Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 135(2): 335-349, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38828596

RESUMO

BACKGROUND: Individuals with type 1 diabetes (T1D) generally have normal or even higher HDL (high-density lipoprotein)-cholesterol levels than people without diabetes yet are at increased risk for atherosclerotic cardiovascular disease (CVD). Human HDL is a complex mixture of particles that can vary in cholesterol content by >2-fold. To investigate if specific HDL subspecies contribute to the increased atherosclerosis associated with T1D, we created mouse models of T1D that exhibit human-like HDL subspecies. We also measured HDL subspecies and their association with incident CVD in a cohort of people with T1D. METHODS: We generated LDL receptor-deficient (Ldlr-/-) mouse models of T1D expressing human APOA1 (apolipoprotein A1). Ldlr-/-APOA1Tg mice exhibited the main human HDL subspecies. We also generated Ldlr-/-APOA1Tg T1D mice expressing CETP (cholesteryl ester transfer protein), which had lower concentrations of large HDL subspecies versus mice not expressing CETP. HDL particle concentrations and sizes and proteins involved in lipoprotein metabolism were measured by calibrated differential ion mobility analysis and targeted mass spectrometry in the mouse models of T1D and in a cohort of individuals with T1D. Endothelial transcytosis was analyzed by total internal reflection fluorescence microscopy. RESULTS: Diabetic Ldlr-/-APOA1Tg mice were severely hyperglycemic and hyperlipidemic and had markedly elevated plasma APOB levels versus nondiabetic littermates but were protected from the proatherogenic effects of diabetes. Diabetic Ldlr-/-APOA1Tg mice expressing CETP lost the atheroprotective effect and had increased lesion necrotic core areas and APOB accumulation, despite having lower plasma APOB levels. The detrimental effects of low concentrations of larger HDL particles in diabetic mice expressing CETP were not explained by reduced cholesterol efflux. Instead, large HDL was more effective than small HDL in preventing endothelial transcytosis of LDL mediated by scavenger receptor class B type 1. Finally, in humans with T1D, increased concentrations of larger HDL particles relative to APOB100 negatively predicted incident CVD independently of HDL-cholesterol levels. CONCLUSIONS: Our results suggest that the balance between APOB lipoproteins and the larger HDL subspecies contributes to atherosclerosis progression and incident CVD in the setting of T1D and that larger HDLs exert atheroprotective effects on endothelial cells rather than by promoting macrophage cholesterol efflux.


Assuntos
Apolipoproteína A-I , Aterosclerose , Diabetes Mellitus Tipo 1 , Receptores de LDL , Animais , Aterosclerose/metabolismo , Aterosclerose/genética , Aterosclerose/sangue , Aterosclerose/patologia , Humanos , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/sangue , Camundongos , Receptores de LDL/genética , Receptores de LDL/deficiência , Receptores de LDL/metabolismo , Apolipoproteína A-I/sangue , Apolipoproteína A-I/metabolismo , Masculino , Proteínas de Transferência de Ésteres de Colesterol/genética , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/sangue , Camundongos Knockout , Feminino , Camundongos Endogâmicos C57BL , Lipoproteínas HDL/sangue , Lipoproteínas HDL/metabolismo , Camundongos Transgênicos , Apolipoproteína B-100/metabolismo , Apolipoproteína B-100/genética , Apolipoproteína B-100/sangue , Pessoa de Meia-Idade , Modelos Animais de Doenças , Adulto
2.
J Atheroscler Thromb ; 31(6): 854-860, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38616110

RESUMO

Atherosclerosis begins with the infiltration of cholesterol-containing lipoproteins into the arterial wall. White blood cell (WBC)-associated inflammation follows. Despite decades of research using genetic and pharmacologic methods to alter WBC function, in humans, the most effective method to prevent the initiation and progression of disease remains low-density lipoprotein (LDL) reduction. However, additional approaches to reducing cardiovascular disease would be useful as residual risk of events continues even with currently effective LDL-reducing treatments. Some of this residual risk may be due to vascular toxicity of triglyceride-rich lipoproteins (TRLs). Another option is that LDL transcytosis continues, albeit at reduced rates due to lower circulating levels of this lipoprotein. This review will address these two topics. The evidence that TRLs promote atherosclerosis and the processes that allow LDL and TRLs to be taken up by endothelial cells leading to their accumulation with the subendothelial space.


Assuntos
Aterosclerose , Lipoproteínas , Humanos , Lipoproteínas/metabolismo , Aterosclerose/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Animais , Células Endoteliais/metabolismo , Triglicerídeos/metabolismo , Lipoproteínas LDL/metabolismo
3.
Curr Opin Lipidol ; 35(2): 85-92, 2024 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-38447014

RESUMO

PURPOSE OF REVIEW: Lipid droplets are increasingly recognized as distinct intracellular organelles that have functions exclusive to the storage of energetic lipids. Lipid droplets modulate macrophage inflammatory phenotype, control the availability of energy for muscle function, store excess lipid, sequester toxic lipids, modulate mitochondrial activity, and allow transfer of fatty acids between tissues. RECENT FINDINGS: There have been several major advances in our understanding of the formation, dissolution, and function of this organelle during the past two years. These include new information on movement and partition of amphipathic proteins between the cytosol and lipid droplet surface, molecular determinants of lipid droplet formation, and pathways leading to lipid droplet hydrophobic lipid formation. Rapid advances in mitochondrial biology have also begun to define differences in their function and partnering with lipid droplets to modulate lipid storage versus oxidation. SUMMARY: This relationship of lipid droplets biology and cellular function provides new understanding of an important cellular organelle that influences muscle function, adipose lipid storage, and diseases of lipotoxicity.


Assuntos
Lipase , Gotículas Lipídicas , Humanos , Ácidos Graxos , Macrófagos , Mitocôndrias
4.
Cardiovasc Res ; 119(10): 1905-1914, 2023 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-37392421

RESUMO

A fine balance between uptake, storage, and the use of high energy fuels, like lipids, is crucial in the homeostasis of different metabolic tissues. Nowhere is this balance more important and more precarious than in the heart. This highly energy-demanding muscle normally oxidizes almost all the available substrates to generate energy, with fatty acids being the preferred source under physiological conditions. In patients with cardiomyopathies and heart failure, changes in the main energetic substrate are observed; these hearts often prefer to utilize glucose rather than oxidizing fatty acids. An imbalance between uptake and oxidation of fatty acid can result in cellular lipid accumulation and cytotoxicity. In this review, we will focus on the sources and uptake pathways used to direct fatty acids to cardiomyocytes. We will then discuss the intracellular machinery used to either store or oxidize these lipids and explain how disruptions in homeostasis can lead to mitochondrial dysfunction and heart failure. Moreover, we will also discuss the role of cholesterol accumulation in cardiomyocytes. Our discussion will attempt to weave in vitro experiments and in vivo data from mice and humans and use several human diseases to illustrate metabolism gone haywire as a cause of or accomplice to cardiac dysfunction.


Assuntos
Insuficiência Cardíaca , Miocárdio , Humanos , Camundongos , Animais , Miocárdio/metabolismo , Metabolismo dos Lipídeos , Coração , Miócitos Cardíacos/metabolismo , Ácidos Graxos/metabolismo , Oxirredução , Metabolismo Energético/fisiologia , Mitocôndrias/metabolismo
5.
J Lipid Res ; 63(11): 100274, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36115595

RESUMO

Lipid accumulation in nonadipose tissues can cause lipotoxicity, leading to cell death and severe organ dysfunction. Adipose triglyceride lipase (ATGL) deficiency causes human neutral lipid storage disease and leads to cardiomyopathy; ATGL deficiency has no current treatment. One possible approach to alleviate this disorder has been to alter the diet and reduce the supply of dietary lipids and, hence, myocardial lipid uptake. However, in this study, when we supplied cardiac Atgl KO mice a low- or high-fat diet, we found that heart lipid accumulation, heart dysfunction, and death were not altered. We next deleted lipid uptake pathways in the ATGL-deficient mice through the generation of double KO mice also deficient in either cardiac lipoprotein lipase or cluster of differentiation 36, which is involved in an lipoprotein lipase-independent pathway for FA uptake in the heart. We show that neither deletion ameliorated ATGL-deficient heart dysfunction. Similarly, we determined that non-lipid-containing media did not prevent lipid accumulation by cultured myocytes; rather, the cells switched to increased de novo FA synthesis. Thus, we conclude that pathological storage of lipids in ATGL deficiency cannot be corrected by reducing heart lipid uptake.


Assuntos
Aciltransferases , Cardiomiopatias , Lipase Lipoproteica , Animais , Humanos , Camundongos , Tecido Adiposo/metabolismo , Cardiomiopatias/metabolismo , Lipase/metabolismo , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Camundongos Knockout , Miocárdio/metabolismo , Triglicerídeos/metabolismo , Aciltransferases/deficiência , Aciltransferases/genética
6.
J Clin Invest ; 132(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34981790

RESUMO

Despite the success of LDL-lowering drugs in reducing cardiovascular disease (CVD), there remains a large burden of residual disease due in part to persistent dyslipidemia characterized by elevated levels of triglyceride-rich lipoproteins (TRLs) and reduced levels of HDL. This form of dyslipidemia is increasing globally as a result of the rising prevalence of obesity and metabolic syndrome. Accumulating evidence suggests that impaired hepatic clearance of cholesterol-rich TRL remnants leads to their accumulation in arteries, promoting foam cell formation and inflammation. Low levels of HDL may associate with reduced cholesterol efflux from foam cells, aggravating atherosclerosis. While fibrates and fish oils reduce TRL, they have not been uniformly successful in reducing CVD, and there is a large unmet need for new approaches to reduce remnants and CVD. Rare genetic variants that lower triglyceride levels via activation of lipolysis and associate with reduced CVD suggest new approaches to treating dyslipidemia. Apolipoprotein C3 (APOC3) and angiopoietin-like 3 (ANGPTL3) have emerged as targets for inhibition by antibody, antisense, or RNAi approaches. Inhibition of either molecule lowers TRL but respectively raises or lowers HDL levels. Large clinical trials of such agents in patients with high CVD risk and elevated levels of TRL will be required to demonstrate efficacy of these approaches.


Assuntos
Aterosclerose , LDL-Colesterol , Dislipidemias , Síndrome Metabólica , Obesidade , Proteína 3 Semelhante a Angiopoietina/antagonistas & inibidores , Proteína 3 Semelhante a Angiopoietina/genética , Proteína 3 Semelhante a Angiopoietina/metabolismo , Animais , Apolipoproteína C-III/antagonistas & inibidores , Apolipoproteína C-III/genética , Apolipoproteína C-III/metabolismo , Aterosclerose/sangue , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , LDL-Colesterol/sangue , LDL-Colesterol/genética , Dislipidemias/sangue , Dislipidemias/tratamento farmacológico , Dislipidemias/genética , Células Espumosas/metabolismo , Variação Genética , Humanos , Lipoproteínas HDL/sangue , Lipoproteínas HDL/genética , Síndrome Metabólica/sangue , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/genética , Obesidade/sangue , Obesidade/tratamento farmacológico , Obesidade/genética
7.
Atherosclerosis ; 329: 1-8, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34130222

RESUMO

Lipids released from circulating lipoproteins by intravascular action of lipoprotein lipase (LpL) reach parenchymal cells in tissues with a non-fenestrated endothelium by transfer through or around endothelial cells. The actions of LpL are controlled at multiple sites, its synthesis and release by myocytes and adipocytes, its transit and association with the endothelial cell luminal surface, and finally its activation and inhibition by a number of proteins and by its product non-esterified fatty acids. Multiple pathways mediate endothelial transit of lipids into muscle and adipose tissues. These include movement of fatty acids via the endothelial cell fatty acid transporter CD36 and movement of whole or partially LpL-hydrolyzed lipoproteins via other apical endothelial cell receptors such as SR-B1and Alk1. Lipids also likely change the barrier function of the endothelium and operation of the paracellular pathway around endothelial cells. This review summarizes in vitro and in vivo support for the key role of endothelial cells in delivery of lipids and highlights incompletely understood processes that are the focus of active investigation.


Assuntos
Células Endoteliais , Ácidos Graxos não Esterificados , Endotélio , Ácidos Graxos , Humanos , Lipase Lipoproteica , Lipoproteínas , Triglicerídeos
8.
J Clin Invest ; 131(12)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34128469

RESUMO

Although tissue uptake of fatty acids from chylomicrons is primarily via lipoprotein lipase (LpL) hydrolysis of triglycerides (TGs), studies of patients with genetic LpL deficiency suggest additional pathways deliver dietary lipids to tissues. Despite an intact endothelial cell (EC) barrier, hyperchylomicronemic patients accumulate chylomicron-derived lipids within skin macrophages, leading to the clinical finding eruptive xanthomas. We explored whether an LpL-independent pathway exists for transfer of circulating lipids across the EC barrier. We found that LpL-deficient mice had a marked increase in aortic EC lipid droplets before and after a fat gavage. Cultured ECs internalized chylomicrons, which were hydrolyzed within lysosomes. The products of this hydrolysis fueled lipid droplet biogenesis in ECs and triggered lipid accumulation in cocultured macrophages. EC chylomicron uptake was inhibited by competition with HDL and knockdown of the scavenger receptor-BI (SR-BI). In vivo, SR-BI knockdown reduced TG accumulation in aortic ECs and skin macrophages of LpL-deficient mice. Thus, ECs internalize chylomicrons, metabolize them in lysosomes, and either store or release their lipids. This latter process may allow accumulation of TGs within skin macrophages and illustrates a pathway that might be responsible for creation of eruptive xanthomas.


Assuntos
Aorta/metabolismo , Quilomícrons/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Gotículas Lipídicas/metabolismo , Triglicerídeos/metabolismo , Xantomatose/metabolismo , Animais , Aorta/patologia , Quilomícrons/genética , Técnicas de Cocultura , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Gotículas Lipídicas/patologia , Lipase Lipoproteica/deficiência , Lipase Lipoproteica/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Triglicerídeos/genética , Xantomatose/genética , Xantomatose/patologia
9.
Circ Res ; 128(3): 433-450, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33539224

RESUMO

Lipid uptake and metabolism are central to the function of organs such as heart, skeletal muscle, and adipose tissue. Although most heart energy derives from fatty acids (FAs), excess lipid accumulation can cause cardiomyopathy. Similarly, high delivery of cholesterol can initiate coronary artery atherosclerosis. Hearts and arteries-unlike liver and adrenals-have nonfenestrated capillaries and lipid accumulation in both health and disease requires lipid movement from the circulation across the endothelial barrier. This review summarizes recent in vitro and in vivo findings on the importance of endothelial cell receptors and uptake pathways in regulating FAs and cholesterol uptake in normal physiology and cardiovascular disease. We highlight clinical and experimental data on the roles of ECs in lipid supply to tissues, heart, and arterial wall in particular, and how this affects organ metabolism and function. Models of FA uptake into ECs suggest that receptor-mediated uptake predominates at low FA concentrations, such as during fasting, whereas FA uptake during lipolysis of chylomicrons may involve paracellular movement. Similarly, in the setting of an intact arterial endothelial layer, recent and historic data support a role for receptor-mediated processes in the movement of lipoproteins into the subarterial space. We conclude with thoughts on the need to better understand endothelial lipid transfer for fuller comprehension of the pathophysiology of hyperlipidemia, and lipotoxic diseases such as some forms of cardiomyopathy and atherosclerosis.


Assuntos
Colesterol/metabolismo , Células Endoteliais/metabolismo , Proteínas de Transporte de Ácido Graxo/metabolismo , Ácidos Graxos/metabolismo , Transtornos do Metabolismo dos Lipídeos/metabolismo , Transcitose , Animais , Antígenos CD36/metabolismo , Quilomícrons/metabolismo , Humanos , Transtornos do Metabolismo dos Lipídeos/patologia , Lipólise , Tamanho da Partícula
10.
EMBO J ; 38(9)2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-30936093

RESUMO

Membrane targeting of autophagy-related complexes is an important step that regulates their activities and prevents their aberrant engagement on non-autophagic membranes. ATG16L1 is a core autophagy protein implicated at distinct phases of autophagosome biogenesis. In this study, we dissected the recruitment of ATG16L1 to the pre-autophagosomal structure (PAS) and showed that it requires sequences within its coiled-coil domain (CCD) dispensable for homodimerisation. Structural and mutational analyses identified conserved residues within the CCD of ATG16L1 that mediate direct binding to phosphoinositides, including phosphatidylinositol 3-phosphate (PI3P). Mutating putative lipid binding residues abrogated the localisation of ATG16L1 to the PAS and inhibited LC3 lipidation. On the other hand, enhancing lipid binding of ATG16L1 by mutating negatively charged residues adjacent to the lipid binding motif also resulted in autophagy inhibition, suggesting that regulated recruitment of ATG16L1 to the PAS is required for its autophagic activity. Overall, our findings indicate that ATG16L1 harbours an intrinsic ability to bind lipids that plays an essential role during LC3 lipidation and autophagosome maturation.


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Autofagia , Membrana Celular/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animais , Proteínas Relacionadas à Autofagia/fisiologia , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Endossomos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Proteínas de Ligação a Fosfato/fisiologia , Enzimas de Conjugação de Ubiquitina/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia
11.
Essays Biochem ; 61(6): 597-607, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29233871

RESUMO

Vesicular trafficking events play key roles in the compartmentalization and proper sorting of cellular components. These events have crucial roles in sensing external signals, regulating protein activities and stimulating cell growth or death decisions. Although mutations in vesicle trafficking players are not direct drivers of cellular transformation, their activities are important in facilitating oncogenic pathways. One such pathway is the sensing of external stimuli and signalling through receptor tyrosine kinases (RTKs). The regulation of RTK activity by the endocytic pathway has been extensively studied. Compelling recent studies have begun to highlight the association between autophagy and RTK signalling. The influence of this interplay on cellular status and its relevance in disease settings will be discussed here.


Assuntos
Autofagia/fisiologia , Endocitose/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Autofagia/genética , Endocitose/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Receptores Proteína Tirosina Quinases/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
12.
J Biol Chem ; 288(39): 27777-88, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-23940052

RESUMO

Cells exposed to stress of different origins synthesize triacylglycerols and generate lipid droplets (LD), but the physiological relevance of this response is uncertain. Using complete nutrient deprivation of cells in culture as a simple model of stress, we have addressed whether LD biogenesis has a protective role in cells committed to die. Complete nutrient deprivation induced the biogenesis of LD in human LN18 glioblastoma and HeLa cells and also in CHO and rat primary astrocytes. In all cell types, death was associated with LD depletion and was accelerated by blocking LD biogenesis after pharmacological inhibition of Group IVA phospholipase A2 (cPLA2α) or down-regulation of ceramide kinase. Nutrient deprivation also induced ß-oxidation of fatty acids that was sensitive to cPLA2α inhibition, and cell survival in these conditions became strictly dependent on fatty acid catabolism. These results show that, during nutrient deprivation, cell viability is sustained by ß-oxidation of fatty acids that requires biogenesis and mobilization of LD.


Assuntos
Ácidos Graxos/química , Regulação da Expressão Gênica , Metabolismo dos Lipídeos , Lipídeos/química , Animais , Astrócitos/citologia , Autofagia , Células CHO , Linhagem Celular Tumoral , Sobrevivência Celular , Cricetulus , Meios de Cultura/metabolismo , Glucose/metabolismo , Células HeLa , Humanos , L-Lactato Desidrogenase/metabolismo , Lipólise , Microscopia de Fluorescência , Oxirredução , Oxigênio/química , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...