1
|
Makhmudova U, Schulze PC, Lorkowski S, März W, Geiling JA, Weingärtner O. Monogenic hypertriglyceridemia and recurrent pancreatitis in a homozygous carrier of a rare APOA5 mutation: a case report. J Med Case Rep 2024; 18:278. [PMID: 38872171 PMCID: PMC11177521 DOI: 10.1186/s13256-024-04532-0] [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] [Received: 01/03/2024] [Accepted: 03/25/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Homozygous mutations in the APOA5 gene constitute a rare cause of monogenic hypertriglyceridemia, or familial chylomicronemia syndrome (FCS). We searched PubMed and identified 16 cases of homozygous mutations in the APOA5 gene. Severe hypertriglyceridemia related to monogenic mutations in triglyceride-regulating genes can cause recurrent acute pancreatitis. Standard therapeutic approaches for managing this condition typically include dietary interventions, fibrates, and omega-3-fatty acids. A novel therapeutic approach, antisense oligonucleotide volanesorsen is approved for use in patients with FCS. CASE PRESENTATION We report a case of a 25-years old Afghani male presenting with acute pancreatitis due to severe hypertriglyceridemia up to 29.8 mmol/L caused by homozygosity in APOA5 (c.427delC, p.Arg143Alafs*57). A low-fat diet enriched with medium-chain TG (MCT) oil and fibrate therapy did not prevent recurrent relapses, and volanesorsen was initiated. Volanesorsen resulted in almost normalized triglyceride levels. No further relapses of acute pancreatitis occurred. Patient reported an improve life quality due to alleviated chronic abdominal pain and headaches. CONCLUSIONS Our case reports a rare yet potentially life-threatening condition-monogenic hypertriglyceridemia-induced acute pancreatitis. The implementation of the antisense drug volanesorsen resulted in improved triglyceride levels, alleviated symptoms, and enhanced the quality of life.
Collapse
Affiliation(s)
- Umidakhon Makhmudova
- Deutsches Herzzentrum der Charité, Hindenburgdamm 30, 12203, Berlin, Germany.
- Friede Springer Cardiovascular Prevention Center @Charité, Hindenburgdamm 30, 12203, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117, Berlin, Germany.
- Klinik für Innere Medizin I, Universitätsklinikum Jena, Am Klinikum 1, 07743, Jena, Germany.
| | - P Christian Schulze
- Klinik für Innere Medizin I, Universitätsklinikum Jena, Am Klinikum 1, 07743, Jena, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Winfried März
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH Mannheim and Augsburg GmbH, Mannheim, Germany
| | - J-A Geiling
- Klinik für Innere Medizin I, Universitätsklinikum Jena, Am Klinikum 1, 07743, Jena, Germany
| | - Oliver Weingärtner
- Klinik für Innere Medizin I, Universitätsklinikum Jena, Am Klinikum 1, 07743, Jena, Germany
| |
Collapse
|
2
|
Mozafari S, Ashoori M, Emami Meybodi SM, Solhi R, Mirjalili SR, Firoozabadi AD, Soltani S. Association between APOA5 polymorphisms and susceptibility to metabolic syndrome: a systematic review and meta-analysis. BMC Genomics 2024; 25:590. [PMID: 38867151 PMCID: PMC11167842 DOI: 10.1186/s12864-024-10493-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND The association between Apolipoprotein A5 (APOA5) genetic polymorphisms and susceptibility to metabolic syndrome (MetS) has been established by many studies, but there have been conflicting results from the literature. We performed a meta-analysis of observational studies to evaluate the association between APOA5 gene polymorphisms and the prevalence of MetS. METHODS PubMed, Web of Science, Embase, and Scopus were searched up to April 2024. The random effects model was used to estimate the odds ratios (ORs) and 95% confidence intervals (CI) of the association between APOA5 gene polymorphisms and the prevalence of MetS development. The potential sources of heterogeneity were evaluated by subgroup analyses and sensitivity analyses. RESULTS A total of 30 studies with 54,986 subjects (25,341 MetS cases and 29,645 healthy controls) were included. The presence of rs662799 and rs651821 polymorphisms is associated with an approximately 1.5-fold higher likelihood of MetS prevalence (OR = 1.42, 95% CI: 1.32, 1.53, p < 0.001; I2 = 67.1%; P-heterogeneity < 0.001; and OR = 1.50, 95% CI: 1.36-1.65, p < 0.001), respectively. MetS is also more prevalent in individuals with the genetic variants rs3135506 and rs2075291. There was no evidence of a connection with rs126317. CONCLUSION The present findings suggest that polymorphisms located in the promoter and coding regions of the APOA5 gene are associated with an increased prevalence of MetS in the adult population. Identifying individuals with these genetic variations could lead to early disease detection and the implementation of preventive strategies to reduce the risk of MetS and its related health issues. However, because the sample size was small and there was evidence of significant heterogeneity for some APOA5 gene polymorphisms, these results need to be confirmed by more large-scale and well-designed studies.
Collapse
Affiliation(s)
- Sima Mozafari
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Afshar Hospital, Jomhouri Blvd., Yazd, 8917945556, Iran
| | - Marziyeh Ashoori
- Rasool Akram Medical Complex, Clinical Research Development Center, Tehran, Iran
| | - Seyed Mahdi Emami Meybodi
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Afshar Hospital, Jomhouri Blvd., Yazd, 8917945556, Iran
| | - Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Seyed Reza Mirjalili
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Afshar Hospital, Jomhouri Blvd., Yazd, 8917945556, Iran
| | - Ali Dehghani Firoozabadi
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Afshar Hospital, Jomhouri Blvd., Yazd, 8917945556, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Afshar Hospital, Jomhouri Blvd., Yazd, 8917945556, Iran.
| |
Collapse
|
3
|
Liu YL, Xiang Z, Zhang BY, Zou YW, Chen GL, Yin L, Shi YL, Xu LL, Bi J, Wang Q. APOA5 alleviates reactive oxygen species to promote oxaliplatin resistance in PIK3CA-mutated colorectal cancer. Aging (Albany NY) 2024; 16:9410-9436. [PMID: 38848145 PMCID: PMC11210231 DOI: 10.18632/aging.205872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/25/2024] [Indexed: 06/09/2024]
Abstract
Although platinum-based chemotherapy is the frontline regimen for colorectal cancer (CRC), drug resistance remains a major challenge affecting its therapeutic efficiency. However, there is limited research on the correlation between chemotherapy resistance and lipid metabolism, including PIK3CA mutant tumors. In this present study, we found that PIK3CA-E545K mutation attenuated cell apoptosis and increased the cell viability of CRC with L-OHP treatment in vitro and in vivo. Mechanistically, PIK3CA-E545K mutation promoted the nuclear accumulation of SREBP1, which promoted the transcription of Apolipoprotein A5 (APOA5). APOA5 activated the PPARγ signaling pathway to alleviate reactive oxygen species (ROS) production following L-OHP treatment, which contributed to cell survival of CRC cells. Moreover, APOA5 overexpression enhanced the stemness-related traits of CRC cells. Increased APOA5 expression was associated with PIK3CA mutation in tumor specimens and poor response to first-line chemotherapy, which was an independent detrimental factor for chemotherapy sensitivity in CRC patients. Taken together, this study indicated that PIK3CA-E545K mutation promoted L-OHP resistance by upregulating APOA5 transcription in CRC, which could be a potent target for improving L-OHP chemotherapeutic efficiency. Our study shed light to improve chemotherapy sensitivity through nutrient management in CRC.
Collapse
Affiliation(s)
- Yu-Lin Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250023, China
- Department of Oncology, Shandong Second Provincial General Hospital, Jinan 250023, China
| | - Zhuo Xiang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250023, China
- Department of Oncology, Shandong Second Provincial General Hospital, Jinan 250023, China
| | - Bo-Ya Zhang
- China Key Laboratory of Marine Drugs, The Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Yu-Wei Zou
- Department of Pathology, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, China
| | - Gui-Lai Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250023, China
- Department of Oncology, Shandong Second Provincial General Hospital, Jinan 250023, China
| | - Li Yin
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250023, China
- Department of Oncology, Shandong Second Provincial General Hospital, Jinan 250023, China
| | - Yan-Long Shi
- Department of Oncology, 960 Hospital of People’s Liberation Army, Jinan 250031, China
| | - Li-Li Xu
- Department of Pathology, Navy 971 People’s Liberation Army Hospital, Qingdao 266071, China
| | - Jingwang Bi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250023, China
- Department of Oncology, Shandong Second Provincial General Hospital, Jinan 250023, China
| | - Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250023, China
- Department of Oncology, Shandong Second Provincial General Hospital, Jinan 250023, China
| |
Collapse
|
4
|
Zhang R, Zhang K. A unified model for regulating lipoprotein lipase activity. Trends Endocrinol Metab 2024; 35:490-504. [PMID: 38521668 DOI: 10.1016/j.tem.2024.02.016] [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: 12/25/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
The regulation of triglyceride (TG) tissue distribution, storage, and utilization, a fundamental process of energy homeostasis, critically depends on lipoprotein lipase (LPL). We review the intricate mechanisms by which LPL activity is regulated by angiopoietin-like proteins (ANGPTL3, 4, 8), apolipoproteins (APOA5, APOC3, APOC2), and the cAMP-responsive element-binding protein H (CREBH). ANGPTL8 functions as a molecular switch, through complex formation, activating ANGPTL3 while deactivating ANGPTL4 in their LPL inhibition. The ANGPTL3-4-8 model integrates the roles of the aforementioned proteins in TG partitioning between white adipose tissue (WAT) and oxidative tissues (heart and skeletal muscles) during the feed/fast cycle. This model offers a unified perspective on LPL regulation, providing insights into TG metabolism, metabolic diseases, and therapeutics.
Collapse
Affiliation(s)
- Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
5
|
Alves M, Laranjeira F, Correia-da-Silva G. Understanding Hypertriglyceridemia: Integrating Genetic Insights. Genes (Basel) 2024; 15:190. [PMID: 38397180 PMCID: PMC10887881 DOI: 10.3390/genes15020190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Hypertriglyceridemia is an exceptionally complex metabolic disorder characterized by elevated plasma triglycerides associated with an increased risk of acute pancreatitis and cardiovascular diseases such as coronary artery disease. Its phenotype expression is widely heterogeneous and heavily influenced by conditions as obesity, alcohol consumption, or metabolic syndromes. Looking into the genetic underpinnings of hypertriglyceridemia, this review focuses on the genetic variants in LPL, APOA5, APOC2, GPIHBP1 and LMF1 triglyceride-regulating genes reportedly associated with abnormal genetic transcription and the translation of proteins participating in triglyceride-rich lipoprotein metabolism. Hypertriglyceridemia resulting from such genetic abnormalities can be categorized as monogenic or polygenic. Monogenic hypertriglyceridemia, also known as familial chylomicronemia syndrome, is caused by homozygous or compound heterozygous pathogenic variants in the five canonical genes. Polygenic hypertriglyceridemia, also known as multifactorial chylomicronemia syndrome in extreme cases of hypertriglyceridemia, is caused by heterozygous pathogenic genetic variants with variable penetrance affecting the canonical genes, and a set of common non-pathogenic genetic variants (polymorphisms, using the former nomenclature) with well-established association with elevated triglyceride levels. We further address recent progress in triglyceride-lowering treatments. Understanding the genetic basis of hypertriglyceridemia opens new translational opportunities in the scope of genetic screening and the development of novel therapies.
Collapse
Affiliation(s)
- Mara Alves
- Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Francisco Laranjeira
- CGM—Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar Universitário de Santo António (CHUdSA), 4099-028 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-346 Porto, Portugal
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-600 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO Applied Molecular Biosciences Unit and Associate Laboratory i4HB—Institute for Health and Bioeconomy Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
6
|
Wheless A, Gunn KH, Neher SB. Macromolecular Interactions of Lipoprotein Lipase (LPL). Subcell Biochem 2024; 104:139-179. [PMID: 38963487 DOI: 10.1007/978-3-031-58843-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Lipoprotein lipase (LPL) is a critical enzyme in humans that provides fuel to peripheral tissues. LPL hydrolyzes triglycerides from the cores of lipoproteins that are circulating in plasma and interacts with receptors to mediate lipoprotein uptake, thus directing lipid distribution via catalytic and non-catalytic functions. Functional losses in LPL or any of its myriad of regulators alter lipid homeostasis and potentially affect the risk of developing cardiovascular disease-either increasing or decreasing the risk depending on the mutated protein. The extensive LPL regulatory network tunes LPL activity to allocate fatty acids according to the energetic needs of the organism and thus is nutritionally responsive and tissue dependent. Multiple pharmaceuticals in development manipulate or mimic these regulators, demonstrating their translational importance. Another facet of LPL biology is that the oligomeric state of the enzyme is also central to its regulation. Recent structural studies have solidified the idea that LPL is regulated not only by interactions with other binding partners but also by self-associations. Here, we review the complexities of the protein-protein and protein-lipid interactions that govern LPL structure and function.
Collapse
Affiliation(s)
- Anna Wheless
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn H Gunn
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Stony Brook University, Stony Brook, USA
| | - Saskia B Neher
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Mirabedini S, Musavi H, Makhlough A, Hashemi-Sooteh MB, Zargari M. Association of S19W polymorphism in APOA5 gene and serum lipid levels in patients with type 2 diabetic nephropathy. Horm Mol Biol Clin Investig 2023; 44:243-249. [PMID: 36855913 DOI: 10.1515/hmbci-2022-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 02/11/2023] [Indexed: 03/02/2023]
Abstract
OBJECTIVES Type 2 diabetic Mellitus (T2DM) is the most common systemic and endocrine disease in humans, and diabetic nephropathy is one of the most serious complications of this disorder. The polymorphisms in the apolipoprotein A5 (ApoA5) gene are strongly related to hypertriglyceridemia and are considered a predisposing factor for diabetic nephropathy. The current study proposed to examine the association of APOA5-S19W polymorphism with serum lipids levels in patients with type 2 diabetic nephropathy in Mazandaran province. METHODS This case-control study was designed to determine the association of APOA5-S19W polymorphism with plasma lipid profile in 161 T2DM patients with nephropathy (DN+), without nephropathy (DN-), and in 58 healthy individuals. Lipid profile values were measured using Pars Azmoun commercial kits. S19W variant, one of the polymorphisms of the APOA5 gene, was determined by PCR-restriction fragment length polymorphism (PCR-RFLP) and Taq1 restriction enzyme. RESULTS In comparison between the three groups, DN+ had a higher mean TG than DN- and the control group (p<0.001). The incidence of the G allele in DN+ was not significant compared to groups of DN-. Comparing the relationship between the mean of biochemical variables with CC and CG genotypes showed that the mean level of TG in people with CC genotype was increased compared to people with CG genotype in diabetic patients. However, this increase was not significant (p=0.19). CONCLUSIONS There was no association between SNP APOA5 S19W and serum lipids in diabetic patients with and without nephropathy.
Collapse
Affiliation(s)
- Shivasadat Mirabedini
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hadis Musavi
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Atieh Makhlough
- Department of Internal Medicine, Diabetes Research Center, Sari Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad-Bagher Hashemi-Sooteh
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehryar Zargari
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
8
|
Ovando Gómez V, Zavaleta Muñiz SA, Ochoa-Díaz-López H, Hernández Contreras JAC, Irecta Nájera CA. Association of rs662799 and rs5070 genetic polymorphisms with hypertriglyceridemia and atherogenic dyslipidemia in pediatric patients in Southeast Mexico. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:53-63. [PMID: 35961839 DOI: 10.1016/j.arteri.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 04/21/2023]
Abstract
BACKGROUND AND AIMS Triglycerides are the initiators of the metabolic changes that lead to atherogenic dyslipidemia (AD). The APOA5 and APOA1 genes are involved in the response and metabolism of serum lipids and lipoproteins, where single nucleotide polymorphisms (SNP) rs662799 (promoter region) and rs5070 (intronic region) have been associated with the susceptibility to dyslipidemia. Until now, few studies evaluate the association of these polymorphisms with the presentation of hypertriglyceridemia and AD among Mexican children. Therefore, the objective was to determine the association between rs662799 and rs5070 with hypertriglyceridemia and AD in a pediatric population of southeastern Mexico. MATERIALS AND METHODS A case-control analysis was performed including 268 infants aged 2-16 years, anthropometric, clinical variables, and serum lipid profiles were analyzed. DNA was extracted from blood samples and genotyping of polymorphisms was executed with the TaqMan SNP genotyping assay. Allele and genotypic frequencies were calculated. For genetic association analysis, logistic regression models were fitted according to models of inheritance. RESULTS The SNP rs662799 (C) was significantly associated with hypertriglyceridemia in the overdominant model (OR=3.89, p=0.001) and AD in the dominant model (OR=4.01, p=0.001). The SNP rs5070 (T) has a protective effect against hypertriglyceridemia in the additive risk model (OR=0.68, p=0.03). CONCLUSION Polymorphism rs662799 was significantly associated with cases of hypertriglyceridemia and AD in minors in southeastern Mexico. On the other hand, rs5070 polymorphism was not associated with cases of hypertriglyceridemia or AD.
Collapse
Affiliation(s)
| | - Soraya Amalí Zavaleta Muñiz
- Health Sciences Faculty, Facultad de Ciencias de la Salud, Universidad Juárez del Estado de Durango, Gómez Palacio, Durango, Mexico
| | - Héctor Ochoa-Díaz-López
- Health Department, El Colegio de la Frontera Sur, San Cristóbal de las Casas, Chiapas, Mexico
| | | | | |
Collapse
|
9
|
Ding K, Zhou Z, Ma Y, Li X, Xiao H, Wu Y, Wu T, Chen D. Identification of Novel Metabolic Subtypes Using Multi-Trait Limited Mixed Regression in the Chinese Population. Biomedicines 2022; 10:biomedicines10123093. [PMID: 36551856 PMCID: PMC9775185 DOI: 10.3390/biomedicines10123093] [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: 10/27/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The aggregation and interaction of metabolic risk factors leads to highly heterogeneous pathogeneses, manifestations, and outcomes, hindering risk stratification and targeted management. To deconstruct the heterogeneity, we used baseline data from phase II of the Fangshan Family-Based Ischemic Stroke Study (FISSIC), and a total of 4632 participants were included. A total of 732 individuals who did not have any component of metabolic syndrome (MetS) were set as a reference group, while 3900 individuals with metabolic abnormalities were clustered into subtypes using multi-trait limited mixed regression (MFMR). Four metabolic subtypes were identified with the dominant characteristics of abdominal obesity, hypertension, hyperglycemia, and dyslipidemia. Multivariate logistic regression showed that the hyperglycemia-dominant subtype had the highest coronary heart disease (CHD) risk (OR: 6.440, 95% CI: 3.177-13.977) and that the dyslipidemia-dominant subtype had the highest stroke risk (OR: 2.450, 95% CI: 1.250-5.265). Exome-wide association studies (EWASs) identified eight SNPs related to the dyslipidemia-dominant subtype with genome-wide significance, which were located in the genes APOA5, BUD13, ZNF259, and WNT4. Functional analysis revealed an enrichment of top genes in metabolism-related biological pathways and expression in the heart, brain, arteries, and kidneys. Our findings provide directions for future attempts at risk stratification and evidence-based management in populations with metabolic abnormalities from a systematic perspective.
Collapse
|
10
|
Jacob J, Boczkowska S, Zaluska W, Buraczynska M. Apolipoprotein A5 gene polymorphism (rs662799) and cardiovascular disease in end-stage kidney disease patients. BMC Nephrol 2022; 23:307. [PMID: 36071387 PMCID: PMC9450442 DOI: 10.1186/s12882-022-02925-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Background Plasma triglyceride (TG) levels are a significant risk factor for cardiovascular disease (CVD). The APOA5 gene is one of the crucial factors in plasma TG metabolism regulation. The rs662799 polymorphism in the APOA5 gene has been reported to be associated with cardiovascular disease. The goal of this study was to evaluate the potential association of this variant with CVD in patients with end-stage kidney disease. Methods In this case–control study the polymorphism was analyzed using the PCR–RFLP method in 800 consecutive patients with ESKD and 500 healthy controls. The genotype and allele distribution was compared between subgroups of patients with CVD (552) versus those without CVD (248). Results The frequency of the minor allele (C) in the healthy individuals was 9% compared to 12% in ESRD group (p = 0.09). The difference between groups was slightly higher for CC homozygote (3.5% versus 1.6%, p = 0.042). The ESKD patient group was analyzed according to the presence or absence of CVD. The significant differences in the polymorphism distribution were revealed in this analysis. The frequency of the C allele in the CVD + subgroup was 14% compared to 6% in CVD- patients (p = 0.001). In the CVD + subgroup the ORs (95% CI) for the C allele and CC genotype were 2.41 (1.61–3.6), p < 0.001 and 3.13 (1.07–9.14), p = 0.036, respectively. This indicates to the association of the variant C allele with cardiovascular disease in ESKD patients. The CC homozygotes have a threefold higher odds of CVD compared to TT homozygotes. The highest frequency of the C allele (18%) was observed in subgroup of patients with diabetic nephropathy, with OR (95% CI) 3.40 (2.13–5.43), p < 0.001.The presence of minor allele (CC and CT genotypes) was significantly associated with increased plasma triglyceride levels (p < 0.001 for both CVD + and CVD- groups). Conclusion The present study demonstrated the effect of rs662799 polymorphism on plasma TG levels and its association with the development of cardiovascular disease in ESKD patients.
Collapse
Affiliation(s)
- Jerry Jacob
- Hope Medical Institute, Newport News, VA, USA
| | - Sylwia Boczkowska
- Department of Nephrology, Medical University of Lublin, Jaczewskiego 8, 20-950, Lublin, Poland
| | - Wojciech Zaluska
- Department of Nephrology, Medical University of Lublin, Jaczewskiego 8, 20-950, Lublin, Poland
| | - Monika Buraczynska
- Department of Nephrology, Medical University of Lublin, Jaczewskiego 8, 20-950, Lublin, Poland.
| |
Collapse
|
11
|
Chen J, Luo J, Qiu H, Tang Y, Yang X, Chen Y, Li Z, Li J. Apolipoprotein A5 ameliorates MCT induced pulmonary hypertension by inhibiting ER stress in a GRP78 dependent mechanism. Lipids Health Dis 2022; 21:69. [PMID: 35941581 PMCID: PMC9358849 DOI: 10.1186/s12944-022-01680-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a chronic, progressive lung vascular disease accompanied by elevated pulmonary vascular pressure and resistance, and it is characterized by increased pulmonary artery smooth muscle cell (PASMC) proliferation. Apolipoprotein A5 (ApoA5) improves monocrotaline (MCT)-induced PAH and right heart failure; however, the underlying mechanism remains unknown. Here we speculate that ApoA5 has a protective effect in pulmonary vessels and aim to evaluate the mechanism. Methods ApoA5 is overexpressed in an MCT-induced PAH animal model and platelet-derived growth factor (PDGF)-BB-induced proliferating PASMCs. Lung vasculature remodeling was measured by immunostaining, and PASMC proliferation was determined by cell counting kit‐8 and 5‐ethynyl‐2'‐deoxyuridine5‐ethynyl‐2'‐deoxyuridine incorporation assays. Coimmunoprecipitation-mass spectrometry was used to investigate the probable mechanism. Next, its role and mechanism were further verified by knockdown studies. Results ApoA5 level was decreased in MCT-induced PAH lung as well as PASMCs. Overexpression of ApoA5 could help to inhibit the remodeling of pulmonary artery smooth muscle. ApoA5 could inhibit PDGF-BB-induced PASMC proliferation and endoplasmic reticulum stress by increasing the expression of glucose-regulated protein 78 (GRP78). After knocking down GRP78, the protecting effects of ApoA5 have been blocked. Conclusion ApoA5 ameliorates MCT-induced PAH by inhibiting endoplasmic reticulum stress in a GRP78 dependent mechanism. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01680-4.
Collapse
Affiliation(s)
- Jingyuan Chen
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha City, Hunan Province, 410011, China
| | - Jun Luo
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha City, Hunan Province, 410011, China
| | - Haihua Qiu
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha City, Hunan Province, 410011, China
| | - Yi Tang
- Department of Cardiology, Clinical Medicine Research Center of Heart Failure of Hunan Province, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Hunan Normal University, Changsha, Hunan, China
| | - Xiaojie Yang
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha City, Hunan Province, 410011, China
| | - Yusi Chen
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha City, Hunan Province, 410011, China
| | - Zilu Li
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha City, Hunan Province, 410011, China
| | - Jiang Li
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha City, Hunan Province, 410011, China.
| |
Collapse
|
12
|
Li X, Shi X, Mesalam NM, Liu L, Chen Z, Yang B. Mechanism of Lysoforte in Improving Jejuna Morphology and Health in Broiler Chickens. Front Vet Sci 2022; 9:946148. [PMID: 35928108 PMCID: PMC9343761 DOI: 10.3389/fvets.2022.946148] [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: 05/17/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Lysoforte (LFT) plays a vital role in maintaining broilers' health and intestinal morphology. However, the mechanism behind the effects of LFT improving intestinal morphology and health is still unclear. Therefore, this study was implemented to explore the central genes linked to the regulatory effect of LFT. Seventy-five newly hatched Cobb 500 male broilers were randomly divided into three groups: control, LFT500, and LFT1000 groups, with 25 chicks per group. The control chicks were provided with the basal diet, and the birds in LFT500 and LFT1000 groups were offered the same basal diet with 500 g/ton and 1,000 g/ton LFT, respectively. GSE94622 dataset consisted of the control and two LFT-treated groups (LFT500 and LFT1000). Jejuna samples were obtained from Gene Expression Omnibus (GEO). Totally 106–344 DEGs were obtained by comparing LFT500 and LFT1000 vs. control and LFT1000 vs. LFT500. Gene ontology (GO) enrichment suggested that the DEGs are mainly related to the phosphatidylethanolamine biosynthetic process and neuron projection extension. KEGG analysis suggested the DEGs were enriched in AGE-RAGE, fatty acid elongation, ECM-receptor interaction (ECMRI), glycerophospholipid metabolism, focal adhesion, unsaturated fatty acids biosynthesis, and ABC transporters. Moreover, 29 genes, such as REG4, GJB1, KAT2A, APOA5, SERPINE2, ELOVL1, ABCC2, ANKRD9, CYP4V2, and PISD, might be closely related to promoting jejuna morphology in broilers. Taken together, our observation enhances the understanding of LFT in maintaining intestinal architecture and the general health of broiler chickens.
Collapse
Affiliation(s)
- Xiaofeng Li
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| | - Xiaoli Shi
- College of Animal Science, Guizhou University, Guiyang, China
| | - Noura M. Mesalam
- Biological Applications Department, Nuclear Research Center, Egyptian Atomic Energy Authority, Abu-Zaabal, Egypt
| | - Lei Liu
- Center of Reproductive Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhihao Chen
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| | - Bing Yang
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
- *Correspondence: Bing Yang
| |
Collapse
|
13
|
Zhang BH, Yin F, Qiao YN, Guo SD. Triglyceride and Triglyceride-Rich Lipoproteins in Atherosclerosis. Front Mol Biosci 2022; 9:909151. [PMID: 35693558 PMCID: PMC9174947 DOI: 10.3389/fmolb.2022.909151] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is still the leading cause of death globally, and atherosclerosis is the main pathological basis of CVDs. Low-density lipoprotein cholesterol (LDL-C) is a strong causal factor of atherosclerosis. However, the first-line lipid-lowering drugs, statins, only reduce approximately 30% of the CVD risk. Of note, atherosclerotic CVD (ASCVD) cannot be eliminated in a great number of patients even their LDL-C levels meet the recommended clinical goals. Previously, whether the elevated plasma level of triglyceride is causally associated with ASCVD has been controversial. Recent genetic and epidemiological studies have demonstrated that triglyceride and triglyceride-rich lipoprotein (TGRL) are the main causal risk factors of the residual ASCVD. TGRLs and their metabolites can promote atherosclerosis via modulating inflammation, oxidative stress, and formation of foam cells. In this article, we will make a short review of TG and TGRL metabolism, display evidence of association between TG and ASCVD, summarize the atherogenic factors of TGRLs and their metabolites, and discuss the current findings and advances in TG-lowering therapies. This review provides information useful for the researchers in the field of CVD as well as for pharmacologists and clinicians.
Collapse
Affiliation(s)
| | | | - Ya-Nan Qiao
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
14
|
Apolipoprotein A-V is a potential target for treating coronary artery disease: evidence from genetic and metabolomic analyses. J Lipid Res 2022; 63:100193. [PMID: 35278410 PMCID: PMC9062431 DOI: 10.1016/j.jlr.2022.100193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 11/22/2022] Open
Abstract
Triglyceride (TG)-lowering LPL variants in combination with genetic LDL-C-lowering variants are associated with reduced risk of coronary artery disease (CAD). Genetic variation in the APOA5 gene encoding apolipoprotein A-V also strongly affects TG levels, but the potential clinical impact and underlying mechanisms are yet to be resolved. Here, we aimed to study the effects of APOA5 genetic variation on CAD risk and plasma lipoproteins through factorial genetic association analyses. Using data from 309,780 European-ancestry participants from the UK Biobank, we evaluated the effects of lower TG levels as a result of genetic variation in APOA5 and/or LPL on CAD risk with or without a background of reduced LDL-C. Next, we compared lower TG levels via APOA5 and LPL variation with over 100 lipoprotein measurements in a combined sample from the Netherlands Epidemiology of Obesity study (N = 4,838) and the Oxford Biobank (N = 6,999). We found that lower TG levels due to combined APOA5 and LPL variation and genetically-influenced lower LDL-C levels afforded the largest reduction in CAD risk (odds ratio: 0.78 (0.73-0.82)). Compared to patients with genetically-influenced lower TG via LPL, genetically-influenced lower TG via APOA5 had similar and independent, but notably larger, effects on the lipoprotein profile. Our results suggest that lower TG levels as a result of APOA5 variation have strong beneficial effects on CAD risk and the lipoprotein profile, which suggest apo A-V may be a potential novel therapeutic target for CAD prevention.
Collapse
|
15
|
Apolipoprotein A5, a unique modulator of fasting and postprandial triglycerides. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159185. [DOI: 10.1016/j.bbalip.2022.159185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/30/2022] [Accepted: 05/13/2022] [Indexed: 11/19/2022]
|
16
|
Bautista-Martínez JS, Mata-Marín JA, Sandoval-Ramírez JL, Chaparro-Sánchez A, Manjarrez-Téllez B, Uribe-Noguez LA, Gaytán-Martínez J, Núñez-Armendáriz M, Cruz-Sánchez A, Núñez-Rodríguez N, Iván MA, Morales-González GS, Álvarez-Mendoza JP, Pérez-Barragán E, Ríos-De Los Ríos J, Contreras-Chávez GG, Tapia-Magallanes DM, Ribas-Aparicio RM, Díaz-López M, Olivares-Labastida A, Gómez-Delgado A, Torres J, Miranda-Duarte A, Zenteno JC, Pompa-Mera EN. Contribution of APOA5, APOC3, CETP, ABCA1 and SIK3 genetic variants to hypertriglyceridemia development in Mexican HIV-patients receiving antiretroviral therapy. Pharmacogenet Genomics 2022; 32:101-110. [PMID: 34693928 DOI: 10.1097/fpc.0000000000000458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the impact of single nucleotide polymorphisms (SNPs) from APOA5, APOC3, CETP, ATP binding cassette transporter A1 and SIK3 genes in the development of hypertriglyceridemia in HIV patients under antiretroviral therapy. MATERIAL AND METHODS A case-control study was developed. Leukocytic genomic DNA was extracted and genotyping for SNPs rs662799, rs964184, rs5128, rs2854116, rs2854117, rs3764261, rs4149310, rs4149267 and rs139961185 was performed by real time-PCR using TaqMan allelic discrimination assays, in Mexican mestizo patients with HIV infection, with hypertriglyceridemia (>1.7 mmol/L) under antiretroviral therapy. Genetic variants were also investigated in a control group of normolipidemic HIV patients (≤ 1.7 mmol/L). Haplotypes and gene interactions were analyzed. RESULTS A total of 602 HIV patients were genotyped (316 cases and 286 controls). Age and antiretroviral regimen based on protease inhibitors were associated with hypertriglyceridemia (P = 0.0001 and P = 0.0002. respectively). SNP rs964184 GG genotype in APOA5 gene exhibited the highest association with hypertriglyceridemia risk (OR, 3.2, 95% CI, 1.7-5.8, P = 0.0001); followed by SNP rs139961185 in SIK3 gene (OR = 2.3; (95% CI, 1.1-4.8; P = 0.03 for AA vs. AG genotype; and APOC3 rs5128 GG genotype, (OR, 2.2; 95% CI, 1.1-4.9; P = 0.04) under codominant models. These associations were maintained in the adjusted analysis by age and protease inhibitors based antiretroviral regimens. CONCLUSIONS This study reveals an association between rs964184 in APOA5; rs5128 in APOC3 and rs139961185 in SIK3 and high triglyceride concentrations in Mexican HIV-patients receiving protease inhibitors. These genetic factors may influence the adverse effects related to antiretroviral therapy.
Collapse
Affiliation(s)
- Jonathan Saúl Bautista-Martínez
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional
| | - José Antonio Mata-Marín
- Servicio de Infectología de Adultos, Hospital de Infectología, Centro Médico Nacional "La Raza"
| | | | | | | | | | - Jesús Gaytán-Martínez
- Servicio de Infectología de Adultos, Hospital de Infectología, Centro Médico Nacional "La Raza"
| | | | | | | | - Martínez-Abarca Iván
- Hospital General Regional 72, Instituto Mexicano del Seguro Social, IMSS. Tlalnepantla, Estado de México
| | | | | | | | - Jussara Ríos-De Los Ríos
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
| | - Gerson Gabriel Contreras-Chávez
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
| | - Denisse Marielle Tapia-Magallanes
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
| | - Rosa Maria Ribas-Aparicio
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional
| | - Mónica Díaz-López
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional
| | - Azucena Olivares-Labastida
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional
| | - Alejandro Gómez-Delgado
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
| | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
| | - Antonio Miranda-Duarte
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra"
| | - Juan C Zenteno
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana"
- Department of Biochemistry, Faculty of Medicine, UNAM, Mexico City, Mexico
| | - Ericka Nelly Pompa-Mera
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, IMSS
| |
Collapse
|
17
|
San-Cristobal R, de Toro-Martín J, Vohl MC. Appraisal of Gene-Environment Interactions in GWAS for Evidence-Based Precision Nutrition Implementation. Curr Nutr Rep 2022; 11:563-573. [PMID: 35948824 PMCID: PMC9750926 DOI: 10.1007/s13668-022-00430-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW This review aims to analyse the currently reported gene-environment (G × E) interactions in genome-wide association studies (GWAS), involving environmental factors such as lifestyle and dietary habits related to metabolic syndrome phenotypes. For this purpose, the present manuscript reviews the available GWAS registered on the GWAS Catalog reporting the interaction between environmental factors and metabolic syndrome traits. RECENT FINDINGS Advances in omics-related analytical and computational approaches in recent years have led to a better understanding of the biological processes underlying these G × E interactions. A total of 42 GWAS were analysed, reporting over 300 loci interacting with environmental factors. Alcohol consumption, sleep time, smoking habit and physical activity were the most studied environmental factors with significant G × E interactions. The implementation of more comprehensive GWAS will provide a better understanding of the metabolic processes that determine individual responses to environmental exposures and their association with the development of chronic diseases such as obesity and the metabolic syndrome. This will facilitate the development of precision approaches for better prevention, management and treatment of these diseases.
Collapse
Affiliation(s)
- Rodrigo San-Cristobal
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| | - Juan de Toro-Martín
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| | - Marie-Claude Vohl
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| |
Collapse
|
18
|
Zwartjes MSZ, Gerdes VEA, Nieuwdorp M. The Role of Gut Microbiota and Its Produced Metabolites in Obesity, Dyslipidemia, Adipocyte Dysfunction, and Its Interventions. Metabolites 2021; 11:531. [PMID: 34436472 PMCID: PMC8398981 DOI: 10.3390/metabo11080531] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is becoming an increasing problem worldwide and is often, but not invariably, associated with dyslipidemia. The gut microbiota is increasingly linked to cardiovascular disease, nonalcoholic fatty liver disease, and type 2 diabetes mellitus. However, relatively little focus has been attributed to the role of gut-microbiota-derived metabolites in the development of dyslipidemia and alterations in lipid metabolism. In this review, we discuss current data involved in these processes and point out the therapeutic potentials. We cover the ability of gut microbiota metabolites to alter lipoprotein lipase action, VLDL secretion, and plasma triglyceride levels, and its effects on reverse cholesterol transport, adipocyte dysfunction, and adipose tissue inflammation. Finally, the current intervention strategies for treatment of obesity and dyslipidemia is addressed with emphasis on the role of gut microbiota metabolites and its ability to predict treatment efficacies.
Collapse
Affiliation(s)
- Max S. Z. Zwartjes
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (V.E.A.G.); (M.N.)
- Department of Internal Medicine, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM Hoofddorp, The Netherlands
| | - Victor E. A. Gerdes
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (V.E.A.G.); (M.N.)
- Department of Internal Medicine, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM Hoofddorp, The Netherlands
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (V.E.A.G.); (M.N.)
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
- Department of Internal Medicine, Diabetes Center, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
19
|
Masjoudi S, Sedaghati-Khayat B, Givi NJ, Bonab LNH, Azizi F, Daneshpour MS. Kernel machine SNP set analysis finds the association of BUD13, ZPR1, and APOA5 variants with metabolic syndrome in Tehran Cardio-metabolic Genetics Study. Sci Rep 2021; 11:10305. [PMID: 33986338 PMCID: PMC8119714 DOI: 10.1038/s41598-021-89509-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
Metabolic syndrome (MetS) is one of the most important risk factors for cardiovascular disease. The 11p23.3 chromosomal region plays a potential role in the pathogenesis of MetS. The present study aimed to assess the association between 18 single nucleotide polymorphisms (SNPs) located at the BUD13, ZPR1, and APOA5 genes with MetS in the Tehran Cardio-metabolic Genetics Study (TCGS). In 5421 MetS affected and non-affected participants, we analyzed the data using two models. The first model (MetS model) examined SNPs' association with MetS. The second model (HTg-MetS Model) examined the association of SNPs with MetS affection participants who had a high plasma triglyceride (TG). The four-gamete rules were used to make SNP sets from correlated nearby SNPs. The kernel machine regression models and single SNP regression evaluated the association between SNP sets and MetS. The kernel machine results showed two sets over three sets of correlated SNPs have a significant joint effect on both models (p < 0.0001). Also, single SNP regression results showed that the odds ratios (ORs) for both models are almost similar; however, the p-values had slightly higher significance levels in the HTg-MetS model. The strongest ORs in the HTg-MetS model belonged to the G allele in rs2266788 (MetS: OR = 1.3, p = 3.6 × 10–7; HTg-MetS: OR = 1.4, p = 2.3 × 10–11) and the T allele in rs651821 (MetS: OR = 1.3, p = 2.8 × 10–7; HTg-MetS: OR = 1.4, p = 3.6 × 10–11). In the present study, the kernel machine regression models could help assess the association between the BUD13, ZPR1, and APOA5 gene variants (11p23.3 region) with lipid-related traits in MetS and MetS affected with high TG.
Collapse
Affiliation(s)
- Sajedeh Masjoudi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box 19195-4763, Tehran, Iran
| | - Bahareh Sedaghati-Khayat
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box 19195-4763, Tehran, Iran
| | - Niloufar Javanrouh Givi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box 19195-4763, Tehran, Iran
| | - Leila Najd Hassan Bonab
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box 19195-4763, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam S Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, PO Box 19195-4763, Tehran, Iran.
| |
Collapse
|
20
|
Affiliation(s)
- Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
21
|
Niforou A, Konstantinidou V, Naska A. Genetic Variants Shaping Inter-individual Differences in Response to Dietary Intakes-A Narrative Review of the Case of Vitamins. Front Nutr 2020; 7:558598. [PMID: 33335908 PMCID: PMC7736113 DOI: 10.3389/fnut.2020.558598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022] Open
Abstract
Recent advances in the field of nutrigenetics have provided evidence on how genetic variations can impact the individuals' response to dietary intakes. An objective and reliable assessment of dietary exposures should rely on combinations of methodologies including frequency questionnaires, short-term recalls or records, together with biological samples to evaluate markers of intake or status and to identify genetic susceptibilities. In an attempt to present current knowledge on how genetic fingerprints contribute to an individual's nutritional status, we present a review of current literature describing associations between genetic variants and levels of well-established biomarkers of vitamin status in free-living and generally healthy individuals. Based on the outcomes of candidate gene, genome-wide-association studies and meta-analyses thereof, we have identified several single nucleotide polymorphisms (SNPs) involved in the vitamins' metabolic pathways. Polymorphisms in genes encoding proteins involved in vitamin metabolism and transport are reported to have an impact on vitamin D status; while genetic variants of vitamin D receptor were most frequently associated with health outcomes. Genetic variations that can influence vitamin E status include SNPs involved in its uptake and transport, such as in SCAR-B1 gene, and in lipoprotein metabolism. Variants of the genes encoding the sodium-dependent vitamin C transport proteins are greatly associated with the body's status on vitamin C. Regarding the vitamins of the B-complex, special reference is made to the widely studied variant in the MTHFR gene. Methodological attributes of genetic studies that may limit the comparability and interpretability of the findings are also discussed. Our understanding of how genes affect our responses to nutritional triggers will enhance our capacity to evaluate dietary exposure and design personalized nutrition programs to sustain health and prevent disease.
Collapse
Affiliation(s)
- Aikaterini Niforou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Androniki Naska
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Association of rs662799 variant and APOA5 gene haplotypes with metabolic syndrome and its components: a meta-analysis in North Africa. Biosci Rep 2020; 40:225924. [PMID: 32725151 PMCID: PMC7426633 DOI: 10.1042/bsr20200706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/14/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Apolipoprotein A5 (APOA5) has been linked to metabolic syndrome (MetS) in several populations. In North Africa, only the Tunisian and Moroccan populations were investigated. Our aim is to assess the association between APOA5 gene variant (rs662799) and haplotypes with MetS in Tunisian population and to perform a meta-analysis in North Africa. A total of 594 Tunisian participants were genotyped for polymorphism rs662799 using KASPar technology. Two polymorphisms rs3135506 and rs651821 in APOA5 gene genotyped in our previous study, were used in addition to rs662799 to assess the haplotype association with MetS. The genotype of 875 participants was used for the meta-analysis. Statistical analyses were performed with R software. The rs662799 increases the risk of MetS under the dominant (P=0.018) and the additive models (P=0.028) in the Tunisian population. After stratification of the cohort following the sex and the geographic origin, a positive association of rs662799 with MetS was found for participant from the Northern region and for the women group. Only the haplotype AGT showed a significant association with MetS by decreasing the risk of the disease. The meta-analysis reported a significant association of rs662799 and rs3135506 with MetS. Our results showed a significant association between the APOA5 gene variants rs662799 and haplotypes with MetS and its traits in Tunisia. An impact of the sex and the geographic origin on the genotype distribution was highlighted. Our funding emphasizes the role of APOA5 in the development of MetS in North Africa.
Collapse
|
23
|
Hanh NTH, Nhung BT, Hop LT, Tuyet LT, Dao DTA, Thu NTT, Binh TQ. The APOA5-rs662799 Polymorphism Is a Determinant of Dyslipidemia in Vietnamese Primary School Children. Lipids 2020; 55:683-691. [PMID: 32777089 DOI: 10.1002/lipd.12270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/10/2020] [Accepted: 07/01/2020] [Indexed: 11/05/2022]
Abstract
Apolipoprotein A-V encoded by apolipoprotein 5 (APOA5) gene plays an important role in lipid metabolism, especially in the regulation of plasma triglycerol levels. The study aimed to evaluate the association of the APOA5-rs662799 polymorphism with dyslipidemia in Vietnamese children and the potential modification of obesity-related traits (body mass index, waist circumference, hip circumference, and waist-to-hip ratio) on this association. A case-control study was conducted with a total of 154 dyslipidemia cases and 389 controls at the age of 6 to 10 recruited at 31 primary schools in Hanoi city of Vietnam. Genotype for APOA5-rs662799 polymorphism was determined by the restriction fragment length polymorphism analysis. The association of APOA5-rs662799 polymorphism with dyslipidemia adjusting for age, sex, residence, and obesity-related traits was analyzed by binary logistic regression analysis. The results showed that in comparison with T/T and T/C carriers, the C/C carriers had a higher concentration of serum TAG in cases (p =0.049). Carriers of the C allele (C/C + T/C) had higher risk for developing dyslipidemia and hypertriglyceridemia than subjects with T/T genotype (odds ratio, OR = 1.7, p =0.0062 and OR = 1.6, p = 0.026, respectively). The association remained significant after adjusting for age, gender, residence, and obesity status (OR = 1.75, p = 0.006 and OR = 1.53, p = 0.049, respectively) or other obesity-related traits. The study suggested that the APOA5-rs662799 polymorphism may be a determinant of dyslipidemia and hypertriglyceridemia in Vietnamese children, independent of obesity-related traits.
Collapse
Affiliation(s)
- Nguyen T H Hanh
- Department of Biology, Hanoi National University of Education, 136 Xuan Thuy Street, Hanoi, 100000, Vietnam
| | - Bui T Nhung
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, 112807, Vietnam
| | - Le T Hop
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, 112807, Vietnam
| | - Le T Tuyet
- Department of Biology, Hanoi National University of Education, 136 Xuan Thuy Street, Hanoi, 100000, Vietnam
| | - Duong T A Dao
- Department of Biology, Hanoi National University of Education, 136 Xuan Thuy Street, Hanoi, 100000, Vietnam
| | - Nguyen T T Thu
- Department of Biology, Hanoi National University of Education, 136 Xuan Thuy Street, Hanoi, 100000, Vietnam
| | - Tran Quang Binh
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, 112807, Vietnam.,Dinh Tien Hoang Institute of Medicine, 20 Cat Linh Street, Hanoi, 100000, Vietnam
| |
Collapse
|
24
|
Morjane I, Charoute H, Ouatou S, Elkhattabi L, Benrahma H, Saile R, Rouba H, Barakat A. Association of c.56C > G (rs3135506) Apolipoprotein A5 Gene Polymorphism with Coronary Artery Disease in Moroccan Subjects: A Case-Control Study and an Updated Meta-Analysis. Cardiol Res Pract 2020; 2020:5981971. [PMID: 32832146 PMCID: PMC7424381 DOI: 10.1155/2020/5981971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Coronary artery diseases (CAD) are clinical cardiovascular events associated with dyslipidemia in common. The interaction between environmental and genetic factors can be responsible for CAD. The present paper aimed to examine the association between c.56C > G (rs3135506) APOA5 gene polymorphism and CAD in Moroccan individuals and to perform an association update meta-analysis. MATERIALS AND METHODS The c.56C > G variant was genotyped in 122 patients with CAD and 134 unrelated controls. Genetic association analysis and comparison of biochemical parameters were performed using R statistical language. In addition, a comprehensive meta-analysis including eleven published studies in addition to our case-control study results was conducted using Review Manager 5.3. Publication bias was examined by Egger's test and funnel plot. RESULTS The case-control study data showed that the c.56C > G polymorphism was associated with CAD susceptibility under codominant (P-value = 0.001), recessive (P-value <0.001) and log-additive (P-value = 0.008) inheritance models. In addition, this polymorphism was significantly associated with increased levels of systolic and diastolic blood pressures, triglycerides, glycemia, and total cholesterol. Furthermore, meta-analysis showed a significant association between the c.56C > G gene polymorphism and increased risk of CAD under recessive (OR = 3.39[1.77-6.50], P value <0.001) and homozygote codominant (OR = 3.96[2.44-6.45], P value <0.001) models. CONCLUSION Our case-control study revealed a significant association between c.56C > G polymorphism and CAD in the Moroccan population. In addition, meta-analysis data supported the implication of this polymorphism in CAD susceptibility.
Collapse
Affiliation(s)
- Imane Morjane
- Laboratory of Genomics and Human Genetics, Institut Pasteur Du Maroc, Casablanca, Morocco
- Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Hicham Charoute
- Laboratory of Genomics and Human Genetics, Institut Pasteur Du Maroc, Casablanca, Morocco
| | - Sanaa Ouatou
- Laboratory of Genomics and Human Genetics, Institut Pasteur Du Maroc, Casablanca, Morocco
| | - Lamiae Elkhattabi
- Laboratory of Genomics and Human Genetics, Institut Pasteur Du Maroc, Casablanca, Morocco
- Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Houda Benrahma
- Laboratory of Genomics and Human Genetics, Institut Pasteur Du Maroc, Casablanca, Morocco
- National Reference Laboratory (LNR), Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Rachid Saile
- Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Hassan Rouba
- Laboratory of Genomics and Human Genetics, Institut Pasteur Du Maroc, Casablanca, Morocco
| | - Abdelhamid Barakat
- Laboratory of Genomics and Human Genetics, Institut Pasteur Du Maroc, Casablanca, Morocco
| |
Collapse
|
25
|
Williams PT. Gene-environment interactions due to quantile-specific heritability of triglyceride and VLDL concentrations. Sci Rep 2020; 10:4486. [PMID: 32161301 PMCID: PMC7066156 DOI: 10.1038/s41598-020-60965-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/17/2020] [Indexed: 12/16/2022] Open
Abstract
"Quantile-dependent expressivity" is a dependence of genetic effects on whether the phenotype (e.g., triglycerides) is high or low relative to its distribution in the population. Quantile-specific offspring-parent regression slopes (βOP) were estimated by quantile regression for 6227 offspring-parent pairs. Quantile-specific heritability (h2), estimated by 2βOP/(1 + rspouse), decreased 0.0047 ± 0.0007 (P = 2.9 × 10-14) for each one-percent decrement in fasting triglyceride concentrations, i.e., h2 ± SE were: 0.428 ± 0.059, 0.230 ± 0.030, 0.111 ± 0.015, 0.050 ± 0.016, and 0.033 ± 0.010 at the 90th, 75th, 50th, 25th, and 10th percentiles of the triglyceride distribution, respectively. Consistent with quantile-dependent expressivity, 11 drug studies report smaller genotype differences at lower (post-treatment) than higher (pre-treatment) triglyceride concentrations. This meant genotype-specific triglyceride changes could not move in parallel when triglycerides were decreased pharmacologically, so that subtracting pre-treatment from post-treatment triglyceride levels necessarily created a greater triglyceride decrease for the genotype with a higher pre-treatment value (purported precision-medicine genetic markers). In addition, sixty-five purported gene-environment interactions were found to be potentially attributable to triglyceride's quantile-dependent expressivity, including gene-adiposity (APOA5, APOB, APOE, GCKR, IRS-1, LPL, MTHFR, PCSK9, PNPLA3, PPARγ2), gene-exercise (APOA1, APOA2, LPL), gene-diet (APOA5, APOE, INSIG2, LPL, MYB, NXPH1, PER2, TNFA), gene-alcohol (ALDH2, APOA5, APOC3, CETP, LPL), gene-smoking (APOC3, CYBA, LPL, USF1), gene-pregnancy (LPL), and gene-insulin resistance interactions (APOE, LPL).
Collapse
Affiliation(s)
- Paul T Williams
- Lawrence Berkeley National Laboratory, Molecular Biophysics & Integrated Bioimaging Division 1 Cyclotron Road, Berkeley, CA, 94720, USA.
| |
Collapse
|
26
|
Abstract
Purpose “Quantile-dependent expressivity” describes an effect of the genotype that depends upon the level of the phenotype (e.g., whether a subject’s triglycerides are high or low relative to its population distribution). Prior analyses suggest that the effect of a genetic risk score (GRS) on fasting plasma triglyceride levels increases with the percentile of the triglyceride distribution. Postprandial lipemia is well suited for testing quantile-dependent expressivity because it exposes each individual’s genotype to substantial increases in their plasma triglyceride concentrations. Ninety-seven published papers were identified that plotted mean triglyceride response vs. time and genotype, which were converted into quantitative data. Separately, for each published graph, standard least-squares regression analysis was used to compare the genotype differences at time t (dependent variable) to average triglyceride concentrations at time t (independent variable) to assess whether the genetic effect size increased in association with higher triglyceride concentrations and whether the phenomenon could explain purported genetic interactions with sex, diet, disease, BMI, and drugs. Results Consistent with the phenomenon, genetic effect sizes increased (P≤0.05) with increasing triglyceride concentrations for polymorphisms associated with ABCA1, ANGPTL4, APOA1, APOA2, APOA4, APOA5, APOB, APOC3, APOE, CETP, FABP2, FATP6, GALNT2, GCKR, HL, IL1b, LEPR, LOX-1, LPL, MC4R, MTTP, NPY, SORT1, SULF2, TNFA, TCF7L2, and TM6SF2. The effect size for these polymorphisms showed a progressively increasing dose-response, with intermediate effect sizes at intermediate triglyceride concentrations. Quantile-dependent expressivity provided an alternative interpretation to their interactions with sex, drugs, disease, diet, and age, which have been traditionally ascribed to gene-environment interactions and genetic predictors of drug efficacy (i.e., personalized medicine). Conclusion Quantile-dependent expressivity applies to the majority of genetic variants affecting postprandial triglycerides, which may arise because the impaired functionalities of these variants increase at higher triglyceride concentrations. Purported gene-drug interactions may be the manifestations of quantile-dependent expressivity, rather than genetic predictors of drug efficacy.
Collapse
|
27
|
Abstract
Hypertriglyceridemia, a commonly encountered phenotype in cardiovascular and metabolic clinics, is surprisingly complex. A range of genetic variants, from single-nucleotide variants to large-scale copy number variants, can lead to either the severe or mild-to-moderate forms of the disease. At the genetic level, severely elevated triglyceride levels resulting from familial chylomicronemia syndrome (FCS) are caused by homozygous or biallelic loss-of-function variants in LPL, APOC2, APOA5, LMF1, and GPIHBP1 genes. In contrast, susceptibility to multifactorial chylomicronemia (MCM), which has an estimated prevalence of ~1 in 600 and is at least 50-100-times more common than FCS, results from two different types of genetic variants: (1) rare heterozygous variants (minor allele frequency <1%) with variable penetrance in the five causal genes for FCS; and (2) common variants (minor allele frequency >5%) whose individually small phenotypic effects are quantified using a polygenic score. There is indirect evidence of similar complex genetic predisposition in other clinical phenotypes that have a component of hypertriglyceridemia, such as combined hyperlipidemia and dysbetalipoproteinemia. Future considerations include: (1) evaluation of whether the specific type of genetic predisposition to hypertriglyceridemia affects medical decisions or long-term outcomes; and (2) searching for other genetic contributors, including the role of genome-wide polygenic scores, novel genes, non-linear gene-gene or gene-environment interactions, and non-genomic mechanisms including epigenetics and mitochondrial DNA.
Collapse
|
28
|
Kelly JM, Ordovas JM, Matuszek G, Smith CE, Huggins GS, Dashti HS, Ichikawa R, Booth SL. The Contribution of Lipids to the Interindividual Response of Vitamin K Biomarkers to Vitamin K Supplementation. Mol Nutr Food Res 2019; 63:e1900399. [PMID: 31533195 PMCID: PMC8815429 DOI: 10.1002/mnfr.201900399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/16/2019] [Indexed: 12/12/2022]
Abstract
SCOPE A better understanding of factors contributing to interindividual variability in biomarkers of vitamin K can enhance the understanding of the equivocal role of vitamin K in cardiovascular disease. Based on the known biology of phylloquinone, the major form of vitamin K, it is hypothesized that plasma lipids contribute to the variable response of biomarkers of vitamin K metabolism to phylloquinone supplementation. METHODS AND RESULTS The association of plasma lipids and 27 lipid-related genetic variants with the response of biomarkers of vitamin K metabolism is examined in a secondary analysis of data from a 3-year phylloquinone supplementation trial in men (n = 66) and women (n = 85). Year 3 plasma triglycerides (TG), but not total cholesterol, LDL-cholesterol, or HDL-cholesterol, are associated with the plasma phylloquinone response (men: β = 1.01, p < 0.001, R2 = 0.34; women: β = 0.61, p = 0.008, R2 = 0.11; sex interaction p = 0.077). Four variants and the TG-weighted genetic risk score are associated with the plasma phylloquinone response in men only. Plasma lipids are not associated with changes in biomarkers of vitamin K function (undercarboxylated osteocalcin and matrix gla protein) in either sex. CONCLUSION Plasma TG are an important determinant of the interindividual response of plasma phylloquinone to phylloquinone supplementation, but changes in biomarkers of vitamin K carboxylation are not influenced by lipids.
Collapse
Affiliation(s)
- Jennifer M. Kelly
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Jose M. Ordovas
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Gregory Matuszek
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Caren E. Smith
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Gordon S. Huggins
- Molecular Cardiology Research Institute Center for Translational Genomics, Tufts Medical Center and Tufts University, Boston, MA
| | - Hassan S. Dashti
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Reiko Ichikawa
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Sarah L. Booth
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| |
Collapse
|
29
|
Tao YC, Wang ML, Wu DB, Luo C, Tang H, Chen EQ. Apolipoprotein A5 alleviates LPS/D-GalN-induced fulminant liver failure in mice by inhibiting TLR4-mediated NF-κB pathway. J Transl Med 2019; 17:151. [PMID: 31077206 PMCID: PMC6511152 DOI: 10.1186/s12967-019-1900-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/29/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Fulminant liver failure (FHF) is a serious clinical problem and liver transplantation is the major intervention. But the overall survival rate of FHF is low owing to the donated organ shortage. Apolipoprotein A-V (ApoA5) is a regulator of triglyceride metabolism and has been reported to act as a predictor for remnant liver growth after preoperative portal vein embolization and liver surgery. This study aimed to investigate the therapeutic effect of ApoA5 on lipopolysaccharide/D-galactosamine (LPS/D-GalN)-induced fulminant liver failure in mice. METHODS FHF mouse model was established using LPS/D-GalN and ApoA5 plasmid was injected by tail vein prior to LPS/D-GalN treatment. The expressions of ApoA5, toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and nuclear factor kappa B p65 (NF-κBp65) were assessed by real-time PCR and western blotting. Serum alanine aminotransferase (ALT) and tumor necrosis factor-α (TNF-α) levels were measured using automatic biochemical analyzer. Histological assessment and immunohistochemical (IHC) staining were conducted. Survival rate after LPS/D-GalN administration was also determined with Kaplan-Meier curve. Meanwhile, the expression of ApoA5 in injured huh7 cells was tested. Cell apoptosis analysis was performed after huh7 cells were transfected with ApoA5 plasmid and stimulated with LPS. RESULTS The expressions of ApoA5 decreased both in injured huh7 cells and FHF mice. ApoA5 overexpression reduced cell death rate using flow cytometry. ApoA5 not only decreased the serum ALT and TNF-α levels but also attenuated hepatic damage in hematoxylin-eosin (HE)-stained liver section. The protein expressions of TLR4, MyD88 and NF-κBp65 were inhibited when ApoA5 overexpressed. But the inhibitory effect would weaken with the increasing concentration of LPS in spite of ApoA5 overexpression. Besides, ApoA5 improved liver injury in a dose-dependent manner and the survival rate in FHF mice increased with increasing concentration of ApoA5. CONCLUSION ApoA5 had a protective effect against LPS/D-GalN-induced fulminant liver failure in mice within a certain range by inhibiting TLR4-mediated NF-κB pathway.
Collapse
Affiliation(s)
- Ya-Chao Tao
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China.,Division of Infectious Diseases, National Key Laboratory of Biotherapy (Sichuan University), West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Meng-Lan Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China.,Division of Infectious Diseases, National Key Laboratory of Biotherapy (Sichuan University), West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Dong-Bo Wu
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China.,Division of Infectious Diseases, National Key Laboratory of Biotherapy (Sichuan University), West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Chen Luo
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China.,Division of Infectious Diseases, National Key Laboratory of Biotherapy (Sichuan University), West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China.,Division of Infectious Diseases, National Key Laboratory of Biotherapy (Sichuan University), West China Hospital of Sichuan University, Chengdu, 610041, China
| | - En-Qiang Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, 610041, China. .,Division of Infectious Diseases, National Key Laboratory of Biotherapy (Sichuan University), West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
30
|
Suzuki R, Warita T, Nakamura Y, Kitamura Y, Aoyama Y, Ogawa Y, Kawada H, Ando K. A case of double-refractory multiple myeloma with both the IgH-MMSET fusion protein and the congenital abnormality t(11;22). Int J Hematol 2019; 109:731-736. [PMID: 30680670 DOI: 10.1007/s12185-019-02603-3] [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: 08/24/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 10/27/2022]
Abstract
A 67-year-old female was referred to our hospital with a sternal fracture in March 2008. She received a diagnosis of multiple myeloma (MM) BJP-κ type (ISS stage III). G-banding karyotype revealed 46, XX, t(11;22)(q23.3;q11.2) (Hubacek, Gene 592:193-9, 2016), which was later confirmed to be congenital. After repeated rounds of chemotherapy with bortezomib and lenalidomide, she obtained a very good partial response in August 2014, and she was followed up with no treatment. However, she relapsed in February 2016. At that time, fluorescence in situ hybridization identified del(13q) and t(4;14)(p16;q32), which are associated with a poor prognosis. Furthermore, PCR analysis showed that the chromosome 11 breakpoint was at the APOA5/APOA4 locus at 11q23.3, which is associated with malignancy, and that the chromosome 22 breakpoint was at the SEPT5 intron 1 locus, which also plays a role in leukemogenesis through formation of a fusion gene with MLL. Although she was treated with three further lines of therapy, she died from disease progression in August 2017. Synergism between t(11;22) and t(4;14) may have induced the double-refractory phenotype to proteasome inhibitor and lenalidomide, at least during the chemorefractory phase. We present a biological analysis of this case and a review of the literature.
Collapse
Affiliation(s)
- Rikio Suzuki
- Department of Hematology/Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan.
| | - Takayuki Warita
- Center for Regenerative Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoshihiko Nakamura
- Center for Regenerative Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yuka Kitamura
- Center for Regenerative Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yasuyuki Aoyama
- Department of Hematology/Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan
| | - Yoshiaki Ogawa
- Department of Hematology/Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan
| | - Hiroshi Kawada
- Department of Hematology/Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan
| | - Kiyoshi Ando
- Department of Hematology/Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan. .,Center for Regenerative Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan.
| |
Collapse
|
31
|
Vitamin E Metabolic Effects and Genetic Variants: A Challenge for Precision Nutrition in Obesity and Associated Disturbances. Nutrients 2018; 10:nu10121919. [PMID: 30518135 PMCID: PMC6316334 DOI: 10.3390/nu10121919] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023] Open
Abstract
Vitamin E (VE) has a recognized leading role as a contributor to the protection of cell constituents from oxidative damage. However, evidence suggests that the health benefits of VE go far beyond that of an antioxidant acting in lipophilic environments. In humans, VE is channeled toward pathways dealing with lipoproteins and cholesterol, underlining its relevance in lipid handling and metabolism. In this context, both VE intake and status may be relevant in physiopathological conditions associated with disturbances in lipid metabolism or concomitant with oxidative stress, such as obesity. However, dietary reference values for VE in obese populations have not yet been defined, and VE supplementation trials show contradictory results. Therefore, a better understanding of the role of genetic variants in genes involved in VE metabolism may be crucial to exert dietary recommendations with a higher degree of precision. In particular, genetic variability should be taken into account in targets concerning VE bioavailability per se or concomitant with impaired lipoprotein transport. Genetic variants associated with impaired VE liver balance, and the handling/resolution of oxidative stress might also be relevant, but the core information that exists at present is insufficient to deliver precise recommendations.
Collapse
|
32
|
Ariza MJ, Rioja J, Ibarretxe D, Camacho A, Díaz-Díaz JL, Mangas A, Carbayo-Herencia JA, Ruiz-Ocaña P, Lamíquiz-Moneo I, Mosquera D, Sáenz P, Masana L, Muñiz-Grijalvo O, Pérez-Calahorra S, Valdivielso P, Suárez Tembra M, Iglesias GP, Carbayo Herencia J, Guerrero Buitrago C, Vila L, Morales Coca C, Llargués Rocabruna E, Perea Castillo V, Pedro-Botet J, Climent E, Mauri Pont M, Pinto X, Ortega Martínez de la Victoria E, Amor J, Zambón Rados D, Blanco Vaca F, Ramiro Lozano J, Fuentes Jiménez F, Soler I, Ferrer C, Zamora Cervantes A, Vila Belmonte A, Novoa Mogollón F, Sanchez-Hernández R, Expósito Montesdeoca A, Romero Jiménez M, González García M, Bueno Díez M, Brea Hernando A, Lahoz C, Mostaza Prieto J, Millán Núñez-Cortés J, Reinares García L, Blanco Echevarría A, Ariza Corbo MJ, Rioja Villodres J, Sánchez-Chaparro M, Jansen Chaparro S, Sáenz Aranzubía P, Martorell Mateu E, Almagro Múgica F, Muñiz Grijalvo O, Masana Martín L, Plana Gil N, Ibarretxe Gerediaga D, Rodríguez Borjabad C, Zabala López S, Hernández Mijares A, Ascaso Gimilio J, Pérez García L, Civeira Murillo F, Pérez-Calahorra S, Lamiquiz-Moneo I, Mateo Gallego R, Marco Benedí V, Ferrando Vela J. Molecular basis of the familial chylomicronemia syndrome in patients from the National Dyslipidemia Registry of the Spanish Atherosclerosis Society. J Clin Lipidol 2018; 12:1482-1492.e3. [DOI: 10.1016/j.jacl.2018.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/09/2018] [Accepted: 07/24/2018] [Indexed: 01/16/2023]
|
33
|
Hannon BA, Khan NA, Teran-Garcia M. Nutrigenetic Contributions to Dyslipidemia: A Focus on Physiologically Relevant Pathways of Lipid and Lipoprotein Metabolism. Nutrients 2018; 10:E1404. [PMID: 30279335 PMCID: PMC6213032 DOI: 10.3390/nu10101404] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/19/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) remains the number one cause of death worldwide, and dyslipidemia is a major predictor of CVD mortality. Elevated lipid concentrations are the result of multiple genetic and environmental factors. Over 150 genetic loci have been associated with blood lipid levels. However, not all variants are present in pathways relevant to the pathophysiology of dyslipidemia. The study of these physiologically relevant variants can provide mechanistic understanding of dyslipidemia and identify potential novel therapeutic targets. Additionally, dietary fatty acids have been evidenced to exert both positive and negative effects on lipid profiles. The metabolism of both dietary and endogenously synthesized lipids can be affected by individual genetic variation to produce elevated lipid concentrations. This review will explore the genetic, dietary, and nutrigenetic contributions to dyslipidemia.
Collapse
Affiliation(s)
- Bridget A Hannon
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
| | - Naiman A Khan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
| | - Margarita Teran-Garcia
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
- Department of Human Development and Family Studies, Cooperative Extension, University of Illinois at Urbana-Champaign, Carle Illinois College of Medicine, Urbana-Champaign, IL 61801, USA.
| |
Collapse
|
34
|
Wójcik C, Fazio S, McIntyre AD, Hegele RA. Co-occurrence of heterozygous CREB3L3 and APOA5 nonsense variants and polygenic risk in a patient with severe hypertriglyceridemia exacerbated by estrogen administration. J Clin Lipidol 2018; 12:1146-1150. [PMID: 29954705 DOI: 10.1016/j.jacl.2018.05.014] [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: 03/28/2018] [Revised: 05/11/2018] [Accepted: 05/22/2018] [Indexed: 02/01/2023]
Abstract
We describe a case of a 36-year-old woman with severe hypertriglyceridemia likely caused by double heterozygosity of a known pathogenic APOA5 nonsense variant (p.Q275X) and a novel CREB3L3 nonsense variant (p.C296X) on a background of very strong polygenic susceptibility. Her clinical course worsened with development of eruptive xanthomata after oral administration of 2 mg estradiol twice daily for 2 weeks as part of a medical protocol for intrauterine embryo transfer following in vitro fertilization. Her triglyceride levels decreased to baseline and xanthomata resolved without treatment after discontinuation of hormonal therapy, which also resulted in termination of pregnancy. Before undergoing a second embryo transfer using her natural cycle and no exogenous hormones, the patient started combination therapy with eicosapentaenoic acid ethyl ester and gemfibrozil, leading to an ∼80% decrease in triglyceride levels. She continued treatment throughout pregnancy, which progressed to term with the delivery of healthy twins.
Collapse
Affiliation(s)
- Cezary Wójcik
- Department of Family Medicine, Oregon Health & Science University, Portland, OR, USA.
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Adam D McIntyre
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
35
|
Hua S, Ma C, Zhang J, Li J, Wu W, Xu N, Luo G, Zhao J. Influence of APOA5 Locus on the Treatment Efficacy of Three Statins: Evidence From a Randomized Pilot Study in Chinese Subjects. Front Pharmacol 2018; 9:352. [PMID: 29695967 PMCID: PMC5904201 DOI: 10.3389/fphar.2018.00352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/27/2018] [Indexed: 12/22/2022] Open
Abstract
Pharmacogenetics or pharmacogenomics approaches are important for addressing the individual variabilities of drug efficacy especially in the era of precision medicine. One particular interesting gene to investigate is APOA5, which has been repeatedly linked with the inter-individual variations of serum triglycerides. Here, we explored APOA5-statin interactions in 195 Chinese subjects randomized to rosuvastatin (5–10 mg/day), atorvastatin (10–20 mg/day), or simvastatin (40 mg/day) for 12 weeks by performing a targeted genotyping analysis of the APOA5 promoter SNP rs662799 (-1131T > C). There were no significant differences between the treatment arms for any of the statin-induced changes in clinical biomarkers. Reductions in LDL cholesterol were influenced by the APOA5 genotype in all three treatment groups. By contrast, changes in HDL cholesterol, and triglycerides were only affected by the APOA5 genotype in the atorvastatin and simvastatin groups and not in the rosuvastatin group. Our results suggest that future studies may need to consider stratifying subjects not only by genetic background but also by prescribed statin type.
Collapse
Affiliation(s)
- Sha Hua
- Department of Cardiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chuanxiang Ma
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jun Zhang
- Comprehensive Laboratory, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jing Li
- Department of Cardiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiwei Wu
- Department of Cardiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ning Xu
- Section of Clinical Chemistry and Pharmacology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Guanghua Luo
- Comprehensive Laboratory, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jianrong Zhao
- Department of Cardiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Fenwick PH, Jeejeebhoy K, Dhaliwal R, Royall D, Brauer P, Tremblay A, Klein D, Mutch DM. Lifestyle genomics and the metabolic syndrome: A review of genetic variants that influence response to diet and exercise interventions. Crit Rev Food Sci Nutr 2018; 59:2028-2039. [PMID: 29400991 DOI: 10.1080/10408398.2018.1437022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Metabolic syndrome (MetS) comprises a cluster of risk factors that includes central obesity, dyslipidemia, impaired glucose homeostasis and hypertension. Individuals with MetS have elevated risk of type 2 diabetes and cardiovascular disease; thus placing significant burdens on social and healthcare systems. Lifestyle interventions (comprised of diet, exercise or a combination of both) are routinely recommended as the first line of treatment for MetS. Only a proportion of people respond, and it has been assumed that psychological and social aspects primarily account for these differences. However, the etiology of MetS is multifactorial and stems, in part, on a person's genetic make-up. Numerous single nucleotide polymorphisms (SNPs) are associated with the various components of MetS, and several of these SNPs have been shown to modify a person's response to lifestyle interventions. Consequently, genetic variants can influence the extent to which a person responds to changes in diet and/or exercise. The goal of this review is to highlight SNPs reported to influence the magnitude of change in body weight, dyslipidemia, glucose homeostasis and blood pressure during lifestyle interventions aimed at improving MetS components. Knowledge regarding these genetic variants and their ability to modulate a person's response will provide additional context for improving the effectiveness of personalized lifestyle interventions that aim to reduce the risks associated with MetS.
Collapse
Affiliation(s)
- Peri H Fenwick
- a Department of Human Health and Nutritional Sciences , University of Guelph , Guelph , Ontario , Canada
| | - Khursheed Jeejeebhoy
- b Emeritus Professor of Medicine and Physician , St. Michael's Hospital , Toronto , Ontario , Canada
| | | | - Dawna Royall
- d Department of Family Relations and Applied Nutrition , University of Guelph , Guelph , Ontario , Canada
| | - Paula Brauer
- d Department of Family Relations and Applied Nutrition , University of Guelph , Guelph , Ontario , Canada
| | - Angelo Tremblay
- e Department of Kinesiology , Faculty of Medicine, Université Laval , Québec City , Québec , Canada
| | - Doug Klein
- f Department of Family Medicine , University of Alberta , Edmonton , Alberta , Canada
| | - David M Mutch
- a Department of Human Health and Nutritional Sciences , University of Guelph , Guelph , Ontario , Canada
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Postprandial lipemia (PPL), the prolonged increase in plasma triglyceride-rich lipoproteins following food consumption, is an independent risk factor for cardiovascular disease. Genetic variation, environment and the interplay between these direct an individual's postprandial lipid response. From such interplay, inducible and reversible epigenetic changes arise. Increasing evidence suggests epigenetic variation contributes to postprandial response in lipids and risk. RECENT FINDINGS Diet and exercise are central agents affecting postprandial lipemia - triglyceride, but heterogeneity of the findings warrant more and larger studies. Several epigenetic loci identified from a human intervention study account for a substantial proportion of PPL phenotype variation, but the burden to conduct an intervention study of postprandial responses likely limits translation to personalized nutrition. SUMMARY The impact of both DNA methylation patterns and environmental factors such as diet, exercise, sleep and medication on PPL is multifaceted. Discovery of interactions that modify the association between CpG (oligodeoxydinucleotide) methylation and postprandial phenotypes is unfolding.
Collapse
Affiliation(s)
| | - Jose M Ordovas
- Jean Mayer-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
- IMDEA Food Institute, CEI UAM + CSIC
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | |
Collapse
|