1
|
Rimbert A, Wargny M, Cariou B. Reply to "The association between primary hypobetalipoproteinemia and hepatic diseases: evidence from genetic studies". J Hepatol 2025; 82:e50-e51. [PMID: 39299457 DOI: 10.1016/j.jhep.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Affiliation(s)
- Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France
| | - 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
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000 Nantes, France.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Ramos Bachiller B, Luque-Ramírez M, Rodríguez-Jiménez C, Arrieta Blanco FJ. Familial ApoB-specific familial hypobetalipoproteinemia in a patient with non-classical congenital adrenal hyperplasia. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:128-132. [PMID: 38195282 DOI: 10.1016/j.arteri.2023.12.002] [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: 09/25/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024]
Abstract
Familial hypobetalipoproteinaemia is a disorder of lipid metabolism characterized by low levels of total cholesterol, low-density lipoprotein cholesterol and apolipoprotein B. ApoB-related familial hypolipoproteinemia is an autosomal condition with a codominance inheritance pattern. Non-classical congenital adrenal hyperplasia is an autosomal recessive disorder due to mutations in the CYP21A2, a gene encoding for the enzyme 21-hydroxylase, which results in an androgen excess production from adrenal source. We here present the case of a 25-year-old woman with NCAH showing decreased levels of total-cholesterol, low-density lipoprotein cholesterol and triglycerides. Her parent had digestive symptoms and severe hepatic steatosis with elevated liver enzymes, as well as decreased levels of total and low-density lipoprotein cholesterol. A genetic-molecular study of the proband identified a mutation in the APOB gene, which allowed a diagnosis of heterozygous ApoB-related hypolipoproteinaemia to be made.
Collapse
Affiliation(s)
- Beatriz Ramos Bachiller
- Servicio de Endocrinología y Nutrición, Complejo Asistencial Universitario de León, Castilla-León, España.
| | - Manuel Luque-Ramírez
- Servicio de Endocrinología y Nutrición, Hospital Universitario Ramón y Cajal, Madrid, España; Grupo de Investigación en Diabetes, Obesidad y Reproducción Humana, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), CIBER de Diabetes y Enfermedades Metabólicas asociadas, y Universidad de Alcalá, Madrid, España
| | - Carmen Rodríguez-Jiménez
- Department of Genetics of Metabolic Diseases, Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, Madrid, España
| | - Francisco J Arrieta Blanco
- Servicio de Endocrinología y Nutrición, Hospital Universitario Ramón y Cajal, Madrid, España; Centro de Referencia Nacional (CSUR) y Europeo (MetabERN) para Enfermedades Metabólicas Hereditarias, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, España
| |
Collapse
|
5
|
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
|
6
|
Molk N, Bitenc M, Urlep D, Zerjav Tansek M, Bertok S, Trebusak Podkrajsek K, Sustar U, Kovac J, Battelino T, Debeljak M, Groselj U. Non-alcoholic fatty liver disease in a pediatric patient with heterozygous familial hypobetalipoproteinemia due to a novel APOB variant: a case report and systematic literature review. Front Med (Lausanne) 2023; 10:1106441. [PMID: 37384046 PMCID: PMC10293746 DOI: 10.3389/fmed.2023.1106441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/09/2023] [Indexed: 06/30/2023] Open
Abstract
Background Familial hypobetalipoproteinemia (FHBL) is an autosomal semi-dominant disorder usually caused by variants in the APOB gene that frequently interferes with protein length. Clinical manifestations include malabsorption, non-alcoholic fatty liver disease, low levels of lipid-soluble vitamins, and neurological, endocrine, and hematological dysfunction. Methods Genomic DNA was isolated from the blood samples of the pediatric patient with hypocholesterolemia and his parents and brother. Next-generation sequencing (NGS) was performed, and an expanded dyslipidemia panel was employed for genetic analysis. In addition, a systematic review of the literature on FHBL heterozygous patients was performed. Case report Genetic investigation revealed the presence of a heterozygous variant in the APOB (NM_000384.3) gene c.6624dup[=], which changes the open reading frame and leads to early termination of translation into the p.Leu2209IlefsTer5 protein (NP_000375.3). The identified variant was not previously reported. Familial segregation analysis confirmed the variant in the mother of the subject, who also has a low level of low-density lipoprotein and non-alcoholic fatty liver disease. We have introduced therapy that includes limiting fats in the diet and adding lipid-soluble vitamins E, A, K, and D and calcium carbonate. We reported 35 individuals with APOB gene variations linked to FHBL in the systematic review. Conclusion We have identified a novel pathogenic variant in the APOB gene causing FHBL in pediatric patients with hypocholesterolemia and fatty liver disease. This case illustrates the importance of genetic testing for dyslipidemias in patients with significant decreases in plasma cholesterol as we can avoid damaging neurological and ophthalmological effects by sufficient vitamin supplementation and regular follow-ups.
Collapse
Affiliation(s)
- Neza Molk
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
| | - Mojca Bitenc
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
| | - Darja Urlep
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Gastroenterology, Hepatology and Nutrition, University Children's Hospital Ljubljana, University Medical Center, Ljubljana, Slovenia
| | - Mojca Zerjav Tansek
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Sara Bertok
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katarina Trebusak Podkrajsek
- Department of Gastroenterology, Hepatology and Nutrition, University Children's Hospital Ljubljana, University Medical Center, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Ursa Sustar
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Jernej Kovac
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Marusa Debeljak
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children's Hospital, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Urh Groselj
- Department of Pediatric Endocrinology, Diabetes and Metabolism, University Medical Center-University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
7
|
Soldatova YV, Faingold II, Poletaeva DA, Kozlov AV, Emel'yanova NS, Khodos II, Chernyaev DA, Kurmaz SV. Design and Investigation of New Water-Soluble Forms of α-Tocopherol with Antioxidant and Antiglycation Activity Using Amphiphilic Copolymers of N-Vinylpyrrolidone. Pharmaceutics 2023; 15:pharmaceutics15051388. [PMID: 37242630 DOI: 10.3390/pharmaceutics15051388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Water-soluble forms of α-tocopherol (TP) as an effective antioxidant were obtained by encapsulating it into nanoparticles (NPs) of amphiphilic copolymers of N-vinylpyrrolidone with triethylene glycol dimethacrylate (CPL1-TP) and N-vinylpyrrolidone with hexyl methacrylate and triethylene glycol dimethacrylate (CPL2-TP) synthesized by radical copolymerization in toluene. The hydrodynamic radii of NPs loaded with TP (3.7 wt% per copolymers) were typically ca. 50 or 80 nm depending on copolymer composition, media, and temperature. Characterization of NPs was accomplished by transmission electron microscopy (TEM), IR-, and 1H NMR spectroscopy. Quantum chemical modeling showed that TP molecules are capable to form hydrogen bonds with donor groups of the copolymer units. High antioxidant activity of both obtained forms of TP has been found by the thiobarbituric acid reactive species and chemiluminescence assays. CPL1-TP and CPL2-TP effectively inhibited the process of spontaneous lipid peroxidation as well as α-tocopherol itself. The IC50 values of luminol chemiluminescence inhibition were determined. Antiglycation activity against vesperlysine and pentosidine-like AGEs of TP water-soluble forms was shown. The developed NPs of TP are promising as materials with antioxidant and antiglycation activity and can be used in various biomedical applications.
Collapse
Affiliation(s)
- Yuliya V Soldatova
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Academician Semenov av., 1, 142432 Chernogolovka, Russia
| | - Irina I Faingold
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Academician Semenov av., 1, 142432 Chernogolovka, Russia
| | - Darya A Poletaeva
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Academician Semenov av., 1, 142432 Chernogolovka, Russia
| | - Alexei V Kozlov
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Academician Semenov av., 1, 142432 Chernogolovka, Russia
| | - Nina S Emel'yanova
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Academician Semenov av., 1, 142432 Chernogolovka, Russia
| | - Igor I Khodos
- Institute of Microelectronics Technology and High-Purity Materials, Russian Academy of Sciences, Institutskaya Street, 6, 142432 Chernogolovka, Russia
| | - Dmitry A Chernyaev
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Academician Semenov av., 1, 142432 Chernogolovka, Russia
| | - Svetlana V Kurmaz
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Academician Semenov av., 1, 142432 Chernogolovka, Russia
| |
Collapse
|
8
|
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
|
9
|
Guidance for the diagnosis and treatment of hypolipidemia disorders. J Clin Lipidol 2022; 16:797-812. [PMID: 36243606 DOI: 10.1016/j.jacl.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 11/15/2022]
|
10
|
Sasaki K, Tada H, Kawashiri MA, Ito T. Case report: Unusual coexistence between familial hypercholesterolemia and familial hypobetalipoproteinemia. Front Cardiovasc Med 2022; 9:942772. [PMID: 36003908 PMCID: PMC9393375 DOI: 10.3389/fcvm.2022.942772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
Type 1 familial hypobetalipoproteinemia (FHBL1), characterized by low levels of apolipoprotein B (ApoB)-containing lipoproteins, elevation of transaminases, and hepatic steatosis, is a rare disease the prevalence of which is 1 in 3,000 among general population. Here we report an extremely rare family where phenotypes of familial hypercholesterolemia (FH) are canceled by coexistence of FHBL1 caused by an truncating mutation in apolipoprotein B (APOB).
Collapse
Affiliation(s)
- Kei Sasaki
- Department of Internal Medicine, Self-Defense Forces Central Hospital, Tokyo, Japan
- Division of Anti-aging, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Hayato Tada
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
- *Correspondence: Hayato Tada
| | - Masa-aki Kawashiri
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Toshimitsu Ito
- Department of Internal Medicine, Self-Defense Forces Central Hospital, Tokyo, Japan
| |
Collapse
|
11
|
Williams L, Baker-Smith CM, Bolick J, Carter J, Kirkpatrick C, Ley SL, Peterson AL, Shah AS, Sikand G, Ware AL, Wilson DP. Nutrition interventions for youth with dyslipidemia an national lipid association clinical perspective. J Clin Lipidol 2022; 16:776-796. [DOI: 10.1016/j.jacl.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022]
|
12
|
Kudo T, Sasaki K, Tada H. Familial hypobetalipoproteinemia caused by homozygous loss-of-function mutations in PCSK9: A case report. J Clin Lipidol 2022; 16:596-600. [DOI: 10.1016/j.jacl.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/07/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022]
|
13
|
Xu R, Pan J, Zhou W, Ji G, Dang Y. Recent advances in lean NAFLD. Biomed Pharmacother 2022; 153:113331. [PMID: 35779422 DOI: 10.1016/j.biopha.2022.113331] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/09/2023] Open
Abstract
As the predominant type of chronic liver disease, the growing prevalence of nonalcoholic fatty liver disease (NAFLD) has become a concern worldwide. Although obesity plays the most pivotal role in NAFLD, approximately 10-20% of individuals with NAFLD who are not overweight or obese (BMI < 25 kg/m2, or BMI < 23 kg/m2 in Asians) have "lean NAFLD." Lean individuals with NAFLD have a lower prevalence of diabetes, hypertension, hypertriglyceridemia, central obesity, and metabolic syndrome than nonlean individuals with NAFLD, but higher fibrosis scores and rates of cardiovascular morbidity and all-cause mortality in advanced stages. The pathophysiological mechanisms of lean NAFLD remain poorly understood. Studies have shown that lean NAFLD is more correlated with factors such as environmental, genetic susceptibility, and epigenetic regulation. This review will examine the way in which the research progress and characteristic of lean NAFLD, and explore the function of epigenetic modification to provide the basis for the clinical treatment and diagnosis of lean NAFLD.
Collapse
Affiliation(s)
- Ruohui Xu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jiashu Pan
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Department of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Yanqi Dang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
14
|
Cefalù AB, Spina R, Noto D, Rabacchi C, Giammanco A, Simone ML, Brucato F, Scrimali C, Gueli-Alletti MG, Barbagallo CM, Tarugi P, Averna MR. Comparison of two polygenic risk score to identify non-monogenic primary hypocholesterolemias in a large cohort of Italian hypocholesterolemic subjects. J Clin Lipidol 2022; 16:530-537. [DOI: 10.1016/j.jacl.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/08/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
|
15
|
Vanhoye X, Janin A, Caillaud A, Rimbert A, Venet F, Gossez M, Dijk W, Marmontel O, Nony S, Chatelain C, Durand C, Lindenbaum P, Rieusset J, Cariou B, Moulin P, Di Filippo M. APOB CRISPR-Cas9 Engineering in Hypobetalipoproteinemia: A Promising Tool for Functional Studies of Novel Variants. Int J Mol Sci 2022; 23:4281. [PMID: 35457099 PMCID: PMC9030618 DOI: 10.3390/ijms23084281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 02/01/2023] Open
Abstract
Hypobetalipoproteinemia is characterized by LDL-cholesterol and apolipoprotein B (apoB) plasma levels below the fifth percentile for age and sex. Familial hypobetalipoproteinemia (FHBL) is mostly caused by premature termination codons in the APOB gene, a condition associated with fatty liver and steatohepatitis. Nevertheless, many families with a FHBL phenotype carry APOB missense variants of uncertain significance (VUS). We here aimed to develop a proof-of-principle experiment to assess the pathogenicity of VUS using the genome editing of human liver cells. We identified a novel heterozygous APOB-VUS (p.Leu351Arg), in a FHBL family. We generated APOB knock-out (KO) and APOB-p.Leu351Arg knock-in Huh7 cells using CRISPR-Cas9 technology and studied the APOB expression, synthesis and secretion by digital droplet PCR and ELISA quantification. The APOB expression was decreased by 70% in the heterozygous APOB-KO cells and almost abolished in the homozygous-KO cells, with a consistent decrease in apoB production and secretion. The APOB-p.Leu351Arg homozygous cells presented with a 40% decreased APOB expression and undetectable apoB levels in cellular extracts and supernatant. Thus, the p.Leu351Arg affected the apoB secretion, which led us to classify this new variant as likely pathogenic and to set up a hepatic follow-up in this family. Therefore, the functional assessment of APOB-missense variants, using gene-editing technologies, will lead to improvements in the molecular diagnosis of FHBL and the personalized follow-up of these patients.
Collapse
Affiliation(s)
- Xavier Vanhoye
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
| | - Alexandre Janin
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Université Claude Bernard Lyon 1, Université de Lyon, F-69008 Lyon, France
| | - Amandine Caillaud
- Institut du Thorax, Nantes Université, CHU Nantes, CNRS, INSERM, F-44000 Nantes, France; (A.C.); (B.C.)
| | - Antoine Rimbert
- Institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (A.R.); (W.D.); (P.L.)
| | - Fabienne Venet
- Laboratoire d’Immunologie, Edouard Herriot Hospital, Hospices Civils de Lyon, F-69437 Lyon, France; (F.V.); (M.G.)
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, F-69364 Lyon, France
| | - Morgane Gossez
- Laboratoire d’Immunologie, Edouard Herriot Hospital, Hospices Civils de Lyon, F-69437 Lyon, France; (F.V.); (M.G.)
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, F-69364 Lyon, France
| | - Wieneke Dijk
- Institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (A.R.); (W.D.); (P.L.)
| | - Oriane Marmontel
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| | - Séverine Nony
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
| | - Charlotte Chatelain
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
| | - Christine Durand
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| | - Pierre Lindenbaum
- Institut du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France; (A.R.); (W.D.); (P.L.)
| | - Jennifer Rieusset
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| | - Bertrand Cariou
- Institut du Thorax, Nantes Université, CHU Nantes, CNRS, INSERM, F-44000 Nantes, France; (A.C.); (B.C.)
| | - Philippe Moulin
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
- Fédération d’Endocrinologie, Maladies Métaboliques, Diabète et Nutrition, Hôpital Louis Pradel, Hospices Civils de Lyon, F-69677 Bron, France
| | - Mathilde Di Filippo
- Service de Biochimie et de Biologie Moléculaire, Laboratoire de Biologie Médicale MultiSites, Hospices Civils de Lyon, F-69677 Bron, France; (X.V.); (A.J.); (O.M.); (S.N.); (C.C.)
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, F-69364 Lyon, France; (C.D.); (J.R.); (P.M.)
| |
Collapse
|
16
|
Welty FK. Familial hypobetalipoproteinemia and abetalipoproteinemia. CHOLESTEROL 2022:465-480. [DOI: 10.1016/b978-0-323-85857-1.00026-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Wilson PWF, Jacobson TA, Martin SS, Jackson EJ, Le NA, Davidson MH, Vesper HW, Frikke-Schmidt R, Ballantyne CM, Remaley AT. Lipid measurements in the management of cardiovascular diseases: Practical recommendations a scientific statement from the national lipid association writing group. J Clin Lipidol 2021; 15:629-648. [PMID: 34802986 DOI: 10.1016/j.jacl.2021.09.046] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/31/2023]
Abstract
Lipoprotein measurements are pivotal in the management of patients at risk for atherosclerotic coronary heart disease (CHD) with myocardial infarction and coronary death as the main outcomes, and for atherosclerotic cardiovascular disease (ASCVD), which includes CHD and stroke. Recent developments and changes in guidelines affect optimization of using lipid measures as cardiovascular biomarkers. This scientific statement reviews the pre-analytical, analytical, post-analytical, and clinical aspects of lipoprotein measurements. Highlights include the following: i) It is acceptable to screen with nonfasting lipids. ii) non-high-density lipoprotein HDL-cholesterol (non-HDL-C) is measured reliably in either the fasting or the nonfasting state and can effectively guide ASCVD prevention. iii) low density lipoprotein cholesterol (LDL-C) can be estimated from total cholesterol, high density lipoprotein cholesterol (HDL-C), and triglyceride (TG) measurements. For patients with LDL-C>100 mg/dL and TG ≤150 mg/dL it is reasonable to use the Friedewald formula. However, for those with TG 150-400 mg/dL the Friedewald formula for LDL-C estimation is less accurate. The Martin/Hopkins method is recommended for LDL-C estimation throughout the range of LDL-C levels and up to TG levels of 399 mg/dL. For TG levels ≥400 mg/dL LDL-C estimating equations are currently not recommended and newer methods are being evaluated. iv) When LDL-C or TG screening results are abnormal the clinician should consider obtaining fasting lipids. v) Advanced lipoprotein tests using apolipoprotein B (apoB), LDL Particle Number (LDL-P) or remnant cholesterol may help to guide therapeutic decisions in select patients, but data are limited for patients already on lipid lowering therapy with low LDL-C levels. Better harmonization of advanced lipid measurement methods is needed. Lipid measurements are recommended 4-12 weeks after a change in lipid treatment. Lipid laboratory reports should denote desirable values and specifically identify extremely elevated LDL-C levels (≥190 mg/dL at any age or ≥160 mg/dL in children) as severe hypercholesterolemia. Potentially actionable abnormal lipid test results, including fasting triglycerides (TG) ≥500 mg/dL, should be reported as hypertriglyceridemia. Appropriate use and reporting of lipid tests should improve their utility in the management of persons at high risk for ASCVD events.
Collapse
Affiliation(s)
- Peter W F Wilson
- Emory University School of Medicine, Atlanta, GA, United States; Atlanta Veterans Affairs Medical Center, Atlanta, GA, United States.
| | | | - Seth S Martin
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | - N-Anh Le
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, United States
| | | | - Hubert W Vesper
- Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Ruth Frikke-Schmidt
- Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Alan T Remaley
- National Heart, Lung and Blood Institute, Bethesda, MD, United States
| |
Collapse
|
18
|
Buryska S, Ahn JC, Allen AM, Simha V, Simonetto DA. Familial Hypobetalipoproteinemia: An Underrecognized Cause of Lean NASH. Hepatology 2021; 74:2897-2898. [PMID: 34091928 DOI: 10.1002/hep.31988] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/01/2021] [Accepted: 05/28/2021] [Indexed: 12/08/2022]
Affiliation(s)
- Seth Buryska
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Joseph C Ahn
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Vinaya Simha
- Division of Endocrinology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
19
|
Successful Genetic Screening and Creating Awareness of Familial Hypercholesterolemia and Other Heritable Dyslipidemias in the Netherlands. Genes (Basel) 2021; 12:genes12081168. [PMID: 34440342 PMCID: PMC8392502 DOI: 10.3390/genes12081168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/17/2022] Open
Abstract
The genetic screening program for familial hypercholesterolemia (FH) in the Netherlands, which was embraced by the Dutch Ministry of Health from 1994 to 2014, has led to twenty years of identification of at least 1500 FH cases per year. Although funding by the government was terminated in 2014, the approach had proven its effectiveness and had built the foundation for the development of more sophisticated diagnostic tools, clinical collaborations, and new molecular-based treatments for FH patients. As such, the community was driven to continue the program, insurance companies were convinced to collaborate, and multiple approaches were launched to find new index cases with FH. Additionally, the screening was extended, now also including other heritable dyslipidemias. For this purpose, a diagnostic next-generation sequencing (NGS) panel was developed, which not only comprised the culprit LDLR, APOB, and PCSK9 genes, but also 24 other genes that are causally associated with genetic dyslipidemias. Moreover, the NGS technique enabled further optimization by including pharmacogenomic genes in the panel. Using such a panel, more patients that are prone to cardiovascular diseases are being identified nowadays and receive more personalized treatment. Moreover, the NGS output teaches us more and more about the dyslipidemic landscape that is less straightforward than we originally thought. Still, continuous progress is being made that underlines the strength of genetics in dyslipidemia, such as discovery of alternative genomic pathogenic mechanisms of disease development and polygenic contribution.
Collapse
|
20
|
Monogenic and polygenic causes of low and extremely low LDL-C levels in patients referred to specialty lipid clinics: Genetics of low LDL-C. J Clin Lipidol 2021; 15:658-664. [PMID: 34340953 DOI: 10.1016/j.jacl.2021.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/14/2021] [Accepted: 07/09/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND In clinical setting, current standard-of-care does not include genetic testing for patients with low (<50 mg/dL) and extremely low (<20 mg/dL) levels of serum low-density lipoprotein-cholesterol (LDL-C). OBJECTIVE We aimed identify the underlying molecular cause - both monogenic and polygenic - of low and extremely low LDL-C levels in a cohort of patients presenting to specialty lipid clinics. METHODS Whole exome sequencing was done in patients with low or extremely low LDL-C not due to any secondary causes. RESULTS Nine patients (4 women), ranging in age from 25 to 63 years old, with low or extremely low LDL-C levels were evaluated. Median LDL-C was 16 mg/dL (range undetectable - 43), total cholesterol 82 mg/dL (42 - 101), triglycerides 35 mg/dL (19-239), and high-density lipoprotein-cholesterol 45 mg/dL (24-81). Of nine patients, two carried known pathogenic variants in APOB (one stop-gain, one deletion; LDL-C range undetectable -10 mg/dL); three patients had novel APOB heterozygous mutations (two frameshift deletions and one splice site; LDL-C range undectable-13 mg/dL); two had heterozygous APOB frameshift deletions previously reported as variants of unknown significance (LDL-C 18 mg/dL in both patients); one (LDL-C 43 mg/dL) had two heterozygous mutations in PCSK9, both previously reported to be benign; and one patient (LDL-C 16 mg/dL) had the APO E2/E2 genotype along with several variants of unknown significance in genes associated with triglycerides. No patients had an LDL-C polygenic risk score below the 5th percentile (range 26th percentile to 93rd percentile). CONCLUSION We found APOB mutations to be the most common molecular defect in patients presenting to lipid clinics with low or extremely low LDL-C . Whether clinical genetic testing and LDL-C polygenic risk scores have any utility - other than diagnostic purposes - for such patients remains unclear. In addition, further efforts may be needed to better reclassify pathogenicity of variants of unknown significance.
Collapse
|
21
|
Sissaoui S, Cochet M, Poinsot P, Bordat C, Collardeau-Frachon S, Lachaux A, Lacaille F, Peretti N. Lipids Responsible for Intestinal or Hepatic Disorder: When to Suspect a Familial Intestinal Hypocholesterolemia? J Pediatr Gastroenterol Nutr 2021; 73:4-8. [PMID: 33853111 DOI: 10.1097/mpg.0000000000003145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ABSTRACT Familial intestinal hypocholesterolemias, such as abetalipoproteinemia, hypobetalipoproteinemia, and chylomicron retention disease, are rare genetic diseases that result in a defect in the synthesis or secretion of lipoproteins containing apolipoprotein B.In children, these conditions present with diarrhoea and growth failure, whereas adults present with neuromuscular, ophthalmological, and hepatic symptoms. Simple laboratory investigations have shown that diagnosis can be made from findings of dramatically decreased cholesterol levels, deficiencies in fat-soluble vitamins (mostly vitamin E), endoscopic findings of the characteristic white intestinal mucosa, and fat-loaded enterocytes in biopsy samples. Genetic analysis is used to confirm the diagnosis. Treatment is based on a low-fat diet with essential fatty acid supplementation, high doses of fat-soluble vitamins, and regular and life-long follow-up.The present study examines cases and literature findings of these conditions, and emphasises the need to explore severe hypocholesterolemia and deficiencies in fat-soluble vitamins to not miss these rare, but easy to diagnose and treat, disorders.
Collapse
Affiliation(s)
- Samira Sissaoui
- Pediatric Hepatology Unit
- Department of Pediatric Gastroenterology-Hepatology-Nutrition, Reference Center for Biliary Atresia and Genetic Cholestasis, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris
| | - Manon Cochet
- Pediatric Hepatology Unit
- Department of Pediatric Gastroenterology-Hepatology-Nutrition, Reference Center for Biliary Atresia and Genetic Cholestasis, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris
| | - Pierre Poinsot
- Univ. Lyon, Hospices Civil de Lyon, Gastro-enterology and Pediatric Nutrition, Reference Center for Intestinal Rare Disease (MaRDi), Hôpital Femme Mere Enfant, Bron
- Univ. Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Pierre-Bénite
| | - Claire Bordat
- Univ. Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Pierre-Bénite
| | - Sophie Collardeau-Frachon
- Univ. Lyon, Hospices Civil de Lyon, Gastro-enterology and Pediatric Nutrition, Reference Center for Intestinal Rare Disease (MaRDi), Hôpital Femme Mere Enfant, Bron
- Univ. Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Pierre-Bénite
- Univ. Lyon, Hospices Civil de Lyon, Institut de pathologie, Groupement Hospitalier Est, Bron, France
| | - Alain Lachaux
- Univ. Lyon, Hospices Civil de Lyon, Gastro-enterology and Pediatric Nutrition, Reference Center for Intestinal Rare Disease (MaRDi), Hôpital Femme Mere Enfant, Bron
- Univ. Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Pierre-Bénite
| | - Florence Lacaille
- Department of Pediatric Gastroenterology-Hepatology-Nutrition, Reference Center for Biliary Atresia and Genetic Cholestasis, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris
| | - Noël Peretti
- Univ. Lyon, Hospices Civil de Lyon, Gastro-enterology and Pediatric Nutrition, Reference Center for Intestinal Rare Disease (MaRDi), Hôpital Femme Mere Enfant, Bron
- Univ. Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Pierre-Bénite
| |
Collapse
|
22
|
Bredefeld C, Peretti N, Hussain MM. New Classification and Management of Abetalipoproteinemia and Related Disorders. Gastroenterology 2021; 160:1912-1916. [PMID: 33275938 DOI: 10.1053/j.gastro.2020.11.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 02/03/2023]
Affiliation(s)
- Cindy Bredefeld
- Department of Medicine, New York University Long Island School of Medicine, NYU Langone Hospital - Long Island, Mineola, New York
| | - Noel Peretti
- Department of Pediatric Gastroenterology-Hepatology and Nutrition, Laboratory INSERM 1060 Cardiovascular Metabolism Endocrinology and Nutrition CarMEN, Lyon, France
| | - M Mahmood Hussain
- Department of Medicine, New York University Long Island School of Medicine, NYU Langone Hospital - Long Island, Mineola, New York
| | | |
Collapse
|
23
|
Montoro-Huguet MA, Belloc B, Domínguez-Cajal M. Small and Large Intestine (I): Malabsorption of Nutrients. Nutrients 2021; 13:1254. [PMID: 33920345 PMCID: PMC8070135 DOI: 10.3390/nu13041254] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Numerous disorders can alter the physiological mechanisms that guarantee proper digestion and absorption of nutrients (macro- and micronutrients), leading to a wide variety of symptoms and nutritional consequences. Malabsorption can be caused by many diseases of the small intestine, as well as by diseases of the pancreas, liver, biliary tract, and stomach. This article provides an overview of pathophysiologic mechanisms that lead to symptoms or complications of maldigestion (defined as the defective intraluminal hydrolysis of nutrients) or malabsorption (defined as defective mucosal absorption), as well as its clinical consequences, including both gastrointestinal symptoms and extraintestinal manifestations and/or laboratory abnormalities. The normal uptake of nutrients, vitamins, and minerals by the gastrointestinal tract (GI) requires several steps, each of which can be compromised in disease. This article will first describe the mechanisms that lead to poor assimilation of nutrients, and secondly discuss the symptoms and nutritional consequences of each specific disorder. The clinician must be aware that many malabsorptive disorders are manifested by subtle disorders, even without gastrointestinal symptoms (for example, anemia, osteoporosis, or infertility in celiac disease), so the index of suspicion must be high to recognize the underlying diseases in time.
Collapse
Affiliation(s)
- Miguel A. Montoro-Huguet
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Ciencias de la Salud y del Deporte, University of Zaragoza, 50009 Zaragoza, Spain
- Unidad de Gastroenterología, Hepatología y Nutrición, Hospital Universitario San Jorge de Huesca, 22004 Huesca, Spain; (B.B.); (M.D.-C.)
- Aragonese Institute of Health Sciences (IACS), 50009 Zaragoza, Spain
| | - Blanca Belloc
- Unidad de Gastroenterología, Hepatología y Nutrición, Hospital Universitario San Jorge de Huesca, 22004 Huesca, Spain; (B.B.); (M.D.-C.)
- Aragonese Institute of Health Sciences (IACS), 50009 Zaragoza, Spain
| | - Manuel Domínguez-Cajal
- Unidad de Gastroenterología, Hepatología y Nutrición, Hospital Universitario San Jorge de Huesca, 22004 Huesca, Spain; (B.B.); (M.D.-C.)
- Aragonese Institute of Health Sciences (IACS), 50009 Zaragoza, Spain
| |
Collapse
|
24
|
Primorac D, Odak L, Perić V, Ćatić J, Šikić J, Radeljić V, Manola Š, Nussbaum R, Vatta M, Aradhya S, Sofrenović T, Matišić V, Molnar V, Skelin A, Mirat J, Brachmann J. Sudden Cardiac Death-A New Insight Into Potentially Fatal Genetic Markers. Front Med (Lausanne) 2021; 8:647412. [PMID: 33829027 PMCID: PMC8019733 DOI: 10.3389/fmed.2021.647412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/01/2021] [Indexed: 01/13/2023] Open
Abstract
Sudden cardiac death (SCD) is an unexpected and dramatic event. It draws special attention especially in young, seemingly healthy athletes. Our scientific paper is based on the death of a young, 23-year-old professional footballer, who died on the football field after a two-year history of cardiac symptoms. In this study we analyzed clinical, ECG and laboratory data, as well as results of genetic testing analysis in family members. To elucidate potential genetic etiology of SCD in this family, our analysis included 294 genes related to various cardiac conditions.
Collapse
Affiliation(s)
- Dragan Primorac
- St. Catherine Specialty Hospital, Zagreb, Croatia.,Eberly College of Science, The Pennsylvania State University, University Park, State College, Philadelphia, PA, United States.,The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT, United States.,Medical School, University of Split, Split, Croatia.,Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Medical School, University of Rijeka, Rijeka, Croatia.,Medical School REGIOMED, Coburg, Germany.,Medical School, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Ljubica Odak
- St. Catherine Specialty Hospital, Zagreb, Croatia.,Children's Hospital Zagreb, Zagreb, Croatia
| | | | - Jasmina Ćatić
- St. Catherine Specialty Hospital, Zagreb, Croatia.,Department of Cardiology, Clinical Hospital Dubrava, Zagreb, Croatia
| | - Jozica Šikić
- Department of Cardiology, Clinical Hospital Sveti Duh, Zagreb, Croatia
| | - Vjekoslav Radeljić
- Department of Cardiology, Clinical Hospital Center Sestre Milosrdnice, Zagreb, Croatia
| | - Šime Manola
- Department of Cardiology, Clinical Hospital Center Sestre Milosrdnice, Zagreb, Croatia
| | | | | | | | | | - Vid Matišić
- St. Catherine Specialty Hospital, Zagreb, Croatia
| | - Vilim Molnar
- St. Catherine Specialty Hospital, Zagreb, Croatia
| | | | - Jure Mirat
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Johannes Brachmann
- Medical School, University of Split, Split, Croatia.,Medical School REGIOMED, Coburg, Germany
| |
Collapse
|
25
|
Vilar-Gomez E, Gawrieh S, Liang T, McIntyre AD, Hegele RA, Chalasani N. Interrogation of selected genes influencing serum LDL-Cholesterol levels in patients with well characterized NAFLD. J Clin Lipidol 2021; 15:275-291. [PMID: 33454241 PMCID: PMC8187295 DOI: 10.1016/j.jacl.2020.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/07/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND The clinical significance of rare mutations in LDL metabolism genes on nonalcoholic fatty liver disease (NAFLD) severity is not well understood. OBJECTIVE To examine the significance of mutations in LDL metabolism genes including apolipoprotein B (APOB), proprotein convertase subtilisin kexin 9 (PCSK9) and LDL receptor (LDLR) in patients with NAFLD. METHODS Patients with biopsy-confirmed NAFLD from the NASH Clinical Research Network studies were stratified into 3 groups of LDL-C (≤50 mg/dL, 130-150 mg/dL, ≥ 190 mg/dL) and then 120 (40 per group) were randomly selected from the strata. We examined the presence of mutations on LDL genes and analyzed its association with selected NAFLD-related features. Multivariable analyses were adjusted for age, race, gender and use of statins. RESULTS Among 40 patients with LDL-C ≤ 50 mg/dL, 7 (18%) patients had heterozygous variants in APOB and 2 had heterozygous variants in PCSK9 (5%). We also found heterozygous mutations in 3 (8%) patients with LDL-C ≥ 190 mg/dL; 2 and 1 located in LDLR and APOE genes, respectively. Compared to wild-type controls with LDL-C ≤ 50, APOB carriers displayed higher levels of alanine aminotransferase (85.86 ± 35.14 U/L vs 45.61 ± 20.84 U/L, Adj. P = 0.002) and steatosis >66% (57% vs 24%, Adj. P = 0.050). These associations remained statistically significant after excluding statin users. Other histological features of NAFLD severity were not different between wild-type controls and APOB mutation carriers. CONCLUSION Mutations in the APOB gene are common among NAFLD patients with very low LDL-C and may be associated with increased aminotransferase levels and steatosis severity.
Collapse
Affiliation(s)
- Eduardo Vilar-Gomez
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Samer Gawrieh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tiebing Liang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adam D McIntyre
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A Hegele
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Naga Chalasani
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
26
|
Surakka I, Hornsby WE, Farhat L, Rubenfire M, Fritsche LG, Hveem K, Chen YE, Brook RD, Willer CJ, Weinberg RL. A Novel Variant in APOB Gene Causes Extremely Low LDL-C Without Known Adverse Effects. JACC Case Rep 2020; 2:775-779. [PMID: 34317346 PMCID: PMC8301695 DOI: 10.1016/j.jaccas.2020.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/09/2020] [Accepted: 03/18/2020] [Indexed: 11/26/2022]
Abstract
A novel frameshift variant was identified in APOB that segregates in a dominant manner with low levels of low-density lipoprotein cholesterol. Affected family members show no apparent clinical complications. There is no consensus regarding clinical management, and the long-term consequences of low levels of low-density lipoprotein cholesterol remain unknown. (Level of Difficulty: Advanced.).
Collapse
Affiliation(s)
- Ida Surakka
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan
| | - Whitney E. Hornsby
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan
| | - Linda Farhat
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan
| | - Melvyn Rubenfire
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan
| | - Lars G. Fritsche
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology and Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway
| | - Y. Eugene Chen
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan
| | - Robert D. Brook
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan
| | - Cristen J. Willer
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics and Department of Human Genetics, University of Michigan, Ann Arbor, Michigan
| | - Richard L. Weinberg
- University of Michigan, Michigan Medicine, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Several mutations in the apolipoprotein (apo) B, proprotein convertase subtilisin kexin 9 (PCSK9) and microsomal triglyceride transfer protein genes result in low or absent levels of apoB and LDL cholesterol (LDL-C) in plasma which cause familial hypobetalipoproteinemia (FHBL) and abetalipoproteinemia (ABL). Mutations in the angiopoietin-like protein 3 ANGPTL3 gene cause familial combined hypolipidemia (FHBL2). Clinical manifestations range from none-to-severe, debilitating and life-threatening disorders. This review summarizes recent genetic, metabolic and clinical findings and management strategies. RECENT FINDINGS Fatty liver, cirrhosis and hepatocellular carcinoma have been reported in FHBL and ABL probably due to decreased triglyceride export from the liver. Loss of function mutations in PCSK-9 and ANGPTL3 cause FHBL but not hepatic steatosis. In 12 case-control studies with 57 973 individuals, an apoB truncation was associated with a 72% reduction in coronary heart disease (odds ratio, 0.28; 95% confidence interval, 0.12-0.64; P = 0.002). PCSK9 inhibitors lowered risk of cardiovascular events in large, randomized trials without apparent adverse sequelae. SUMMARY Mutations causing low LDL-C and apoB have provided insight into lipid metabolism, disease associations and the basis for drug development to lower LDL-C in disorders causing high levels of cholesterol. Early diagnosis and treatment is necessary to prevent adverse sequelae from FHBL and ABL.
Collapse
Affiliation(s)
- Francine K Welty
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Di Taranto MD, Giacobbe C, Fortunato G. Familial hypercholesterolemia: A complex genetic disease with variable phenotypes. Eur J Med Genet 2020; 63:103831. [DOI: 10.1016/j.ejmg.2019.103831] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/01/2019] [Accepted: 12/21/2019] [Indexed: 12/21/2022]
|
29
|
Zhou H, Gong Y, Wu Q, Ye X, Yu B, Lu C, Jiang W, Ye J, Fu Z. Rare Diseases Related with Lipoprotein Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:171-188. [PMID: 32705600 DOI: 10.1007/978-981-15-6082-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rare diseases are gathering increasing attention in last few years, not only for its effects on innovation scientific research, but also for its propounding influence on common diseases. One of the most famous milestones made by Michael Brown and Joseph Goldstein in metabolism field is the discovery of the defective gene in familial hypercholesterolemia, a rare human genetic disease manifested with extreme high level of serum cholesterol (Goldstein JL, Brown MS, Proc Natl Acad Sci USA 70:2804-2808, 1973; Brown MS, Dana SE, Goldstein JL, J Biol Chem 249:789-796, 1974). Follow-up work including decoding the gene function, mapping-related pathways, and screening therapeutic targets are all based on the primary finding (Goldstein JL, Brown MS Arterioscler Thromb Vasc Biol 29:431-438, 2009). A series of succession win the two brilliant scientists the 1985 Nobel Prize, and bring about statins widely used for lipid management and decreasing cardiovascular disease risks. Translating the clinical extreme phenotypes into laboratory bench work has turned out to be the first important step in the paradigm conducting translational and precise medical research. Here we review the main categories of rare disorders related with lipoprotein metabolism, aiming to strengthen the notion that human rare inheritable genetic diseases would be the window to know ourselves better, to treat someone more efficiently, and to lead a healthy life longer. Few rare diseases related with lipoprotein metabolism were clustered into six sections based on changes in lipid profile, namely, hyper- or hypocholesterolemia, hypo- or hyperalphalipoproteinemia, abetalipoproteinemia, hypobetalipoproteinemia, and sphingolipid metabolism diseases. Each section consists of a brief introduction, followed by a summary of well-known disease-causing genes in one table, and supplemented with one or two diseases as example for detailed description. Here we aimed to raise more attention on rare lipoprotein metabolism diseases, calling for more work from basic research and clinical trials.
Collapse
Affiliation(s)
- Hongwen Zhou
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yingyun Gong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qinyi Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Ye
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Baowen Yu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenyan Lu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wanzi Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingya Ye
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenzhen Fu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
In vitro functional characterization of splicing variants of the APOB gene found in familial hypobetalipoproteinemia. J Clin Lipidol 2019; 13:960-969. [PMID: 31629702 DOI: 10.1016/j.jacl.2019.09.003] [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: 06/15/2019] [Revised: 08/22/2019] [Accepted: 09/06/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Familial hypobetalipoproteinemia type 1 (FHBL-1) is a codominant disorder characterized by greatly reduced plasma levels of total cholesterol, low-density lipoprotein cholesterol, and apolipoprotein B. Rare exonic pathogenic variants of APOB gene (nonsense variants, minute deletions/insertions and nonsynonymous variants) have been frequently reported in subjects with FHBL-1. Also, rare intronic variants of APOB located at intron/exon junctions and assumed to affect splicing have been reported. However, the pathogenicity of most of these intronic variants remains to be established. OBJECTIVE The objective of this study was the in vitro functional characterization of six splicing variants of APOB gene identified in seven putative FHBL-1 heterozygotes. METHODS ApoB minigenes harboring each variant were expressed in COS-1 cells and their transcripts were sequenced. RESULTS Four novel variants (c.237+1G>A, c.818+5G>A, c.3000-1G>T, and c.3842+1G>A), predicted in silico to obliterate splice site activity, were found to generate abnormal transcripts. The abnormal transcripts were generated by the activation of cryptic splice sites or exon skipping. All these transcripts harbored a premature termination codon and were predicted to encode truncated apoBs devoid of function. The predicted translation products were: i) p.(Lys41Serfs*2) and p.(Val80Ilefs*10) for c.237+1G>A; ii) p.(Asn274*) for c.818+5G>A; iii) p.(Leu1001Alafs*10) for c.3000-1G>T, and iv) p.(Ser1281Argfs*2) for c.3842+1G>A. Two previously annotated rare variants (c.905-15C>G and c.1618-4G>A) with uncertain effect in silico were found to generate only wild-type transcripts. CONCLUSIONS These in vitro minigene expression studies support the assignment of pathogenicity to four novel splice site variants of APOB gene found in FHBL-1.
Collapse
|
31
|
Simone ML, Rabacchi C, Kuloglu Z, Kansu A, Ensari A, Demir AM, Hizal G, Di Leo E, Bertolini S, Calandra S, Tarugi P. Novel mutations of SAR1B gene in four children with chylomicron retention disease. J Clin Lipidol 2019; 13:554-562. [DOI: 10.1016/j.jacl.2019.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/30/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
|
32
|
Hartz J, Hegele RA, Wilson DP. Low LDL cholesterol-Friend or foe? J Clin Lipidol 2019; 13:367-373. [PMID: 31130490 PMCID: PMC11789202 DOI: 10.1016/j.jacl.2019.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 01/19/2023]
Affiliation(s)
- Jacob Hartz
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Robert A Hegele
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Don P Wilson
- Department of Pediatric Endocrinology, Cook Children's Medical Center, Ft Worth, TX, USA.
| |
Collapse
|
33
|
Sánchez-Chaparro MÁ, Pérez-Martínez P, Ibarretxe D, Suárez-Tembra M, Valdivielso P. Criteria for referring patients to Spanish Atherosclerosis Society lipid units. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2019; 31:26-30. [PMID: 30257791 DOI: 10.1016/j.arteri.2018.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 11/16/2022]
Abstract
The Spanish Arteriosclerosis Society has accredited more than 70 lipid units across the country. The main criteria for patients to be referred to these units are presented. These are not only grouped by the type of dyslipidaemia or the lipid levels, but also on certain clinical characteristics suggesting primary hyperlipidaemia, a complex diagnosis, or difficult management due to inefficacy, or side effects.
Collapse
Affiliation(s)
- Miguel Ángel Sánchez-Chaparro
- Unidad de Lípidos, Departamento de Medicina y Dermatología, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, España
| | - Pablo Pérez-Martínez
- Unidad de Lípidos y Arteriosclerosis, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Universidad de Córdoba, Córdoba, España; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, España
| | - Diana Ibarretxe
- Unidad de Metabolismo y Medicina Vascular, Unidad de Investigación en Lípidos y Arteriosclerosis, Hospital Universitario Sant Joan, Universidad Rovira i Virgili, Reus, Tarragona, España; Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, España
| | | | - Pedro Valdivielso
- Unidad de Lípidos, Departamento de Medicina y Dermatología, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, España.
| | | |
Collapse
|
34
|
Soran H, Ho JH, Durrington PN. Acquired low cholesterol: diagnosis and relevance to safety of low LDL therapeutic targets. Curr Opin Lipidol 2018; 29:318-326. [PMID: 29746303 DOI: 10.1097/mol.0000000000000526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Acquired hypocholesterolaemia occurs more commonly than inherited hypocholesterolaemia but has received little attention in the literature. In this review, we discuss the causes and underlying mechanisms of acquired hypocholesterolaemia and its relevance to safety of therapeutically induced decreased LDL cholesterol levels. RECENT FINDINGS Hypocholesterolaemia is increasingly identified as cholesterol testing becomes more widespread in the assessment of cardiovascular risk. Lower therapeutic targets for LDL cholesterol are also being achieved more regularly with the introduction of more intensive cholesterol-lowering regimens. Acquired hypocholesterolaemia may be the presenting feature of treatable diseases. Understanding its mechanisms may also provide new treatment approaches for neoplastic disease, such as breast cancer, and infections, such as tuberculosis. SUMMARY When hypocholesterolaemia is discovered, it is important to identify its cause. Further research into the pathogenesis of hypocholesterolaemia may provide new therapies for primary diseases underlying it.
Collapse
Affiliation(s)
- Handrean Soran
- Lipoprotein Research Group, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jan Hoong Ho
- Lipoprotein Research Group, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Paul N Durrington
- Lipoprotein Research Group, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
| |
Collapse
|
35
|
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: 2.0] [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
|
36
|
Marmontel O, Charrière S, Simonet T, Bonnet V, Dumont S, Mahl M, Jacobs C, Nony S, Chabane K, Bozon D, Janin A, Peretti N, Lachaux A, Bardel C, Millat G, Moulin P, Marçais C, Di Filippo M. Single, short in-del, and copy number variations detection in monogenic dyslipidemia using a next-generation sequencing strategy. Clin Genet 2018; 94:132-140. [DOI: 10.1111/cge.13250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 01/08/2023]
Affiliation(s)
- O. Marmontel
- Service de Biochimie et Biologie moléculaire Grand Est, GHE; Hospices Civils de Lyon; Bron France
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397; Université Claude Bernard Lyon 1, INSA Lyon; Villeurbanne France
| | - S. Charrière
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397; Université Claude Bernard Lyon 1, INSA Lyon; Villeurbanne France
- Fédération d'endocrinologie, maladies métaboliques, diabète et nutrition, GHE; Hospices Civils de Lyon; Bron France
| | - T. Simonet
- Service de Biostatistique-Bioinformatique; Hospices Civils de Lyon; Lyon France
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Evolutive UMR5558; Villeurbanne France
| | - V. Bonnet
- Service de Biochimie et Biologie moléculaire Grand Est, GHE; Hospices Civils de Lyon; Bron France
| | - S. Dumont
- Service de Biochimie et Biologie moléculaire Grand Est, GHE; Hospices Civils de Lyon; Bron France
| | - M. Mahl
- Centre de Biologie Sud, Laboratoire de Biochimie moléculaire et métabolique, GHS; Hospices Civils de Lyon; Pierre-Benite France
| | - C. Jacobs
- Service de Biochimie et Biologie moléculaire Grand Est, GHE; Hospices Civils de Lyon; Bron France
| | - S. Nony
- Service de Biochimie et Biologie moléculaire Grand Est, GHE; Hospices Civils de Lyon; Bron France
| | - K. Chabane
- Laboratoire d'hématologie, Biologie Moléculaire; Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon; Pierre-Benite France
| | - D. Bozon
- Plateforme NGS CHU Lyon, GHE; Hospices Civils de Lyon; Bron France
| | - A. Janin
- Plateforme NGS CHU Lyon, GHE; Hospices Civils de Lyon; Bron France
| | - N. Peretti
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397; Université Claude Bernard Lyon 1, INSA Lyon; Villeurbanne France
- Service de Gastroentérologie Hépatologie et Nutrition Pédiatrique, GHE; Hospices Civils de Lyon; Bron France
| | - A. Lachaux
- Service de Gastroentérologie Hépatologie et Nutrition Pédiatrique, GHE; Hospices Civils de Lyon; Bron France
- INSERM U 1111, Faculté de médecine Lyon Est; Université Lyon 1; Lyon France
| | - C. Bardel
- Service de Biostatistique-Bioinformatique; Hospices Civils de Lyon; Lyon France
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Evolutive UMR5558; Villeurbanne France
| | - G. Millat
- Plateforme NGS CHU Lyon, GHE; Hospices Civils de Lyon; Bron France
| | - P. Moulin
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397; Université Claude Bernard Lyon 1, INSA Lyon; Villeurbanne France
- Fédération d'endocrinologie, maladies métaboliques, diabète et nutrition, GHE; Hospices Civils de Lyon; Bron France
| | - C. Marçais
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397; Université Claude Bernard Lyon 1, INSA Lyon; Villeurbanne France
- Centre de Biologie Sud, Laboratoire de Biochimie moléculaire et métabolique, GHS; Hospices Civils de Lyon; Pierre-Benite France
- CENS, Centre de Recherche en Nutrition Humaine Rhône-Alpes; Oullins France
| | - M. Di Filippo
- Service de Biochimie et Biologie moléculaire Grand Est, GHE; Hospices Civils de Lyon; Bron France
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397; Université Claude Bernard Lyon 1, INSA Lyon; Villeurbanne France
| |
Collapse
|
37
|
Abstract
BACKGROUND Abetalipoproteinemia and homozygous hypobetalipoproteinemia are classical Mendelian autosomal recessive and co-dominant conditions, respectively, which are phenotypically similar and are usually caused by bi-allelic mutations in MTTP and APOB genes, respectively. Instances of more complex patterns of genomic variants resulting in this distinct phenotype have not been reported. METHODS A 43 year-old male had a longstanding severe deficiency of apolipoprotein (apo) B-containing lipoproteins and circulating fat soluble vitamins consistent with either abetalipoproteinemia or homozygous familial hypobetalipoproteinemia (FHBL). He also had acanthocytosis, a long term history of fat malabsorption, and mild retinopathy, but was free from coagulopathy, myopathy and neuropathy. He had taken high dose oral fat soluble vitamins since childhood. RESULTS Targeted next generation DNA sequencing revealed several rare heterozygous missense variants in both MTTP and APOB genes known or predicted to be deleterious, in addition to a novel heterozygous missense variant in SAR1B, which encodes the gene causing chylomicron retention disease. Evaluation of first degree relatives with mild FHBL clarified the segregation of variants. CONCLUSIONS The proband's characteristic phenotype likely resulted from an oligogenic interaction involving multiple rare variants in MTTP and APOB, and related genes, each of which individually was associated with a milder or minimal clinical and biochemical phenotype.
Collapse
Affiliation(s)
- Linda R Wang
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A - 1151 Richmond Street North, London, ON, N6A 5B7, Canada
| | - Adam D McIntyre
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A - 1151 Richmond Street North, London, ON, N6A 5B7, Canada
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A - 1151 Richmond Street North, London, ON, N6A 5B7, Canada.
| |
Collapse
|
38
|
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: 3.9] [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
|
39
|
Kipp S, Segelke D, Schierenbeck S, Reinhardt F, Reents R, Wurmser C, Pausch H, Fries R, Thaller G, Tetens J, Pott J, Haas D, Raddatz BB, Hewicker-Trautwein M, Proios I, Schmicke M, Grünberg W. Identification of a haplotype associated with cholesterol deficiency and increased juvenile mortality in Holstein cattle. J Dairy Sci 2016; 99:8915-8931. [PMID: 27614835 DOI: 10.3168/jds.2016-11118] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022]
Abstract
Over the last decades, several genetic disorders have been discovered in cattle. However, the genetic background of disorders in calves is less reported. Recently, German cattle farmers reported on calves from specific matings with chronic diarrhea and retarded growth of unknown etiology. Affected calves did not respond to any medical treatment and died within the first months of life. These calves were underdeveloped in weight and showed progressive and severe emaciation despite of normal feed intake. Hallmark findings of the blood biochemical analysis were pronounced hypocholesterolemia and deficiency of fat-soluble vitamins. Results of the clinical and blood biochemical examination had striking similarities with findings reported in human hypobetalipoproteinemia. Postmortem examination revealed near-complete atrophy of the body fat reserves including the spinal canal and bone marrow. To identify the causal region, we performed a genome-wide association study with 9 affected and 21,077 control animals genotyped with the Illumina BovineSNP50 BeadChip (Illumina Inc., San Diego, CA), revealing a strong association signal on BTA 11. Subsequent autozygosity mapping identified a disease-associated haplotype encompassing 1.01 Mb. The segment of extended homozygosity contains 6 transcripts, among them the gene APOB, which is causal for cholesterol disorders in humans. However, results from multi-sample variant calling of 1 affected and 47 unaffected animals did not detect any putative causal mutation. The disease-associated haplotype has an important adverse effect on calf mortality in the homozygous state when comparing survival rates of risk matings vs. non-risk matings. Blood cholesterol values of animals are significantly associated with the carrier status indicating a codominant inheritance. The frequency of the haplotype in the current Holstein population was estimated to be 4.2%. This study describes the identification and phenotypic manifestation of a new Holstein haplotype characterized by pronounced hypocholesterolemia, chronic emaciation, growth retardation, and increased mortality in young cattle, denominated as cholesterol deficiency haplotype. Our genomic investigations and phenotypic examinations provide additional evidence for a mutation within the APOB gene causing cholesterol deficiency in Holstein cattle.
Collapse
Affiliation(s)
- S Kipp
- Vereinigte Informationssysteme Tierhaltung w.V. (vit), 27283 Verden, Germany.
| | - D Segelke
- Vereinigte Informationssysteme Tierhaltung w.V. (vit), 27283 Verden, Germany
| | - S Schierenbeck
- Vereinigte Informationssysteme Tierhaltung w.V. (vit), 27283 Verden, Germany
| | - F Reinhardt
- Vereinigte Informationssysteme Tierhaltung w.V. (vit), 27283 Verden, Germany
| | - R Reents
- Vereinigte Informationssysteme Tierhaltung w.V. (vit), 27283 Verden, Germany
| | - C Wurmser
- Chair of Animal Breeding, Technische Universitaet Muenchen, 85354 Freising, Germany
| | - H Pausch
- Chair of Animal Breeding, Technische Universitaet Muenchen, 85354 Freising, Germany
| | - R Fries
- Chair of Animal Breeding, Technische Universitaet Muenchen, 85354 Freising, Germany
| | - G Thaller
- Chair of Animal Breeding, Christian-Albrechts-Universitaet zu Kiel, 24098 Kiel, Germany
| | - J Tetens
- Chair of Animal Breeding, Christian-Albrechts-Universitaet zu Kiel, 24098 Kiel, Germany
| | - J Pott
- Masterrind GmbH, 27283 Verden, Germany
| | - D Haas
- University Children's Hospital Heidelberg, Division of Neuropediatrics and Metabolic Diseases, Im Neuenheimer Feld 699, 69120 Heidelberg, Germany
| | - B B Raddatz
- Department of Pathology, University of Veterinary Medicine, Hannover, Foundation, Bünteweg 17, 30559 Hanover, Germany
| | - M Hewicker-Trautwein
- Department of Pathology, University of Veterinary Medicine, Hannover, Foundation, Bünteweg 17, 30559 Hanover, Germany
| | - I Proios
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hanover, Germany
| | - M Schmicke
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hanover, Germany
| | - W Grünberg
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hanover, Germany
| |
Collapse
|
40
|
Lonardo A, Sookoian S, Pirola CJ, Targher G. Non-alcoholic fatty liver disease and risk of cardiovascular disease. Metabolism 2016; 65:1136-50. [PMID: 26477269 DOI: 10.1016/j.metabol.2015.09.017] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 08/17/2015] [Accepted: 09/19/2015] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the leading cause of chronic liver diseases worldwide, causing considerable liver-related mortality and morbidity. During the past decade, it has also become increasingly evident that NAFLD is a multisystem disease that affects many extra-hepatic organ systems, including the heart and the vascular system. In this updated clinical review, we discuss the rapidly expanding body of clinical and epidemiological evidence that supports a strong association of NAFLD with cardiovascular diseases (CVDs) and other functional and structural myocardial abnormalities. We also discuss some recently published data that correlate NAFLD due to specific genetic polymorphisms with the risk of CVDs. Finally, we briefly examine the assessment tools for estimating the global CVD risk in patients with NAFLD as well as the conventional and the more innovative pharmacological approaches for the treatment of CVD risk in this group of patients.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Outpatient Liver Clinic and Division of Internal Medicine, Department of Biomedical, Metabolic and Neural Sciences, NOCSAE, Baggiovara, Azienda USL and University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Sookoian
- Department of Clinical and Molecular Hepatology, Institute of Medical Research A Lanari-IDIM, University of Buenos Aires-National Scientific and Technical Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlos J Pirola
- Department of Molecular Genetics and Biology of Complex Diseases, Institute of Medical Research A Lanari-IDIM, University of Buenos Aires-National Scientific and Technical Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy.
| |
Collapse
|
41
|
Jiang ZG, Tapper EB, Connelly MA, Pimentel CFMG, Feldbrügge L, Kim M, Krawczyk S, Afdhal N, Robson SC, Herman MA, Otvos JD, Mukamal KJ, Lai M. Steatohepatitis and liver fibrosis are predicted by the characteristics of very low density lipoprotein in nonalcoholic fatty liver disease. Liver Int 2016; 36:1213-20. [PMID: 26815314 PMCID: PMC4942367 DOI: 10.1111/liv.13076] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/14/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS A major challenge in the management of nonalcoholic fatty liver disease (NAFLD) is to identify patients with nonalcoholic steatohepatitis (NASH) and early liver fibrosis. The progression of NAFLD is accompanied by distinctive changes in very low density lipoprotein (VLDL), a lipoprotein particle produced exclusively in the liver. Herein, we sought to determine the characteristics of VLDL profiles associated with NASH and liver fibrosis. METHODS We evaluated VLDL profiles of 128 patients from a single centre NAFLD registry, and examined VLDL size, total and subclass VLDL concentrations in relation to NAFLD activity score (NAS), steatohepatitis and liver fibrosis as determined by liver biopsy. RESULTS A near linear relationship was observed between mean VLDL particle size and NAFLD activity score (NAS). In multivariate models, VLDL particle size was significantly associated with both NAS and NASH, after adjustment for BMI and diabetes. A decrease in small VLDL particle concentration was associated with more advanced liver fibrosis. In receiver operative characteristic analyses, mean VLDL size performed similarly to cytokeratin 18 in predicting NASH, whereas small VLDL particle concentration had similar performance to NAFLD fibrosis score in predicting stage 2 or above liver fibrosis. CONCLUSIONS The increase in mean VLDL size in NASH and decrease in small VLDL particle concentration in liver fibrosis likely reflect changes in the number and state of hepatocytes associated with NASH and fibrosis. In addition to its value in risk stratification of cardiovascular diseases, circulating VLDL profile may provide information for the staging of NAFLD disease severity.
Collapse
Affiliation(s)
- Zhenghui G. Jiang
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Elliot B. Tapper
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Carolina F. M. G. Pimentel
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Linda Feldbrügge
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Misung Kim
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sarah Krawczyk
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nezam Afdhal
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C. Robson
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark A. Herman
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - James D. Otvos
- LipoScience, Laboratory Corporation of America Holdings, Raleigh, NC, USA
| | - Kenneth J. Mukamal
- Division of General Medicine and Primary Care, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michelle Lai
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Rabacchi C, Bigazzi F, Puntoni M, Sbrana F, Sampietro T, Tarugi P, Bertolini S, Calandra S. Phenotypic variability in 4 homozygous familial hypercholesterolemia siblings compound heterozygous for LDLR mutations. J Clin Lipidol 2016; 10:944-952.e1. [DOI: 10.1016/j.jacl.2016.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/31/2022]
|
43
|
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: 0.2] [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]
|
44
|
Microsomal triglyceride transfer protein gene mutations in Turkish children: A novel mutation and clinical follow up. Indian J Gastroenterol 2016; 35:236-41. [PMID: 27160094 DOI: 10.1007/s12664-016-0654-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/27/2016] [Indexed: 02/04/2023]
Abstract
Abetalipoproteinemia (ABL; OMIM 200100) is a rare autosomal recessive disease that affects the absorption of dietary fats and fat soluble vitamins. Here, we describe the clinical and genetic characteristics of three patients with ABL. Two patients (patients 1 and 2) who were carriers of the c.398-399delAA mutation (previously known mutation) had developmental delay and hepatic steatosis developed at the age of five in patient 1. Patient 3 was the carrier of a novel mutation (g.10886-10902delAAGgtaagtttgtgttg in intron 3 and c.506A>T exon 5) in microsomal triglyceride transfer protein (MTP) gene and had hepatic steatosis.
Collapse
|
45
|
Ponziani FR, Pecere S, Gasbarrini A, Ojetti V. Physiology and pathophysiology of liver lipid metabolism. Expert Rev Gastroenterol Hepatol 2016; 9:1055-67. [PMID: 26070860 DOI: 10.1586/17474124.2015.1056156] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Liver lipid metabolism and its modulation are involved in many pathologic conditions, such as obesity, non-alcoholic fatty liver disease, diabetes mellitus, atherosclerosis and cardiovascular disease. Metabolic disorders seem to share a similar background of low-grade chronic inflammation, even if the pathophysiological mechanisms leading to tissue and organ damage have not been completely clarified yet. The accumulation of neutral lipids in the liver is now recognized as a beneficial and protective mechanism; on the other hand, lipoperoxidation is involved in the development and progression of non-alcoholic steatohepatitis. The role of the gut microbiota in liver lipid metabolism has been the object of recent scientific investigations. It is likely that the gut microbiota is involved in a complex metabolic modulation and the translocation of gut microflora may also contribute to maintaining the low-grade inflammatory status of metabolic syndrome. Therefore, lipid metabolism pathology has vague limits and complex mechanisms, and the knowledge of these is essential to guide diagnostic and therapeutic decisions.
Collapse
|
46
|
Magnolo L, Noto D, Cefalù AB, Averna M, Calandra S, Yao Z, Tarugi P. Characterization of a mutant form of human apolipoprotein B (Thr26_Tyr27del) associated with familial hypobetalipoproteinemia. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:371-9. [DOI: 10.1016/j.bbalip.2016.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/14/2015] [Accepted: 01/24/2016] [Indexed: 10/22/2022]
|
47
|
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: 0.2] [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
|
48
|
Yilmaz BS, Mungan NO, Di Leo E, Magnolo L, Artuso L, Bernardis I, Tumgor G, Kor D, Tarugi P. Homozygous familial hypobetalipoproteinemia: A Turkish case carrying a missense mutation in apolipoprotein B. Clin Chim Acta 2016; 452:185-90. [PMID: 26612772 DOI: 10.1016/j.cca.2015.11.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 11/15/2022]
|
49
|
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: 0.6] [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
|
50
|
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: 0.9] [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
|