Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Lipid Res ; 65(1): 100482, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38052254

RESUMO

Excess cholesterol originating from nonhepatic tissues is transported within HDL particles to the liver for metabolism and excretion. Cholesterol efflux is initiated by lipid-free or lipid-poor apolipoprotein A1 interacting with the transmembrane protein ABCA1, a key player in cholesterol homeostasis. Defective ABCA1 results in reduced serum levels of HDL cholesterol, deposition of cholesterol in arteries, and an increased risk of early onset CVD. Over 300 genetic variants in ABCA1 have been reported, many of which are associated with reduced HDL cholesterol levels. Only a few of these have been functionally characterized. In this study, we have analyzed 51 previously unclassified missense variants affecting the extracellular domains of ABCA1 using a sensitive, easy, and low-cost fluorescence-based assay. Among these, only 12 variants showed a distinct loss-of-function phenotype, asserting their direct association with severe HDL disorders. These findings emphasize the crucial role of functional characterization of genetic variants in pathogenicity assessment and precision medicine. The functional rescue of ABCA1 loss-of-function variants through proteasomal inhibition or by the use of the chemical chaperone 4-phenylbutyric acid was genotype specific. Genotype-specific responses were also observed for the ability of apolipoprotein A1 to stabilize the different ABCA1 variants. In view of personalized medicine, this could potentially form the basis for novel therapeutic strategies.


Assuntos
Apolipoproteína A-I , Colesterol , HDL-Colesterol , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Fluorescência , Transportador 1 de Cassete de Ligação de ATP/genética , Colesterol/metabolismo , Mutação de Sentido Incorreto
2.
J Lipid Res ; 65(5): 100539, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38556050

RESUMO

Flavonoids, polyphenolic compounds found in plant-based diets, are associated with reduced risk of cardiovascular disease and longevity. These components are reported to reduce plasma levels of low-density lipoprotein (LDL) through an upregulation of the LDL receptor (LDLR), but the mechanism is still largely unknown. In this study, we have systematically screened the effect of 12 flavonoids from six different flavonoid subclasses on the effect on LDLR. This paper provides an in-depth analysis on how these flavonoids affect LDLR regulation and functionality. We found that most but not all of the tested flavonoids increased LDLR mRNA levels. Surprisingly, this increase was attributed to different regulatory mechanisms, such as enhanced LDLR promoter activity, LDLR mRNA stabilization, or LDLR protein stabilization, of which specific effectual parts of the flavonoid molecular structure could be assigned. These types of comparative analysis of various flavonoids enhance clarity and deepen the understanding of how the different structures of flavonoids affect LDLR regulation. Our data offer useful insights that may guide future research in developing therapeutic approaches for cardiovascular health.


Assuntos
Flavonoides , Receptores de LDL , Flavonoides/farmacologia , Flavonoides/química , Receptores de LDL/metabolismo , Receptores de LDL/genética , Humanos , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Regiões Promotoras Genéticas
3.
Hum Mol Genet ; 29(8): 1229-1238, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31600776

RESUMO

The cell-surface low-density lipoprotein receptor (LDLR) internalizes low-density lipoprotein (LDL) by receptor-mediated endocytosis and plays a key role in the regulation of plasma cholesterol levels. The ligand-binding domain of the LDLR contains seven ligand-binding repeats of approximately 40 residues each. Between ligand-binding repeats 4 and 5, there is a 10-residue linker region that is subject to enzymatic cleavage. The cleaved LDLR is unable to bind LDL. In this study, we have screened a series of enzyme inhibitors in order to identify the enzyme that cleaves the linker region. These studies have identified bone morphogenetic protein 1 (BMP1) as being the cleavage enzyme. This conclusion is based upon the use of the specific BMP1 inhibitor UK 383367, silencing of the BMP1 gene by the use of siRNA or CRISPR/Cas9 technology and overexpression of wild-type BMP1 or the loss-of-function mutant E214A-BMP1. We have also shown that the propeptide of BMP1 has to be cleaved at RSRR120↓ by furin-like proprotein convertases for BMP1 to have an activity towards the LDLR. Targeting BMP1 could represent a novel strategy to increase the number of functioning LDLRs in order to lower plasma LDL cholesterol levels. However, a concern by using BMP1 inhibitors as cholesterol-lowering drugs could be the risk of side effects based on the important role of BMP1 in collagen assembly.


Assuntos
Proteína Morfogenética Óssea 1/genética , LDL-Colesterol/genética , Colesterol/genética , Furina/genética , Receptores de LDL/genética , Animais , Proteína Morfogenética Óssea 1/antagonistas & inibidores , Células CHO , Sistemas CRISPR-Cas/genética , LDL-Colesterol/antagonistas & inibidores , LDL-Colesterol/sangue , Cricetulus , Endocitose/genética , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidrazinas/farmacologia , Ligantes , Lipoproteínas LDL/genética , Pró-Proteína Convertases/genética , Proteólise/efeitos dos fármacos , RNA Interferente Pequeno/genética , Receptores de LDL/antagonistas & inibidores , Tioureia/análogos & derivados , Tioureia/farmacologia
4.
Hum Mol Genet ; 28(18): 3043-3052, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31131398

RESUMO

Hydrolysis of cholesteryl esters and triglycerides in the lysosome is performed by lysosomal acid lipase (LAL). In this study we have investigated how 23 previously identified missense mutations in the LAL gene (LIPA) (OMIM# 613497) affect the structure of the protein and thereby disrupt LAL activity. Moreover, we have performed transfection studies to study intracellular transport of the 23 mutants. Our main finding was that most pathogenic mutations result in defective enzyme activity by affecting the normal folding of LAL. Whereas, most of the mutations leading to reduced stability of the cap domain did not alter intracellular transport, nearly all mutations that affect the stability of the core domain gave rise to a protein that was not efficiently transported from the endoplasmic reticulum (ER) to the Golgi apparatus. As a consequence, ER stress was generated that is assumed to result in ER-associated degradation of the mutant proteins. The two LAL mutants Q85K and S289C were selected to study whether secretion-defective mutants could be rescued from ER-associated degradation by the use of chemical chaperones. Of the five chemical chaperones tested, only the proteasomal inhibitor MG132 markedly increased the amount of mutant LAL secreted. However, essentially no increased enzymatic activity was observed in the media. These data indicate that the use of chemical chaperones to promote the exit of folding-defective LAL mutants from the ER, may not have a great therapeutic potential as long as these mutants appear to remain enzymatically inactive.


Assuntos
Mutação de Sentido Incorreto , Esterol Esterase/genética , Esterol Esterase/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Biologia Computacional/métodos , Estresse do Retículo Endoplasmático , Ativação Enzimática , Humanos , Modelos Moleculares , Conformação Proteica , Transporte Proteico , Proteólise , Esterol Esterase/biossíntese , Esterol Esterase/química , Relação Estrutura-Atividade
5.
Hum Mol Genet ; 28(22): 3734-3741, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31332430

RESUMO

A main strategy for lowering plasma low-density lipoprotein (LDL) cholesterol levels is to increase the number of cell-surface LDL receptors (LDLRs). This can be achieved by increasing the synthesis or preventing the degradation of the LDLR. One mechanism by which an LDLR becomes non-functional is enzymatic cleavage within the 10 residue linker region between ligand-binding repeats 4 and 5. The cleaved LDLR has only three ligand-binding repeats and is unable to bind LDL. In this study, we have performed cell culture experiments to identify strategies to prevent this cleavage. As a part of these studies, we found that Asp193 within the linker region is critical for cleavage to occur. Moreover, both 14-mer synthetic peptides and antibodies directed against the linker region prevented cleavage. As a consequence, more functional LDLRs were observed on the cell surface. The observation that the cleaved LDLR was present in extracts from the human adrenal gland indicates that cleavage of the linker region takes place in vivo. Thus, preventing cleavage of the LDLR by pharmacological measures could represent a novel lipid-lowering strategy.


Assuntos
Lipoproteínas LDL/metabolismo , Receptores de LDL/genética , Receptores de LDL/fisiologia , Animais , Anticorpos/imunologia , Células CHO , Membrana Celular/metabolismo , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Cricetulus , Humanos , Ligantes , Metabolismo dos Lipídeos/genética , Lipoproteínas LDL/genética , Peptídeos/metabolismo , Ligação Proteica/genética , Ligação Proteica/fisiologia
6.
Hum Mol Genet ; 26(9): 1634-1642, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28334946

RESUMO

Familial hypercholesterolemia (FH) is caused by mutations in the low density lipoprotein receptor (LDLR) gene. To study the impact of mutations affecting the hydrophobic transmembrane domain of the LDLR, each of the 22 amino acids of the transmembrane domain was individually mutated to arginine. The more centrally in the transmembrane domain an arginine was located, the lower amounts of the 120 kDa precursor LDLR in the endoplasmic reticulum were observed. This led to lower amounts of the 160 kDa mature LDLR on the cell surface. For the mutants V797R-LDLR, L798R-LDLR and L799R-LDLR a proportion of full-length receptors including the transmembrane and cytoplasmic domains, was secreted into the endoplasmic reticulum lumen to appear in the culture medium. When the transmembrane domain of the epidermal growth factor receptor (EGFR) was replaced by that of the mutant L799R-LDLR, similar effects were observed for the EGFR as for L799R-LDLR. Introducing arginines in the transmembrane domain of the LDLR also affected metalloproteinase cleavage of the ectodomain and γ-secretase cleavage within the transmembrane domain. The most likely explanation for the low amounts of the 120 kDa precursor is that a basic residue in the hydrophobic transmembrane domain prevents the mutant LDLR from being inserted in the endoplasmic reticulum membrane from the Sec61 translocon complex. As a consequence, quality control systems could be activated. However, our data indicate that proteasomal degradation, lysosomal degradation, autophagy or ectodomain cleavage were not the underlying mechanism for degradation of these mutant LDLRs.


Assuntos
Receptores de LDL/genética , Receptores de LDL/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Endocitose , Retículo Endoplasmático/metabolismo , Células Hep G2 , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Domínios Proteicos , Transporte Proteico , Proteólise
7.
Mol Genet Metab ; 123(2): 169-176, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29196158

RESUMO

Lysosomal acid lipase hydrolyzes cholesteryl esters and triglycerides contained in low density lipoprotein. Patients who are homozygous or compound heterozygous for mutations in the lysosomal acid lipase gene (LIPA), and have some residual enzymatic activity, have cholesteryl ester storage disease. One of the clinical features of this disease is hypercholesterolemia. Thus, patients with hypercholesterolemia who do not carry a mutation as a cause of autosomal dominant hypercholesterolemia, may actually have cholesteryl ester storage disease. In this study we have performed DNA sequencing of LIPA in 3027 hypercholesterolemic patients who did not carry a mutation as a cause of autosomal dominant hypercholesterolemia. Functional analyses of possibly pathogenic mutations and of all mutations in LIPA listed in The Human Genome Mutation Database were performed to determine the pathogenicity of these mutations. For these studies, HeLa T-REx cells were transiently transfected with mutant LIPA plasmids and Western blot analysis of cell lysates was performed to determine if the mutants were synthesized in a normal fashion. The enzymatic activity of the mutants was determined in lysates of the transfected cells using 4-methylumbelliferone-palmitate as the substrate. A total of 41 mutations in LIPA were studied, of which 32 mutations were considered pathogenic by having an enzymatic activity <10% of normal. However, none of the 3027 hypercholesterolemic patients were homozygous or compound heterozygous for a pathogenic mutation. Thus, cholesteryl ester storage disease must be a very rare cause of hypercholesterolemia in Norway.


Assuntos
Doença do Armazenamento de Colesterol Éster/epidemiologia , Doença do Armazenamento de Colesterol Éster/genética , Hipercolesterolemia/fisiopatologia , Mutação , Esterol Esterase/genética , Adulto , Doença do Armazenamento de Colesterol Éster/enzimologia , Feminino , Células HeLa , Homozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Noruega/epidemiologia , Fenótipo , Prevalência
8.
Hum Mol Genet ; 24(20): 5836-44, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26220972

RESUMO

Mutations in the low-density lipoprotein receptor (LDLR) gene cause familial hypercholesterolemia (FH). The mechanism by which mutations in the LDLR affecting the transmembrane domain of the receptor cause FH has not been thoroughly investigated. In this study, we have selected 12 naturally occurring mutations affecting the transmembrane domain and studied their effect on the LDLR. The main strategy has been to transiently transfect HepG2 cells with mutant LDLR plasmids and to study the mutant LDLRs in cell lysates and in media by western blot analysis. The most striking finding was that mutation p.L799R led to secretion of the entire 160 kDa mature L799R-LDLR. Residue 799Leu is in the middle of the 22-residue transmembrane domain, and introduction of a basic residue in the hydrophobic core of the transmembrane domain could prevent L799R-LDLR from being correctly recognized and integrated in the membrane by the Sec61 translocon complex. This would then lead to translocation of the entire L799R-LDLR into the lumen of the endoplasmic reticulum. Mutation p.L799R should be considered a member of a separate class of FH-causing mutations that affects the insertion of the LDLR in the cell membrane.


Assuntos
Hiperlipoproteinemia Tipo II/genética , Mutação , Receptores de LDL/genética , Motivos de Aminoácidos , Membrana Celular/metabolismo , Células Hep G2 , Humanos , Hiperlipoproteinemia Tipo II/metabolismo , Receptores de LDL/química , Receptores de LDL/metabolismo , Alinhamento de Sequência , Transfecção
9.
Biochem J ; 457(1): 99-105, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24144304

RESUMO

PCSK9 (proprotein convertase subtilisin/kexin type 9) binds to the LDLR (low-density lipoprotein receptor) at the cell surface and disrupts recycling of the LDLR. However, PCSK9 also interacts with the LDLR in the ER (endoplasmic reticulum). In the present study we have investigated the role of PCSK9 for the transport of the LDLR from the ER to the cell membrane. A truncated LDLR consisting of the ectodomain (ED-LDLR) was used for these studies to avoid PCSK9-mediated degradation of the LDLR. The amount of secreted ED-LDLR was used as a measure of the amount of ED-LDLR transported from the ER. From co-transfection experiments of various PCSK9 and ED-LDLR plasmids, PCSK9 increased the amount of WT (wild-type) ED-LDLR in the medium, but not of an ED-LDLR lacking the EGF (epidermal growth factor)-A repeat or of a Class 2a mutant ED-LDLR which fails to exit the ER. Mutant PCSK9s which failed to undergo autocatalytic cleavage or failed to exit the ER, failed to increase the amount of WT-ED-LDLR in the medium. These mutants also reduced the amount of WT-ED-LDLR intracellularly, which could partly be prevented by the proteasome inhibitor lactacystine. WT-ED-LDLR promoted autocatalytic cleavage of pro-PCSK9. The findings of the present study indicate that the binding of WT-ED-LDLR to pro-PCSK9 in the ER promotes autocatalytic cleavage of PCSK9, and autocatalytically cleaved PCSK9 acts as a chaperone to promote the exit of WT-ED-LDLR from the ER.


Assuntos
Retículo Endoplasmático/metabolismo , Chaperonas Moleculares/fisiologia , Pró-Proteína Convertases/fisiologia , Receptores de LDL/metabolismo , Serina Endopeptidases/fisiologia , Catálise , Membrana Celular/metabolismo , Células Hep G2 , Homeostase , Humanos , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/química , Domínios e Motivos de Interação entre Proteínas/fisiologia , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Precursores de Proteínas/química , Precursores de Proteínas/fisiologia , Transporte Proteico , Proteólise , Receptores de LDL/química , Serina Endopeptidases/química
10.
Hum Mol Genet ; 21(6): 1402-9, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22156580

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the epidermal growth factor homology domain repeat A of the low-density lipoprotein receptor (LDLR) at the cell surface and disrupts recycling of the internalized LDLR. As a consequence, the LDLR is rerouted to the lysosomes for degradation. Although PCSK9 may bind to an LDLR lacking the ligand-binding domain, at least three ligand-binding repeats of the ligand-binding domain are required for PCSK9 to reroute the LDLR to the lysosomes. In this study, we have studied the binding of PCSK9 to an LDLR with or without the ligand-binding domain at increasingly acidic conditions in order to mimic the milieu of the LDLR:PCSK9 complex as it translocates from the cell membrane to the sorting endosomes. These studies have shown that PCSK9 is rapidly released from an LDLR lacking the ligand-binding domain at pH in the range of 6.9-6.1. A similar pattern of release at acidic pH was also observed for the binding to the normal LDLR of mutant PCSK9 lacking the C-terminal domain. Together these data indicate that an interaction between the negatively charged ligand-binding domain of the LDLR and the positively charged C-terminal domain of PCSK9 is required for PCSK9 to remain bound to the LDLR during the early phase of endosomal acidification as the LDLR translocates from the cell membrane to the sorting endosome.


Assuntos
Ácidos/metabolismo , Endossomos/metabolismo , Pró-Proteína Convertases/metabolismo , Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Animais , Western Blotting , Células CHO , Membrana Celular/metabolismo , Células Cultivadas , Cricetinae , Humanos , Concentração de Íons de Hidrogênio , Rim/citologia , Rim/metabolismo , Lisossomos/metabolismo , Mutagênese Sítio-Dirigida , Mutação/genética , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/genética , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores de LDL/genética , Serina Endopeptidases/genética , Transdução de Sinais
11.
J Clin Lipidol ; 18(1): e80-e89, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37981531

RESUMO

BACKGROUND: Severe hypertriglyceridemia can be caused by pathogenic variants in genes encoding proteins involved in the metabolism of triglyceride-rich lipoproteins. A key protein in this respect is lipoprotein lipase (LPL) which hydrolyzes triglycerides in these lipoproteins. Another important protein is glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1) which transports LPL to the luminal side of the endothelial cells. OBJECTIVE: Our objective was to identify a genetic cause of hypertriglyceridemia in 459 consecutive unrelated subjects with levels of serum triglycerides ≥20 mmol/l. These patients had been referred for molecular genetic testing from 1998 to 2021. In addition, we wanted to study whether GPIHBP1 autoantibodies also were a cause of hypertriglyceridemia. METHODS: Molecular genetic analyses of the genes encoding LPL, GPIHBP1, apolipoprotein C2, lipase maturation factor 1 and apolipoprotein A5 as well as apolipoprotein E genotyping, were performed in all 459 patients. Serum was obtained from 132 of the patients for measurement of GPIHBP1 autoantibodies approximately nine years after molecular genetic testing was performed. RESULTS: A monogenic cause was found in four of the 459 (0.9%) patients, and nine (2.0%) patients had dyslipoproteinemia due to homozygosity for apolipoprotein E2. One of the 132 (0.8%) patients had GPIHBP1 autoantibody syndrome. CONCLUSION: Only 0.9% of the patients had monogenic hypertriglyceridemia, and only 0.8% had GPIHBP1 autoantibody syndrome. The latter figure is most likely an underestimate because serum samples were obtained approximately nine years after hypertriglyceridemia was first identified. There is a need to implement measurement of GPIHBP1 autoantibodies in clinical medicine to secure that proper therapeutic actions are taken.


Assuntos
Hipertrigliceridemia , Receptores de Lipoproteínas , Humanos , Autoanticorpos , Células Endoteliais , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Lipoproteínas , Hipertrigliceridemia/genética , Triglicerídeos/metabolismo , Biologia Molecular , Apolipoproteínas
12.
PLoS One ; 18(12): e0294764, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38039300

RESUMO

BACKGROUND: Cholesteryl ester transfer protein (CETP) transfers cholesteryl esters in plasma from high density lipoprotein (HDL) to very low density lipoprotein and low density lipoprotein. Loss-of-function variants in the CETP gene cause elevated levels of HDL cholesterol. In this study, we have determined the functional consequences of 24 missense variants in the CETP gene. The 24 missense variants studied were the ones reported in the Human Gene Mutation Database and in the literature to affect HDL cholesterol levels, as well as two novel variants identified at the Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital in subjects with hyperalphalipoproteinemia. METHODS: HEK293 cells were transiently transfected with mutant CETP plasmids. The amounts of CETP protein in lysates and media were determined by Western blot analysis, and the lipid transfer activities of the CETP variants were determined by a fluorescence-based assay. RESULTS: Four of the CETP variants were not secreted. Five of the variants were secreted less than 15% compared to the WT-CETP, while the other 15 variants were secreted in varying amounts. There was a linear relationship between the levels of secreted protein and the lipid transfer activities (r = 0.96, p<0.001). Thus, the secreted variants had similar specific lipid transfer activities. CONCLUSION: The effect of the 24 missense variants in the CETP gene on the lipid transfer activity was mediated predominantly by their impact on the secretion of the CETP protein. The four variants that prevented CETP secretion cause autosomal dominant hyperalphalipoproteinemia. The five variants that markedly reduced secretion of the respective variants cause mild hyperalphalipoproteinemia. The majority of the remaining 15 variants had minor effects on the secretion of CETP, and are considered neutral genetic variants.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol , Ésteres do Colesterol , Humanos , Proteínas de Transferência de Ésteres de Colesterol/genética , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , HDL-Colesterol , Células HEK293 , Transporte Biológico , Ésteres do Colesterol/metabolismo
13.
J Clin Lipidol ; 17(6): 800-807, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37718180

RESUMO

BACKGROUND: Molecular genetic testing of patients with hypobetalipoproteinemia may identify a genetic cause that can form the basis for starting proper therapy. Identifying a genetic cause may also provide novel data on the structure-function relationship of the mutant protein. OBJECTIVE: To identify a genetic cause of hypobetalipoproteinemia in a patient with levels of low density lipoprotein cholesterol at the detection limit of 0.1 mmol/l. METHODS: DNA sequencing of the translated exons with flanking intron sequences of the genes adenosine triphosphate-binding cassette transporter 1, angiopoietin-like protein 3, apolipoprotein B, apolipoprotein A1, lecithin-cholesterol acyltransferase, microsomal triglyceride transfer protein and proprotein convertase subtilisin/kexin type 9. RESULTS: The patient was homozygous for mutation Q384K (c.1150C>A) in the apolipoprotein B gene, and this mutation segregated with hypobetalipoproteinemia in the family. Residue Gln384 is located in the large lipid transfer module of apoB that has been suggested to be important for lipidation of apolipoprotein B through interaction with microsomal triglyceride transfer protein. Based on measurements of serum levels of triglycerides and apolipoprotein B-48 after an oral fat load, we conclude that the patient was able to synthesize apolipoprotein B-48 in the intestine in a seemingly normal fashion. CONCLUSION: Our data indicate that mutation Q384K severely reduces the secretion of apolipoprotein B-100 in the liver without reducing the secretion of apolipoprotein B-48 in the intestine. Possible mechanisms for the different effects of this and other missense mutations affecting the large lipid transfer module on the two forms of apoB are discussed.


Assuntos
Hipobetalipoproteinemias , Mutação de Sentido Incorreto , Humanos , Apolipoproteína B-100/genética , Apolipoproteína B-48 , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Intestinos , Hipobetalipoproteinemias/genética , Mutação , Fígado/metabolismo
14.
Mol Genet Metab ; 105(1): 149-54, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22078455

RESUMO

In this study we have characterized a naturally occurring truncated form of the low density lipoprotein receptor (LDLR). Western blot analysis of transfected cells indicated that the truncated form (∆N-LDLR) is a degradation product of the full-length LDLR generated by cleavage in the linker region between ligand-binding repeats 4 and 5 of the ligand-binding domain. The cleavage of the linker was not caused by components of the culture media, as heat inactivation of the media did not prevent cleavage. Rather, it is assumed that cleavage was caused by an enzyme secreted from the cells. Biotinylation experiments showed that ∆N-LDLR is located on the cell surface and is detectable approximately 5 h after synthesis of the full-length LDLR. Flow cytometric analysis showed that ∆N-LDLR was not able to bind and internalize low density lipoprotein (LDL). ∆N-LDLR appeared to be equally stable as the full-length LDLR. Thus, generation of ∆N-LDLR does not appear to be the first signal for degradation of the LDLR. The existence of two functionally different populations of LDLRs on the cell surface, of which ∆N-LDLR constitutes 28%, must be taken into account when interpreting results of experiments to study LDLRs on the cell surface. Furthermore, if the cleavage of the linker between ligand-binding repeats 4 and 5 could be prevented by an enzyme inhibitor, this could represent a novel therapeutic strategy to increase the number of functioning LDLRs and thereby decrease the levels of plasma LDL cholesterol.


Assuntos
Proteólise , Receptores de LDL/metabolismo , Animais , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Meios de Cultura , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Humanos , Ligantes , Proteínas Mutantes/metabolismo , Inibidores de Proteases/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Receptores de LDL/antagonistas & inibidores , Receptores de LDL/química , Deleção de Sequência
15.
J Lipid Res ; 52(10): 1787-94, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21771976

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the low density lipoprotein receptor (LDLR) at the cell surface and disrupts the normal recycling of the LDLR. In this study, we investigated the role of the C-terminal domain for the activity of PCSK9. Experiments in which conserved residues and histidines on the surface of the C-terminal domain were mutated indicated that no specific residues of the C-terminal domain, apart from those responsible for maintaining the overall structure, are required for the activity of PCSK9. Rather, the net charge of the C-terminal domain is important. The more positively charged the C-terminal domain, the higher the activity toward the LDLR. Moreover, replacement of the C-terminal domain with an unrelated protein of comparable size led to significant activity of the chimeric protein. We conclude that the role of the evolutionary, poorly conserved C-terminal domain for the activity of PCSK9 reflects its overall positive charge and size and not the presence of specific residues involved in protein-protein interactions.


Assuntos
Endossomos/metabolismo , Receptores de LDL/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Alanina/química , Alanina/metabolismo , Sequência de Aminoácidos , Endossomos/química , Células Hep G2 , Histidina/química , Histidina/metabolismo , Humanos , Dados de Sequência Molecular , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Ligação Proteica , Receptores de LDL/química , Células Tumorais Cultivadas
16.
Biochem Biophys Res Commun ; 406(2): 234-8, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21324305

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the low density lipoprotein receptor (LDLR) at the cell surface and mediates intracellular degradation of the LDLR. The amino-terminus of mature PCSK9, residues 31-53 of the prodomain, has an inhibitory effect on this function of PCSK9, but the underlying mechanism is not fully understood. In this study, we have identified two highly conserved negatively charged segments (residues 32-40 and 48-50, respectively) within this part of the prodomain and performed deletions and substitutions to study their importance for degradation of the LDLRs. Deletion of the acidic residues of the longest negatively charged segment increased PCSK9's ability to degrade the LDLR by 31%, whereas a modest 8% increase was observed when these residues were mutated to uncharged amino acids. Thus, both the length and the charge of this part of the prodomain were important for its inhibitory effect. Deletion of the residues of the shorter second negatively charged segment only increased PCSK9's activity by 8%. Substitution of the amino acids of both charged segments to uncharged residues increased PCSK9's activity by 36%. These findings indicate that the inhibitory effect of residues 31-53 of the prodomain is due to the negative charge of this segment. The underlying mechanism could involve the binding of this peptide segment to positively charged structures which are important for PCSK9's activity. One possible candidate could be the histidine-rich C-terminal domain of PCSK9.


Assuntos
Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Sequência Conservada , Humanos , Dados de Sequência Molecular , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Estrutura Terciária de Proteína , Deleção de Sequência , Serina Endopeptidases/química , Serina Endopeptidases/genética
17.
Biochem Biophys Res Commun ; 408(4): 642-6, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21531209

RESUMO

The low density lipoprotein receptor (LDLR) binds and internalizes low density lipoprotein (LDL). At the mildly acidic pH of the sorting endosomes, LDL is released from the receptor and the receptor recycles back to the cell membrane. Mutations in the LDLR gene may disrupt the normal function of the LDLR in different ways. Class 5 mutations result in receptors that are able to bind and internalize LDL, but they fail to release LDL in the sorting endosomes and fail to recycle. Instead they are rerouted to the lysosomes for degradation. However, the underlying mechanism remains to be determined. To study the role of the cytoplasmic domain of the LDLR for rerouting Class 5 mutants to the lysosomes, we have performed studies to determine whether Class 5 mutants caused by mutations E387K or V408M are degraded when the cytoplasmic domain has been altered or deleted. As determined by confocal laser-scanning microscopy, these mutant LDLR were inserted into the cell membrane and were able to internalize LDL. As determined by Western blot analysis, Class 5 mutants without a cytoplasmic domain still were degraded after binding LDL. Thus, the cytoplasmic domain does not play a role in rerouting Class 5 mutant LDLR to the lysosomes. Rather, one may speculate that sterical hindrance may prevent Class 5 mutants with bound LDL from entering the narrow recycling tubules of the sorting endosome.


Assuntos
Citoplasma/metabolismo , Lisossomos/metabolismo , Receptores de LDL/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Mutação , Estrutura Terciária de Proteína , Receptores de LDL/genética
18.
Biochem Biophys Res Commun ; 415(4): 642-5, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-22079632

RESUMO

Newly synthesized low density lipoprotein receptors (LDLRs) exit the endoplasmic reticulum (ER) as the first step in the secretory pathway. In this study we have generated truncating deletions and substitutions within the 50 amino acid cytoplasmic domain of the LDLR in order to identify residues required for the exit from the ER. Western blot analysis was used to determine the relative amounts of the 120 kDa precursor form of the LDLR located in the ER and the 160 kDa mature form that has exited the ER. These studies have shown that the exit of an LDLR lacking the cytoplasmic domain, is markedly reduced. Moreover, the longer the cytoplasmic domain, the more efficient is the exit from the ER. At least 30 residues were required for the LDLR to efficiently exit the ER. Mutations in the two di-acidic motifs ExE(814) and/or ExD(837) had only a small effect on the exit from the ER. The requirement for a certain length of the cytoplasmic domain for efficient exit from the ER, could reflect the distance needed to interact with the COPII complex of the ER membrane or the requirement for the LDLR to undergo dimerization.


Assuntos
Citoplasma/metabolismo , Retículo Endoplasmático/metabolismo , Receptores de LDL/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Células CHO , Cricetinae , Humanos , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Transporte Proteico , Receptores de LDL/química , Receptores de LDL/genética , Deleção de Sequência
19.
Mol Genet Metab ; 99(2): 149-56, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19828345

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the extracellular domain of the low density lipoprotein receptor (LDLR) at the cell surface, and disrupts the normal recycling of the LDLR. However, the exact mechanism by which the LDLR is re-routed for lysosomal degradation remains to be determined. To clarify the role of the cytoplasmic domain of the LDLR for re-routing to the lysosomes, we have studied the ability of PCSK9 to degrade a chimeric receptor which contains the extracellular and transmembrane domains of the LDLR and the cytoplasmic domain of the transferrin receptor. These studies were performed in CHO T-REx cells stably transfected with a plasmid encoding the chimeric receptor and a novel assay was developed to study the effect of PCSK9 on the LDLR in these cells. Localization, function and stability of the chimeric receptor were similar to that of the wild-type LDLR. The addition of purified gain-of-function mutant D374Y-PCSK9 to the culture medium of stably transfected CHO T-REx cells showed that the chimeric receptor was degraded, albeit to a lower extent than the wild-type LDLR. In addition, a mutant LDLR, which has the three lysines in the intracellular domain substituted with arginines, was also degraded by D374Y-PCSK9. Thus, the mechanism for the PCSK9-mediated degradation of the LDLR does not appear to involve an interaction between the endosomal sorting machinery and LDLR-specific motifs in the cytoplasmic domain. Moreover, ubiquitination of lysines in the cytoplasmic domain does not appear to play a critical role in the PCSK9-mediated degradation of the LDLR.


Assuntos
Processamento de Proteína Pós-Traducional , Receptores de LDL/metabolismo , Receptores da Transferrina/química , Receptores da Transferrina/metabolismo , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Endocitose , Ensaios Enzimáticos , Humanos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Estabilidade Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores de LDL/química , Transfecção
20.
Mol Genet Metab ; 101(1): 76-80, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20659812

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) post-translationally regulates the number of cell-surface low density lipoprotein receptors (LDLR). This is accomplished by the ability of PCSK9 to mediate degradation of the LDLR. The underlying mechanism involves binding of secreted PCSK9 to the epidermal growth factor-like repeat A of the extracellular domain of the LDLR at the cell surface, followed by lysosomal degradation of the internalized LDLR:PCSK9 complex. However, the mechanism by which the normal recycling of the LDLR is disrupted by PCSK9, remains to be determined. In this study we have investigated the role of the cytoplasmic domain of the LDLR for this process. This has been done by studying the ability of a mutant LDLR (K811X-LDLR) which lacks the cytoplasmic domain, to be degraded by PCSK9. We show that this mutant receptor is degraded by PCSK9. Thus, the machinery which directs the LDLR:PCSK9 complex to the lysosomes for degradation, does not interact with the cytoplasmic domain of the LDLR.


Assuntos
Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Citoplasma/metabolismo , Estrutura Terciária de Proteína , Receptores de LDL/análise , Receptores de LDL/química , Serina Endopeptidases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA