201
|
Hovingh GK, Davidson MH, Kastelein JJ, O'Connor AM. Diagnosis and treatment of familial hypercholesterolaemia. Eur Heart J 2013; 34:962-71. [DOI: 10.1093/eurheartj/eht015] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
202
|
Xu H, Xu G, Wang D, Zheng C, Wan L. Molecular cloning and tissue distribution of the phosphotyrosine interaction domain containing 1 (PID1) gene in Tianfu goat. Gene 2013; 515:71-7. [DOI: 10.1016/j.gene.2012.11.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/28/2012] [Accepted: 11/27/2012] [Indexed: 10/27/2022]
|
203
|
Sorrentino V, Fouchier SW, Motazacker MM, Nelson JK, Defesche JC, Dallinga-Thie GM, Kastelein JJP, Kees Hovingh G, Zelcer N. Identification of a loss-of-function inducible degrader of the low-density lipoprotein receptor variant in individuals with low circulating low-density lipoprotein. Eur Heart J 2013; 34:1292-7. [PMID: 23324548 DOI: 10.1093/eurheartj/ehs472] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIMS Recent genome-wide association studies suggest that IDOL (also known as MYLIP) contributes to variation in circulating levels of low-density lipoprotein cholesterol (LDL-C). IDOL, an E3-ubiquitin ligase, is a recently identified post-transcriptional regulator of LDLR abundance. Briefly, IDOL promotes degradation of the LDLR thereby limiting LDL uptake. Yet the exact role of IDOL in human lipoprotein metabolism is unclear. Therefore, this study aimed at identifying and functionally characterizing IDOL variants in the Dutch population and to assess their contribution to circulating levels of LDL-C. METHODS AND RESULTS We sequenced the IDOL coding region in 677 individuals with LDL-C above the 95th percentile adjusted for age and gender (high-LDL-C cohort) in which no mutations in the LDLR, APOB, and PCSK9 could be identified. In addition, IDOL was sequenced in 560 individuals with baseline LDL-C levels below the 20th percentile adjusted for age and gender (low-LDL-C cohort). We identified a total of 14 IDOL variants (5 synonymous, 8 non-synonymous, and 1 non-sense). Functional characterization of these variants demonstrated that the p.Arg266X variant represents a complete loss of IDOL function unable to promote ubiquitylation and subsequent degradation of the LDLR. Consistent with loss of IDOL function, this variant was identified in individuals with low circulating LDL-C. CONCLUSION Our results support the notion that IDOL contributes to variation in circulating levels of LDL-C. Strategies to inhibit IDOL activity may therefore provide a novel therapeutic venue to treating dyslipidaemia.
Collapse
Affiliation(s)
- Vincenzo Sorrentino
- Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, Meibergdreef 15, Amsterdam 1105AZ, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Guardamagna O, Cagliero P, Abello F. Management of Inherited Atherogenic Dyslipidemias in Children. Ther Apher Dial 2012; 17:150-61. [DOI: 10.1111/j.1744-9987.2012.01146.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
205
|
Kang YL, Yochem J, Bell L, Sorensen EB, Chen L, Conner SD. Caenorhabditis elegans reveals a FxNPxY-independent low-density lipoprotein receptor internalization mechanism mediated by epsin1. Mol Biol Cell 2012; 24:308-18. [PMID: 23242996 PMCID: PMC3564534 DOI: 10.1091/mbc.e12-02-0163] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A genome-wide RNA interference screen using Caenorhabditis elegans LRP-1/megalin as a model for LDLR transport was employed to identify factors critical to LDLR uptake. We provide evidence that epsin1 promotes LDLR internalization via a FxNPxY-independent pathway. We complement C. elegans in vivo approaches with loss-of-function and biochemical analyses, using mammalian cell culture systems to evaluate epsin1’s mode of action in LDLR endocytosis. Low-density lipoprotein receptor (LDLR) internalization clears cholesterol-laden LDL particles from circulation in humans. Defects in clathrin-dependent LDLR endocytosis promote elevated serum cholesterol levels and can lead to atherosclerosis. However, our understanding of the mechanisms that control LDLR uptake remains incomplete. To identify factors critical to LDLR uptake, we pursued a genome-wide RNA interference screen using Caenorhabditis elegans LRP-1/megalin as a model for LDLR transport. In doing so, we discovered an unanticipated requirement for the clathrin-binding endocytic adaptor epsin1 in LDLR endocytosis. Epsin1 depletion reduced LDLR internalization rates in mammalian cells, similar to the reduction observed following clathrin depletion. Genetic and biochemical analyses of epsin in C. elegans and mammalian cells uncovered a requirement for the ubiquitin-interaction motif (UIM) as critical for receptor transport. As the epsin UIM promotes the internalization of some ubiquitinated receptors, we predicted LDLR ubiquitination as necessary for endocytosis. However, engineered ubiquitination-impaired LDLR mutants showed modest internalization defects that were further enhanced with epsin1 depletion, demonstrating epsin1-mediated LDLR endocytosis is independent of receptor ubiquitination. Finally, we provide evidence that epsin1-mediated LDLR uptake occurs independently of either of the two documented internalization motifs (FxNPxY or HIC) encoded within the LDLR cytoplasmic tail, indicating an additional internalization mechanism for LDLR.
Collapse
Affiliation(s)
- Yuan-Lin Kang
- Department of Genetics, Cell Biology, and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
206
|
|
207
|
Pećin I, Whittall R, Futema M, Sertić J, Reiner Ž, Leigh SEA, Humphries SE. Mutation detection in Croatian patients with Familial Hypercholesterolemia. Ann Hum Genet 2012; 77:22-30. [DOI: 10.1111/j.1469-1809.2012.00735.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 08/25/2012] [Indexed: 02/03/2023]
Affiliation(s)
- Ivan Pećin
- Department of Internal Medicine; University Hospital Center Zagreb; Croatia
| | - Ros Whittall
- Centre for Cardiovascular Genetics; British Heart Foundation Laboratories, The Rayne Building; Royal Free and University College London Medical School; London; WC1E 6JJ; UK
| | - Marta Futema
- Centre for Cardiovascular Genetics; British Heart Foundation Laboratories, The Rayne Building; Royal Free and University College London Medical School; London; WC1E 6JJ; UK
| | - Jadranka Sertić
- Center for Clinical and Laboratory Diagnostics; University Hospital Center Zagreb; Croatia
| | - Željko Reiner
- Department of Internal Medicine; University Hospital Center Zagreb; Croatia
| | - Sarah E. A. Leigh
- Centre for Cardiovascular Genetics; British Heart Foundation Laboratories, The Rayne Building; Royal Free and University College London Medical School; London; WC1E 6JJ; UK
| | - Steve E. Humphries
- Centre for Cardiovascular Genetics; British Heart Foundation Laboratories, The Rayne Building; Royal Free and University College London Medical School; London; WC1E 6JJ; UK
| |
Collapse
|
208
|
Ferri N. Proprotein convertase subtilisin/kexin type 9: from the discovery to the development of new therapies for cardiovascular diseases. SCIENTIFICA 2012; 2012:927352. [PMID: 24278757 PMCID: PMC3820617 DOI: 10.6064/2012/927352] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 08/28/2012] [Indexed: 06/02/2023]
Abstract
The identification of the HMG-CoA reductase inhibitors, statins, has represented a dramatic innovation of the pharmacological modulation of hypercholesterolemia and associated cardiovascular diseases. However, not all patients receiving statins achieve guideline-recommended low density lipoprotein (LDL) cholesterol goals, particularly those at high risk. There remains, therefore, an unmet medical need to develop additional well-tolerated and effective agents to lower LDL cholesterol levels. The discovery of proprotein convertase subtilisin/kexin type 9 (PCSK9), a secretory protein that posttranscriptionally regulates levels of low density lipoprotein receptor (LDLR) by inducing its degradation, has opened a new era of pharmacological modulation of cholesterol homeostasis. This paper summarizes the current knowledge of the basic molecular mechanism underlying the regulatory effect of LDLR expression by PCSK9 obtained from in vitro cell-cultured studies and the analysis of the crystal structure of PCSK9. It also describes the epidemiological and experimental evidences of the regulatory effect of PCSK9 on LDL cholesterol levels and cardiovascular diseases and summarizes the different pharmacological approaches under development for inhibiting PCSK9 expression, processing, and the interaction with LDLR.
Collapse
Affiliation(s)
- Nicola Ferri
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| |
Collapse
|
209
|
Wang Y, Huang Y, Hobbs HH, Cohen JC. Molecular characterization of proprotein convertase subtilisin/kexin type 9-mediated degradation of the LDLR. J Lipid Res 2012; 53:1932-43. [PMID: 22764087 PMCID: PMC3413232 DOI: 10.1194/jlr.m028563] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that promotes degradation of cell surface LDL receptors (LDLRs) in selected cell types. Here we used genetic and pharmacological inhibitors to define the pathways involved in PCSK9-mediated LDLR degradation. Inactivating mutations in autosomal recessive hypercholesterolemia (ARH), an endocytic adaptor, blocked PCSK9-mediated LDLR degradation in lymphocytes but not in fibroblasts. Thus, ARH is not specifically required for PCSK9-mediated LDLR degradation. Knockdown of clathrin heavy chain with siRNAs prevented LDLR degradation. In contrast, prevention of ubiquitination of the LDLR cytoplasmic tail, inhibition of proteasomal activity, or disruption of proteins required for lysosomal targeting via macroautophagy (autophagy related 5 and 7) or the endosomal sorting complex required for trafficking (ESCRT) pathway (hepatocyte growth factor-regulated Tyr-kinase substrate and tumor suppressor gene 101) failed to block PCSK9-mediated LDLR degradation. These findings are consistent with a model in which the LDLR-PCSK9 complex is internalized via clathrin-mediated endocytosis and then routed to lysosomes via a mechanism that does not require ubiquitination and is distinct from the autophagy and proteosomal degradation pathways. Finally, the PCSK9-LDLR complex appears not to be transported by the canonical ESCRT pathway.
Collapse
Affiliation(s)
- Yan Wang
- Department of Molecular Genetics, Howard Hughes Medical Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | |
Collapse
|
210
|
Hymel D, Peterson BR. Synthetic cell surface receptors for delivery of therapeutics and probes. Adv Drug Deliv Rev 2012; 64:797-810. [PMID: 22401875 PMCID: PMC3359398 DOI: 10.1016/j.addr.2012.02.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/18/2012] [Accepted: 02/20/2012] [Indexed: 11/17/2022]
Abstract
Receptor-mediated endocytosis is a highly efficient mechanism for cellular uptake of membrane-impermeant ligands. Cells use this process to acquire nutrients, initiate signal transduction, promote development, regulate neurotransmission, and maintain homeostasis. Natural receptors that participate in receptor-mediated endocytosis are structurally diverse, ranging from large transmembrane proteins to small glycolipids embedded in the outer leaflet of cellular plasma membranes. Despite their vast structural differences, these receptors share common features of binding to extracellular ligands, clustering in dynamic membrane regions that pinch off to yield intracellular vesicles, and accumulation of receptor-ligand complexes in membrane-sealed endosomes. Receptors typically dissociate from ligands in endosomes and cycle back to the cell surface, whereas internalized ligands are usually delivered into lysosomes, where they are degraded, but some can escape and penetrate into the cytosol. Here, we review efforts to develop synthetic cell surface receptors, defined as nonnatural compounds, exemplified by mimics of cholesterol, that insert into plasma membranes, bind extracellular ligands including therapeutics, probes, and endogenous proteins, and engage endocytic membrane trafficking pathways. By mimicking natural mechanisms of receptor-mediated endocytosis, synthetic cell surface receptors have the potential to function as prosthetic molecules capable of seamlessly augmenting the endocytic uptake machinery of living mammalian cells.
Collapse
Affiliation(s)
- David Hymel
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, United States
| | - Blake R. Peterson
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, United States
| |
Collapse
|
211
|
Sorrentino V, Zelcer N. Post-transcriptional regulation of lipoprotein receptors by the E3-ubiquitin ligase inducible degrader of the low-density lipoprotein receptor. Curr Opin Lipidol 2012; 23:213-219. [PMID: 22510808 DOI: 10.1097/mol.0b013e3283532947] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW The hepatic low-density lipoprotein receptor (LDLR) pathway is essential for clearing circulating LDL and is an important therapeutic target for treating cardiovascular disease. Abundance of the LDLR is subject to both transcriptional and nontranscriptional control. Here, we highlight a new post-transcriptional mechanism for controlling LDLR function via ubiquitination of the receptor by the E3-ubiquitin ligase inducible degrader of the LDLR (IDOL). RECENT FINDINGS IDOL is a recently identified transcriptional target of the liver X receptors. Acting as an E3-ubiquitin ligase IDOL promotes ubiquitination of the LDLR, thereby marking it for lysosomal degradation. The determinants required for degradation of the LDLR by IDOL have been largely identified. IDOL also targets two related lipoprotein receptors, the very low-density lipoprotein receptor and apolipoprotein E receptor 2. Despite several similarities, the IDOL, and PCSK9 pathways for controlling LDLR abundance seem independent of each other. Genome-wide association studies have recently identified IDOL as a locus influencing variability in circulating levels of LDL, thereby highlighting the possible role of IDOL in human lipoprotein metabolism. SUMMARY Transcriptional induction of IDOL by liver X receptor defines a new post-transcriptional pathway for controlling LDLR abundance and LDL uptake independent of sterol regulatory element binding proteins. Targeting IDOL activity may offer a novel therapeutic approach complementary to statins for treating cardiovascular disease.
Collapse
Affiliation(s)
- Vincenzo Sorrentino
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
212
|
Yamamoto T, Harada-Shiba M, Nakatani M, Wada S, Yasuhara H, Narukawa K, Sasaki K, Shibata MA, Torigoe H, Yamaoka T, Imanishi T, Obika S. Cholesterol-lowering Action of BNA-based Antisense Oligonucleotides Targeting PCSK9 in Atherogenic Diet-induced Hypercholesterolemic Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2012; 1:e22. [PMID: 23344002 PMCID: PMC3393380 DOI: 10.1038/mtna.2012.16] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent findings in molecular biology implicate the involvement of proprotein convertase
subtilisin/kexin type 9 (PCSK9) in low-density lipoprotein receptor (LDLR) protein
regulation. The cholesterol-lowering potential of anti-PCSK9 antisense oligonucleotides
(AONs) modified with bridged nucleic acids (BNA-AONs) including 2′,4′-BNA
(also called as locked nucleic acid (LNA)) and 2′,4′-BNANC
chemistries were demonstrated both in vitro and in vivo. An in
vitro transfection study revealed that all of the BNA-AONs induce dose-dependent
reductions in PCSK9 messenger RNA (mRNA) levels concomitantly with increases in LDLR
protein levels. BNA-AONs were administered to atherogenic diet-fed C57BL/6J mice twice
weekly for 6 weeks; 2′,4′-BNA-AON that targeted murine PCSK9 induced a
dose-dependent reduction in hepatic PCSK9 mRNA and LDL cholesterol (LDL-C); the 43%
reduction of serum LDL-C was achieved at a dose of 20 mg/kg/injection with only
moderate increases in toxicological indicators. In addition, the serum high-density
lipoprotein cholesterol (HDL-C) levels increased. These results support antisense
inhibition of PCSK9 as a potential therapeutic approach. When compared with
2′,4′-BNA-AON, 2′,4′-BNANC-AON showed an earlier
LDL-C–lowering effect and was more tolerable in mice. Our results validate the
optimization of 2′,4′-BNANC-based anti-PCSK9 antisense molecules to
produce a promising therapeutic agent for the treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Tsuyoshi Yamamoto
- 1] Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan [2] Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Abstract
Cardiovascular disease encompasses a range of conditions extending from myocardial infarction to congenital heart disease, most of which are heritable. Enormous effort has been invested in understanding the genes and specific DNA sequence variants that are responsible for this heritability. Here, we review the lessons learned for monogenic and common, complex forms of cardiovascular disease. We also discuss key challenges that remain for gene discovery and for moving from genomic localization to mechanistic insights, with an emphasis on the impact of next-generation sequencing and the use of pluripotent human cells to understand the mechanism by which genetic variation contributes to disease.
Collapse
Affiliation(s)
- Sekar Kathiresan
- Center for Human Genetic Research and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | |
Collapse
|
214
|
Brown WV, Breslow J, Ballantyne C. Clinical use of genetic typing in human lipid disorders†. J Clin Lipidol 2012; 6:199-207. [DOI: 10.1016/j.jacl.2012.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 02/09/2012] [Accepted: 02/13/2012] [Indexed: 10/28/2022]
|
215
|
Atomic structure of the autosomal recessive hypercholesterolemia phosphotyrosine-binding domain in complex with the LDL-receptor tail. Proc Natl Acad Sci U S A 2012; 109:6916-21. [PMID: 22509010 DOI: 10.1073/pnas.1114128109] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hypercholesterolemia, high serum cholesterol in the form of LDL, is a major risk factor for atherosclerosis. LDL is mostly degraded in the liver after its cellular internalization with the LDL receptor (LDLR). This clathrin-mediated endocytosis depends on the protein autosomal recessive hypercholesterolemia (ARH), which binds the LDLR cytoplasmic tail. Mutations in either the LDLR tail or in ARH lead to hypercholesterolemia and premature atherosclerosis. Despite the significance of this interaction for cholesterol homeostasis, no structure of either ARH or the LDLR tail is available to determine its molecular basis. We report the crystal structure at 1.37-Å resolution of the phosphotyrosine-binding (PTB) domain of ARH in complex with an LDLR tail peptide containing the FxNPxY(0) internalization signal. Surprisingly, ARH interacts with a longer portion of the tail than previously recognized, which extends to I(-7)xF(-5)xNPxY(0)QK(+2). The LDLR tail assumes a unique "Hook"-like structure with a double β-turn conformation, which is accommodated in distinctive ARH structural determinants (i.e., an extended backbone hydrogen-bonding platform, three hydrophobic helical grooves, and a hydrophobic pocket for Y(0)). This unique complementarity differs significantly in related PTB proteins and may account for the unique physiological role of these partners in the hepatic uptake of cholesterol LDL. Moreover, the unusual hydrophobic pocket for Y(0) explains the distinctive ability of ARH to internalize proteins containing either FxNPxY(0) or FxNPxF(0) sequences. Biophysical measurements reveal how mutations associated with hypercholesterolemia destabilize ARH and its complex with LDLR and illuminate LDL internalization defects seen in patients.
Collapse
|
216
|
Braamskamp MJ, Wijburg FA, Wiegman A. Drug Therapy of Hypercholesterolaemia in Children and Adolescents. Drugs 2012; 72:759-72. [DOI: 10.2165/11632810-000000000-00000] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
217
|
Sigismund S, Confalonieri S, Ciliberto A, Polo S, Scita G, Di Fiore PP. Endocytosis and signaling: cell logistics shape the eukaryotic cell plan. Physiol Rev 2012; 92:273-366. [PMID: 22298658 DOI: 10.1152/physrev.00005.2011] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Our understanding of endocytosis has evolved remarkably in little more than a decade. This is the result not only of advances in our knowledge of its molecular and biological workings, but also of a true paradigm shift in our understanding of what really constitutes endocytosis and of its role in homeostasis. Although endocytosis was initially discovered and studied as a relatively simple process to transport molecules across the plasma membrane, it was subsequently found to be inextricably linked with almost all aspects of cellular signaling. This led to the notion that endocytosis is actually the master organizer of cellular signaling, providing the cell with understandable messages that have been resolved in space and time. In essence, endocytosis provides the communications and supply routes (the logistics) of the cell. Although this may seem revolutionary, it is still likely to be only a small part of the entire story. A wealth of new evidence is uncovering the surprisingly pervasive nature of endocytosis in essentially all aspects of cellular regulation. In addition, many newly discovered functions of endocytic proteins are not immediately interpretable within the classical view of endocytosis. A possible framework, to rationalize all this new knowledge, requires us to "upgrade" our vision of endocytosis. By combining the analysis of biochemical, biological, and evolutionary evidence, we propose herein that endocytosis constitutes one of the major enabling conditions that in the history of life permitted the development of a higher level of organization, leading to the actuation of the eukaryotic cell plan.
Collapse
Affiliation(s)
- Sara Sigismund
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | | | | | | | | | | |
Collapse
|
218
|
Genetic susceptibility to atherosclerosis. Stroke Res Treat 2012; 2012:362941. [PMID: 22550613 PMCID: PMC3329672 DOI: 10.1155/2012/362941] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 01/21/2012] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a complex multifocal arterial disease involving interactions of multiple genetic and environmental factors. Advances in techniques of molecular genetics have revealed that genetic ground significantly influences susceptibility to atherosclerotic vascular diseases. Besides further investigations of monogenetic diseases, candidate genes, genetic polymorphisms, and susceptibility loci associated with atherosclerotic diseases have been identified in recent years, and their number is rapidly increasing. This paper discusses main genetic investigations fields associated with human atherosclerotic vascular diseases. The paper concludes with a discussion of the directions and implications of future genetic research in arteriosclerosis with an emphasis on prospective prediction from an early age of individuals who are predisposed to develop premature atherosclerosis as well as to facilitate the discovery of novel drug targets.
Collapse
|
219
|
Tada H, Kawashiri MA, Ikewaki K, Terao Y, Noguchi T, Nakanishi C, Tsuchida M, Takata M, Miwa K, Konno T, Hayashi K, Nohara A, Inazu A, Kobayashi J, Mabuchi H, Yamagishi M. Altered Metabolism of Low-Density Lipoprotein and Very-Low-Density Lipoprotein Remnant in Autosomal Recessive Hypercholesterolemia. ACTA ACUST UNITED AC 2012; 5:35-41. [DOI: 10.1161/circgenetics.111.960948] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Autosomal recessive hypercholesterolemia (ARH) exhibits different responsiveness to statins compared with that in homozygous familial hypercholesterolemia (FH). However, few data exist regarding lipoprotein metabolism of ARH. Therefore, we aimed to clarify lipoprotein metabolism, especially the remnant lipoprotein fractions of ARH before and after statin therapy.
Methods and Results—
We performed a lipoprotein kinetic study in an ARH patient and 7 normal control subjects, using stable isotope methodology (10 mg/kg of [
2
H
3
]-leucine). These studies were performed at baseline and after the 20 mg daily dose of atorvastatin. Tracer/tracee ratio of apolipoprotein B (apoB) was determined by gas chromatography/mass spectrometry and fractional catabolic rates (FCR) were determined by multicompartmental modeling, including remnant lipoprotein fractions. FCR of low-density lipoprotein (LDL) apoB of ARH was significantly lower than those of control subjects (0.109 versus 0.450±0.122 1/day). In contrast, the direct removal of very-low-density lipoprotein remnant was significantly greater in ARH than those in control subjects (47.5 versus 2±2%). Interestingly, FCR of LDL apoB in ARH dramatically increased to 0.464 1/day, accompanying reduction of LDL cholesterol levels from 8.63 to 4.22 mmol/L after treatment with atorvastatin of 20 mg/d for 3 months.
Conclusions—
These results demonstrate that ARH exhibits decreased LDL clearance associated with decreased FCR of LDL apoB and increased clearance for very-low-density lipoprotein remnant. We suggest that increased clearance of remnant lipoprotein fractions could contribute to the great responsiveness to statins, providing new insights into the lipoprotein metabolism of ARH and the novel pharmacological target for LDLRAP1.
Collapse
Affiliation(s)
- Hayato Tada
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Masa-aki Kawashiri
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Katsunori Ikewaki
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Yoshio Terao
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Tohru Noguchi
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Chiaki Nakanishi
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Masayuki Tsuchida
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Mutsuko Takata
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Kenji Miwa
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Tetsuo Konno
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Kenshi Hayashi
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Atsushi Nohara
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Akihiro Inazu
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Junji Kobayashi
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Hiroshi Mabuchi
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| | - Masakazu Yamagishi
- From the Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (H.T., M.-a.K., C.N., M. Tsuchida, M. Takata, T.K., K.H., M.Y.); the Division of Anti-Aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan (K.I.); the Division of Cardiology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan (Y.T.); the Department of Lipidology, Kanazawa University Graduate School of Medicine, Kanazawa,
| |
Collapse
|
220
|
Faiz F, Hooper AJ, van Bockxmeer FM. Molecular pathology of familial hypercholesterolemia, related dyslipidemias and therapies beyond the statins. Crit Rev Clin Lab Sci 2012; 49:1-17. [PMID: 22214202 DOI: 10.3109/10408363.2011.646942] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The development of the statin class of cholesterol-lowering drugs is one of the most significant success stories of modern pharmacotherapy. World-wide there are an estimated 150 million people on statins, with the emerging economies of India and China predicted to contribute significantly to that number. Notwithstanding their success, a significant number of people cannot tolerate statins because of serious side effects; of equal concern, a substantial proportion of high risk patients fail to reach cholesterol-lowering targets. For these subjects there is an urgent need for new cholesterol-lowering agents to be used alone or in combination with statins. The success of statins has been largely underpinned by knowledge of cholesterol homeostasis at a molecular level, knowledge that was first gleaned in the 1980s from Brown and Goldstein's pioneering studies of familial hypercholesterolemia (FH, OMIM 143890). Follow-up work that has identified a number of intracellular and circulating factors, all capable of disrupting LDL clearance, has revealed that the low-density lipoprotein receptor- (LDLR) mediated clearance pathway is substantially more complex than previously thought. These factors were discovered in studies of individuals with very rare inherited conditions that lead to either hypo- or hypercholesterolemia. These investigations, besides providing clearer insight into the molecular mechanisms regulating plasma LDL concentrations, have also revealed a number of novel therapeutic targets independent from statins. Consequently, a number of novel therapeutic approaches that are based on small interfering bio-molecules, including antisense oligonucleotides, are now in clinical development. These are aimed at impairing the assembly, synthesis and secretion of apolipoprotein B-containing lipoproteins and/or accelerating their hepatic catabolism. The aim of this article is to focus on these recent advances in the understanding of the molecular basis of cholesterol metabolism that should herald novel cholesterol-lowering agents beyond the statins.
Collapse
Affiliation(s)
- Fathimath Faiz
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | | | | |
Collapse
|
221
|
Chan DC, Watts GF. Postprandial lipoprotein metabolism in familial hypercholesterolemia: thinking outside the box. Metabolism 2012; 61:3-11. [PMID: 21945105 DOI: 10.1016/j.metabol.2011.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/14/2011] [Accepted: 07/26/2011] [Indexed: 10/17/2022]
Abstract
Familial hypercholesterolemia (FH) is a dominantly inherited disorder principally due to mutations in the low-density lipoprotein (LDL) receptor that classically cause markedly elevated plasma LDL cholesterol concentrations and premature coronary heart disease (CHD). However, elevated plasma LDL cholesterol alone does not fully account for the increase or variation in risk of CHD. We propose a hypothetical model for the role of postprandial dyslipoproteinemia based on the overproduction and decreased catabolism of triglyceride-rich lipoproteins, which may be a consequence of LDL receptor deficiency. Expression of postprandial dyslipoproteinemia in FH may also depend on the type of pathogenic gene variants and on coexistent conditions, particularly obesity and insulin resistance. Further research is required to investigate our model proposed and to test whether treating postprandial dyslipoproteinemia decreases CHD risk in FH incremental to standard therapy.
Collapse
Affiliation(s)
- Dick C Chan
- Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | | |
Collapse
|
222
|
Palacios L, Grandoso L, Cuevas N, Olano-Martín E, Martinez A, Tejedor D, Stef M. Molecular characterization of familial hypercholesterolemia in Spain. Atherosclerosis 2011; 221:137-42. [PMID: 22244043 DOI: 10.1016/j.atherosclerosis.2011.12.021] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 11/25/2011] [Accepted: 12/16/2011] [Indexed: 11/29/2022]
Abstract
Familial hypercholesterolemia (FH), characterized by isolated elevation of plasmatic low-density lipoprotein (LDL) cholesterol and premature coronary heart disease (CHD), is associated with mutations in three major genes: LDL receptor (LDLR), apolipoprotein B (APOB) and proprotein convertase subtilisin/kexin 9 (PCSK9). We have analyzed 5430 Spanish index cases and 2223 relatives since 2004 with LIPOchip(®) genetic diagnostic platform, a microarray for the detection of Spanish common mutations in these three genes, including copy number variation (CNV) in LDLR, followed by sequencing analysis of the coding regions of LDLR and exon 26 of APOB, when the result is negative. Samples were received from hospitals of all around Spain. The preferred clinical criterion to diagnose FH was Dutch Lipid Clinic Network (DLCN) score. Our results show that there is a broad spectrum of mutations in the LDLR gene in Spain since about 400 different mutations were detected, distributed along almost the whole LDLR gene. Mutations in APOB (mainly p.Arg3527Gln) covered 6.5% of positive cases and only one PCSK9 mutation was detected. We found correlation between more severe mutations and the clinical diagnosis but also that 28% of FH patients harboring mutations do not have a definite clinical diagnosis. This study analyzes the mutation spectrum in Spain, remarks the importance of genetic diagnosis of FH patients, as well as the cascade screening, and shows how it is being carried out in Spain.
Collapse
|
223
|
Sun XM, Patel DD, Acosta JC, Gil J, Soutar AK. Premature senescence in cells from patients with autosomal recessive hypercholesterolemia (ARH): evidence for a role for ARH in mitosis. Arterioscler Thromb Vasc Biol 2011; 31:2270-7. [PMID: 21778424 DOI: 10.1161/atvbaha.111.232223] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The defective gene causing autosomal recessive hypercholesterolemia (ARH) encodes ARH, a clathrin-associated adaptor protein required for low-density-lipoprotein receptor endocytosis in most cells but not in skin fibroblasts. The aim here was to elucidate why ARH fibroblasts grow slowly and undergo premature senescence. METHODS AND RESULTS Knockdown of ARH by RNA interference in IMR90 cells produces the same phenotype, indicated by increased p16 expression, γ-H2AX-positive foci, and enlarged flattened morphology. We showed that ARH contributes to several aspects of mitosis: it localizes to mitotic microtubules, with lamin B1 on the nuclear envelope and spindle matrix, and with clathrin heavy chain on mitotic spindles. Second, ARH is phosphorylated in G(2)/M phase by a roscovitine-sensitive kinase, probably cdc2. Third, cells lacking ARH show disfigured nuclei and defective mitotic spindles. Defects are most marked in ARH W22X cells, where translation starts at Met46, so the protein lacks a phosphorylation site at Ser14, identified by mass spectrometry of wild-type ARH. CONCLUSIONS The ARH protein is involved in cell cycle progression, possibly by affecting nuclear membrane formation through interaction with lamin B1 or other mitotic proteins, and its absence affects cell proliferation and induces premature senescence, which may play a role in the development of atherosclerosis in ARH.
Collapse
Affiliation(s)
- Xi-Ming Sun
- Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, United Kingdom
| | | | | | | | | |
Collapse
|
224
|
Evans D, Aberle J, Beil FU. The relative importance of common and rare genetic variants in the development of hypertriglyceridemia. Expert Rev Cardiovasc Ther 2011; 9:637-44. [PMID: 21615327 DOI: 10.1586/erc.11.53] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Plasma lipid levels are a complex trait with a genetic and an environmental component. There are two models for the genetic basis of complex traits: the common-disease common-variant hypothesis, in which susceptibility is due to variants occurring at relatively high frequency but low effect size; and the common-disease rare-variant hypothesis, where disease is due to multiple rare variants each occurring at low frequency but with high effect size. Genome-wide association studies have identified a number of common variants associated with plasma lipid levels. However, they account for only a proportion of the genetic variance. Resequencing studies are revealing the importance of rare variants in accounting for the missing variance. Next-generation sequencing will allow the relative importance of the two hypotheses to be assessed.
Collapse
Affiliation(s)
- David Evans
- Endokrinologie und Stoffwechsel, Medizinische Klinik III, Zentrum für Innere Medizin, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | |
Collapse
|
225
|
Zhang L, Hou D, Chen X, Li D, Zhu L, Zhang Y, Li J, Bian Z, Liang X, Cai X, Yin Y, Wang C, Zhang T, Zhu D, Zhang D, Xu J, Chen Q, Ba Y, Liu J, Wang Q, Chen J, Wang J, Wang M, Zhang Q, Zhang J, Zen K, Zhang CY. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: evidence of cross-kingdom regulation by microRNA. Cell Res 2011; 22:107-26. [PMID: 21931358 DOI: 10.1038/cr.2011.158] [Citation(s) in RCA: 778] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Our previous studies have demonstrated that stable microRNAs (miRNAs) in mammalian serum and plasma are actively secreted from tissues and cells and can serve as a novel class of biomarkers for diseases, and act as signaling molecules in intercellular communication. Here, we report the surprising finding that exogenous plant miRNAs are present in the sera and tissues of various animals and that these exogenous plant miRNAs are primarily acquired orally, through food intake. MIR168a is abundant in rice and is one of the most highly enriched exogenous plant miRNAs in the sera of Chinese subjects. Functional studies in vitro and in vivo demonstrated that MIR168a could bind to the human/mouse low-density lipoprotein receptor adapter protein 1 (LDLRAP1) mRNA, inhibit LDLRAP1 expression in liver, and consequently decrease LDL removal from mouse plasma. These findings demonstrate that exogenous plant miRNAs in food can regulate the expression of target genes in mammals.
Collapse
Affiliation(s)
- Lin Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Calandra S, Tarugi P, Speedy HE, Dean AF, Bertolini S, Shoulders CC. Mechanisms and genetic determinants regulating sterol absorption, circulating LDL levels, and sterol elimination: implications for classification and disease risk. J Lipid Res 2011; 52:1885-926. [PMID: 21862702 DOI: 10.1194/jlr.r017855] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This review integrates historical biochemical and modern genetic findings that underpin our understanding of the low-density lipoprotein (LDL) dyslipidemias that bear on human disease. These range from life-threatening conditions of infancy through severe coronary heart disease of young adulthood, to indolent disorders of middle- and old-age. We particularly focus on the biological aspects of those gene mutations and variants that impact on sterol absorption and hepatobiliary excretion via specific membrane transporter systems (NPC1L1, ABCG5/8); the incorporation of dietary sterols (MTP) and of de novo synthesized lipids (HMGCR, TRIB1) into apoB-containing lipoproteins (APOB) and their release into the circulation (ANGPTL3, SARA2, SORT1); and receptor-mediated uptake of LDL and of intestinal and hepatic-derived lipoprotein remnants (LDLR, APOB, APOE, LDLRAP1, PCSK9, IDOL). The insights gained from integrating the wealth of genetic data with biological processes have important implications for the classification of clinical and presymptomatic diagnoses of traditional LDL dyslipidemias, sitosterolemia, and newly emerging phenotypes, as well as their management through both nutritional and pharmaceutical means.
Collapse
Affiliation(s)
- Sebastiano Calandra
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | | | |
Collapse
|
227
|
Tada H, Kawashiri MA, Ohtani R, Noguchi T, Nakanishi C, Konno T, Hayashi K, Nohara A, Inazu A, Kobayashi J, Mabuchi H, Yamagishi M. A novel type of familial hypercholesterolemia: double heterozygous mutations in LDL receptor and LDL receptor adaptor protein 1 gene. Atherosclerosis 2011; 219:663-6. [PMID: 21872251 DOI: 10.1016/j.atherosclerosis.2011.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 08/01/2011] [Accepted: 08/01/2011] [Indexed: 11/28/2022]
Abstract
BACKGROUND Autosomal recessive hypercholesterolemia (ARH) is an extremely rare inherited hypercholesterolemia, the cause of which is mutations in low-density lipoprotein (LDL) receptor adaptor protein 1 (LDLRAP1) gene. METHODS A total of 146 heterozygous familial hypercholesterolemic (FH) patients with a mutation in LDLR gene were screened for genes encoding proprotein convertase subtilisin/kexin type 9 (PCSK9) and LDLRAP1. RESULTS Among the 146 subjects, we identified a 79-year-old Japanese female with double mutations in LDLR gene (c.2431A>T) and LDLRAP1 gene (c.606dup). Two other relatives with double mutations in those genes in her family were also identified. Although the proband exhibited massive Achilles tendon xanthoma and coronary and aortic valvular disease, serum LDL-C level of subjects with double mutations was similar with that of subjects with single LDLR mutation (284.0±43.5 versus 265.1 ± 57.4 mg/dl). CONCLUSION Additional mutation in LDLRAP1 may account for severer phenotype in terms of xanthoma and atherosclerotic cardiovascular disease in FH patients.
Collapse
Affiliation(s)
- Hayato Tada
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
McMahon HT, Boucrot E. Molecular mechanism and physiological functions of clathrin-mediated endocytosis. Nat Rev Mol Cell Biol 2011; 12:517-33. [PMID: 21779028 DOI: 10.1038/nrm3151] [Citation(s) in RCA: 1613] [Impact Index Per Article: 115.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
229
|
Abstract
Leonardo da Vinci was born in Italy. Among the researchers and scientists, he is favourably known for his remarkable efforts in scientific work. His investigations of atherosclerosis judiciously combine three separate fields of research. In 1506, he finished his masterpiece, painting of Mona Lisa. A careful clinical examination of the famous painting reveals a yellow irregular leather-like spot at the inner end of the left upper eyelid and a soft bumpy well-defined swelling of the dorsum of the right hand beneath the index finger about 3 cm long. This is probably the first case of familial hypercholesterolemia (FH). The FH code of Leonardo da Vinci was given immense consideration by scientists like Carl Muller, who described the xanthomas tuberosum and angina pectoris. On the contrary, Akira Endo searched for microbial metabolites that would inhibit HMG-CoA reductase, the rate-limiting enzyme in the synthesis of cholesterol and finally, Michael Brown and Joseph Goldstein published a remarkable series of elegant and insightful papers in the 70s and 80s. They established that the cellular uptake of low-density lipoprotein (LDL) essentially requires the LDL receptor. In conclusion: this was the real Code of Leonardo da Vinci.
Collapse
Affiliation(s)
- Leiv Ose
- Lipid Clinic, Medical Department, Rikshospitalet HF, NO 0027 Oslo Norway
| |
Collapse
|
230
|
Sorrentino V, Scheer L, Santos A, Reits E, Bleijlevens B, Zelcer N. Distinct functional domains contribute to degradation of the low density lipoprotein receptor (LDLR) by the E3 ubiquitin ligase inducible Degrader of the LDLR (IDOL). J Biol Chem 2011; 286:30190-9. [PMID: 21734303 DOI: 10.1074/jbc.m111.249557] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We recently identified the liver X receptor-regulated E3 ubiquitin ligase inducible degrader of the LDL receptor (IDOL) as a modulator of lipoprotein metabolism. Acting as an E3 ubiquitin ligase, IDOL triggers ubiquitination and subsequent degradation of the low density lipoprotein receptor (LDLR). We demonstrate here that this outcome requires the conserved FERM and RING domains present in IDOL. The RING domain promotes ubiquitination in vitro and Lys-63-specific ubiquitination of the LDLR in vivo in response to IDOL or liver X receptor activation. We further identify RING residues that differentially influence ubiquitination of the LDLR or stability of IDOL. The FERM domain interacts with the LDLR and in living cells co-localizes with the receptor at the plasma membrane. Homology modeling revealed a phosphotyrosine-binding element embedded in the FERM domain. Mutating residues within this region or residues in the LDLR preceding the NPVY endocytosis motif abrogate LDLR degradation by IDOL. Collectively, our results indicate that both the FERM and RING domains are required for promoting lysosomal degradation of the LDLR by IDOL. Our findings may facilitate development of structure-based IDOL inhibitors aimed at increasing LDLR abundance in therapeutic strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- Vincenzo Sorrentino
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
231
|
Wang K, Edmondson AC, Li M, Gao F, Qasim AN, Devaney JM, Burnett MS, Waterworth DM, Mooser V, Grant SFA, Epstein SE, Reilly MP, Hakonarson H, Rader DJ. Pathway-Wide Association Study Implicates Multiple Sterol Transport and Metabolism Genes in HDL Cholesterol Regulation. Front Genet 2011; 2:41. [PMID: 22303337 PMCID: PMC3268595 DOI: 10.3389/fgene.2011.00041] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 06/21/2011] [Indexed: 12/30/2022] Open
Abstract
Pathway-based association methods have been proposed to be an effective approach in identifying disease genes, when single-marker association tests do not have sufficient power. The analysis of quantitative traits may be benefited from these approaches, by sampling from two extreme tails of the distribution. Here we tested a pathway association approach on a small genome-wide association study (GWAS) on 653 subjects with extremely high high-density lipoprotein cholesterol (HDL-C) levels and 784 subjects with low HDL-C levels. We identified 102 genes in the sterol transport and metabolism pathways that collectively associate with HDL-C levels, and replicated these association signals in an independent GWAS. Interestingly, the pathways include 18 genes implicated in previous GWAS on lipid traits, suggesting that genuine HDL-C genes are highly enriched in these pathways. Additionally, multiple biologically relevant loci in the pathways were not detected by previous GWAS, including genes implicated in previous candidate gene association studies (such as LEPR, APOA2, HDLBP, SOAT2), genes that cause Mendelian forms of lipid disorders (such as DHCR24), and genes expressing dyslipidemia phenotypes in knockout mice (such as SOAT1, PON1). Our study suggests that sampling from two extreme tails of a quantitative trait and examining genetic pathways may yield biological insights from smaller samples than are generally required using single-marker analysis in large-scale GWAS. Our results also implicate that functionally related genes work together to regulate complex quantitative traits, and that future large-scale studies may benefit from pathway-association approaches to identify novel pathways regulating HDL-C levels.
Collapse
Affiliation(s)
- Kai Wang
- Center for Applied Genomics, Children's Hospital of Philadelphia Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Familial hypercholesterolemia: the lipids or the genes? Nutr Metab (Lond) 2011; 8:23. [PMID: 21513517 PMCID: PMC3104361 DOI: 10.1186/1743-7075-8-23] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 04/22/2011] [Indexed: 02/05/2023] Open
Abstract
Familial Hypercholesterolemia (FH) is a common cause of premature cardiovascular disease and is often undiagnosed in young people. Although the disease is diagnosed clinically by high LDL cholesterol levels and family history, to date there are no single internationally accepted criteria for the diagnosis of FH. Several genes have been shown to be involved in FH; yet determining the implications of the different mutations on the phenotype remains a hard task. The polygenetic nature of FH is being enhanced by the discovery of new genes that serve as modifiers. Nevertheless, the picture is still unclear and many unknown genes contributing to the phenotype are most likely involved. Because of this evolving polygenetic nature, the diagnosis of FH by genetic testing is hampered by its cost and effectiveness. In this review, we reconsider the clinical versus genetic nomenclature of FH in the literature. After we describe each of the genetic causes of FH, we summarize the known correlation with phenotypic measures so far for each genetic defect. We then discuss studies from different populations on the genetic and clinical diagnoses of FH to draw helpful conclusions on cost-effectiveness and suggestions for diagnosis.
Collapse
|
233
|
Familial hypercholesterolemias: prevalence, genetics, diagnosis and screening recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol 2011; 5:S9-17. [PMID: 21600530 DOI: 10.1016/j.jacl.2011.03.452] [Citation(s) in RCA: 240] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 03/29/2011] [Indexed: 11/20/2022]
|
234
|
Fahed AC, Safa RM, Haddad FF, Bitar FF, Andary RR, Arabi MT, Azar ST, Nemer G. Homozygous familial hypercholesterolemia in Lebanon: a genotype/phenotype correlation. Mol Genet Metab 2011; 102:181-8. [PMID: 21145767 DOI: 10.1016/j.ymgme.2010.11.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 11/05/2010] [Indexed: 11/28/2022]
Abstract
Familial hypercholesterolemia (FH) is an inherited disease characterized by the deposition of LDL in tissues causing premature atherosclerosis. Many genes are implicated in FH resulting in a large variability in the phenotype. DNA sequencing of the LDLR gene was done for forty patients clinically diagnosed with homozygous FH and forty family members variably affected. Patients underwent noninvasive heart and vascular studies. Statistical and pedigree analyses were used to correlate the different genotypes with the phenotypes. The prevalence of homozygosity at the Lebanese allele (2043C>A) is 45%. However, 27.5% of the patients have no mutations at all in the LDLR gene, and 27.5% are either heterozygous for the 2043C>A mutation, heterozygous for a mutation in another exon of the LDLR gene, or combined heterozygous for two different mutations. We confirm previous reports on the higher prevalence of FH in Lebanon. Our results do, however contradict previous reports on an assumed higher prevalence among the Christian Lebanese. Mutations in the LDLR especially combined heterozygosity can cause a severe phenotype similar to the homozygous mutation in the Lebanese allele. This information is particularly important in targeting the more prevalent heterozygotes in the general population with early diagnosis and intervention.
Collapse
Affiliation(s)
- Akl C Fahed
- Department of Biochemistry, American University of Beirut, Bliss Street, Beirut, Lebanon
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Cao XL, Yin RX, Wu DF, Miao L, Aung LHH, Hu XJ, Li Q, Yan TT, Lin WX, Pan SL. Genetic variant of V825I in the ATP-binding cassette transporter A1 gene and serum lipid levels in the Guangxi Bai Ku Yao and Han populations. Lipids Health Dis 2011; 10:14. [PMID: 21247457 PMCID: PMC3034691 DOI: 10.1186/1476-511x-10-14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 01/19/2011] [Indexed: 12/04/2022] Open
Abstract
Background Several genetic variants in the ATP-binding cassette transporter A1 (ABCA1) gene have associated with modifications of serum high-density lipoprotein cholesterol (HDL-C) levels and the susceptibility for coronary heart disease, but the findings are still controversial in diverse racial/ethnic groups. Bai Ku Yao is an isolated subgroup of the Yao minority in southern China. The present study was undertaken to detect the possible association of V825I (rs2066715) polymorphism in the ABCA1 gene and several environmental factors with serum lipid levels in the Guangxi Bai Ku Yao and Han populations. Methods A total of 677 subjects of Bai Ku Yao and 646 participants of Han Chinese were randomly selected from our previous stratified randomized cluster samples. Polymerase chain reaction and restriction fragment length polymorphism assay combined with gel electrophoresis were performed for the genotyping of V825I variant, and then confirmed by direct sequencing. Results The levels of serum total cholesterol (TC), HDL-C, apolipoprotein (Apo) AI and ApoB were lower in Bai Ku Yao than in Han (P < 0.01 for all). The frequency of G and A alleles was 57.4% and 42.6% in Bai Ku Yao, and 57.7% and 42.3% in Han (P > 0.05); respectively. The frequency of GG, GA and AA genotypes was 33.7%, 47.4% and 18.9% in Bai Ku Yao, and 33.4%, 48.6% and 18.0% in Han (P > 0.05); respectively. There was no difference in the genotypic and allelic frequencies between males and females in the both ethnic groups. The subjects with AA genotype in Bai Ku Yao had higher serum TC levels than the subjects with GG and GA genotypes (P < 0.05). The participants with AA genotype in Han had lower serum HDL-C and ApoAI levels than the participants with GG and GA genotypes (P < 0.05 for each), but these results were found in males but not in females. Multivariate linear regression analysis showed that the levels of TC in Bai Ku Yao and HDL-C and ApoAI in male Han were correlated with genotypes (P < 0.05 for all). Serum lipid parameters were also correlated with sex, age, body mass index, alcohol consumption, and blood pressure in both ethnic groups (P < 0.05-0.001). Conclusion The present study suggests that the V825I polymorphism in the ABCA1 gene is associated with male serum HDL-C and ApoAI levels in the Han, and serum TC levels in the Bai Ku Yao populations. The difference in the association of V825I polymorphism and serum lipid levels between the two ethnic groups might partly result from different ABCA1 gene-enviromental interactions.
Collapse
Affiliation(s)
- Xiao-Li Cao
- Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Marduel M, Carrié A, Sassolas A, Devillers M, Carreau V, Di Filippo M, Erlich D, Abifadel M, Marques-Pinheiro A, Munnich A, Junien C, Boileau C, Varret M, Rabès JP. Molecular spectrum of autosomal dominant hypercholesterolemia in France. Hum Mutat 2010; 31:E1811-24. [PMID: 20809525 PMCID: PMC3152176 DOI: 10.1002/humu.21348] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Autosomal Dominant Hypercholesterolemia (ADH), characterized by isolated elevation of plasmatic LDL cholesterol and premature cardiovascular complications, is associated with mutations in 3 major genes: LDLR (LDL receptor), APOB (apolipoprotein B) and PCSK9(proprotein convertase subtilisin-kexin type 9). Through the French ADH Research Network, we collected molecular data from 1358 French probands from eleven different regions in France.Mutations in the LDLR gene were identified in 1003 subjects representing 391 unique events with 46.0% missense, 14.6% frameshift, 13.6% splice, and 11.3% nonsense mutations, 9.7% major rearrangements, 3.8% small in frame deletions/insertions, and 1.0% UTR mutations. Interestingly,175 are novel mutational events and represent 45% of the unique events we identified, highlighting a specificity of the LDLR mutation spectrum in France. Furthermore, mutations in the APOB gene were identified in 89 probands and in the PCSK9 gene in 10 probands. Comparison of available clinical and biochemical data showed a gradient of severity for ADH-causing mutations:FH=PCSK9>FDB>«Others» genes. The respective contribution of each known gene to ADH inthis French cohort is: LDLR 73.9%, APOB 6.6%, PCSK9 0.7%. Finally, in 19.0% of the probands,no mutation was found, thus underscoring the existence of ADH mutations located in still unknown genes.
Collapse
Affiliation(s)
- Marie Marduel
- Institut National de la Santé et de la Recherche Médicale, U781, 75015, Paris, France; 2 Université Paris Descartes, 75006, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Izar MC, Machado VA, Fonseca FA. Genetic screening for homozygous and heterozygous familial hypercholesterolemia. Appl Clin Genet 2010; 3:147-57. [PMID: 23776359 PMCID: PMC3681171 DOI: 10.2147/tacg.s13490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Familial hypercholesterolemia (FH) is a common inherited disorder that results in premature atherosclerosis. Diagnosis of FH is suspected on the basis of clinical criteria, but confirmation requires genetic testing. In the era of statins, early diagnosis and initiation of treatment can modify disease progression and outcomes. Therefore, cascade screening with a combination of lipid concentration measurements and DNA testing should be used to identify relatives of index cases with a clinical diagnosis of FH. Autosomal dominant FH is related to mutations in the low-density lipoprotein receptor (LDLR), apolipoprotein B-100 (APOB), or proprotein convertase subtilisin/kexin type 9 (PCSK9) genes. Genetic screening of the LDLR gene is challenging to achieve at a feasible cost, especially in people who do not have a founder effect. Nucleotide sequencing of all exons and flanking splicing regions in combination with multiplex ligation probe amplification to detect large insertions or deletions is considered the gold-standard approach to screen for LDLR mutations. Alternatively, the cDNA can be sequenced; however, this procedure is not suitable for use in large populations, because of the need of RNA extraction. Multiplex analysis can be appropriate for population with founder effects or a low number of different mutations. Finally, there are many techniques for a mutation scanning approach, which have some benefits over sequencing, and also with the potential for detecting known and novel mutations. Familial defective Apo B is amenable to genetic diagnosis by screening for a few mutations. Recently, gain-of-function mutations in PCSK9 gene have been demonstrated to cause FH phenotype. Strategies for population screening, cost-effectiveness of genetic screening, ethical aspects, and insurance policies are discussed and need implementation worldwide.
Collapse
Affiliation(s)
- Maria C Izar
- Cardiology Division, Department of Medicine, Federal University of São Paulo, UNIFESP, São Paulo, SP, Brazil
| | - Valéria A Machado
- Cardiology Division, Department of Medicine, Federal University of São Paulo, UNIFESP, São Paulo, SP, Brazil
| | - Francisco A Fonseca
- Cardiology Division, Department of Medicine, Federal University of São Paulo, UNIFESP, São Paulo, SP, Brazil
| |
Collapse
|
238
|
|
239
|
Abstract
Coronary artery disease and its clinical manifestations, including myocardial infarction, are heritable traits, consistent with a role for inherited DNA sequence variation in conferring risk for disease. Knowledge of the new sequence variations in the genome that confer risk has the potential to illuminate new causal biologic pathways in humans and to thereby further improve diagnosis and treatment. Here, we review recent progress in mapping genetic loci related to coronary disease and risk factor phenotypes, including plasma lipoprotein concentrations. Genome-wide linkage (in families) and association (in populations) studies have identified more than a dozen genetic loci related to coronary disease. A key challenge now is to move from mapping loci to pinpointing causal genes and variants, and to develop a molecular understanding of how these genes lead to coronary disease.
Collapse
Affiliation(s)
- Kiran Musunuru
- Center for Human Genetic Research and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02108, USA
| | | |
Collapse
|
240
|
Clinical Implications of Lipid Genetics for Cardiovascular Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2010; 4:461-468. [PMID: 21853159 DOI: 10.1007/s12170-010-0131-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in the developed world. Epidemiologic data support a strong relationship of atherosclerotic cardiovascular disease (ASCVD) with both elevated low-density lipoprotein cholesterol (LDL-C), and reduced high-density lipoprotein cholesterol (HDL-C). The study of the human genetics of plasma lipid traits, both rare Mendelian disorders as well as common variants, has illuminated multiple genes and pathways involved in the regulation of LDL-C and HDL-C levels. Mendelian disorders of extremes of LDL-C and Mendelian randomization studies of common gene variants associated with LDL-C strongly support a causal relationship between LDL-C and ASCVD, independent of mechanism. In contrast, Mendelian disorders of extremes of HDL-C and Mendelian randomization studies of common genetic variants for HDL-C are inconsistent in their support of a causal relationship between HDL-C and ASCVD. In contrast to LDL-C, a causal relationship between HDL-C and ASCVD may be dependent on the specific mechanism leading to variation in HDL-C levels.
Collapse
|
241
|
Rios J, Stein E, Shendure J, Hobbs HH, Cohen JC. Identification by whole-genome resequencing of gene defect responsible for severe hypercholesterolemia. Hum Mol Genet 2010; 19:4313-8. [PMID: 20719861 PMCID: PMC2957323 DOI: 10.1093/hmg/ddq352] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Whole-genome sequencing is a potentially powerful tool for the diagnosis of genetic diseases. Here, we used sequencing-by-ligation to sequence the genome of an 11-month-old breast-fed girl with xanthomas and very high plasma cholesterol levels (1023 mg/dl). Her parents had normal plasma cholesterol levels and reported no family history of hypercholesterolemia, suggesting either an autosomal recessive disorder or a de novo mutation. Known genetic causes of severe hypercholesterolemia were ruled out by sequencing the responsible genes (LDLRAP, LDLR, PCSK9, APOE and APOB), and sitosterolemia was ruled out by documenting a normal plasma sitosterol:cholesterol ratio. Sequencing revealed 3 797 207 deviations from the reference sequence, of which 9726 were nonsynonymous single-nucleotide substitutions. A total of 9027 of the nonsynonymous substitutions were present in dbSNP or in 21 additional individuals from whom complete exonic sequences were available. The 699 novel nonsynonymous substitutions were distributed among 604 genes, 23 of which were single-copy genes that each contained 2 nonsynonymous substitutions consistent with an autosomal recessive model. One gene, ABCG5, had two nonsense mutations (Q16X and R446X). This finding indicated that the infant has sitosterolemia. Thus, whole-genome sequencing led to the diagnosis of a known disease with an atypical presentation. Diagnosis was confirmed by the finding of severe sitosterolemia in a blood sample obtained after the infant had been weaned. These findings demonstrate that whole-genome (or exome) sequencing can be a valuable aid to diagnose genetic diseases, even in individual patients.
Collapse
Affiliation(s)
- Jonathan Rios
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas,TX 75390, USA
| | | | | | | | | |
Collapse
|
242
|
Marques-Pinheiro A, Marduel M, Rabès JP, Devillers M, Villéger L, Allard D, Weissenbach J, Guerin M, Zair Y, Erlich D, Junien C, Munnich A, Krempf M, Abifadel M, Jaïs JP, Boileau C, Varret M. A fourth locus for autosomal dominant hypercholesterolemia maps at 16q22.1. Eur J Hum Genet 2010; 18:1236-42. [PMID: 20571503 DOI: 10.1038/ejhg.2010.94] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Autosomal dominant hypercholesterolemia (ADH) is characterized by isolated increase in plasmatic low-density lipoprotein (LDL) cholesterol levels associated with high risk of premature cardiovascular disease. Mutations in LDLR, APOB, and PCSK9 genes have been shown to cause ADH. We now report further genetic heterogeneity of ADH through the study of a large French family in which the involvement of these three genes was excluded. A genome-wide scan mapped the disease-causing gene, named HCHOLA4, at 16q22.1 in a 7.89-Mb interval containing 154 genes with a maximum LOD score of 3.9. To reduce the linked region, we genotyped 18 smaller non-LDLR/non-APOB/non-PCSK9-ADH families at the HCHOLA4 locus. Six families did not exclude linkage to the locus, but none allowed reduction of the disease interval. The 154 regional genes were sorted according to the function of the encoded protein and tissue expression profiles, and 57 genes were analyzed through sequencing of their coding region and close flanking intronic parts. No disease-causing mutation was identified in these families, particularly in the LCAT gene. Finally, our results also show the existence of other ADH genes as nine families were neither linked to LDLR, APOB, and PCSK9 genes nor to the new HCHOLA4 locus.
Collapse
|
243
|
Hong C, Duit S, Jalonen P, Out R, Scheer L, Sorrentino V, Boyadjian R, Rodenburg KW, Foley E, Korhonen L, Lindholm D, Nimpf J, van Berkel TJC, Tontonoz P, Zelcer N. The E3 ubiquitin ligase IDOL induces the degradation of the low density lipoprotein receptor family members VLDLR and ApoER2. J Biol Chem 2010; 285:19720-6. [PMID: 20427281 PMCID: PMC2888382 DOI: 10.1074/jbc.m110.123729] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have previously identified the E3 ubiquitin ligase-inducible degrader of the low density lipoprotein receptor (LDLR) (Idol) as a post-translational modulator of LDLR levels. Idol is a direct target for regulation by liver X receptors (LXRs), and its expression is responsive to cellular sterol status independent of the sterol-response element-binding proteins. Here we demonstrate that Idol also targets two closely related LDLR family members, VLDLR and ApoE receptor 2 (ApoER2), proteins implicated in both neuronal development and lipid metabolism. Idol triggers ubiquitination of the VLDLR and ApoER2 on their cytoplasmic tails, leading to their degradation. We further show that the level of endogenous VLDLR is sensitive to cellular sterol content, Idol expression, and activation of the LXR pathway. Pharmacological activation of the LXR pathway in mice leads to increased Idol expression and to decreased Vldlr levels in vivo. Finally, we establish an unexpected functional link between LXR and Reelin signaling. We demonstrate that LXR activation results in decreased Reelin binding to VLDLR and reduced Dab1 phosphorylation. The identification of VLDLR and ApoER2 as Idol targets suggests potential roles for this LXR-inducible E3 ligase in the central nervous system in addition to lipid metabolism.
Collapse
Affiliation(s)
- Cynthia Hong
- Department of Pathology and Laboratory Medicine and the Howard Hughes Medical Institute, University of California, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Marat AL, McPherson PS. The connecdenn family, Rab35 guanine nucleotide exchange factors interfacing with the clathrin machinery. J Biol Chem 2010; 285:10627-37. [PMID: 20154091 PMCID: PMC2856271 DOI: 10.1074/jbc.m109.050930] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 12/15/2009] [Indexed: 11/06/2022] Open
Abstract
Rabs constitute the largest family of monomeric GTPases, yet for the majority of Rabs relatively little is known about their activation and recruitment to vesicle-trafficking pathways. We recently identified connecdenn (DENND1A), which contains an N-terminal DENN (differentially expressed in neoplastic versus normal cells) domain, a common and evolutionarily ancient protein module. Through its DENN domain, connecdenn functions enzymatically as a guanine-nucleotide exchange factor (GEF) for Rab35. Here we identify two additional connecdenn family members and demonstrate that all connecdenns function as Rab35 GEFs, albeit with different levels of activity. The DENN domain of connecdenn 1 and 2 binds Rab35, whereas connecdenn 3 does not, indicating that Rab35 binding and activation are separable functions. Through their highly divergent C termini, each of the connecdenns binds to clathrin and to the clathrin adaptor AP-2. Interestingly, all three connecdenns use different mechanisms to bind AP-2. Characterization of connecdenn 2 reveals binding to the beta2-ear of AP-2 on a site that overlaps with that used by the autosomal recessive hypercholesterolemia protein and betaarrestin, although the sequence used by connecdenn 2 is unique. Loss of connecdenn 2 function through small interference RNA knockdown results in an enlargement of early endosomes, similar to what is observed upon loss of Rab35 activity. Our studies reveal connecdenn DENN domains as generalized GEFs for Rab35 and identify a new AP-2-binding motif, demonstrating a complex link between the clathrin machinery and Rab35 activation.
Collapse
Affiliation(s)
- Andrea L. Marat
- From the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Peter S. McPherson
- From the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
245
|
Mettlen M, Loerke D, Yarar D, Danuser G, Schmid SL. Cargo- and adaptor-specific mechanisms regulate clathrin-mediated endocytosis. ACTA ACUST UNITED AC 2010; 188:919-33. [PMID: 20231386 PMCID: PMC2845073 DOI: 10.1083/jcb.200908078] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Clathrin-coated pit size and dynamic behavior varies with low density lipoprotein receptor (LDLR) expression levels in a manner dependent on the LDLR-specific adaptors, Dab2 and ARH. Clathrin-mediated endocytosis of surface receptors and their bound ligands (i.e., cargo) is highly regulated, including by the cargo itself. One of the possible sources of the observed heterogeneous dynamics of clathrin-coated pits (CCPs) might be the different cargo content. Consistent with this, we show that CCP size and dynamic behavior varies with low density lipoprotein receptor (LDLR) expression levels in a manner dependent on the LDLR-specific adaptors, Dab2 and ARH. In Dab2-mCherry–expressing cells, varying LDLR expression leads to a progressive increase in CCP size and to the appearance of nonterminal endocytic events. In LDLR and ARH-mCherry–expressing cells in addition to an increase in CCP size, turnover of abortive CCPs increases, and the rate of CCP maturation decreases. Altogether, our results underscore the highly dynamic and cargo-responsive nature of CCP assembly and suggest that the observed heterogeneity is, in part, related to compositional differences (e.g., cargo and adaptors) between CCPs.
Collapse
Affiliation(s)
- Marcel Mettlen
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
246
|
Pedersen GA, Chakraborty S, Steinhauser AL, Traub LM, Madsen M. AMN directs endocytosis of the intrinsic factor-vitamin B(12) receptor cubam by engaging ARH or Dab2. Traffic 2010; 11:706-20. [PMID: 20088845 DOI: 10.1111/j.1600-0854.2010.01042.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cubam is a multi-ligand receptor involved in dietary uptake of intrinsic factor-vitamin B(12) in the small intestine and reabsorption of various low-molecular-weight proteins (such as albumin, transferrin, apolipoprotein A-I and vitamin D-binding protein) in the kidney. Cubam is composed of two proteins: cubilin and amnionless. Cubilin harbors ligand binding capabilities, while amnionless provides membrane anchorage and potential endocytic capacity via two FXNPXF signals within the cytosolic domain. These signals are similar to the FXNPXY signals found in members of the low-density lipoprotein receptor superfamily, which associate with clathrin-associated sorting proteins, including Disabled-2 (Dab2) and autosomal recessive hypercholesterolemia (ARH), during endocytosis. We therefore investigated the functionality of each amnionless FXNPXF signal and their respective interaction with sorting proteins. By sequential mutation and expression of a panel of amnionless mutants combined with yeast two-hybrid analyses, we demonstrate that the signals are functionally redundant and both are able to mediate endocytosis of cubam through interaction with Dab2 and ARH.
Collapse
Affiliation(s)
- Gitte Albinus Pedersen
- Department of Medical Biochemistry, University of Aarhus, Ole Worms Allé, Bldg. 1170-1171, Aarhus C, Denmark
| | | | | | | | | |
Collapse
|
247
|
Harada K, Miyamoto Y, Morisaki H, Ohta N, Yamanaka I, Kokubo Y, Makino H, Harada-Shiba M, Okayama A, Tomoike H, Tomonori O, Saito Y, Yoshimasa Y, Morisaki T. A Novel Thr56Met Mutation of the Autosomal Recessive Hypercholesterolemia Gene Associated with Hypercholesterolemia. J Atheroscler Thromb 2010; 17:131-40. [DOI: 10.5551/jat.2873] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
248
|
Soutar AK. Regulation of the LDL receptor in familial hypercholesterolemia. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/clp.09.60] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
249
|
Mousavi SA, Berge KE, Leren TP. The unique role of proprotein convertase subtilisin/kexin 9 in cholesterol homeostasis. J Intern Med 2009; 266:507-19. [PMID: 19930098 DOI: 10.1111/j.1365-2796.2009.02167.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The LDL receptor (LDLR) plays an essential role in the regulation of plasma (LDL) cholesterol concentrations by virtue of its ability to clear plasma LDL. Down-regulation of the LDLR by proprotein convertase subtilisin/kexin 9 (PCSK9) has recently emerged as a regulatory mechanism that controls plasma LDL cholesterol concentrations. Studies in which PCSK9 is over-expressed in mice, have demonstrated that PCSK9, by enhancing hepatic LDLR degradation, decreases the availability of the LDLR for LDL uptake, resulting in increased plasma LDL cholesterol levels. However, PCSK9 has also recently been shown to mediate down-regulation of surface receptors other than the LDLR, suggesting that it may have much broader roles than initially thought.
Collapse
Affiliation(s)
- S A Mousavi
- Medical Genetics Laboratory, Department of Medical Genetics, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | | |
Collapse
|
250
|
Fang L, Garuti R, Kim BY, Wade JB, Welling PA. The ARH adaptor protein regulates endocytosis of the ROMK potassium secretory channel in mouse kidney. J Clin Invest 2009; 119:3278-89. [PMID: 19841541 DOI: 10.1172/jci37950] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Accepted: 08/26/2009] [Indexed: 11/17/2022] Open
Abstract
Renal outer medullary potassium (ROMK) channels are exquisitely regulated to adjust renal potassium excretion and maintain potassium balance. Clathrin-dependent endocytosis plays a critical role, limiting urinary potassium loss in potassium deficiency. In renal disease, aberrant ROMK endocytosis may contribute to potassium retention and hyperkalemia. Previous work has indicated that ROMK endocytosis is stimulated by with-no-lysine (WNK) kinases, but the endocytotic signal and the internalization machinery have not been defined. Here, we found that ROMK bound directly to the clathrin adaptor molecule autosomal recessive hypercholesterolemia (ARH), and this interaction was mediated by what we believe to be a novel variant of the canonical "NPXY" endocytotic signal, YxNPxFV. ARH recruits ROMK to clathrin-coated pits for constitutive and WNK1-stimuated endocytosis, and ARH knockdown decreased basal rates of ROMK endocytosis, in a heterologous expression system, COS-7 cells. We found that ARH was predominantly expressed in the distal nephron where it coimmunoprecipitated and colocalized with ROMK. In mice, the abundance of kidney ARH protein was modulated by dietary potassium and inversely correlated with changes in ROMK. Furthermore, ARH-knockout mice exhibited an altered ROMK response to potassium intake. These data suggest that ARH marks ROMK for clathrin-dependent endocytosis, in concert with the demands of potassium homeostasis.
Collapse
Affiliation(s)
- Liang Fang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | |
Collapse
|