Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Atherosclerosis ; 393: 117558, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38703417

ABSTRACT

BACKGROUND: Carriers of the E40K loss-of-function variant in Angiopoietin-like 4 (ANGPTL4), have lower plasma triglyceride levels as well as lower rates of coronary artery disease (CAD) and type 2 diabetes (T2D). These genetic data suggest ANGPTL4 inhibition as a potential therapeutic target for cardiometabolic diseases. However, it is unknown whether the association between E40K and human diseases is due to linkage disequilibrium confounding. The broader impact of genetic ANGPTL4 inhibition is also unknown, raising uncertainties about the safety and validity of this target. METHODS: To assess the impact of ANGPLT4 inhibition, we evaluated whether E40K and other loss-of-function variants in ANGPTL4 influenced a wide range of health markers and diseases using 29 publicly available genome-wide association meta-analyses of cardiometabolic traits and diseases, as well as 1589 diseases assessed in electronic health records within FinnGen (n = 309,154). To determine whether these relationships were likely causal, and not driven by other correlated variants, we used the Bayesian fine mapping algorithm CoPheScan. RESULTS: The CoPheScan posterior probability of E40K being the causal variant for triglyceride levels was 99.99 %, validating the E40K to proxy lifelong lower activity of ANGPTL4. The E40K variant was associated with lower risk of CAD (odds ratio [OR] = 0.84, 95 % CI = 0.81 to 0.87, p=3.6e-21) and T2D (OR = 0.91, 95 % CI = 0.87 to 0.95, p=2.8e-05) in GWAS meta-analyses, with results replicated in FinnGen. These significant results were also replicated using other rare loss-of-function variants identified through whole exome sequencing in 488,278 participants of the UK Biobank. Using a Mendelian randomization study design, the E40K variant effect on cardiometabolic diseases was concordant with lipoprotein lipase enhancement (r = 0.82), but not hepatic lipase enhancement (r = -0.10), suggesting that ANGPTL4 effects on cardiometabolic diseases are potentially mainly mediated through lipoprotein lipase. After correction for multiple testing, the E40K variant did not significantly increase the risk of any of the 1589 diseases tested in FinnGen. CONCLUSIONS: ANGPTL4 inhibition may represent a potentially safe and effective target for cardiometabolic diseases prevention or treatment.

2.
Atherosclerosis ; : 117546, 2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38692978

ABSTRACT

The worldwide prevalence of individuals with an elevated body weight has increased steadily over the past five decades. Billions of research dollars have been invested to improve our understanding of the causes and consequences of having an elevated body weight. All this knowledge has, however, failed to influence populational body weight trajectories of most countries around the world. Research on the definition of "obesity" has also evolved. Body mass index (BMI), the most commonly used tool to make its diagnosis, has major limitations. In this review article, we will highlight evidence from observational studies, genetic association studies and randomized clinical trials that have shown the remarkable inter-individual differences in the way humans store energy as body fat. Increasing evidence also suggests that, as opposed to weight inclusive, lifestyle-based approaches, weight-centric approaches advising people to simply eat less and move more are not sustainable for most people for long-term weight loss and maintenance. It is time to recognize that this outdated approach may have produced more harm than good. On the basis of pathophysiological, genetic and clinical evidence presented in this review, we propose that it may be time to shift away from the traditional clinical approach, which is BMI-centric. Rather, emphasis should be placed on actionable lifestyle-related risk factors aiming at improving overall diet quality and increasing physical activity level in the general population.

3.
Atherosclerosis ; 391: 117501, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38547584

ABSTRACT

BACKGROUND AND AIMS: Inhibitors of apolipoprotein C-III (apoC3) are currently approved for the reduction of triglyceride levels in patients with Familial Chylomicronemia Syndrome. We used drug target Mendelian randomization (MR) to assess the effect of genetically predicted decrease in apoC3 blood protein levels on cardiometabolic traits and diseases. METHODS: We quantified lifelong reductions in apoC3 blood levels by selecting all genome wide significant and independent (r2<0.1) single nucleotide polymorphisms (SNPs) in the APOC3 gene region ±1 Mb, from three genome-wide association studies (GWAS) of apoC3 blood protein levels (deCODE, n = 35,378, Fenland, n = 10,708 and ARIC, n = 7213). We included the largest GWASes on 18 cardiometabolic traits and 9 cardiometabolic diseases as study outcomes. RESULTS: A one standard deviation lowering in apoC3 blood protein levels was associated with lower triglycerides, apolipoprotein B, low-density lipoprotein cholesterol, alanine aminotransferase, and glomerular filtration rate as well as higher high-density lipoprotein cholesterol levels. ApoC3 lowering was also associated with lower risk of acute pancreatitis (odds ratio [OR] = 0.91 95% CI = 0.82 to 1.00), aortic stenosis (OR = 0.82 95% CI = 0.73 to 0.93), and coronary artery disease (OR = 0.86 95% CI = 0.80 to 0.93), and was associated with increased parental lifespan (0.06 95% CI = 0.03-0.09 years). These results were concordant across robust MR methods, the three protein datasets and upon adjustment for APOA1, APOA4 and APOA5 using a multivariable MR framework. CONCLUSIONS: These results provide evidence that apoC3 lowering could result in widespread benefits for cardiometabolic health and encourage the launch of trials on apoC3 inhibition for coronary artery disease prevention.


Subject(s)
Apolipoprotein C-III , Cardiovascular Diseases , Humans , Acute Disease , Apolipoprotein C-III/genetics , Blood Proteins , Cardiovascular Diseases/genetics , Cardiovascular Diseases/prevention & control , Cholesterol, LDL , Coronary Artery Disease/genetics , Genome-Wide Association Study , Mendelian Randomization Analysis , Pancreatitis , Polymorphism, Single Nucleotide , Triglycerides
5.
Nat Commun ; 15(1): 2407, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38494474

ABSTRACT

There is currently no medical therapy to prevent calcific aortic valve stenosis (CAVS). Multi-omics approaches could lead to the identification of novel molecular targets. Here, we perform a genome-wide association study (GWAS) meta-analysis including 14,819 cases among 941,863 participants of European ancestry. We report 32 genomic loci, among which 20 are novel. RNA sequencing of 500 human aortic valves highlights an enrichment in expression regulation at these loci and prioritizes candidate causal genes. Homozygous genotype for a risk variant near TWIST1, a gene involved in endothelial-mesenchymal transition, has a profound impact on aortic valve transcriptomics. We identify five genes outside of GWAS loci by combining a transcriptome-wide association study, colocalization, and Mendelian randomization analyses. Using cross-phenotype and phenome-wide approaches, we highlight the role of circulating lipoproteins, blood pressure and inflammation in the disease process. Our findings pave the way for the development of novel therapies for CAVS.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Aortic Valve/pathology , Calcinosis , Humans , Aortic Valve/metabolism , Genome-Wide Association Study , Aortic Valve Stenosis/genetics , Genomics
6.
Eur Heart J ; 45(9): 707-721, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38243829

ABSTRACT

BACKGROUND AND AIMS: RNA-based, antibody-based, and genome editing-based therapies are currently under investigation to determine if the inhibition of angiopoietin-like protein-3 (ANGPTL3) could reduce lipoprotein-lipid levels and atherosclerotic cardiovascular disease (ASCVD) risk. Mendelian randomisation (MR) was used to determine whether genetic variations influencing ANGPTL3 liver gene expression, blood levels, and protein structure could causally influence triglyceride and apolipoprotein B (apoB) levels as well as coronary artery disease (CAD), ischaemic stroke (IS), and other cardiometabolic diseases. METHODS: RNA sequencing of 246 explanted liver samples and genome-wide genotyping was performed to identify single-nucleotide polymorphisms (SNPs) associated with liver expression of ANGPTL3. Genome-wide summary statistics of plasma protein levels of ANGPTL3 from the deCODE study (n = 35 359) were used. A total of 647 carriers of ANGPTL3 protein-truncating variants (PTVs) associated with lower plasma triglyceride levels were identified in the UK Biobank. Two-sample MR using SNPs that influence ANGPTL3 liver expression or ANGPTL3 plasma protein levels as exposure and cardiometabolic diseases as outcomes was performed (CAD, IS, heart failure, non-alcoholic fatty liver disease, acute pancreatitis, and type 2 diabetes). The impact of rare PTVs influencing plasma triglyceride levels on apoB levels and CAD was also investigated in the UK Biobank. RESULTS: In two-sample MR studies, common genetic variants influencing ANGPTL3 hepatic or blood expression levels of ANGPTL3 had a very strong effect on plasma triglyceride levels, a more modest effect on low-density lipoprotein cholesterol, a weaker effect on apoB levels, and no effect on CAD or other cardiometabolic diseases. In the UK Biobank, the carriers of rare ANGPTL3 PTVs providing lifelong reductions in median plasma triglyceride levels [-0.37 (interquartile range 0.41) mmol/L] had slightly lower apoB levels (-0.06 ± 0.32 g/L) and similar CAD event rates compared with non-carriers (10.2% vs. 10.9% in carriers vs. non-carriers, P = .60). CONCLUSIONS: PTVs influencing ANGPTL3 protein structure as well as common genetic variants influencing ANGPTL3 hepatic expression and/or blood protein levels exhibit a strong effect on circulating plasma triglyceride levels, a weak effect on circulating apoB levels, and no effect on ASCVD. Near-complete inhibition of ANGPTL3 function in patients with very elevated apoB levels may be required to reduce ASCVD risk.


Subject(s)
Atherosclerosis , Brain Ischemia , Coronary Artery Disease , Diabetes Mellitus, Type 2 , Pancreatitis , Stroke , Humans , Acute Disease , Coronary Artery Disease/genetics , Angiopoietin-Like Protein 3 , Antibodies , Apolipoproteins B/genetics , Triglycerides
7.
Neurology ; 102(4): e209128, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38261980

ABSTRACT

The Mendelian randomization (MR) paradigm allows for causal inferences to be drawn using genetic data. In recent years, the expansion of well-powered publicly available genetic association data related to phenotypes such as brain tissue gene expression, brain imaging, and neurologic diseases offers exciting opportunities for the application of MR in the field of neurology. In this review, we discuss the basic principles of MR, its myriad applications to research in neurology, and potential pitfalls of injudicious applications. Throughout, we provide examples where MR-informed findings have shed light on long-standing epidemiologic controversies, provided insights into the pathophysiology of neurologic conditions, prioritized drug targets, and informed drug repurposing opportunities. With the ever-expanding availability of genome-wide association data, we project MR to become a key driver of progress in the field of neurology. It is therefore paramount that academics and clinicians within the field are familiar with the approach.


Subject(s)
Genome-Wide Association Study , Neurology , Humans , Mendelian Randomization Analysis , Brain , Drug Repositioning
8.
Atherosclerosis ; 386: 117371, 2023 12.
Article in English | MEDLINE | ID: mdl-38029505

ABSTRACT

BACKGROUND AND AIMS: Individuals with a higher abdominal adipose tissue accumulation are at higher risk of developing cardiometabolic diseases. For a given body mass index (BMI), women typically present lower abdominal adipose tissue accumulation compared to men. Whether abdominal adiposity is a causal driver of cardiometabolic risk, or a mere marker of ectopic fat deposition is debated. METHODS: We investigated the sex-specific and sex-combined impact of height and BMI-adjusted gluteofemoral adipose tissue (GFATadj) adjusted abdominal subcutaneous adipose tissue (ASATadj) and adjusted visceral adipose tissue (VATadj) on cardiometabolic traits and diseases using Mendelian randomization. RESULTS: Leveraging genome-wide summary statistics on GFATadj, ASATadj and VATadj from 39,076 UK Biobank participants with full-body magnetic resonance imaging available, we found that GFATadj is associated with a more favourable cardiometabolic risk profile including lower low density lipoprotein (LDL) cholesterol, triglycerides, fasting glucose, fasting insulin, liver enzyme levels and blood pressure as well as higher high density lipoprotein (HDL) cholesterol levels. GFATadj also is negatively associated with ischemic stroke, coronary artery disease, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD). ASATadj is not associated with cardiometabolic traits and diseases, whereas VATadj is associated with liver fat accumulation but not with NAFLD or other cardiometabolic traits or diseases. Although the absolute effect sizes of GFATadj on LDL cholesterol were more pronounced in women compared to men, most associations did not differ by sex. CONCLUSIONS: The inability of subcutaneous fat depots to efficiently store energy substrates could be the causal factor underlying the association of visceral lipid deposition with cardiometabolic health.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Male , Humans , Female , Intra-Abdominal Fat/diagnostic imaging , Intra-Abdominal Fat/pathology , Diabetes Mellitus, Type 2/pathology , Mendelian Randomization Analysis , Body Mass Index , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cholesterol
9.
Circ Genom Precis Med ; 16(5): 470-477, 2023 10.
Article in English | MEDLINE | ID: mdl-37753708

ABSTRACT

BACKGROUND: Lp(a) (lipoprotein[a]) is a highly atherogenic lipoprotein subfraction that may contribute to polygenic risk of coronary artery disease (CAD), but the extent of this contribution is unknown. Our objective was to estimate the contribution of Lp(a) to polygenic risk of CAD and to evaluate the respective contributions of Lp(a) and a CAD polygenic risk score (PRS) to CAD. METHODS: A total of 372 385 UK Biobank participants of European ancestry free of CAD at baseline were included. Plasma Lp(a) levels were measured and a CAD-PRS was calculated using the LDpred2 algorithm. Over the median follow-up of 12.6 years, 13 538 participants had incident CAD (myocardial infarction, coronary artery bypass grafting, or coronary angioplasty). RESULTS: The LPA region contribution to the CAD-PRS-mediated CAD risk was modest (7.2% [95% CI, 6.1-8.3]). Lp(a) levels significantly increased the predictive performance of a CAD-PRS including age and sex in Cox regression (C statistic 0.751 versus 0.746, difference, 0.005 [95% CI, 0.004-0.006]). Compared with participants in the bottom CAD-PRS quintile with Lp(a) levels <25 nmol/L (CAD event rate, 1.4%), the hazard ratio for incident CAD in participants in the top CAD-PRS quintile with Lp(a) levels ≥125 nmol/L was 5.45 (95% CI, 4.93-6.03; P=9.35×10-242, CAD event rate 6.6%). CONCLUSIONS: Compared with individuals with a low genetic risk of CAD (low CAD-PRS and low Lp[a] levels), those with a high genetic risk (high CAD-PRS and high Lp[a] levels) had a 5-fold higher CAD risk. These results highlight a substantial contribution of genetic risk factors to CAD and that accurate estimation of genetic risk of CAD may need to consider blood levels of Lp(a).


Subject(s)
Coronary Artery Disease , Humans , Coronary Artery Disease/genetics , Prospective Studies , Lipoprotein(a)/genetics , Biological Specimen Banks , Risk Factors , United Kingdom/epidemiology
10.
iScience ; 26(7): 107127, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37456853

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent disease with no specific drug therapy. High-throughput metabolomics present an unprecedented opportunity to identify biomarkers and potentially causal risk factors for NAFLD. Here, we determined the impact of 21 circulating metabolites, 17 lipids, and 132 lipoprotein particle characteristics on NAFLD combining prospective observational and two-sample Mendelian randomization (MR) analyses in 121,032 UK Biobank participants. We identified several metabolic factors associated with NAFLD risk in observational and MR analyses including triglyceride-rich and high-density lipoprotein particles composition, as well as the ratio of polyunsaturated fatty acids to total fatty acids. This study, is one of the largest to investigate incident NAFLD, provides concordant observational and genetic evidence that therapies aimed at reducing circulating triglycerides and increasing large HDL particles, as well as interventions aimed at increasing polyunsaturated fatty acid content may warrant further investigation into NAFLD prevention and treatment.

11.
J Am Heart Assoc ; 12(13): e028502, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37345817

ABSTRACT

Background The impact of an elevated body mass index (BMI) on atherosclerotic cardiovascular disease (ASCVD) risk in individuals who are metabolically healthy is debated. We investigated the respective contributions of BMI as well as lifestyle and cardiometabolic risk factors combined to ASCVD incidence in 319 866 UK Biobank participants. Methods and Results We developed a cardiovascular health score (CVHS) based on 4 lifestyle and 6 cardiometabolic parameters. The impact of the CVHS on incident ASCVD (15 699 events) alone and in BMI and waist-to-hip ratio categories was assessed using Cox proportional hazards in women and men separately. In participants with a high CVHS (8-10), those with a BMI ≥35.0 kg/m2 had a nonsignificantly higher ASCVD risk (hazard ratio [HR], 1.20 [95% CI, 0.84-1.70]; P=0.32) compared with those with a BMI of 18.5 to 24.9 kg/m2. In participants with a BMI of 18.5 to 24.9 kg/m2, those with a lower CVHS (0-2) had a higher ASCVD risk (HR, 4.06 [95% CI, 3.23-5.10]; P<0.001) compared with those with a higher CVHS (8-10). When we used the waist-to-hip ratio instead of the BMI, a dose-response relationship between the waist-to-hip ratio and ASCVD risk was obtained in healthier participants. Results were similar in women compared with men. Conclusions In women and men in the UK Biobank, the relationship between the BMI and ASCVD incidence in healthy individuals was inconsistent, whereas cardiovascular risk factors strongly predicted ASCVD incidence in all BMI categories. Assessing lifestyle and cardiometabolic risk factors as well as body fat distribution indices may help identify individuals at high ASCVD risk, regardless of body weight.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Male , Humans , Female , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Incidence , Body Mass Index , Atherosclerosis/diagnosis , Atherosclerosis/epidemiology , Body Weight , Risk Factors
12.
Atherosclerosis ; 374: 107-120, 2023 06.
Article in English | MEDLINE | ID: mdl-37188555

ABSTRACT

In 2022, the European Atherosclerosis Society (EAS) published a new consensus statement on lipoprotein(a) [Lp(a)], summarizing current knowledge about its causal association with atherosclerotic cardiovascular disease (ASCVD) and aortic stenosis. One of the novelties of this statement is a new risk calculator showing how Lp(a) influences lifetime risk for ASCVD and that global risk may be underestimated substantially in individuals with high or very high Lp(a) concentration. The statement also provides practical advice on how knowledge about Lp(a) concentration can be used to modulate risk factor management, given that specific and highly effective mRNA-targeted Lp(a)-lowering therapies are still in clinical development. This advice counters the attitude: "Why should I measure Lp(a) if I can't lower it?". Subsequent to publication, questions have arisen relating to how the recommendations of this statement impact everyday clinical practice and ASCVD management. This review addresses 30 of the most frequently asked questions about Lp(a) epidemiology, its contribution to cardiovascular risk, Lp(a) measurement, risk factor management and existing therapeutic options.


Subject(s)
Aortic Valve Stenosis , Atherosclerosis , Cardiovascular Diseases , Humans , Lipoprotein(a) , Risk Factors , Risk Assessment , Aortic Valve Stenosis/complications , Atherosclerosis/diagnosis , Atherosclerosis/epidemiology , Atherosclerosis/prevention & control , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control
14.
iScience ; 26(4): 106376, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37013190

ABSTRACT

Genome-wide association studies (GWAS) have identified hundreds of genetic variants associated with body weight but the biological relevance of most remains unexplored. Given the critical role of the brain in body weight regulation, we set out to determine whether genetic variants linked with body mass index (BMI) could be mapped to brain proteins. Using genetic colocalization, we mapped 25 loci from the largest BMI GWAS (n = 806,834) to brain protein concentrations obtained from publicly available datasets. We also performed a proteome-wide Mendelian randomization on 696 brain proteins followed by genetic colocalization and identified 35 additional brain proteins. Only a minority of these proteins (<30%) had a colocalization signal with cortex gene expression levels, highlighting the value of moving beyond gene expression levels and examining brain protein levels. In conclusion, we identified 60 unique proteins expressed in the brain that may be critical regulators of body weight in humans.

15.
Eur Heart J Open ; 3(2): oead032, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37077580

ABSTRACT

Aims: Elevated lipoprotein(a) [Lp(a)] levels are associated with the risk of coronary artery disease (CAD) and calcific aortic valve stenosis (CAVS). Observational studies revealed that Lp(a) and C-reactive protein (CRP) levels, a biomarker of systemic inflammation, may jointly predict CAD risk. Whether Lp(a) and CRP levels also jointly predict CAVS incidence and progression is unknown. Methods and results: We investigated the association of Lp(a) with CAVS according to CRP levels in the European Prospective Investigation into Cancer and Nutrition (EPIC)-Norfolk study (n = 18 226, 406 incident cases) and the UK Biobank (n = 438 260, 4582 incident cases), as well as in the ASTRONOMER study (n = 220), which assessed the haemodynamic progression rate of pre-existing mild-to-moderate aortic stenosis. In EPIC-Norfolk, in comparison to individuals with low Lp(a) levels (<50 mg/dL) and low CRP levels (<2.0 mg/L), those with elevated Lp(a) (>50 mg/dL) and low CRP levels (<2.0 mg/L) and those with elevated Lp(a) (>50 mg/dL) and elevated CRP levels (>2.0 mg/L) had a higher CAVS risk [hazard ratio (HR) = 1.86 (95% confidence intervals, 1.30-2.67) and 2.08 (1.44-2.99), respectively]. A comparable predictive value of Lp(a) in patients with vs. without elevated CRP levels was also noted in the UK Biobank. In ASTRONOMER, CAVS progression was comparable in patients with elevated Lp(a) levels with or without elevated CRP levels. Conclusion: Lp(a) predicts the incidence and possibly progression of CAVS regardless of plasma CRP levels. Lowering Lp(a) levels may warrant further investigation in the prevention and treatment of CAVS, regardless of systemic inflammation.

16.
Gastroenterology ; 164(6): 953-965.e3, 2023 05.
Article in English | MEDLINE | ID: mdl-36736436

ABSTRACT

BACKGROUND & AIMS: Acute pancreatitis (AP) is a complex disease and the leading cause of gastrointestinal disease-related hospital admissions. Few therapeutic options exist for AP prevention. Blood proteins with causal evidence may represent promising drug targets, but few have been causally linked with AP. Our objective was to identify blood proteins linked with AP by combining genome-wide association meta-analysis and proteome-wide Mendelian randomization (MR) studies. METHODS: We performed a genome-wide association meta-analysis totalling 10,630 patients with AP and 844,679 controls and a series of inverse-variance weighted MR analyses using cis-acting variants on 4719 blood proteins from the deCODE study (N = 35,559) and 4979 blood proteins from the Fenland study (N = 10,708). RESULTS: The meta-analysis identified genome-wide significant variants (P <5 × 10-8) at 5 loci (ABCG5/8, TWIST2, SPINK1, PRSS2 and MORC4). The proteome-wide MR analyses identified 68 unique blood proteins that may causally be associated with AP, including 29 proteins validated in both data sets. Functional annotation of these proteins confirmed expression of many proteins in metabolic tissues responsible for digestion and energy metabolism, such as the esophagus, adipose tissue, and liver as well as acinar cells of the pancreas. Genetic colocalization and investigations into the druggable genome also identified potential drug targets for AP. CONCLUSIONS: This large genome-wide association study meta-analysis for AP identified new variants linked with AP as well as several blood proteins that may be causally associated with AP. This study provides new information on the genetic architecture of this disease and identified pathways related to AP, which may be further explored as possible therapeutic targets for AP.


Subject(s)
Pancreatitis , Proteome , Humans , Proteome/genetics , Mendelian Randomization Analysis , Genome-Wide Association Study , Acute Disease , Pancreatitis/genetics , Blood Proteins , Polymorphism, Single Nucleotide , Trypsin/genetics , Trypsinogen/genetics , Trypsin Inhibitor, Kazal Pancreatic/genetics , Nuclear Proteins/genetics
17.
J Transl Med ; 21(1): 60, 2023 01 30.
Article in English | MEDLINE | ID: mdl-36717893

ABSTRACT

Features of the gut microbiota have been associated with several chronic diseases and longevity in preclinical models as well as in observational studies. Whether these relations underlie causal effects in humans remains to be established. We aimed to determine whether the gut microbiota influences cardiometabolic traits as well as the risk of chronic diseases and human longevity using a comprehensive 2-Sample Mendelian randomization approach. We included as exposures 10 gut-associated metabolites and pathways and 57 microbial taxa abundance. We included as outcomes nine cardiometabolic traits (fasting glucose, fasting insulin, systolic blood pressure, diastolic blood pressure, HDL cholesterol, LDL cholesterol, triglycerides, estimated glomerular filtration rate, body mass index [BMI]), eight chronic diseases previously linked with the gut microbiota in observational studies (Alzheimer's disease, depression, type 2 diabetes, non-alcoholic fatty liver disease, coronary artery disease (CAD), stroke, osteoporosis and chronic kidney disease), as well as parental lifespan and longevity. We found 7 associations with evidence of causality before and after sensitivity analyses, but not after multiple testing correction (1198 tests). Most effect sizes (4/7) were small. The two largest exposure-outcome effects were markedly attenuated towards the null upon inclusion of BMI or alcohol intake frequency in multivariable MR analyses. While finding robust genetic instruments for microbiota features is challenging hence potentially inflating type 2 errors, these results do not support a large causal impact of human gut microbita features on cardiometabolic traits, chronic diseases or longevity. These results also suggest that the previously documented associations between gut microbiota and human health outcomes may not always underly causal relations.


Subject(s)
Coronary Artery Disease , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Humans , Longevity/genetics , Gastrointestinal Microbiome/genetics , Mendelian Randomization Analysis , Coronary Artery Disease/genetics , Body Mass Index , Chronic Disease , Polymorphism, Single Nucleotide , Genome-Wide Association Study
18.
J Clin Endocrinol Metab ; 108(6): 1308-1317, 2023 05 17.
Article in English | MEDLINE | ID: mdl-36585897

ABSTRACT

CONTEXT: Hyperinsulinemia and adiposity are associated with one another, but the directionality of this relation is debated. OBJECTIVE: Here, we tested the direction of the causal effects of fasting insulin (FI) levels and body fat accumulation/distribution using 2-sample bidirectional Mendelian randomization (MR). METHODS: We included summary statistics from large-scale genome-wide association studies for body mass index (BMI, n = 806 834), waist to hip ratio adjusted for BMI (WHRadjBMI, n = 694 649), abdominal subcutaneous, visceral and gluteofemoral adipose tissue (n = 38 965), FI levels (n = 98 210), pancreatic islets gene expression (n = 420), and hypothalamus gene expression (n = 155). We used inverse variance-weighted and robust MR methods that relied on statistically and biologically driven genetic instruments. RESULTS: Both BMI and WHRadjBMI were positively associated with FI. Results were consistent across all robust MR methods and when variants mapped to the hypothalamus (presumably associated with food behavior) were included. In multivariable MR analyses, when waist circumference and BMI were mutually adjusted, the direct effect of waist circumference on FI was 2.43 times larger than the effect of BMI on FI. FI was not associated with adiposity. By contrast, using genetic instruments mapped to gene expression in pancreatic islets (presumably more specific to insulin secretion), insulin was positively associated with BMI and abdominal subcutaneous and gluteofemoral adipose tissue, but not with visceral adipose tissue. CONCLUSION: Although these results will need to be supported by experimental investigations, results of this MR study suggest that abdominal adiposity may be a key determinant of circulating insulin levels. Alternatively, insulin secretion may promote peripheral adipose tissue accumulation.


Subject(s)
Genome-Wide Association Study , Mendelian Randomization Analysis , Humans , Insulin Secretion , Body Fat Distribution , Obesity/genetics , Obesity/complications , Insulin , Fasting , Body Mass Index , Polymorphism, Single Nucleotide
19.
Commun Med (Lond) ; 2: 130, 2022.
Article in English | MEDLINE | ID: mdl-36249462

ABSTRACT

Background: Observational studies have linked adiposity and especially abdominal adiposity to liver fat accumulation and non-alcoholic fatty liver disease. These traits are also associated with type 2 diabetes and coronary artery disease but the causal factor(s) underlying these associations remain unexplored. Methods: We used a multivariable Mendelian randomization study design to determine whether body mass index and waist circumference were causally associated with non-alcoholic fatty liver disease using publicly available genome-wide association study summary statistics of the UK Biobank (n = 461,460) and of non-alcoholic fatty liver disease (8434 cases and 770,180 control). A multivariable Mendelian randomization study design was also used to determine the respective causal contributions of waist circumference and liver fat (n = 32,858) to type 2 diabetes and coronary artery disease. Results: Using multivariable Mendelian randomization we show that waist circumference increase non-alcoholic fatty liver disease risk even when accounting for body mass index (odd ratio per 1-standard deviation increase = 2.35 95% CI = 1.31-4.22, p = 4.2e-03), but body mass index does not increase non-alcoholic fatty liver disease risk when accounting for waist circumference (0.86 95% CI = 0.54-1.38, p = 5.4e-01). In multivariable Mendelian randomization analyses accounting for liver fat, waist circumference remains strongly associated with both type 2 diabetes (3.27 95% CI = 2.89-3.69, p = 3.8e-80) and coronary artery disease (1.66 95% CI = 1.54-1.8, p = 3.4e-37). Conclusions: These results identify waist circumference as a strong, independent, and causal contributor to non-alcoholic fatty liver disease, type 2 diabetes and coronary artery disease, thereby highlighting the importance of assessing body fat distribution for the prediction and prevention of cardiometabolic diseases.

20.
Am J Clin Nutr ; 116(6): 1748-1758, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36124644

ABSTRACT

BACKGROUND: Canada's Food Guide (CFG) was profoundly revised in 2019, but the extent to which adherence to recommendations on healthy food choices reduces the risk of cardiovascular disease (CVD) is unknown. OBJECTIVES: The aim of this study was to examine how greater adherence to the 2019 CFG's recommendations on healthy food choices influences the risk of incident CVD. METHODS: Participants were a sample of adults without history of CVD, diabetes, or cancer from the UK Biobank prospective cohort study. Usual dietary intakes were estimated by modeling data from repeated Web-based 24-h dietary recalls using the National Cancer Institute multivariate method. Adherence to key CFG recommendations on healthy food choices was assessed using the Healthy Eating Food Index (HEFI)-2019, which has a maximum of 80 points. The CVD outcome was a composite of fatal and nonfatal myocardial infarction and ischemic stroke. Cox regression models adjusted via inverse probability weighting were used to estimate CVD risks. Counterfactual models were used to interpret risks of hypothetical changes in the HEFI-2019 score. RESULTS: A total of 136,698 participants met the eligibility criteria (55% females; mean age: 57.2 y; range: 40-75 y). During the 11-y follow-up, there were 2843 cases of incident CVD. Compared with no change in the HEFI-2019 score, increasing the HEFI-2019 score of all participants to the 90th percentile of the score distribution (58.1 points) hypothetically reduced the risk of CVD by 24% (RR: 0.76; 95% CI: 0.58, 0.94; absolute risk difference: -0.58%). CONCLUSIONS: These results suggest that greater adherence to the 2019 CFG recommendations on healthy food choices reduces the 11-y risk of CVD in middle-aged and older adults.


Subject(s)
Cardiovascular Diseases , Middle Aged , Female , Humans , Aged , Male , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Prospective Studies , Biological Specimen Banks , Canada/epidemiology , United Kingdom/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...