in the regulation of cholesterol homeostasis. By binding to hepatic low-density lipoprotein (LDL) receptors and promoting their lysosomal degradation, PCSK9 reduces LDL uptake, leading to an increase in LDL cholesterol concentrations. Gain-of-function mutations in PCSK9 associated with high LDL cholesterol and premature cardiovascular disease have been causally implicated in the pathophysiology of autosomal-dominant familial hypercholesterolemia. In contrast, the more commonly expressed loss-of-function mutations in PCSK9 are associated with reduced LDL cholesterol and cardiovascular disease risk. The development of therapeutic approaches that inhibit PCSK9 function has therefore attracted considerable attention from clinicians and the pharmaceutical industry for the management of hypercholesterolemia and its associated cardiovascular disease risk. This review summarizes the effects of PCSK9 on hepatic and intestinal lipid metabolism and the more recently explored functions of PCSK9 in extrahepatic tissues. Therapeutic approaches that prevent interaction of PCSK9 with hepatic LDL receptors (monoclonal antibodies, mimetic peptides), inhibit PCSK9 synthesis in the endoplasmic reticulum (antisense oligonucleotides, siRNAs), and interfere with PCSK9 function (small molecules) are also described. Finally, clinical trials testing the safety and efficacy of monoclonal antibodies to PCSK9 are reviewed. These have shown dose-dependent decreases in LDL cholesterol (44%-65%), apolipoprotein B (48%-59%), and lipoprotein(a) (27%-50%) without major adverse effects in various high-risk patient categories, including those with statin intolerance. Initial reports from 2 of these trials have indicated the expected reduction in cardiovascular events. Hence, inhibition of PCSK9 holds considerable promise as a therapeutic option for decreasing cardiovascular disease risk.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Terapia de Alvo Molecular , Pró-Proteína Convertases/antagonistas & inibidores , Adipócitos/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Encéfalo/metabolismo , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/genética , LDL-Colesterol/metabolismo , Ensaios Clínicos como Assunto , Predisposição Genética para Doença , Humanos , Hiperlipoproteinemias/genética , Incidência , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Camundongos , Mutação , Oligonucleotídeos Antissenso/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/química , Pró-Proteína Convertases/deficiência , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/fisiologia , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico , Receptores de LDL/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/deficiência , Serina Endopeptidases/genética , Serina Endopeptidases/fisiologia , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...