1
|
Hartz J. Low LDL-C: Is It all Good News? Curr Atheroscler Rep 2024; 26:673-681. [PMID: 39254830 DOI: 10.1007/s11883-024-01238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE OF REVIEW This review presents the risks and benefits of very low LDL cholesterol and the safety of using lipid-lowering therapy to achieve these levels. RECENT FINDINGS A growing body of literature suggests that lower LDL cholesterol levels are associated with a reduced risk of cardiovascular disease. Further, achieving these levels with pharmaceuticals is remarkably safe. Although statins may slightly increase the risk of diabetes mellitus and hemorrhagic stroke, the benefits outweigh the risks. While recommendations from professional societies are increasingly aggressive, additional risk reduction could be achieved by setting more even ambitious LDL cholesterol goals.
Collapse
Affiliation(s)
- Jacob Hartz
- Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
2
|
Wargny M, Goronflot T, Rimbert A, Boursier J, Kab S, Henny J, Lainé A, Leux C, Smati S, Hadjadj S, Le May C, Goldberg M, Zins M, Cariou B. Primary hypocholesterolemia is associated with an increased risk of hepatic complications in the general population. J Hepatol 2024; 80:846-857. [PMID: 38331324 DOI: 10.1016/j.jhep.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 01/22/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND & AIMS Beyond cardiovascular disease protection, the health consequences of very low concentrations of low-density lipoprotein-cholesterol (LDL-C) remain a matter of debate. In primary hypobetalipoproteinemia (HBL), liver steatosis and cirrhosis have occasionally been reported. Here, we aimed to investigate the association between HBL and the risk of hepatic complications (cirrhosis complications and/or primary liver cancer) in the general population. METHODS A cohort study was conducted in the French population-based cohort CONSTANCES. Participants with primary HBL (LDL-C <5th percentile for age and sex, [HBL]) were compared with those with normal LDL-C concentrations (40th-60th percentile, [Control]). Participants on lipid-lowering therapies were excluded. For hepatic complications, follow-up events were compared by calculating the incidence density ratio (IDR). The same analyses were replicated in the UK Biobank (UKBB) cohort. RESULTS In the CONSTANCES and UKBB cohorts, 34,653 and 94,666 patients were analyzed, with median ages of 45 and 56 years, mean LDL-C concentrations (HBL vs. control) of 71 vs. 128 mg/dl and 86 vs. 142 mg/dl, and mean follow-up durations of 5.0 and 11.5 years, respectively. The HBL group presented a higher incidence of hepatic complications than the control group: 0.32/ vs. 0.07/1,000 person-years (IDR = 4.50, 95% CI 1.91-10.6) in CONSTANCES, and 0.69/ vs. 0.21/1,000 person-years (IDR = 3.27, 95% CI 2.63-4.06) in the UKBB. This risk proved to be independent of classic risk factors for liver disease (obesity, alcohol consumption, diabetes, viral hepatitis), including in a 5-year landmark analysis excluding early events. Sensitivity analyses based on apoliprotein-B levels (instead of LDL-C levels) or genetically defined HBL showed similar results. CONCLUSIONS HBL is associated with a markedly increased risk of hepatic complications. HBL must be considered as a substantial independent risk factor for liver diseases which justifies specific prevention and screening. IMPACT AND IMPLICATIONS Hypobetalipoproteinemia (HBL) is a lipid disorder characterized by permanent, inherited low levels (below the 5th percentile) of low-density lipoprotein-cholesterol. While HBL is associated with a lower risk of cardiovascular events, some studies suggest that it may be associated with a potential risk of hepatic steatosis and hepatic complications. Here, we studied the association between HBL and hepatic complications (defined as cirrhosis complications and/or primary liver cancer) in two populations of several hundred thousand people, both in France (CONSTANCES cohort) and the United Kingdom (UKBB). The results show that HBL is associated with a significant and independent excess risk of hepatic complications, including primary liver cancer. Thus, in people with HBL, the value of regular liver monitoring must be studied.
Collapse
Affiliation(s)
- Matthieu Wargny
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France; Nantes Université, CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, INSERM, CIC 1413, F-44000 Nantes, France
| | - Thomas Goronflot
- Nantes Université, CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, INSERM, CIC 1413, F-44000 Nantes, France
| | - Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France
| | - Jérôme Boursier
- Service d'Hépato-Gastroentérologie et Oncologie Digestive, Hôpital Universitaire d'Angers, Angers, France; Laboratoire HIFIH UPRES EA3859, SFR ICAT 4208, Université d'Angers, Angers, France
| | - Sofiane Kab
- Université Paris Cité, Paris Saclay University, UVSQ, Inserm UMS 011, Villejuif, France
| | - Joseph Henny
- Université Paris Cité, Paris Saclay University, UVSQ, Inserm UMS 011, Villejuif, France
| | - Antoine Lainé
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France
| | - Christophe Leux
- Nantes Université, CHU Nantes, Service d'information médicale, F-44000 Nantes, France
| | - Sarra Smati
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France
| | - Samy Hadjadj
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France
| | - Cédric Le May
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France
| | - Marcel Goldberg
- Université Paris Cité, Paris Saclay University, UVSQ, Inserm UMS 011, Villejuif, France
| | - Marie Zins
- Université Paris Cité, Paris Saclay University, UVSQ, Inserm UMS 011, Villejuif, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France.
| |
Collapse
|
3
|
Bremova-Ertl T, Hofmann J, Stucki J, Vossenkaul A, Gautschi M. Inborn Errors of Metabolism with Ataxia: Current and Future Treatment Options. Cells 2023; 12:2314. [PMID: 37759536 PMCID: PMC10527548 DOI: 10.3390/cells12182314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
A number of hereditary ataxias are caused by inborn errors of metabolism (IEM), most of which are highly heterogeneous in their clinical presentation. Prompt diagnosis is important because disease-specific therapies may be available. In this review, we offer a comprehensive overview of metabolic ataxias summarized by disease, highlighting novel clinical trials and emerging therapies with a particular emphasis on first-in-human gene therapies. We present disease-specific treatments if they exist and review the current evidence for symptomatic treatments of these highly heterogeneous diseases (where cerebellar ataxia is part of their phenotype) that aim to improve the disease burden and enhance quality of life. In general, a multimodal and holistic approach to the treatment of cerebellar ataxia, irrespective of etiology, is necessary to offer the best medical care. Physical therapy and speech and occupational therapy are obligatory. Genetic counseling is essential for making informed decisions about family planning.
Collapse
Affiliation(s)
- Tatiana Bremova-Ertl
- Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland; (J.H.); (J.S.)
- Center for Rare Diseases, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland
| | - Jan Hofmann
- Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland; (J.H.); (J.S.)
| | - Janine Stucki
- Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland; (J.H.); (J.S.)
| | - Anja Vossenkaul
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (A.V.); (M.G.)
| | - Matthias Gautschi
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (A.V.); (M.G.)
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
4
|
Validation of Knock-Out Caco-2 TC7 Cells as Models of Enterocytes of Patients with Familial Genetic Hypobetalipoproteinemias. Nutrients 2023; 15:nu15030505. [PMID: 36771214 PMCID: PMC9921550 DOI: 10.3390/nu15030505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/21/2023] Open
Abstract
Abetalipoproteinemia (FHBL-SD1) and chylomicron retention disease (FHBL-SD3) are rare recessive disorders of lipoprotein metabolism due to mutations in MTTP and SAR1B genes, respectively, which lead to defective chylomicron formation and secretion. This results in lipid and fat-soluble vitamin malabsorption, which induces severe neuro-ophthalmic complications. Currently, treatment combines a low-fat diet with high-dose vitamin A and E supplementation but still fails in normalizing serum vitamin E levels and providing complete ophthalmic protection. To explore these persistent complications, we developed two knock-out cell models of FHBL-SD1 and FHBL-SD3 using the CRISPR/Cas9 technique in Caco-2/TC7 cells. DNA sequencing, RNA quantification and Western blotting confirmed the introduction of mutations with protein knock-out in four clones associated with i) impaired lipid droplet formation and ii) defective triglyceride (-57.0 ± 2.6% to -83.9 ± 1.6%) and cholesterol (-35.3 ± 4.4% to -60.6 ± 3.5%) secretion. A significant decrease in α-tocopherol secretion was also observed in these clones (-41.5 ± 3.7% to -97.2 ± 2.8%), even with the pharmaceutical forms of vitamin E: tocopherol-acetate and tocofersolan (α-tocopheryl polyethylene glycol succinate 1000). MTTP silencing led to a more severe phenotype than SAR1B silencing, which is consistent with clinical observations. Our cellular models thus provide an efficient tool to experiment with therapeutic strategies and will allow progress in understanding the mechanisms involved in lipid metabolism.
Collapse
|
5
|
Rodríguez de Vera-Gómez P, del Pino-Bellido P, García-González JJ, Sánchez-Jiménez F, Oliva-Rodríguez R, Arrobas-Velilla T, Martínez-Brocca MA. Novel APOB nonsense variant related to familial hypobetalipoproteinemia and hepatic steatosis: A case report and review. J Clin Lipidol 2022; 16:601-607. [DOI: 10.1016/j.jacl.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/29/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022]
|
6
|
A new phenotypic classification system for dyslipidemias based on the standard lipid panel. Lipids Health Dis 2021; 20:170. [PMID: 34838008 PMCID: PMC8627634 DOI: 10.1186/s12944-021-01585-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
Background Dyslipoproteinemias can be classified by their distinct lipoprotein patterns, which helps determine atherosclerotic cardiovascular disease (ASCVD) risk and directs lipid management but this has required advanced laboratory testing. Objective To develop a new algorithm for classifying lipoprotein disorders that only relies on the standard lipid panel. Methods Lipid thresholds for defining the different lipoprotein phenotypes were derived for Non-High-Density Lipoprotein-Cholesterol (NonHDL-C) and Triglycerides (TG) to be concordant when possible with the current US Multi-Society guidelines for blood cholesterol management. Results The new classification method categorizes patients into all the classical Fredrickson-like phenotypes except for Type III dysbetalipoproteinemia. In addition, a new hypolipidemic phenotype (Type VI) due to genetic mutations in apoB-metabolism is described. The validity of the new algorithm was confirmed by lipid analysis by NMR (N = 11,365) and by concordance with classification by agarose gel electrophoresis/beta-quantification (N = 5504). Furthermore, based on the Atherosclerosis Risk in Communities (ARIC) cohort (N = 14,742), the lipoprotein phenotypes differ in their association with ASCVD (TypeV>IIb > IVb > IIa > IVa > normolipidemic) and can be used prognostically as risk enhancer conditions in the management of patients. Conclusions We describe a clinically useful lipoprotein phenotyping system that is only dependent upon the standard lipid panel. It, therefore, can be easily implemented for increasing compliance with current guidelines and for improving the care of patients at risk for ASCVD. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01585-8. A new algorithm is described for categorizing dyslipidemic patients into Fredrickson-like lipoprotein phenotypes except for Type III. The new lipoprotein phenotypes were validated by NMR-lipoprotein analysis and by agarose gel electrophoresis/beta-quantification in a large number of subjects. The new lipoprotein phenotyping system identifies high-risk cardiovascular patients and helps direct clinical management. A major advance is that the new lipoprotein phenotypes are based on just the standard lipid panel, and thus can be automatically calculated by the clinical laboratory and widely implemented.
Collapse
|
7
|
Domenech M, Llano-Rivas I, Arroyo V, Ortega E. Novel APOB mutation in familial hypobetalipoproteinemia. J Clin Lipidol 2021; 16:28-32. [PMID: 34852964 DOI: 10.1016/j.jacl.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Affiliation(s)
- M Domenech
- Lipid and Vascular Risk Unit, Endocrinology and Nutrition Department, Hospital Clinic of Barcelona, Spain; Faculty of Medicine and Health Sciences. University of Barcelona. Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN). Institute of Health Carlos III, ISCIII. Spain
| | - Isabel Llano-Rivas
- Clinical Genetics, Genetic Service. Hospital Universitario Cruces, Basque Country, Spain
| | - Vicente Arroyo
- EF Clif, EASL-CLIF Consortium and Grifols Chair, Barcelona, Spain
| | - Emilio Ortega
- Lipid and Vascular Risk Unit, Endocrinology and Nutrition Department, Hospital Clinic of Barcelona, Spain; Faculty of Medicine and Health Sciences. University of Barcelona. Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN). Institute of Health Carlos III, ISCIII. Spain.
| |
Collapse
|
8
|
Ayoub C, Azar Y, Abou-Khalil Y, Ghaleb Y, Elbitar S, Halaby G, Jambart S, Gannagé-Yared MH, Yaghi C, Saade Riachy C, El Khoury R, Rabès JP, Varret M, Boileau C, El Khoury P, Abifadel M. Identification of a Variant in APOB Gene as a Major Cause of Hypobetalipoproteinemia in Lebanese Families. Metabolites 2021; 11:564. [PMID: 34564380 PMCID: PMC8469161 DOI: 10.3390/metabo11090564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/03/2022] Open
Abstract
Familial hypobetalipoproteinemia (FHBL) is a codominant genetic disorder characterized by reduced plasma levels of low-density lipoprotein cholesterol and apolipoprotein B. To our knowledge, no study on FHBL in Lebanon and the Middle East region has been reported. Therefore, we conducted genetic studies in unrelated families and probands of Lebanese origin presenting with FHBL, in order to identify the causes of this disease. We found that 71% of the recruited probands and their affected relatives were heterozygous for the p.(Arg490Trp) variant in the APOB gene. Haplotype analysis showed that these patients presented the same mutant haplotype. Moreover, there was a decrease in plasma levels of PCSK9 in affected individuals compared to the non-affected and a significant positive correlation between circulating PCSK9 and ApoB levels in all studied probands and their family members. Some of the p.(Arg490Trp) carriers suffered from diabetes, hepatic steatosis or neurological problems. In conclusion, the p.(Arg490Trp) pathogenic variant seems a cause of FHBL in patients from Lebanese origin, accounting for approximately 70% of the probands with FHBL presumably as a result of a founder mutation in Lebanon. This study is crucial to guide the early diagnosis, management and prevention of the associated complications of this disease.
Collapse
Affiliation(s)
- Carine Ayoub
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
| | - Yara Azar
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
- Centre Hospitalo-Universitaire Xavier Bichat, Université de Paris, F-75018 Paris, France
| | - Yara Abou-Khalil
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
- Centre Hospitalo-Universitaire Xavier Bichat, Université de Paris, F-75018 Paris, France
| | - Youmna Ghaleb
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
| | - Sandy Elbitar
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
| | - Georges Halaby
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
| | - Selim Jambart
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
| | - Marie-Hélène Gannagé-Yared
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Hotel Dieu de France of Beirut University Hospital, Beirut 166830, Lebanon
| | - Cesar Yaghi
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Hotel Dieu de France of Beirut University Hospital, Beirut 166830, Lebanon
| | - Carole Saade Riachy
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
| | - Ralph El Khoury
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
| | - Jean-Pierre Rabès
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
- Biochemistry and Molecular Genetics Laboratory, AP-HP, Université Paris-Saclay, Ambroise Paré Hospital, Boulogne Billancourt, UVSQ, UFR Simone Veil-Santé, F-78180 Montigny-Le-Bretonneux, France
| | - Mathilde Varret
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
- Centre Hospitalo-Universitaire Xavier Bichat, Université de Paris, F-75018 Paris, France
| | - Catherine Boileau
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
- Centre Hospitalo-Universitaire Xavier Bichat, Université de Paris, F-75018 Paris, France
- Genetics Department, AP-HP, Bichat Hospital, F-75018 Paris, France
| | - Petra El Khoury
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
| | - Marianne Abifadel
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut 17-5208, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Bichat Hospital, F-75018 Paris, France
| |
Collapse
|
9
|
Yang YX, Li P, Wang P, Zhu BT. 17β-Estradiol-Induced Conformational Changes of Human Microsomal Triglyceride Transfer Protein: A Computational Molecular Modelling Study. Cells 2021; 10:cells10071566. [PMID: 34206252 PMCID: PMC8304645 DOI: 10.3390/cells10071566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/19/2021] [Accepted: 05/05/2021] [Indexed: 11/23/2022] Open
Abstract
Human microsomal triglyceride transfer protein (hMTP) plays an essential role in the assembly of apoB-containing lipoproteins, and has become an important drug target for the treatment of several disease states, such as abetalipoproteinemia, fat malabsorption and familial hypercholesterolemia. hMTP is a heterodimer composed of a larger hMTPα subunit and a smaller hMTPβ subunit (namely, protein disulfide isomerase, hPDI). hPDI can interact with 17β-estradiol (E2), an endogenous female sex hormone. It has been reported that E2 can significantly reduce the blood levels of low-density lipoprotein, cholesterol and triglyceride, and modulate liver lipid metabolism in vivo. However, some of the estrogen’s actions on lipid metabolism are not associated with estrogen receptors (ER), and the exact mechanism underlying estrogen’s ER-independent lipid-modulating action is still not clear at present. In this study, the potential influence of E2 on the stability of the hMTP complex is investigated by jointly using multiple molecular dynamics analyses based on available experimental structures. The molecular dynamics analyses indicate that the hMTP complex in the presence of E2 has reduced interface contacts and surface areas. A steered molecular dynamics analysis shows that the forces required to separate the two subunits (namely, hPDI and hMTPα subunit) of the hMTP complex in the absence of E2 are significantly higher than the forces required to separate the complex in which its hPDI is already bound with E2. E2 makes the interface between hMTPα and hPDI subunits more flexible and less stable. The results of this study suggest that E2-induced conformational changes of the hMTP complex might be a novel mechanism partly accounting for the ER-independent lipid-modulating effect of E2.
Collapse
Affiliation(s)
- Yong-Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; (Y.-X.Y.); (P.L.); (P.W.)
| | - Peng Li
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; (Y.-X.Y.); (P.L.); (P.W.)
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; (Y.-X.Y.); (P.L.); (P.W.)
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Bao-Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; (Y.-X.Y.); (P.L.); (P.W.)
- Shenzhen Bay Laboratory, Shenzhen 518055, China
- Correspondence: ; Tel.: +86-755-84273851
| |
Collapse
|
10
|
Martínez-Hervás S, Real-Collado JT, Ascaso-Gimilio JF. Hypotriglyceridemias/hypolipidemias. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2021; 33 Suppl 2:63-68. [PMID: 34006356 DOI: 10.1016/j.arteri.2020.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 12/31/2020] [Indexed: 06/12/2023]
Abstract
Hypolipoproteinemias are characterized by a decrease in the plasma concentration of lipoproteins. Within them, we find two groups: hypobetalipoproteinemias (HBL), due to a decrease in the plasma concentration of lipoproteins containing apolipoprotein B, and hypoalphalipoproteinemias. Hypolipoproteinemias can be classified according to their origin, into primary and secondary. Primary HBLs are rare entities produced by mutations in different genes. So far, more than 140 mutations have been identified in the APOB, PCSK9, ANGPTL3, MTTP, and SAR1 genes. Early diagnosis and treatment are essential to avoid the development of serious complications. In this review we address the diagnosis and treatment of HBL, especially those in which there is hypotriglyceridemia.
Collapse
Affiliation(s)
- Sergio Martínez-Hervás
- Servicio de Endocrinología y Nutrición, Hospital Clínico Universitario de Valencia-INCLIVA, Valencia, España; Departamento de Medicina, Universitat de Valencia, Valencia, España; CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Valencia, España.
| | - José Tomás Real-Collado
- Servicio de Endocrinología y Nutrición, Hospital Clínico Universitario de Valencia-INCLIVA, Valencia, España; Departamento de Medicina, Universitat de Valencia, Valencia, España; CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Valencia, España
| | - Juan Francisco Ascaso-Gimilio
- Departamento de Medicina, Universitat de Valencia, Valencia, España; CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Valencia, España
| |
Collapse
|
11
|
Takaeko Y, Kajikawa M, Kishimoto S, Yamaji T, Harada T, Han Y, Kihara Y, Hida E, Chayama K, Goto C, Aibara Y, Yusoff FM, Maruhashi T, Nakashima A, Higashi Y. Low Levels of Low-Density Lipoprotein Cholesterol and Endothelial Function in Subjects without Lipid-Lowering Therapy. J Clin Med 2020; 9:E3796. [PMID: 33255270 PMCID: PMC7761134 DOI: 10.3390/jcm9123796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/10/2020] [Accepted: 11/21/2020] [Indexed: 11/16/2022] Open
Abstract
An elevation of serum low-density lipoprotein cholesterol (LDL-C) levels has been associated with endothelial dysfunction in statin naïve subjects. However, there is no information on endothelial function in subjects with extremely low levels of LDL-C. The purpose of the present study was to determine the relationship of LDL-C levels, especially low levels of LDL-C, with endothelial function. Endothelial function assessed by flow-mediated vasodilation (FMD) measurement and LDL-C levels were evaluated in 7120 subjects without lipid-lowering therapy. We divided the subjects into five groups by LDL-C levels: <70 mg/dL, 70-99 mg/dL, 100-119 md/dL, 120-139 mg/dL, and ≥140 mg/dL. FMD values were significantly smaller in subjects with LDL-C levels of ≥140 mg/dL than in those with LDL-C levels of 70-99 mg/dL and 100-119 mg/dL (p < 0.001 and p = 0.004, respectively). The FMD values in the LDL-C of <70 mg/dL group were not significantly different from those in the other groups. To evaluate the relationship of extremely low LDL-C levels with endothelial function, we divided the subjects with LDL-C of <70 mg/dL into those with LDL-C levels of <50 mg/dL and 50-69 mg/dL. FMD values were similar in the LDL-C <50 mg/dL group and ≥50 mg/dL group in the propensity score-matched population (p = 0.570). A significant benefit was not found in subjects with low LDL-C levels from the aspect of endothelial function.
Collapse
Affiliation(s)
- Yuji Takaeko
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Faculty of Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.T.); (S.K.); (T.Y.); (T.H.); (Y.H.); (Y.K.)
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 734-8551, Japan;
| | - Shinji Kishimoto
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Faculty of Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.T.); (S.K.); (T.Y.); (T.H.); (Y.H.); (Y.K.)
| | - Takayuki Yamaji
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Faculty of Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.T.); (S.K.); (T.Y.); (T.H.); (Y.H.); (Y.K.)
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Faculty of Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.T.); (S.K.); (T.Y.); (T.H.); (Y.H.); (Y.K.)
| | - Yiming Han
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Faculty of Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.T.); (S.K.); (T.Y.); (T.H.); (Y.H.); (Y.K.)
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Faculty of Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.T.); (S.K.); (T.Y.); (T.H.); (Y.H.); (Y.K.)
| | - Eisuke Hida
- Department of Biostatistics and Data Science, Graduate School of Medicine, Faculty of Medicine, Osaka University, Osaka 565-0871, Japan;
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University, Hiroshima 739-2695, Japan;
| | - Yoshiki Aibara
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.A.); (F.M.Y.); (T.M.)
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.A.); (F.M.Y.); (T.M.)
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.A.); (F.M.Y.); (T.M.)
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical and Health Sciences, Faculty of Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 734-8551, Japan;
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (Y.A.); (F.M.Y.); (T.M.)
| |
Collapse
|
12
|
Hegele RA, Borén J, Ginsberg HN, Arca M, Averna M, Binder CJ, Calabresi L, Chapman MJ, Cuchel M, von Eckardstein A, Frikke-Schmidt R, Gaudet D, Hovingh GK, Kronenberg F, Lütjohann D, Parhofer KG, Raal FJ, Ray KK, Remaley AT, Stock JK, Stroes ES, Tokgözoğlu L, Catapano AL. Rare dyslipidaemias, from phenotype to genotype to management: a European Atherosclerosis Society task force consensus statement. Lancet Diabetes Endocrinol 2020; 8:50-67. [PMID: 31582260 DOI: 10.1016/s2213-8587(19)30264-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/23/2019] [Accepted: 07/27/2019] [Indexed: 12/18/2022]
Abstract
Genome sequencing and gene-based therapies appear poised to advance the management of rare lipoprotein disorders and associated dyslipidaemias. However, in practice, underdiagnosis and undertreatment of these disorders are common, in large part due to interindividual variability in the genetic causes and phenotypic presentation of these conditions. To address these challenges, the European Atherosclerosis Society formed a task force to provide practical clinical guidance focusing on patients with extreme concentrations (either low or high) of plasma low-density lipoprotein cholesterol, triglycerides, or high-density lipoprotein cholesterol. The task force also recognises the scarcity of quality information regarding the prevalence and outcomes of these conditions. Collaborative registries are needed to improve health policy for the care of patients with rare dyslipidaemias.
Collapse
Affiliation(s)
- Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henry N Ginsberg
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Marcello Arca
- Department of Internal Medicine and Allied Sciences, Center for Rare Disorders of Lipid Metabolism, Sapienza University of Rome, Rome, Italy
| | - Maurizio Averna
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialities, University of Palermo, Palermo, Italy
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Laura Calabresi
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M John Chapman
- National Institute for Health and Medical Research (INSERM), Sorbonne University and Pitié-Salpétrière University Hospital, Paris, France
| | - Marina Cuchel
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ruth Frikke-Schmidt
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| | - Daniel Gaudet
- Clinical Lipidology and Rare Lipid Disorders Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal, Montreal, QC, Canada; ECOGENE, Clinical and Translational Research Center, Chicoutimi, QC, Canada; Lipid Clinic, Chicoutimi Hospital, Chicoutimi, QC, Canada
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Klaus G Parhofer
- Medizinische Klinik IV-Grosshadern, University of Munich, Munich, Germany
| | - Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg, South Africa
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College London, London, UK
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jane K Stock
- European Atherosclerosis Society, Gothenburg, Sweden
| | - Erik S Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
13
|
Wilmshurst JM, Ouvrier RA, Ryan MM. Peripheral nerve disease secondary to systemic conditions in children. Ther Adv Neurol Disord 2019; 12:1756286419866367. [PMID: 31447934 PMCID: PMC6691669 DOI: 10.1177/1756286419866367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/07/2019] [Indexed: 12/17/2022] Open
Abstract
This review is an overview of systemic conditions that can be associated with peripheral nervous system dysfunction. Children may present with neuropathic symptoms for which, unless considered, a causative systemic condition may not be recognized. Similarly, some systemic conditions may be complicated by comorbid peripheral neuropathies, surveillance for which is indicated. The systemic conditions addressed in this review are critical illness polyneuropathy, chronic renal failure, endocrine disorders such as insulin-dependent diabetes mellitus and multiple endocrine neoplasia type 2b, vitamin deficiency states, malignancies and reticuloses, sickle cell disease, neurofibromatosis, connective tissue disorders, bowel dysmotility and enteropathy, and sarcoidosis. In some disorders presymptomatic screening should be undertaken, while in others there is no benefit from early detection of neuropathy. In children with idiopathic peripheral neuropathies, systemic disorders such as celiac disease should be actively excluded. While management is predominantly focused on symptomatic care through pain control and rehabilitation, some neuropathies improve with effective control of the underlying etiology and in a small proportion a more targeted approach is possible. In conclusion, peripheral neuropathies can be associated with a diverse range of medical conditions and unless actively considered may not be recognized and inadequately managed.
Collapse
Affiliation(s)
- Jo M. Wilmshurst
- Department of Paediatric Neurology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s, Hospital Neuroscience Institute, University of Cape Town, Klipfontein Road, Cape Town, Western Cape, 7700, South Africa
| | - Robert A. Ouvrier
- The Institute of Neuroscience and Muscle Research, The Children’s Hospital at Westmead, Sydney, Australia
| | - Monique M. Ryan
- Department of Neurology, Royal Children’s Hospital, Murdoch Children’s Research Institute and University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Abstract
This study provides a structure for microsomal triglyceride transfer protein, a key protein in lipid metabolism and transport. Microsomal triglyceride transfer protein is linked to a human disease state, abetalipoproteinemia. The structure helps us to understand how this protein functions and gives a rationale for how previously reported mutations result in loss of function of the protein and hence, cause disease. The structure also provides a means for rational drug design to treat cardiovascular disease, hypercholesterolemia, and obesity. Microsomal triglyceride transfer protein is composed of 2 subunits. The β-subunit, protein disulfide isomerase (PDI), also acts independently as a protein folding catalyst. The structure that we present here gives insights into how PDI functions in protein folding. Microsomal triglyceride transfer protein (MTP) plays an essential role in lipid metabolism, especially in the biogenesis of very low-density lipoproteins and chylomicrons via the transfer of neutral lipids and the assembly of apoB-containing lipoproteins. Our understanding of the molecular mechanisms of MTP has been hindered by a lack of structural information of this heterodimeric complex comprising an MTPα subunit and a protein disulfide isomerase (PDI) β-subunit. The structure of MTP presented here gives important insights into the potential mechanisms of action of this essential lipid transfer molecule, structure-based rationale for previously reported disease-causing mutations, and a means for rational drug design against cardiovascular disease and obesity. In contrast to the previously reported structure of lipovitellin, which has a funnel-like lipid-binding cavity, the lipid-binding site is encompassed in a β-sandwich formed by 2 β-sheets from the C-terminal domain of MTPα. The lipid-binding cavity of MTPα is large enough to accommodate a single lipid. PDI independently has a major role in oxidative protein folding in the endoplasmic reticulum. Comparison of the mechanism of MTPα binding by PDI with previously published structures gives insights into large protein substrate binding by PDI and suggests that the previous structures of human PDI represent the “substrate-bound” and “free” states rather than differences arising from redox state.
Collapse
|
15
|
Benachenhou S, Etcheverry A, Galarneau L, Dubé J, Çaku A. Implication of hypocholesterolemia in autism spectrum disorder and its associated comorbidities: A retrospective case-control study. Autism Res 2019; 12:1860-1869. [PMID: 31385649 DOI: 10.1002/aur.2183] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/17/2019] [Indexed: 12/26/2022]
Abstract
Autism spectrum disorder (ASD) has been associated with low cholesterol levels in a limited number of studies. However, the prevalence of hypocholesterolemia as well as the degree of association with ASD remains to be elucidated. We therefore sought to investigate the lipid profiles of a group of French-Canadian ASD individuals. The medical records of 79 ASD individuals and 79 age and gender-matched healthy controls were retrospectively reviewed. The fasting lipid profiles including total cholesterol (TC), high-density lipoprotein, triglycerides, and low-density lipoprotein were extracted for individuals of both groups along with the following clinical data: anthropometric measurements, medication use and associated disorders. Lipid parameters were compared to age and gender-based normative population and categorized in centile groups. The prevalence of hypocholesterolemia was revealed to be more than threefold higher in ASD individuals as compared to the general population (23%; P = 0.005). The 25th centile was determined as a potential TC threshold that could best predict the ASD (odds ratio [OR] = 3.04; 95% confidence interval [CI]: 1.58-6.65; P < 0.001). This study identified specific ASD comorbidities associated with hypocholesterolemia: TC levels below the 10th centile were associated with a higher rate of ASD-associated intellectual disability (OR = 3.33; 95% CI: 1.26-8.00) and anxiety/depression (OR = 4.74; 95% CI: 1.40-15.73). Overall, these results support a potential association between hypocholesterolemia and ASD occurrence. Application of this study to larger populations is urging to provide more extensive data that may further elucidate the association between hypocholesterolemia and ASD. Autism Res 2019, 12: 1860-1869. © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Association of autism spectrum disorder (ASD) with abnormally low levels of cholesterol (hypocholesterolemia) has been documented before. These studies were incomplete, and the conclusion remains speculative. Here, we reviewed the medical records of 79 French-Canadian ASD individuals and compared their total cholesterol (TC) levels to healthy individuals matched for age and gender. We observed four times more hypocholesterolemia in ASD than in the general population. Furthermore, low TC in ASD was associated with higher rates of ASD-associated intellectual disability and anxiety/depression. Our results support an association between hypocholesterolemia and ASD and open novel opportunities for the diagnosis and treatment of specific forms of ASD.
Collapse
Affiliation(s)
- Sérine Benachenhou
- Biochemistry Department, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Antoine Etcheverry
- Biochemistry Department, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Luc Galarneau
- Biochemistry Department, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean Dubé
- Biochemistry Department, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Artuela Çaku
- Biochemistry Department, Faculty of Medicine and Health Sciences, Universite de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
16
|
Lima Pessoa E, Costa Vilella Dos Reis M, Sayuri Yamamoto T, Ribeiro Neto M, Ferraro O, Alves MJ, Guedes Coelho Lopes R. Familial heterozygous hypobetalipoproteinemia and breast cancer risk: A systematic review and suggestions for further research. Breast J 2019; 25:763-765. [PMID: 31111608 DOI: 10.1111/tbj.13341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Eduardo Lima Pessoa
- Department of Radiation Oncology, Hospital do Servidor Público Estadual de São Paulo- Iamspe, São Paulo, Brazil
| | | | - Tatiana Sayuri Yamamoto
- Department of Radiation Oncology, Hospital do Servidor Público Estadual de São Paulo- Iamspe, São Paulo, Brazil
| | - Mario Ribeiro Neto
- Department of Radiation Oncology, Hospital do Servidor Público Estadual de São Paulo- Iamspe, São Paulo, Brazil
| | - Odair Ferraro
- Department of Gynecology and Obstetrics, Hospital do Servidor Público Estadual de São Paulo- Iamspe, São Paulo, Brazil
| | - Maria José Alves
- Department of Radiation Oncology, Hospital do Servidor Público Estadual de São Paulo- Iamspe, São Paulo, Brazil
| | - Reginaldo Guedes Coelho Lopes
- Department of Gynecology and Obstetrics, Hospital do Servidor Público Estadual de São Paulo- Iamspe, São Paulo, Brazil
| |
Collapse
|
17
|
Blanco-Vaca F, Martin-Campos JM, Beteta-Vicente Á, Canyelles M, Martínez S, Roig R, Farré N, Julve J, Tondo M. Molecular analysis of APOB, SAR1B, ANGPTL3, and MTTP in patients with primary hypocholesterolemia in a clinical laboratory setting: Evidence supporting polygenicity in mutation-negative patients. Atherosclerosis 2019; 283:52-60. [PMID: 30782561 DOI: 10.1016/j.atherosclerosis.2019.01.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/17/2019] [Accepted: 01/24/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Primary hypobetalipoproteinemia is generally considered a heterogenic group of monogenic, inherited lipoprotein disorders characterized by low concentrations of LDL cholesterol and apolipoprotein B in plasma. Lipoprotein disorders include abetalipoproteinemia, familial hypobetalipoproteinemia, chylomicron retention disease, and familial combined hypolipidemia. Our aim was to review and analyze the results of the molecular analysis of hypolipidemic patients studied in our laboratory over the last 15 years. METHODS The study included 44 patients with clinical and biochemical data. Genomic studies were performed and genetic variants were characterized by bioinformatics analysis. A weighted LDL cholesterol gene score was calculated to evaluate common variants associated with impaired lipid concentrations and their distribution among patients. RESULTS Twenty-three patients were genetically confirmed as affected by primary hypobetalipoproteinemia. In this group of patients, the most prevalent mutated genes were APOB (in 17 patients, with eight novel mutations identified), SAR1B (in 3 patients, with one novel mutation identified), ANGPTL3 (in 2 patients), and MTTP (in 1 patient). The other 21 patients could not be genetically diagnosed with hypobetalipoproteinemia despite presenting suggestive clinical and biochemical features. In these patients, two APOB genetic variants associated with lower LDL cholesterol were more frequent than in controls. Moreover, the LDL cholesterol gene score, calculated with 11 SNPs, was significantly lower in mutation-negative patients. CONCLUSIONS Around half of the patients could be genetically diagnosed. The results suggest that, in at least some of the patients without an identified mutation, primary hypobetalipoproteinemia may have a polygenic origin.
Collapse
Affiliation(s)
- Francisco Blanco-Vaca
- Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica - IIB Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain.
| | - Jesús M Martin-Campos
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau- IIB Sant Pau, Barcelona, Spain
| | - Ángel Beteta-Vicente
- Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica - IIB Sant Pau, Barcelona, Spain
| | - Marina Canyelles
- Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica - IIB Sant Pau, Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain
| | - Susana Martínez
- Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica - IIB Sant Pau, Barcelona, Spain
| | - Rosa Roig
- Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica - IIB Sant Pau, Barcelona, Spain
| | - Núria Farré
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau- IIB Sant Pau, Barcelona, Spain
| | - Josep Julve
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau- IIB Sant Pau, Barcelona, Spain
| | - Mireia Tondo
- Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica - IIB Sant Pau, Barcelona, Spain.
| |
Collapse
|
18
|
Cariou B, Challet-Bouju G, Bernard C, Marrec M, Hardouin JB, Authier C, Bach-Ngohou K, Leux C, Pichelin M, Grall-Bronnec M. Prevalence of hypobetalipoproteinemia and related psychiatric characteristics in a psychiatric population: results from the retrospective HYPOPSY Study. Lipids Health Dis 2018; 17:249. [PMID: 30400945 PMCID: PMC6220563 DOI: 10.1186/s12944-018-0892-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 10/15/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Hypobetalipoproteinemia (HBL) is defined by plasma concentrations of LDL-cholesterol (LDL-C) lower than the fifth percentile for age and sex. Several psychiatric symptoms have been reported in association with HBL. The objective was to assess the prevalence of primary HBL in patients hospitalized in a psychiatric population and to better characterize the related psychiatric disorders. METHODS HYPOPSY is a retrospective study including 839 adults hospitalized in the Psychiatry department of Nantes University Hospital during the year 2014, except patients with eating disorders. The prevalence of primary HBL was defined by a plasma LDL-C concentration ≤ 50 mg/dL. Secondary causes of HBL were excluded after a review of medical records (n=2). Related-psychiatric disorders in patients with and without HBL were recorded using the ICD-10 classification. RESULTS Twenty cases of primary HBL (mean [SD] LDL-C: 42 [7] mg/dL) were diagnosed, leading to a prevalence of 2.39%. In comparison, the prevalence of HBL in a healthy control population was 0.57%. Psychiatric patients with HBL were characterized by a higher frequency of schizophrenia (p=0.044), hetero-aggression (p=0.015) and pervasive and specific developmental disorders (including autism) (p=0.011). CONCLUSIONS The prevalence of HBL is 4-fold higher in psychiatric than in general population. More specifically, some statistically significant associations were found between low LDL-C concentrations and schizophrenia, autism and hetero-aggression. These data reinforce the hypothesis for a link between genetically low LDL-C levels and psychiatric disorders.
Collapse
Affiliation(s)
- Bertrand Cariou
- CHU Nantes, l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, F-44000, Nantes, France. .,CHU de Nantes, CIC Endocrino-Nutrition INSERM UMR 1413, l'nstitut du thorax, F-44000, Nantes, France. .,Clinique d'Endocrinologie, Maladies Métaboliques et Nutrition, Hôpital Guillaume & René Laennec, Boulevard Jacques Monod, Saint-Herblain, 44093, Nantes Cedex 1, France.
| | - Gaëlle Challet-Bouju
- CHU Nantes, Service d'Addictologie et de Psychiatrie, F-44000, Nantes, France.,Université de Nantes, Université de Tours, Inserm UMR 1246-SPHERE, F-44000, Nantes, France
| | - Céline Bernard
- Clinique d'Endocrinologie, Maladies Métaboliques et Nutrition, Hôpital Guillaume & René Laennec, Boulevard Jacques Monod, Saint-Herblain, 44093, Nantes Cedex 1, France.,Present address: CHU Sud Reunion, Site Saint-Pierre, Avenue président F Mitterrand, BP 350, 97448 ST, Pierre Cedex, La Reunion, France
| | - Marie Marrec
- CHU de Nantes, CIC Endocrino-Nutrition INSERM UMR 1413, l'nstitut du thorax, F-44000, Nantes, France
| | - Jean-Benoit Hardouin
- Université de Nantes, Université de Tours, Inserm UMR 1246-SPHERE, F-44000, Nantes, France.,CHU Nantes, DRCi, Plateforme de Méthodologie et de Biostatistique, F-44000, Nantes, France
| | - Charlotte Authier
- Centre d'examens de santé de la Caisse Primaire d'Assurance Maladie de Loire-Atlantique, St Nazaire, F-44600, Saint-Nazaire, France
| | - Kalyane Bach-Ngohou
- CHU Nantes, Service de Biochimie, F-44000, Nantes, France.,INSERM1235, Université de Nantes, Institut des Maladies de l'Appareil Digestif, F-44000, Nantes, France
| | - Christophe Leux
- CHU Nantes, Service d'Information Médicale, F-44000, Nantes, France
| | - Matthieu Pichelin
- CHU Nantes, l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, F-44000, Nantes, France.,CHU de Nantes, CIC Endocrino-Nutrition INSERM UMR 1413, l'nstitut du thorax, F-44000, Nantes, France.,Clinique d'Endocrinologie, Maladies Métaboliques et Nutrition, Hôpital Guillaume & René Laennec, Boulevard Jacques Monod, Saint-Herblain, 44093, Nantes Cedex 1, France
| | - Marie Grall-Bronnec
- CHU Nantes, Service d'Addictologie et de Psychiatrie, F-44000, Nantes, France. .,Université de Nantes, Université de Tours, Inserm UMR 1246-SPHERE, F-44000, Nantes, France.
| |
Collapse
|
19
|
Wu Z, Yang F, Jiang S, Sun X, Xu J. Induction of Liver Steatosis in BAP31-Deficient Mice Burdened with Tunicamycin-Induced Endoplasmic Reticulum Stress. Int J Mol Sci 2018; 19:ijms19082291. [PMID: 30081561 PMCID: PMC6121476 DOI: 10.3390/ijms19082291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/24/2018] [Accepted: 08/02/2018] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is highly associated with liver steatosis. B-cell receptor-associated protein 31 (BAP31) has been reported to be involved in ER homeostasis, and plays key roles in hepatic lipid metabolism in high-fat diet-induced obese mice. However, whether BAP31 modulates hepatic lipid metabolism via regulating ER stress is still uncertain. In this study, wild-type and liver-specific BAP31-depleted mice were administrated with ER stress activator of Tunicamycin, the markers of ER stress, liver steatosis, and the underlying molecular mechanisms were determined. BAP31 deficiency increased Tunicamycin-induced hepatic lipid accumulation, aggravated liver dysfunction, and increased the mRNA levels of ER stress markers, including glucose-regulated protein 78 (GRP78), X-box binding protein 1 (XBP1), inositol-requiring protein-1α (IRE1α) and C/EBP homologous protein (CHOP), thus promoting ER stress in vivo and in vitro. Hepatic lipid export via very low-density lipoprotein (VLDL) secretion was impaired in BAP31-depleted mice, accompanied by reduced Apolipoprotein B (APOB) and microsomal triglyceride transfer protein (MTTP) expression. Exogenous lipid clearance was also inhibited, along with impaired gene expression related to fatty acid transportation and fatty acid β-oxidation. Finally, BAP31 deficiency increased Tunicamycin-induced hepatic inflammatory response. These results demonstrate that BAP31 deficiency increased Tunicamycin-induced ER stress, impaired VLDL secretion and exogenous lipid clearance, and reduced fatty acid β-oxidation, which eventually resulted in liver steatosis.
Collapse
Affiliation(s)
- Zhenhua Wu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| | - Fan Yang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| | - Shan Jiang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| | - Xiaoyu Sun
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| | - Jialin Xu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| |
Collapse
|
20
|
Cuerq C, Henin E, Restier L, Blond E, Drai J, Marçais C, Di Filippo M, Laveille C, Michalski MC, Poinsot P, Caussy C, Sassolas A, Moulin P, Reboul E, Charriere S, Levy E, Lachaux A, Peretti N. Efficacy of two vitamin E formulations in patients with abetalipoproteinemia and chylomicron retention disease. J Lipid Res 2018; 59:1640-1648. [PMID: 30021760 DOI: 10.1194/jlr.m085043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
Abetalipoproteinemia (ABL) and chylomicron retention disease (CMRD) are extremely rare recessive forms of hypobetalipoproteinemia characterized by intestinal lipid malabsorption and severe vitamin E deficiency. Vitamin E is often supplemented in the form of fat-soluble vitamin E acetate, but fat malabsorption considerably limits correction of the deficiency. In this crossover study, we administered two different forms of vitamin E, tocofersolan (a water-soluble derivative of RRR-α-tocopherol) and α-tocopherol acetate, to three patients with ABL and four patients with CMRD. The aims of this study were to evaluate the intestinal absorption characteristics of tocofersolan versus α-tocopherol acetate by measuring the plasma concentrations of α-tocopherol over time after a single oral load and to compare efficacy by evaluating the ability of each formulation to restore vitamin E storage after 4 months of treatment. In patients with ABL, tocofersolan and α-tocopherol acetate bioavailabilities were extremely low (2.8% and 3.1%, respectively). In contrast, bioavailabilities were higher in patients with CMRD (tocofersolan, 24.7%; α-tocopherol acetate, 11.4%). Plasma concentrations of α-tocopherol at 4 months were not significantly different by formulation type in ABL or CMRD. This study provides new insights about vitamin E status in ABL and CMRD and suggests the potential of different formulations as treatment options.
Collapse
Affiliation(s)
- Charlotte Cuerq
- Biochemistry Department, Lyon Sud Hospital, Hospices Civils de Lyon, Lyon, France; INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | | | - Lioara Restier
- Pediatric Hepato-Gastroenterology and Nutrition Unit, Hôpital Femme Mère Enfant de Lyon, Dyslipidemia Unity Hospices Civils de Lyon, Lyon, Bron, France
| | - Emilie Blond
- Biochemistry Department, Lyon Sud Hospital, Hospices Civils de Lyon, Lyon, France; INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | - Jocelyne Drai
- Biochemistry Department, Lyon Sud Hospital, Hospices Civils de Lyon, Lyon, France; INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | - Christophe Marçais
- Biochemistry Department, Lyon Sud Hospital, Hospices Civils de Lyon, Lyon, France; INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | - Mathilde Di Filippo
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France; Department of Biochemistry and Molecular Biology, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Lyon, Bron, France
| | | | | | - Pierre Poinsot
- Pediatric Hepato-Gastroenterology and Nutrition Unit, Hôpital Femme Mère Enfant de Lyon, Dyslipidemia Unity Hospices Civils de Lyon, Lyon, Bron, France
| | - Cyrielle Caussy
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | - Agnès Sassolas
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France; Department of Biochemistry and Molecular Biology, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Lyon, Bron, France
| | - Philippe Moulin
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France; Fédération d'Endocrinologie, Maladies Métaboliques, Diabète et Nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | | | - Sybil Charriere
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France; Fédération d'Endocrinologie, Maladies Métaboliques, Diabète et Nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Emile Levy
- Research Centre, CHU Sainte-Justine, and Department of Nutrition, Université de Montréal, Montréal, Québec, Canada
| | - Alain Lachaux
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France; Pediatric Hepato-Gastroenterology and Nutrition Unit, Hôpital Femme Mère Enfant de Lyon, Dyslipidemia Unity Hospices Civils de Lyon, Lyon, Bron, France
| | - Noël Peretti
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France; Pediatric Hepato-Gastroenterology and Nutrition Unit, Hôpital Femme Mère Enfant de Lyon, Dyslipidemia Unity Hospices Civils de Lyon, Lyon, Bron, France.
| |
Collapse
|
21
|
Balder JW, Rimbert A, Zhang X, Viel M, Kanninga R, van Dijk F, Lansberg P, Sinke R, Kuivenhoven JA. Genetics, Lifestyle, and Low-Density Lipoprotein Cholesterol in Young and Apparently Healthy Women. Circulation 2018; 137:820-831. [DOI: 10.1161/circulationaha.117.032479] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/08/2018] [Indexed: 01/02/2023]
Abstract
Background:
Atherosclerosis starts in childhood but low-density lipoprotein cholesterol (LDL-C), a causal risk factor, is mostly studied and dealt with when clinical events have occurred. Women are usually affected later in life than men and are underdiagnosed, undertreated, and understudied in cardiovascular trials and research. This study aims at a better understanding of lifestyle and genetic factors that affect LDL-C in young women.
Methods:
We randomly selected for every year of age 8 women with LDL-C ≤1st percentile (≤50 mg/dL) and 8 women with LDL-C ≥99th percentile (≥186 mg/dL) from 28 000 female participants aged between 25 to 40 years of a population-based cohort study. The resulting groups include 119 and 121 women, respectively, of an average 33 years of age. A gene-sequencing panel was used to assess established monogenic and polygenic origins of these phenotypes. Information on lifestyle was extracted from questionnaires. A healthy lifestyle score was allocated based on a recently developed algorithm.
Results:
Of the women with LDL-C ≤1st percentile, 19 (15.7%) carried mutations that are causing monogenic hypocholesterolemia and 60 (49.6%) were genetically predisposed to low LDL-C on the basis of an extremely low weighted genetic risk score. In comparison with control groups, a healthier lifestyle was not associated with low LDL-C in women without genetic predispositions. Among women with LDL-C ≥99th percentile, 20 women (16.8%) carried mutations that cause familial hypercholesterolemia, whereas 25 (21%) were predisposed to high LDL-C on the basis of a high-weighted genetic risk score. The women in whom no genetic origin for hypercholesterolemia could be identified were found to exhibit a significantly unfavorable lifestyle in comparison with controls.
Conclusions:
This study highlights the need for early assessment of the cardiovascular risk profile in apparently healthy young women to identify those with LDL-C ≥99th percentile for their age: first, because, in this study, 17% of the cases were molecularly diagnosed with familial hypercholesterolemia, which needs further attention; second, because our data indicate that an unfavorable lifestyle is significantly associated with severe hypercholesterolemia in genetically unaffected women, which may also need further attention.
Collapse
Affiliation(s)
- Jan-Willem Balder
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
- Department of Vascular Medicine, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B.)
| | - Antoine Rimbert
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
| | - Xiang Zhang
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands (X.Z.)
| | - Martijn Viel
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Roan Kanninga
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Freerk van Dijk
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Peter Lansberg
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
| | - Richard Sinke
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
| |
Collapse
|
22
|
Lee CJ, Lee Y, Park S, Kang SM, Jang Y, Lee JH, Lee SH. Rare and common variants of APOB and PCSK9 in Korean patients with extremely low low-density lipoprotein-cholesterol levels. PLoS One 2017; 12:e0186446. [PMID: 29036232 PMCID: PMC5643101 DOI: 10.1371/journal.pone.0186446] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 10/02/2017] [Indexed: 01/03/2023] Open
Abstract
Background Screening of variants, related to lipid metabolism in patients with extreme cholesterol levels, is a tool used to identify targets affecting cardiovascular outcomes. The aim of this study was to examine the prevalence and characteristics of rare and common variants of APOB and PCSK9 in Korean patients with extremely low low-density lipoprotein-cholesterol (LDL-C) levels. Methods Among 13,545 participants enrolled in a cardiovascular genome cohort, 22 subjects, whose LDL-C levels without lipid-lowering agents were ≤1 percentile (48 mg/dL) of Korean population, were analyzed. Two target genes, APOB and PCSK9, were sequenced by targeted next-generation sequencing. Prediction of functional effects was conducted using SIFT, PolyPhen-2, and Mutation Taster, and matched against a public database of variants. Results Eight rare variants of the two candidate genes (five in APOB and three in PCSK9) were found in nine subjects. Two subjects had more than two different rare variants of either gene (one subject in APOB and another subject in APOB/PCSK9). Conversely, 12 common variants (nine in APOB and three in PCSK9) were discovered in 21 subjects. Among all variants, six in APOB and three in PCSK9 were novel. Several variants previously reported functional, including c.C277T (p.R93C) and c.G2009A (p.G670E) of PCSK9, were found in our population. Conclusions Rare variants of APOB or PCSK9 were identified in nine of the 22 study patients with extremely low LDL-C levels, whereas most of them had common variants of the two genes. The common novelty of variants suggested polymorphism of the two genes among them. Our results provide rare genetic information associated with this lipid phenotype in East Asian people.
Collapse
Affiliation(s)
- Chan Joo Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yunbeom Lee
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Sungha Park
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Seok-Min Kang
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yangsoo Jang
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Lee
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
- * E-mail: (SHL); (JHL)
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (SHL); (JHL)
| |
Collapse
|
23
|
Ellis KL, Boffa MB, Sahebkar A, Koschinsky ML, Watts GF. The renaissance of lipoprotein(a): Brave new world for preventive cardiology? Prog Lipid Res 2017; 68:57-82. [DOI: 10.1016/j.plipres.2017.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/24/2022]
|
24
|
Di Costanzo A, Di Leo E, Noto D, Cefalù AB, Minicocci I, Polito L, D'Erasmo L, Cantisani V, Spina R, Tarugi P, Averna M, Arca M. Clinical and biochemical characteristics of individuals with low cholesterol syndromes: A comparison between familial hypobetalipoproteinemia and familial combined hypolipidemia. J Clin Lipidol 2017; 11:1234-1242. [PMID: 28733173 DOI: 10.1016/j.jacl.2017.06.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/27/2017] [Accepted: 06/17/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The most frequent monogenic causes of low plasma cholesterol are familial hypobetalipoproteinemia (FHBL1) because of truncating mutations in apolipoprotein B coding gene (APOB) and familial combined hypolipidemia (FHBL2) due to loss-of-function mutations in ANGPTL3 gene. OBJECTIVE A direct comparison of lipid phenotypes of these 2 conditions has never been carried out. In addition, although an increased prevalence of liver steatosis in FHBL1 has been consistently reported, the hepatic consequences of FHBL2 are not well established. METHODS We investigated 350 subjects, 67 heterozygous carriers of APOB mutations, 63 carriers of the p.S17* mutation in ANGPTL3 (57 heterozygotes and 6 homozygotes), and 220 noncarrier normolipemic controls. Prevalence and degree of hepatic steatosis were assessed by ultrasonography. RESULTS A steady decrease of low-density lipoprotein cholesterol levels were observed from heterozygous to homozygous FHBL2 and to FHBL1 individuals, with the lowest levels in heterozygous FHBL1 carrying truncating mutations in exons 1 to 25 of APOB (P for trend <.001). Plasma triglycerides levels were similar in heterozygous FHBL1 and homozygous FHBL2 individuals, but higher in heterozygous FHBL2. The lowest high-density lipoprotein cholesterol levels were detected in homozygous FHBL2 (P for trend <.001). Compared with controls, prevalence and severity of hepatic steatosis were increased in heterozygous FHBL1 (P < .001), but unchanged in FHBL2 individuals. CONCLUSION Truncating APOB mutations showed the more striking low-density lipoprotein cholesterol lowering effect compared with p.S17* mutation in ANGPTL3. Reduced high-density lipoprotein cholesterol levels were the unique lipid characteristic associated with FHBL2. Mutations impairing liver synthesis or secretion of apolipoprotein B are crucial to increase the risk of liver steatosis.
Collapse
Affiliation(s)
- Alessia Di Costanzo
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy.
| | - Enza Di Leo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Davide Noto
- Department of Biomedicine, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Angelo Baldassare Cefalù
- Department of Biomedicine, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Ilenia Minicocci
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Luca Polito
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Laura D'Erasmo
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Vito Cantisani
- Department of Radiological Sciences, Sapienza University of Rome, Rome, Italy
| | - Rossella Spina
- Department of Biomedicine, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Patrizia Tarugi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maurizio Averna
- Department of Biomedicine, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Marcello Arca
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Bays HE, Jones PH, Orringer CE, Brown WV, Jacobson TA. National Lipid Association Annual Summary of Clinical Lipidology 2016. J Clin Lipidol 2016; 10:S1-43. [PMID: 26891998 DOI: 10.1016/j.jacl.2015.08.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/03/2015] [Indexed: 01/25/2023]
Abstract
The National Lipid Association (NLA) Annual Summary of Clinical Lipidology is a yearly updated summary of principles important to the patient-centered evaluation, management, and care of patients with dyslipidemia. This summary is intended to be a "living document," with future annual updates based on emerging science, clinical considerations, and new NLA Position, Consensus, and Scientific Statements, thus providing an ongoing resource that applies the latest in medical science towards the clinical management of patients with dyslipidemia. Topics include the NLA Recommendations for Patient-Centered Management of Dyslipidemia, genetics, Familial Hypercholesterolemia, secondary causes of dyslipidemia, biomarkers and advanced lipid testing, nutrition, physical activity, obesity, adiposopathy, metabolic syndrome, diabetes mellitus, lipid pharmacotherapy, lipid-altering drug interactions, lipoprotein apheresis, dyslipidemia management and treatment based upon age (children, adolescents, and older individuals), dyslipidemia considerations based upon race, ethnicity and gender, dyslipidemia and human immune virus infection, dyslipidemia and immune disorders, adherence strategies and collaborative care, and lipid-altering drugs in development. Hyperlinks direct the reader to sentinel online tables, charts, and figures relevant to lipidology, access to online atherosclerotic cardiovascular disease risk calculators, worldwide lipid guidelines, recommendations, and position/scientific statements, as well as links to online audio files, websites, slide shows, applications, continuing medical education opportunities, and patient information.
Collapse
Affiliation(s)
- Harold E Bays
- Louisville Metabolic and Atherosclerosis Research Center, Louisville, KY, USA.
| | | | - Carl E Orringer
- University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | | | | |
Collapse
|
26
|
Cuerq C, Restier L, Drai J, Blond E, Roux A, Charriere S, Michalski MC, Di Filippo M, Levy E, Lachaux A, Peretti N. Establishment of reference values of α-tocopherol in plasma, red blood cells and adipose tissue in healthy children to improve the management of chylomicron retention disease, a rare genetic hypocholesterolemia. Orphanet J Rare Dis 2016; 11:114. [PMID: 27520363 PMCID: PMC4982212 DOI: 10.1186/s13023-016-0498-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/03/2016] [Indexed: 01/04/2023] Open
Abstract
Background Chylomicron retention disease (CMRD), a rare genetic hypocholesterolemia, results in neuro-ophtalmologic damages, which can be prevented by high doses of vitamin E during infancy. In these patients, plasma vitamin E concentration is significantly reduced due to defects of chylomicron secretion. Vitamin E in adipose tissue (AT) and red blood cells (RBC) have been proposed as potential relevant biomarkers of vitamin E status but no reference values in children are available. The objectives were (i) to establish age-reference intervals in healthy children for α-tocopherol in plasma, red blood cells (RBC) and adipose tissue (AT) and (ii) to determine the variations of α-tocopherol in patients with CMRD after oral treatment with vitamin E. Methods This prospective study included 166 healthy children (1 month - 18 years) and 4 patients with CMRD. Blood and AT were collected in healthy children during a scheduled surgery and in patients before and after a 4-month treatment with α-tocopherol acetate. Results The reference ranges for α-tocopherol were 11.9 - 30 μmol/L in plasma, 2.0 - 7.8 μmol/L packed cells in RBC and 60 - 573 nmol/g in AT. α-tocopherol levels in plasma correlated with those of RBC (r = 0.31; p < 0.01). In patients with CMRD after 4 months treatment, α-tocopherol concentrations remained less than 70 % of the control values in plasma, increased by 180 % to reach normal values in RBC, and remained stable in the normal range in AT. Conclusion This study establishes pediatric reference intervals for α-tocopherol in plasma, RBC and AT. These values will be beneficial in assessing accurate α-tocopherol status in children and to optimize the monitoring of rare diseases such as CMRD. Our data suggest that RBC α-tocopherol, appears as a relevant biomarker to appreciate the effectiveness of treatment with α-tocopherol in patients with a rare primary hypocholesterolemia. The biopsy of AT could be used at diagnosis to assess the severity of the vitamin E deficiency and periodically after a long duration of vitamin E therapy to assess whether the treatment is effective, based on reference intervals defined in this study.
Collapse
Affiliation(s)
- Charlotte Cuerq
- Biochemistry Department, Lyon Sud Hospital, Hospices Civils de Lyon, Lyon, France.,INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | - Lioara Restier
- Pediatric Hepato-Gastroenterology and Nutrition Unit, Hôpital Femme Mère Enfant de Lyon, Hospices Civils de Lyon, Lyon, Bron, France
| | - Jocelyne Drai
- Biochemistry Department, Lyon Sud Hospital, Hospices Civils de Lyon, Lyon, France.,INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | - Emilie Blond
- Biochemistry Department, Lyon Sud Hospital, Hospices Civils de Lyon, Lyon, France.,INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France
| | - Adeline Roux
- Hospices Civils de Lyon, Pole IMER, Lyon, France
| | - Sybil Charriere
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France.,Fédération d'endocrinologie, maladies métaboliques, diabète et nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | | | - Mathilde Di Filippo
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France.,Dyslipidemia Unity, Department of Biochemistry and Molecular Biology, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Lyon, Bron, France
| | - Emile Levy
- Research Centre, CHU Sainte-Justine, Université de Montréal, Montréal, Québec, H3T 1C5, Canada.,Department of Nutrition, Université de Montréal, Montréal, Québec, H3T 1A8, Canada
| | - Alain Lachaux
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France.,Pediatric Hepato-Gastroenterology and Nutrition Unit, Hôpital Femme Mère Enfant de Lyon, Hospices Civils de Lyon, Lyon, Bron, France
| | - Noël Peretti
- INSERM U1060, INRA UMR 1397, INSA-Lyon, CarMeN Laboratory, Université Lyon 1, Lyon, France. .,Pediatric Hepato-Gastroenterology and Nutrition Unit, Hôpital Femme Mère Enfant de Lyon, Hospices Civils de Lyon, Lyon, Bron, France.
| |
Collapse
|
27
|
Hooper AJ, Robertson K, Champain D, Hua J, Song S, Parhofer KG, Barrett PHR, van Bockxmeer FM, Burnett JR. Lipoprotein metabolism in an apoB-80 familial hypobetalipoproteinemia heterozygote. Clin Biochem 2016; 49:720-722. [DOI: 10.1016/j.clinbiochem.2016.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 02/17/2016] [Accepted: 02/19/2016] [Indexed: 11/30/2022]
|
28
|
Rimbert A, Pichelin M, Lecointe S, Marrec M, Le Scouarnec S, Barrak E, Croyal M, Krempf M, Le Marec H, Redon R, Schott JJ, Magré J, Cariou B. Identification of novel APOB mutations by targeted next-generation sequencing for the molecular diagnosis of familial hypobetalipoproteinemia. Atherosclerosis 2016; 250:52-6. [PMID: 27179706 DOI: 10.1016/j.atherosclerosis.2016.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/08/2016] [Accepted: 04/08/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND AIMS Familial hypobetalipoproteinemia (FHBL) is a co-dominant disorder characterized by decreased plasma levels of LDL-cholesterol and apolipoprotein B (ApoB). Currently, genetic diagnosis in FHBL relies largely on Sanger sequencing to identify APOB and PCSK9 gene mutations and on western blotting to detect truncated ApoB species. METHODS Here, we applied targeted enrichment and next-generation sequencing (NGS) on a panel of three FHBL genes and two abetalipoproteinemia genes (APOB, PCSK9, ANGPTL3, MTTP and SAR1B). RESULTS In this study, we identified five likely pathogenic heterozygous rare variants. These include four novel nonsense mutations in APOB (p.Gln845*, p.Gln2571*, p.Cys2933* and p.Ser3718*) and a rare variant in PCSK9 (Minor Allele Frequency <0.1%). The affected family members tested were shown to be carriers, suggesting co-segregation with low LDL-C. CONCLUSIONS Our study further demonstrates that NGS is a reliable and practical approach for the molecular screening of FHBL-causative genes that may provide a mean for deciphering the genetic basis in FHBL.
Collapse
Affiliation(s)
- Antoine Rimbert
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France
| | - Matthieu Pichelin
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France; CIC Thorax, CHU Nantes, l'institut du Thorax, Nantes, F-44000, France
| | - Simon Lecointe
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France
| | - Marie Marrec
- CHU Nantes, l'institut du Thorax, Nantes, F-44000, France; CIC Thorax, CHU Nantes, l'institut du Thorax, Nantes, F-44000, France
| | - Solena Le Scouarnec
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France
| | - Elias Barrak
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France
| | - Mikael Croyal
- Centre de Recherche en Nutrition Humaine de l'Ouest (CRNHO, INRA UMR1280), Nantes, F-44093, France
| | - Michel Krempf
- Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France; Centre de Recherche en Nutrition Humaine de l'Ouest (CRNHO, INRA UMR1280), Nantes, F-44093, France
| | - Hervé Le Marec
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France
| | - Richard Redon
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France
| | - Jean-Jacques Schott
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France.
| | - Jocelyne Magré
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France
| | - Bertrand Cariou
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France; CNRS, UMR 6291, Nantes, F-44000, France; Université de Nantes, Nantes, F-44000, France; CHU Nantes, l'institut du Thorax, Nantes, F-44000, France; CIC Thorax, CHU Nantes, l'institut du Thorax, Nantes, F-44000, France.
| |
Collapse
|
29
|
Miller SA, Hooper AJ, Mantiri GA, Marais D, Tanyanyiwa DM, McKnight J, Burnett JR. Novel APOB missense variants, A224T and V925L, in a black South African woman with marked hypocholesterolemia. J Clin Lipidol 2016; 10:604-9. [PMID: 27206948 DOI: 10.1016/j.jacl.2016.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/06/2016] [Accepted: 01/25/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND One genetic cause of markedly low plasma concentrations of apolipoprotein (apo) B and low density lipoprotein (LDL)-cholesterol is familial hypobetalipoproteinemia. OBJECTIVE We aimed to determine the molecular basis for the marked hypocholesterolemia consistent with heterozygous familial hypobetalipoproteinemia in a black female subject of Xhosa lineage. METHODS Coding regions of APOB, MTTP, PCSK9,ANGPTL3, SAR1B and APOC3 were sequenced, and APOE was genotyped. COS-7 cells were transfected with plasmids containing apoB variants. Western blotting was used to detect cellular and secreted apoB, and co-immunoprecipitation performed to assess binding with the microsomal triglyceride transfer protein (MTP). RESULTS Sequence analysis of the APOB gene revealed her to be heterozygous for two novel variants, c.751G>A (A224T) and c.2854G>C (V925L). She was also homozygous for the APOEε2 allele, and did not carry a PCSK9 loss-of-function mutation. Although Ala(224) is within the postulated MTP binding region in apoB, it is not conserved among mammalian species. Subsequent genotyping showed that Ala224Thr is found in a southern African population (n=654) with an allele frequency of 1.15% and is not associated with plasma lipid levels. Val(925), like Ala(224), is within the N-terminal 1000 amino acids required for lipoprotein assembly, but was not found in the population screen. However, in vitro studies showed that apoB V925L did not affect apoB48 production or secretion nor have a deleterious effect on MTP interaction with apoB. CONCLUSION Taken together, this suggests that the hypocholesterolemia in our case may be a result of being homozygous for APOEε2 with a low baseline cholesterol.
Collapse
Affiliation(s)
- Sharon A Miller
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Amanda J Hooper
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, Australia; Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital and Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - George A Mantiri
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - David Marais
- Division of Chemical Pathology, University of Cape Town, National Health Laboratory Service and MRC Cape Heart Group, Cape Town, South Africa
| | - Donald M Tanyanyiwa
- University of Witwatersrand and National Health Laboratory Service and Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - James McKnight
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA
| | - John R Burnett
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia; Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital and Fiona Stanley Hospital, Perth, Western Australia, Australia.
| |
Collapse
|
30
|
Hooper AJ, Heeks L, Robertson K, Champain D, Hua J, Song S, Parhofer KG, Barrett PHR, van Bockxmeer FM, Burnett JR. Lipoprotein Metabolism in APOB L343V Familial Hypobetalipoproteinemia. J Clin Endocrinol Metab 2015; 100:E1484-90. [PMID: 26323024 DOI: 10.1210/jc.2015-2731] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
CONTEXT Familial hypobetalipoproteinemia (FHBL) is a codominant disorder of lipoprotein metabolism characterized by decreased plasma concentrations of low-density lipoprotein (LDL)-cholesterol and apolipoprotein B (apoB). OBJECTIVE The objective was to examine the effect of heterozygous APOB L343V FHBL on postprandial triglyceride-rich lipoprotein (TRL) and fasting lipoprotein metabolism. METHODS Plasma incremental area under the curve apoB-48 and apoB-48 kinetics were determined after ingestion of a standardized oral fat load using compartmental modeling. Very low-density lipoprotein (VLDL)-, intermediate-density lipoprotein (IDL)-, and LDL-apoB kinetics were determined in the fasting state using stable isotope methods and compartmental modeling. RESULTS The postprandial incremental area under the curve (0-10 h) in FHBL subjects (n = 3) was lower for large TRL-triglyceride (-77%; P < .0001), small TRL-cholesterol (-83%; P < .001), small TRL-triglyceride (-88%; P < .001), and for plasma triglyceride (-70%; P < .01) and apoB (-63%; P < .0001) compared with controls. Compartmental analysis showed that apoB-48 production was lower (-91%; P < .05) compared with controls. VLDL-apoB concentrations in FHBL subjects (n = 2) were lower by more than 75% compared with healthy, normolipidemic control subjects (P < .01). The VLDL-apoB fractional catabolic rate (FCR) was more than 5-fold higher in the FHBL subjects (P = .07). ApoB production rates and IDL- and LDL-apoB FCRs were not different between FHBL subjects and controls. CONCLUSIONS We conclude that when compared to controls, APOB L343V FHBL heterozygotes show lower TRL production with normal postprandial TRL particle clearance. In contrast, VLDL-apoB production was normal, whereas the FCR was higher in heterozygotes compared with lean control subjects. These mechanisms account for the marked hypolipidemic state observed in these FHBL subjects.
Collapse
MESH Headings
- Adult
- Amino Acid Substitution
- Apolipoprotein B-48/blood
- Apolipoprotein B-48/metabolism
- Apolipoproteins B/blood
- Apolipoproteins B/genetics
- Apolipoproteins B/metabolism
- Diet, High-Fat/adverse effects
- Down-Regulation
- Female
- Heterozygote
- Humans
- Hypobetalipoproteinemia, Familial, Apolipoprotein B/blood
- Hypobetalipoproteinemia, Familial, Apolipoprotein B/genetics
- Hypobetalipoproteinemia, Familial, Apolipoprotein B/metabolism
- Lipoproteins/blood
- Lipoproteins/metabolism
- Lipoproteins, IDL/blood
- Lipoproteins, IDL/metabolism
- Lipoproteins, VLDL/blood
- Lipoproteins, VLDL/metabolism
- Male
- Meals
- Middle Aged
- Models, Biological
- Mutation
- Postprandial Period
- Triglycerides/blood
- Triglycerides/metabolism
Collapse
Affiliation(s)
- Amanda J Hooper
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - Liesl Heeks
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - Ken Robertson
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - Danie Champain
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - Jianmin Hua
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - Swithin Song
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - Klaus G Parhofer
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - P Hugh R Barrett
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - Frank M van Bockxmeer
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| | - John R Burnett
- Department of Clinical Biochemistry (A.J.H., L.H., K.R., F.M.v.B., J.R.B.), PathWest Laboratory Medicine WA, Royal Perth Hospital, Perth WA 6000, Australia; School of Medicine and Pharmacology (A.J.H., D.C., P.H.R.B., J.R.B.), and School of Pathology and Laboratory Medicine (A.J.H., K.R.), University of Western Australia, Crawley WA 6009, Australia; Department of Radiology (J.H., S.S.), Royal Perth Hospital, Perth WA 6000, Australia; Medical Department II (K.G.P.), Grosshadern, University of Munich, 81377 Munich, Germany; and School of Surgery (F.M.v.B.), University of Western Australia, Crawley WA 6009, Australia
| |
Collapse
|
31
|
Burnett JR, Hooper AJ. Vitamin E and oxidative stress in abetalipoproteinemia and familial hypobetalipoproteinemia. Free Radic Biol Med 2015; 88:59-62. [PMID: 26086616 DOI: 10.1016/j.freeradbiomed.2015.05.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/07/2015] [Accepted: 05/26/2015] [Indexed: 01/13/2023]
Abstract
Abetalipoproteinemia (ABL) and familial hypobetalipoproteinemia (FHBL) are genetic diseases characterized by low density lipoprotein deficiency. ABL presents early in life with the gastroenterological manifestations of fat malabsorption, steatorrhea, and failure to thrive, and later in life, with progressive ophthalmopathy and neuropathy as a result of deficiency of the fat-soluble vitamins A and E. Heterozygous FHBL subjects are usually asymptomatic, but may develop fatty liver disease. In homozygous (compound heterozygous) FHBL, the clinical and biochemical features are indistinguishable from those of ABL and treatment recommendations are the same: dietary fat restriction to prevent steatorrhea, and long-term high-dose vitamin E and A supplementation to prevent or at least slow the progression of neuromuscular and retinal degenerative disease. Despite their low plasma vitamin E levels, individuals with heterozygous FHBL do not require vitamin E supplementation. There are conflicting reports on whether increased oxidative stress is seen in ABL; these differences may relate to the small size of patient groups as well as differences in patient age and dose of vitamin E supplementation, or the contribution from dietary sources of vitamin E. High density lipoproteins in ABL appear to be severely oxidized yet able to inhibit platelet aggregation by binding to scavenger receptor B1. We review the role of vitamin E and oxidative stress in ABL and FHBL.
Collapse
Affiliation(s)
- John R Burnett
- Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Australia; School of Medicine & Pharmacology, University of Western Australia, Perth, Australia.
| | - Amanda J Hooper
- Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Australia; School of Medicine & Pharmacology, University of Western Australia, Perth, Australia; School of Pathology & Laboratory Medicine, University of Western Australia, Perth, Australia
| |
Collapse
|
32
|
Agarwala A, Jones P, Nambi V. The role of antisense oligonucleotide therapy in patients with familial hypercholesterolemia: risks, benefits, and management recommendations. Curr Atheroscler Rep 2015; 17:467. [PMID: 25398643 DOI: 10.1007/s11883-014-0467-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antisense oligonucleotide therapy is a promising approach for the treatment of a broad variety of medical conditions. It functions at the cellular level by interfering with RNA function, often leading to degradation of specifically targeted abnormal gene products implicated in the disease process. Mipomersen is a novel antisense oligonucleotide directed at apolipoprotein (apoB)-100, the primary apolipoprotein associated with low-density lipoprotein cholesterol (LDL-C), which has recently been approved for the treatment of familial hypercholesterolemia. A number of clinical studies have demonstrated its efficacy in lowering LDL-C and apoB levels in patients with elevated LDL-C despite maximal medical therapy using conventional lipid-lowering agents. This review outlines the risks and benefits of therapy and provides recommendations on the use of mipomersen.
Collapse
Affiliation(s)
- Anandita Agarwala
- Department of Medicine, Baylor College of Medicine, Michael E. DeBakey Veterans Affairs Medical Center, One Baylor Plaza, Houston, TX, 77030, USA,
| | | | | |
Collapse
|
33
|
Kawashiri MA, Tada H, Hashimoto M, Taniyama M, Nakano T, Nakajima K, Inoue T, Mori M, Nakanishi C, Konno T, Hayashi K, Nohara A, Inazu A, Koizumi J, Ishihara H, Kobayashi J, Hirano T, Mabuchi H, Yamagishi M. Extreme Contrast of Postprandial Remnant-Like Particles Formed in Abetalipoproteinemia and Homozygous Familial Hypobetalipoproteinemia. JIMD Rep 2015; 22:85-94. [PMID: 25763510 DOI: 10.1007/8904_2015_415] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/25/2015] [Accepted: 01/27/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Familial hypobetalipoproteinemia (FHBL) and abetalipoproteinemia (ABL) are rare inherited forms of hypolipidemia. Their differential diagnosis is important for predicting of the prognosis and selecting appropriate therapy. MATERIALS AND METHODS Genetic analysis was performed in two patients with primary hypocholesterolemia born from consanguineous parents. The oral fat tolerance test (OFTT) was performed in one patient with FHBL (apoB-87.77) and one with ABL as well as in four normal control subjects. After overnight fasting, blood samples were drawn. Serum lipoprotein and remnant-like particle (RLP) fractions were determined by HPLC analysis. RESULTS Both patients with homozygous FHBL were asymptomatic probably because of preserved levels of fat-soluble vitamins, especially vitamin E. The patients with FHBL were homozygous because of novel apoB-83.52 and apoB-87.77 mutations, and although one of them (apoB-87.77) had fatty liver disease, microscopic findings suggesting nonalcoholic steatohepatitis were absent. Fasting apoB-48 and RLP-triglyceride levels in the patient with homozygous FHBL, which were similar to those in normal control subjects, increased after OFTT both in normal control subjects and the patient with FHBL but not in the patient with ABL, suggesting that the fat load administered was absorbed only in the patient with FHBL. CONCLUSION Although lipid levels in the patients with homozygous FHBL and ABL were comparable, fasting, postoral fat loading of apoB-48, as well as RLP-triglyceride levels, may help in the differential diagnosis of FHBL and ABL and provide a prompt diagnosis using genetic analysis in the future.
Collapse
Affiliation(s)
- Masa-Aki Kawashiri
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, 13-1 Takara-machi, Kanazawa, 920-8641, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hooper AJ, Burnett JR, Watts GF. Contemporary Aspects of the Biology and Therapeutic Regulation of the Microsomal Triglyceride Transfer Protein. Circ Res 2015; 116:193-205. [DOI: 10.1161/circresaha.116.304637] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Amanda J. Hooper
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA (A.J.H., J.R.B.), School of Medicine and Pharmacology (A.J.H., J.R.B., G.F.W.), School of Pathology and Laboratory Medicine (A.J.H), and Lipid Disorders Clinic, Cardiovascular Medicine (J.R.B., G.F.W), Royal Perth Hospital, University of Western Australia, Perth, Western Australia, Australia
| | - John R. Burnett
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA (A.J.H., J.R.B.), School of Medicine and Pharmacology (A.J.H., J.R.B., G.F.W.), School of Pathology and Laboratory Medicine (A.J.H), and Lipid Disorders Clinic, Cardiovascular Medicine (J.R.B., G.F.W), Royal Perth Hospital, University of Western Australia, Perth, Western Australia, Australia
| | - Gerald F. Watts
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA (A.J.H., J.R.B.), School of Medicine and Pharmacology (A.J.H., J.R.B., G.F.W.), School of Pathology and Laboratory Medicine (A.J.H), and Lipid Disorders Clinic, Cardiovascular Medicine (J.R.B., G.F.W), Royal Perth Hospital, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
35
|
Burnett JR, Bell DA, Hooper AJ, Hegele RA. Clinical utility gene card for: Familial hypobetalipoproteinaemia (APOB)--Update 2014. Eur J Hum Genet 2014; 23:ejhg2014225. [PMID: 25335495 DOI: 10.1038/ejhg.2014.225] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/08/2014] [Accepted: 09/19/2014] [Indexed: 01/08/2023] Open
Affiliation(s)
- John R Burnett
- 1] Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia [2] School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Damon A Bell
- 1] Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia [2] School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Amanda J Hooper
- 1] Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia [2] School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia [3] School of Pathology & Laboratory Medicine, University of Western Australia, Perth, Western Australia, Australia
| | | |
Collapse
|
36
|
Clinical utility gene card for: Abetalipoproteinaemia--Update 2014. Eur J Hum Genet 2014; 23:ejhg2014224. [PMID: 25335492 DOI: 10.1038/ejhg.2014.224] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/08/2014] [Accepted: 09/19/2014] [Indexed: 11/08/2022] Open
|