Next Article in Journal
RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review
Next Article in Special Issue
Downregulation of Circulating Hsa-miR-200c-3p Correlates with Dyslipidemia in Patients with Stable Coronary Artery Disease
Previous Article in Journal
Enhanced Bactericidal Effect of Calcinated Mg–Fe Layered Double Hydroxide Films Driven by the Fenton Reaction
Previous Article in Special Issue
De Novo Asp219Val Mutation in Cardiac Tropomyosin Associated with Hypertrophic Cardiomyopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Circulating Amino Acids and Risk of Peripheral Artery Disease in the PREDIMED Trial

by
Cristina Razquin
1,2,
Miguel Ruiz-Canela
1,2,
Estefania Toledo
1,2,
Clary B. Clish
3,
Marta Guasch-Ferré
4,5,6,
Jesús F. García-Gavilán
2,7,8,
Clemens Wittenbecher
4,9,
Angel Alonso-Gómez
2,10,
Montse Fitó
2,11,
Liming Liang
12,
Dolores Corella
2,13,
Enrique Gómez-Gracia
14,
Ramon Estruch
2,15,
Miquel Fiol
2,16,
Jose M. Santos-Lozano
2,17,
Luis Serra-Majem
2,18,
Emilio Ros
2,19,
Fernando Aros
2,10,
Jordi Salas-Salvadó
2,7,8,
Frank B. Hu
4,20 and
Miguel A. Martínez-González
1,2,4,*
add Show full author list remove Hide full author list
1
Department of Preventive Medicine and Public Health, IdiSNA (Instituto de Investigación Sanitaria de Navarra), University of Navarra, 31008 Pamplona, Spain
2
CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
3
Broad Institute and MIT, Harvard University, Cambridge, MA 02142, USA
4
Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
5
Department of Public Health, Section of Epidemiology, University of Copenhagen, 1165 Copenhagen, Denmark
6
Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 1353 Copenhagen, Denmark
7
Unitat de Nutrició Humana, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43201 Reus, Spain
8
Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari San Joan de Reus, 43204 Reus, Spain
9
SciLifeLab, Food and Nutrition Science Division, Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
10
Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
11
Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain
12
Department of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
13
Department of Preventive Medicine, University of Valencia, 46100 Valencia, Spain
14
Department of Preventive Medicine, University of Málaga, 29071 Málaga, Spain
15
Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
16
Plataforma de Ensayos Clínicos, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
17
Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013 Sevilla, Spain
18
Nutrition Research Group, Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
19
Lipid Clinic, Department of Endocrinology and Nutrition, Agust Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
20
Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 270; https://doi.org/10.3390/ijms24010270
Submission received: 7 November 2022 / Revised: 16 December 2022 / Accepted: 19 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Molecular Research in Cardiovascular and Cerebrovascular Diseases)

Abstract

:
Effective prevention and risk prediction are important for peripheral artery disease (PAD) due to its poor prognosis and the huge disease burden it produces. Circulating amino acids (AA) and their metabolites may serve as biomarkers of PAD risk, but they have been scarcely investigated. The objective was to prospectively analyze the associations of baseline levels of plasma AA (and their pathways) with subsequent risk of PAD and the potential effect modification by a nutritional intervention with the Mediterranean diet (MedDiet). A matched case-control study was nested in the PREDIMED trial, in which participants were randomized to three arms: MedDiet with tree nut supplementation group, MedDiet with extra-virgin olive oil (EVOO) supplementation group or control group (low-fat diet). One hundred and sixty-seven PAD cases were matched with 250 controls. Plasma AA was measured with liquid chromatography/mass spectrometry at the Broad Institute. Baseline tryptophan, serine and threonine were inversely associated with PAD (ORfor 1 SD increase = 0.78 (0.61–0.99); 0.67 (0.51–0.86) and 0.75 (0.59–0.95), respectively) in a multivariable-adjusted conditional logistic regression model. The kynurenine/tryptophan ratio was directly associated with PAD (ORfor 1 SD increase = 1.50 (1.14–1.98)). The nutritional intervention with the MedDiet+nuts modified the association between threonine and PAD (p-value interaction = 0.018) compared with the control group. However, subjects allocated to the MedDiet+EVOO group were protected against PAD independently of baseline threonine. Plasma tryptophan, kynurenine/tryptophan ratio, serine and threonine might serve as early biomarkers of future PAD in subjects at a high risk of cardiovascular disease. The MedDiet supplemented with EVOO exerted a protective effect, regardless of baseline levels of threonine.

1. Introduction

Peripheral artery disease (PAD) is defined as the occlusion of the arteries that supply the lower extremities, and it is usually caused by atherosclerosis and associated thrombosis [1]. PAD is one of the main causes of atherosclerotic cardiovascular morbidity. The global burden of PAD has increased over the last decades [2,3].
Patients with PAD have a considerably increased risk of major cardiovascular and cerebrovascular events, together with functional impairment. In fact, PAD has been associated with heart failure (HF), atrial fibrillation (AF), myocardial infarction and stroke [4,5,6,7]. Consequently, patients with PAD show a poorer quality of life and elevated mortality rates [2,4,6,8,9,10].
Although symptoms of PAD may appear, such as intermittent claudication or atypical leg symptoms [11], PAD is still commonly underdiagnosed in primary care, with a consequent increase in the aforementioned risks of other hard cardiovascular events and premature death [7]. Its huge disease burden and the substantial proportion of underdiagnosed PAD cases highlight the need to improve early predictions of PAD beyond the use of traditional risk factors [12]. The discovery of new biomarkers of PAD may contribute to designing more effective preventive strategies.
Amino acids (AA) are the building blocks for the formation of proteins. Among the 20 amino acids contributing to protein formation, nine cannot be synthesized either at all or in sufficient amounts by the human body, so they can only be acquired by dietary intake (i.e., they are essential AA), which include: valine, leucine, isoleucine, threonine, lysine, histidine, phenylalanine, tryptophan and methionine. Other AAs can be endogenously synthesized [13]. Circulating branched-chain AA [14], glutamine and glutamate [15,16], and AA in the tryptophan-kynurenine pathway [17] have been previously associated with the risk of atherosclerotic cardiovascular events. Consequently, it is likely that circulating levels of AA may be related to the risk of PAD. Studying the association between plasma AA and future PAD may help identify the potential role of these metabolites in the pathogenesis of PAD. Some of these AAs may contribute to the design of combined biomarkers for the early prediction of PAD.
Using a case-control study nested within the PREDIMED trial, the aim of this study was to assess the prospective association of circulating AA or their metabolites with the risk of developing PAD. A secondary aim was to examine whether these associations might be modified by a dietary intervention based on the Mediterranean diet.

2. Results

Table 1 shows the baseline characteristics of the participants according to PAD incidence. PAD cases were more likely to have type 2 diabetes (T2D) and belong to the control group of the trial.
The first step in the present case-control study was to analyze the association between baseline AA and PAD incidence in conditional logistic models. In the crude models, we observed that tryptophan (ORfor 1 SD increase = 0.80 (0.64–0.99)), serine (ORfor 1 SD increase = 0.72 (0.57–0.90)) and threonine (ORfor 1 SD increase = 0.74 (0.59–0.91)) were inversely associated with PAD risk.
When the models were further adjusted for the intervention group, smoking status, BMI, leisure-time physical activity, prevalent hypertension, prevalent T2D, prevalent dyslipidemia and educational level (Table 2), tryptophan maintained its inverse association with PAD (ORfor 1 SD increase = 0.78 (0.61–0.99)) although no consistent association across increasing quartiles was observed (p-Value for Linear trend = 0.216). Moreover, both serine and threonine were inversely associated with PAD (ORfor 1 SD increase = 0.67 (0.51–0.86) and 0.75 (0.59–0.95), respectively). These inverse associations were also observed across increasing quartiles of serine (p-Value for Linear trend = 0.015) and threonine (p-Value for Linear trend = 0.026). Finally, an inverse association between baseline glutamate (quartiles) and PAD risk (p-Value for Linear trend = 0.029) was observed, but when tested as a continuous variable (per each SD increase), the association of glutamate was only marginally significant (ORfor 1 SD increase = 0.79 (0.60–1.03)).
After adjusting for multiple comparisons (FDR), only the association between serine and PAD was borderline significant (p-value = 0.065).
The second step was to analyze the additional metabolites involved in the degradation pathways for AA that appeared to be associated with PAD development. First, the metabolites implicated in the tryptophan/kynurenine and tryptophan/serine pathways were analyzed. In Table 3, it was shown that higher values for the kynurenine to tryptophan ratio (Kyn/Trp) were associated with a higher risk of PAD. This association was observed across increasing quartiles of the Kyn/Trp ratio (p-Value for Linear trend = 0.012) and also when the ratio was introduced as a continuous variable (ORfor 1 SD increase = 1.50 (1.14–1.98)). On the other hand, baseline serotonin was associated with a higher risk of PAD across increasing quartiles (ORQ4vsQ1 = 2.30 (1.12–4.73); p-Value for Linear trend= 0.049), but this association was considerably weaker and non-significant when serotonin was introduced as a continuous variable (ORfor 1 SD increase = 1.18 (0.91–1.53)) (Table 3).
No metabolite related to the degradation pathways of serine and threonine was significantly associated with the risk of PAD (Table 4). The glutamine-to-glutamate ratio was not significantly associated with PAD (Table 5).
In addition, potential interactions between baseline levels of metabolites that appeared to be associated with PAD and the intervention group were analyzed. The interaction terms of the Kyn/Trp ratio and serine with the intervention group were not statistically significant (p-value = 0.553 (Figure S1) and p-value = 0.651 (Figure S2), respectively). However, an effect modification by the intervention for the association between threonine and PAD (p-value = 0.018) was found. The subjects allocated to either the control or the MedDiet+nuts groups and with high baseline levels of threonine showed a lower risk of PAD compared with those in the control group and with low baseline threonine levels (Figure 1). However, compared with those of low threonine levels allocated to the control group, subjects allocated to the MedDiet+EVOO group showed a lower risk of PAD regardless of their baseline threonine (Figure 1). Consequently, the beneficial effects of MedDiet+EVOO did not depend on threonine levels, while in the MedDiet+nuts group, threonine levels played a role in the association between the intervention and PAD.

3. Discussion

In this case-control study nested in the PREDIMED study, certain baseline AAs were prospectively associated with the risk of PAD. High circulating baseline levels of tryptophan, glutamate, serine and threonine were inversely associated with the risk of PAD. On the contrary, the ratio of kynurenine/tryptophan was prospectively associated with a higher risk of PAD.
Moreover, we found that subjects allocated to the MedDiet+EVOO group were protected against PAD regardless of their baseline AA profile. However, only subjects allocated to the control or to the MedDiet+nuts groups and with high baseline levels of threonine showed a lower risk of PAD than those with low levels of threonine.
Metabolites of the tryptophan–kynurenine degradation pathway and, specifically, the Kyn/Trp ratio have been previously associated with a higher risk of atherosclerosis [18] and coronary artery disease [19], aside from other CVDs. Indeed, in previous assessments in the PREDIMED trial, we found that the plasma Kyn/Trp ratio was associated with an increased risk of heart failure [20], and some kynurenine-related metabolites were associated with a higher risk of CVD [21]. The kynurenine pathway is intimately related to inflammation; Cytokines regulate the tryptophan–kynurenine pathway at several steps [22], and metabolites implicated in this pathway have a regulatory role in inflammatory responses [22]. In this context, inflammation is one of the processes which, in combination with endothelial dysfunction, interact with conventional risk factors leading to the onset and clinical manifestations of atherosclerosis and PAD [23,24]. Thus, it is coherent that both inflammation and the kynurenines pathway could be activated in PAD subjects even before its diagnosis or clinical manifestations. However, we cannot establish whether an activated tryptophan–kynurenine pathway may trigger the inflammatory response or whether the over-expressed cytokines activated the kynurenine pathway.
On the other hand, we found that baseline plasma serine was inversely associated with the risk of PAD. In concordance with our results, serine has been implicated in the production of antioxidants and nitric oxide in animal/cell models [25,26], which may lead to anti-atherogenic effects [25]. Serine was inversely associated with hypertension, a recognized risk factor for PAD [23] in the EPIC-Potsdam study and others supporting these findings [27].
We also found that higher circulating levels of baseline threonine were prospectively and inversely related to PAD. Similarly, threonine was previously reported to be inversely associated with some atherogenic lipids (small dense, low-density lipoprotein cholesterol, remnant-like particle cholesterol and triglycerides (TG)) in a healthy, community-based Chinese cohort [28]. In the Heart Study Framingham cohort, threonine was associated with lower levels of plasma TG [29]. Thus, the association between threonine and PAD risk might be at least partially explained by its anti-atherogenic properties.
The inverse association between MedDiet+EVOO and PAD independent of baseline levels of threonine is consistent with previous reports in the PREDIMED trial. In this trial, a MedDiet enriched with EVOO protected against PAD [30]. Surprisingly, we observed that subjects allocated to the MedDiet+nuts or the control group benefited from the intervention depending on threonine baseline levels; only those with high baseline threonine (either belonging to the MedDiet+nuts or the control group) showed a lower PAD risk as compared with control group subjects with low levels of threonine. We hypothesize that the anti-atherogenic effects of a low-fat diet or the MedDiet+nuts could act in synergy with the anti-atherogenic effects of threonine.
Finally, a potential inverse and non-linear association was observed between glutamate and PAD, which contrasts with previous findings that associated glutamate with a higher risk of atheroma plaque or CVD [16,31]. However, we did not observe any associations with glutamine or the ratio of glutamine to glutamate with PAD, which did not support the glutamate finding.
Our study presents some limitations that need to be addressed. First of all, the sample size was not large regarding the number of adjudicated incident PAD cases; thus, the results should be interpreted with caution and replicated in larger studies. Unfortunately, we were not able to replicate our results in other cohorts. Nevertheless, the results on serine and threonine (sharing metabolic pathways) seemed to be robust. On the other hand, although we have adjusted our statistical models for several confounders, residual confounding may still be present. Finally, it is not possible to generalize our results to other populations with a lower prevalence of cardiovascular risk factors, younger populations or populations pertaining to ethnic groups other than Caucasians.
In conclusion, the results of our matched case-control study nested in the PREDIMED study suggest that baseline circulating AAs, especially serine, threonine, tryptophan and the ratio kynurenine/tryptophan, may contribute to the prospective prediction of the risk of PAD in populations with high cardiovascular risk.

4. Materials and Methods

4.1. Study Design

The design and methods of the PREDIMED trial have been previously described [32]. From 2003 to 2009, 7447 Spanish participants with high cardiovascular risk were recruited in eleven different sites. Participants were female or male aged 55–80 years, with type 2 diabetes (T2D) or with at least three of the following cardiovascular risk factors: body mass index (BMI) ≥ 25 kg/m2, currently smoking, hypertension, high levels of LDL cholesterol, low levels of HDL cholesterol or a family history of early coronary artery disease. Participants were randomly allocated 1:1:1 to one of the three intervention groups: Mediterranean diet (MedDiet) supplemented with extra virgin olive oil (EVOO), MedDiet supplemented with mixed tree nuts or advice to adhere to a low-fat control diet. The median follow-up was 4.8 years. The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of all PREDIMED recruiting centers.
For the molecular phenotyping studies, a matched case-control study (a retrospective study designed after the completion of the main trial) was designed, including all the PAD incident cases identified from 2003 to 2017 with available blood samples. Incidence density sampling with replacement was used as the control sampling method [20]. Thus, controls were randomly selected from all participants at risk at the time of the occurrence of the incident case, and matched controls could be selected again as a control for another index case and they could become later a case [33]. One to three controls per case were matched by their recruitment center, year of birth (± five years) and sex. Initially, 170 cases and 254 controls were included in the analyses. However, there were four controls and three cases that were excluded because of missing data for the studied metabolites. Therefore, the final analyzed sample consisted of 167 PAD cases and 250 controls.

4.2. Ascertainment of PAD

PAD was pre-specified as a secondary endpoint in the PREDIMED trial protocol. Participants’ medical records were systematically and periodically examined during the follow-up period to identify PAD cases [30]. The adjudication of a PAD case by the Clinical Events Committee was required to meet at least one of the following documented criteria: an ankle-brachial index lower than 0.9 at rest, clinical evidence of arterial occlusive disease or an endovascular or open surgical revascularization (or amputation) occurrence. All PAD and other major cardiovascular disease-related (CVD) events (myocardial infarction, stroke and CVD death) were confirmed by the Central Endpoint Adjudication Committee, who were blinded to the interventions.

4.3. Sample Collection and Metabolomic Analysis

At baseline, participants provided fasting blood samples which were processed at each recruiting center no later than 2 h after collection and stored at −80 °C until analysis. Samples for this study were shipped and assayed in the same analytical run and randomly sorted to reduce bias and inter-assay variability.
Liquid chromatography-tandem mass spectrometry (LC-MS) was used to measure polar plasma metabolites. All AA were measured using hydrophilic interaction liquid chromatography (HILIC) coupled with high-resolution positive ion mode MS detection (18 out of the 20 AA were available among the annotated metabolomics data). For the specific analyses of the tryptophan pathway, kynurenine and quinolinic acid were measured using HILIC and targeted negative ion mode MS detection. Both methods were previously described [34,35].

4.4. Covariates

Baseline questionnaires were used to collect sociodemographic lifestyle variables, prevalent and family history of disease and medication use. Leisure-time physical activity was measured with the validated version of the Minnesota Leisure-Time Physical Activity questionnaire [36].

4.5. Statistical Analysis

Individual metabolite values were normalized and scaled in multiples of 1 SD with Blom’s inverse normal transformation [37]. Means and standard deviations (SD) were used to describe quantitative traits, and percentages were used to describe categorical variables.
Conditional logistic regression models were fitted to account for the matching between cases and controls. In these conditional logistic models, matched odds ratios (OR) and their 95% CIs for PAD were calculated while considering the first quartile as the reference category. Quartile cut-off points were calculated according to the distribution of metabolites among controls (participants without PAD through the follow-up). The matched odds ratios for the SD of each metabolite, including them as continuous variables, were also calculated. Crude and multivariable models adjusted for the intervention group (MedDiet + EVOO/MedDiet + nuts/control), smoking status (never/current/former), BMI (kg/m2), leisure-time physical activity (metabolic equivalent task [MET]-min/day), prevalent chronic conditions at baseline hypertension, T2D and dyslipidemia- and educational level (primary school or lower/secondary school or higher) were fitted. The p-values were penalized for multiple comparisons using the false discovering rate described by Simes [38].
For AAs that were prospectively associated with PAD incidence in the individual metabolite analyses explained above, additional analyses, including all the annotated metabolites available for each related pathway, were performed. Specifically, conditional logistic regression models adjusted for the same confounders for the available metabolites implicated in the degradation pathways of tryptophan (kynurenine, kynurenic acid, 3-hydroxyanthranilic acid and quinolinic acid), serine, threonine and glycine (choline, betaine and dimethylglycine) were performed. Ratios between implicated metabolites (kynurenine/tryptophan, serine/tryptophan and glutamine/glutamate) were also calculated to evaluate specific steps of each pathway and included in the same conditional logistic regression models.
Interactions (2 degrees of freedom) between each plasma AA as a dichotomous trait (below/above the median) and considering the three arms of the intervention (control/MedDiet + EVOO/MedDiet + nuts) were tested using conditional logistic models adjusted for smoking status (never/current/former), BMI (kg/m2), leisure-time physical activity (metabolic equivalent task [MET]-min/day), prevalent chronic diseases such as hypertension, T2D and dyslipidemia–, family history of premature coronary heart disease (CHD) and educational level (primary school or lower/secondary school or higher). In addition, adjustment for propensity scores that used 30 baseline variables to estimate the probability of assignment to each of the intervention groups and robust variance estimators were used considering that a small percentage of participants were non-individually randomized to the intervention groups and minor imbalances in baseline covariates existed in the trial [32]. A detailed description of how propensity scores were calculated can be found in the supplement of the main paper of PREDIMED [32]. The p-values for interactions were calculated using the likelihood ratio test for each assessment of potential effect modifiers.
Moreover, as an ancillary analysis, if the interaction of any of the metabolites and intervention results were statistically significant, a new logistic conditional model including a joint variable that combined the metabolite as dichotomous (below/above the median) and the intervention group (MedDiet+EVOO and MedDiet+nuts vs. control) was conducted.
A p-value < 0.05 was deemed statistically significant for each performed test.
All the statistical procedures were carried out with STATA 16 software, 2.1.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24010270/s1.

Author Contributions

Conceptualization, M.A.M.-G., F.B.H. and C.R.; Methodology, M.R.-C. and E.T.; Software, C.B.C.; Validation, C.R., E.T. and M.R.-C.; Formal analysis, C.R., E.T., L.L., C.W., M.G.-F. and M.R.-C.; Investigation, F.B.H., J.S.-S. and M.A.M.-G.; Resources, F.B.H., J.S.-S. and M.A.M.-G.; Data curation, C.B.C.; Writing—original draft preparation, C.R.; Writing—review and editing, all authors; Visualization, C.R.; Supervision, M.A.M.-G., E.T. and M.R.-C.; Project administration, M.A.M.-G. and F.B.H.; Funding acquisition, M.F. (Miquel Fiol), R.E., E.R., J.S.-S., D.C., M.F. (Montse Fitó), E.G.-G., F.A., L.S.-M., A.A.-G., J.M.S.-L., M.A.M.-G. and F.B.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health research grant R01HL118264 (1R01HL118264-01/2R01HL118264-05/2R01HL118264-09). The PREDIMED trial was supported by the official funding agency for biomedical research of the Spanish government, Instituto de Salud Carlos III (ISCIII), through grants provided to research networks specifically developed for the trial (RTIC G03/140 to Ramón Estruch during 2003–2005; RTIC RD 06/0045 to Miguel A. Martínez-González during 2006–2013 and through the Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición [CIBEROBN]), and by grants from the Centro Nacional de Investigaciones Cardiovasculares (CNIC 06/2007), the Fondo de Investigación Sanitaria–Fondo Europeo de Desarrollo Regional (PI04–2239, PI 05/2584, CP06/00100, PI07/0240, PI07/1138, PI07/0954, PI 07/0473, PI10/01407, PI10/02658, PI11/01647, P11/02505, PI13/00462 and JR17/00022), the Ministerio de Ciencia e Innovación (AGL-2009–13906-C02, AGL2010–22319-C03 and SAF2016–80532-R), the Fundación Mapfre 2010 (no role in the design, implementation, analysis or interpretation of the data), the Consejería de Salud de la Junta de Andalucía (PI0105/2007), the Public Health Division of the Department of Health of the Autonomous Government of Catalonia, Generalitat Valenciana (ACOMP06109, GVA-COMP2010–181, GVACOMP2011–151, CS2010-AP-111, PROMETEO 17/2017 and CS2011-AP-042), the Fundació La Marató-TV3 (grants 294/C/2015 and 538/U/2016 (no role in the design, implementation, analysis or interpretation of the data)) and the Regional Government of Navarra (P27/2011). Marta Guasch-Ferré was supported by the American Diabetes Association, grant #1-18-PMF-029. Jordi Salas-Salvadó was partially supported by ICREA under the ICREA Academia programme. Wittenbecher was supported by the SciLifeLab & Wallenberg Data Driven Life Science Program (grant: KAW 2020.0239). Marta Guasch-Ferré was supported by NNF18CC0034900.

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of all PREDIMED recruiting centers.

Informed Consent Statement

Informed consent was obtained from all subjects involved in this study.

Data Availability Statement

We will be happy to provide access to the Predimed dataset (including data dictionaries), making possible the replication of the main analyses used for the present article. Due to the restrictions imposed by the Informed Consent and the Institutional Review Board, bona fide investigators interested in analyzing the Predimed dataset used for the present article may submit a brief proposal and statistical analysis plan to the corresponding author. Upon approval from the Predimed Steering Committee and Institutional Review Boards, the data will be made available to them using an onsite secure access data enclave.

Acknowledgments

We thank all the volunteers for their participation and personnel for their contributions to the PREDIMED trial. We also thank all the investigators of the PREDIMED study.

Conflicts of Interest

J. Salas-Salvadó was an advisor for the International Nut and Dried Fruit Foundation and E. Ros for the California Walnut Commission, International Nut and Dried Fruit Council. J. Salas-Salvadó was a non-paid member of the International Advisory Board of the project “Effect of cashew nut supplementation on glycemic status and lipid profile in type 2 diabetes subjects”, a member of the Institute Danone Spain Advisory Board and of the Scientific Committee of Danone Institute International and World forum for nutrition research and Dissemination. E Ros received honoraria from Alexion and J. Salas-Salvadó from Mundipharma.

References

  1. Golledge, J.; Fernando, M.E.; Armstrong, D.G. Current Management of Peripheral Artery Disease: Focus on Pharmacotherapy. Drugs 2022, 82, 1165–1177. [Google Scholar] [CrossRef] [PubMed]
  2. Fowkes, F.G.R.; Rudan, D.; Rudan, I.; Aboyans, V.; Denenberg, J.O.; McDermott, M.M.; Norman, P.E.; Sampson, U.K.A.; Williams, L.J.; Mensah, G.A.; et al. Comparison of Global Estimates of Prevalence and Risk Factors for Peripheral Artery Disease in 2000 and 2010: A Systematic Review and Analysis. Lancet 2013, 382. [Google Scholar] [CrossRef] [PubMed]
  3. Kyu, H.H.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Disability-Adjusted Life-Years (DALYs) for 359 Diseases and Injuries and Healthy Life Expectancy (HALE) for 195 Countries and Territories, 1990–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1859–1922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gupta, D.K.; Skali, H.; Claggett, B.; Kasabov, R.; Cheng, S.; Shah, A.M.; Loehr, L.R.; Heiss, G.; Nambi, V.; Aguilar, D.; et al. Heart Failure Risk Across the Spectrum of Ankle-Brachial Index: The ARIC Study (Atherosclerosis Risk In Communities). JACC Heart Fail 2014, 2, 447–454. [Google Scholar] [CrossRef] [PubMed]
  5. Emdin, C.A.; Wong, C.X.; Hsiao, A.J.; Altman, D.G.; Peters, S.A.E.; Woodward, M.; Odutayo, A.A. Atrial Fibrillation as Risk Factor for Cardiovascular Disease and Death in Women Compared with Men: Systematic Review and Meta-Analysis of Cohort Studies. BMJ 2016, 352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. O’Neal, W.T.; Efird, J.T.; Nazarian, S.; Alonso, A.; Heckbert, S.R.; Soliman, E.Z. Peripheral Arterial Disease and Risk of Atrial Fibrillation and Stroke: The Multi-Ethnic Study of Atherosclerosis. J. Am. Heart Assoc. 2014, 3. [Google Scholar] [CrossRef] [Green Version]
  7. Hirsch, A.T.; Criqui, M.H.; Treat-Jacobson, D.; Regensteiner, J.G.; Creager, M.A.; Olin, J.W.; Krook, S.H.; Hunninghake, D.B.; Comerota, A.J.; Walsh, M.E.; et al. Peripheral Arterial Disease Detection, Awareness, and Treatment in Primary Care. JAMA 2001, 286, 1317–1324. [Google Scholar] [CrossRef]
  8. Sigvant, B.; Lundin, F.; Wahlberg, E. The Risk of Disease Progression in Peripheral Arterial Disease Is Higher than Expected: A Meta-Analysis of Mortality and Disease Progression in Peripheral Arterial Disease. Eur. J. Vasc. Endovasc. Surg. 2016, 51, 395–403. [Google Scholar] [CrossRef] [Green Version]
  9. Breek, J.C.; Hamming, J.F.; de Vries, J.; Aquarius, A.E.A.M.; van Berge Henegouwen, D.P. Quality of Life in Patients with Intermittent Claudication Using The World Health Organisation (WHO) Questionnaire. Eur. J. Vasc. Endovasc. Surg. 2001, 21, 118–122. [Google Scholar] [CrossRef] [Green Version]
  10. McDermott, M.M.G.; Fried, L.; Simonsick, E.; Ling, S.; Guralnik, J.M. Asymptomatic Peripheral Arterial Disease Is Independently Associated With Impaired Lower Extremity Functioning. Circulation 2000, 101, 1007–1012. [Google Scholar] [CrossRef]
  11. Tendera, M.; Aboyans, V.; Bartelink, M.L.; Baumgartner, I.; Clment, D.; Collet, J.P.; Cremonesi, A.; de Carlo, M.; Erbel, R.; Fowkes, F.G.R.; et al. ESC Guidelines on the Diagnosis and Treatment of Peripheral Artery DiseasesDocument Covering Atherosclerotic Disease of Extracranial Carotid and Vertebral, Mesenteric, Renal, Upper and Lower Extremity ArteriesThe Task Force on the Diagnosis and Treatment of Peripheral Artery Diseases of the European Society of Cardiology (ESC). Eur. Heart J. 2011, 32, 2851–2906. [Google Scholar] [CrossRef] [Green Version]
  12. Fowkes, F.G.R.; Aboyans, V.; Fowkes, F.J.I.; McDermott, M.M.; Sampson, U.K.A.; Criqui, M.H. Peripheral Artery Disease: Epidemiology and Global Perspectives. Nat. Rev. Cardiol. 2016, 14, 156–170. [Google Scholar] [CrossRef] [PubMed]
  13. Hutson, S.M.; Sweatt, A.J.; LaNoue, K.F. Branched-Chain Amino Acid Metabolism: Implications for Establishing Safe Intakes. J. Nutr. 2005, 135, 1557S–1564S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ruiz-Canela, M.; Toledo, E.; Clish, C.B.; Hruby, A.; Liang, L.; Salas-Salvado, J.; Razquin, C.; Corella, D.; Estruch, R.; Ros, E.; et al. Plasma Branched-Chain Amino Acids and Incident Cardiovascular Disease in the PREDIMED Trial. Clin. Chem. 2016, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wang, X.; Yang, R.; Zhang, W.; Wang, S.; Mu, H.; Li, H.; Dong, J.; Chen, W.; Yu, X.; Ji, F. Serum Glutamate and Glutamine-to-Glutamate Ratio Are Associated with Coronary Angiography Defined Coronary Artery Disease. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 186–194. [Google Scholar] [CrossRef] [PubMed]
  16. Zheng, Y.; Hu, F.B.; Ruiz-Canela, M.; Clish, C.B.; Dennis, C.; Salas-Salvado, J.; Hruby, A.; Liang, L.; Toledo, E.; Corella, D.; et al. Metabolites of Glutamate Metabolism Are Associated With Incident Cardiovascular Events in the PREDIMED PREvención Con DIeta MEDiterránea (PREDIMED) Trial. J. Am. Heart Assoc. 2016, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Sulo, G.; Vollset, S.E.; Nygard, O.; Midttun, O.; Ueland, P.M.; Eussen, S.J.P.M.; Pedersen, E.R.; Tell, G.S. Neopterin and Kynurenine-Tryptophan Ratio as Predictors of Coronary Events in Older Adults, the Hordaland Health Study. Int. J. Cardiol. 2013, 168, 1435–1440. [Google Scholar] [CrossRef]
  18. Liu, J.J.; Movassat, J.; Portha, B. Emerging Role for Kynurenines in Metabolic Pathologies. Curr. Opin. Clin. Nutr. Metab. Care 2019, 22, 82–90. [Google Scholar] [CrossRef]
  19. Gáspár, R.; Halmi, D.; Demján, V.; Berkecz, R.; Pipicz, M.; Csont, T. Kynurenine Pathway Metabolites as Potential Clinical Biomarkers in Coronary Artery Disease. Front. Immunol. 2022, 12, 5874. [Google Scholar] [CrossRef]
  20. Razquin, C.; Ruiz-Canela, M.; Toledo, E.; Hernández-Alonso, P.; Clish, C.B.; Guasch-Ferré, M.; Li, J.; Wittenbecher, C.; Dennis, C.; Alonso-Gómez, A.; et al. Metabolomics of the Tryptophan–Kynurenine Degradation Pathway and Risk of Atrial Fibrillation and Heart Failure: Potential Modification Effect of Mediterranean Diet. Am. J. Clin. Nutr. 2021, 114, 1646. [Google Scholar] [CrossRef]
  21. Yu, E.; Ruiz-Canela, M.; Guasch-Ferré, M.; Zheng, Y.; Toledo, E.; Clish, C.B.; Salas-Salvadó, J.; Liang, L.; Wang, D.D.; Corella, D.; et al. Increases in Plasma Tryptophan Are Inversely Associated with Incident Cardiovascular Disease in the Prevención Con Dieta Mediterránea (PREDIMED) Study. J. Nutr. 2017, 147, 249–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Baumgartner, R.; Forteza, M.J.; Ketelhuth, D.F.J. The Interplay between Cytokines and the Kynurenine Pathway in Inflammation and Atherosclerosis. Cytokine 2019, 122, 154148. [Google Scholar] [CrossRef] [PubMed]
  23. Poredoš, P.; Šabovič, M.; Božič Mijovski, M.; Nikolajević, J.; Antignani, P.L.; Paraskevas, K.I.; Mikhailidis, D.P.; Blinc, A. Inflammatory and Prothrombotic Biomarkers, DNA Polymorphisms, MicroRNAs and Personalized Medicine for Patients with Peripheral Arterial Disease. Int. J. Mol. Sci. 2022, 23, 12054. [Google Scholar] [CrossRef]
  24. Brevetti, G.; Giugliano, G.; Brevetti, L.; Hiatt, W.R. Inflammation in Peripheral Artery Disease. Circulation 2010, 122, 1862–1875. [Google Scholar] [CrossRef] [PubMed]
  25. Naderi Maralani, M.; Movahedian, A.; Haghjooy Javanmard, S. Antioxidant and Cytoprotective Effects of L-Serine on Human Endothelial Cells. Res. Pharm. Sci. 2012, 7, 209. [Google Scholar]
  26. Mishra, R.C.; Tripathy, S.; Desai, K.M.; Quest, D.; Lu, Y.; Akhtar, J.; Gopalakrishnan, V. Nitric Oxide Synthase Inhibition Promotes Endothelium-Dependent Vasodilatation and the Antihypertensive Effect of L-Serine. Hypertension 2008, 51, 791–796. [Google Scholar] [CrossRef] [Green Version]
  27. Shi, M.; He, J.; Li, C.; Lu, X.; He, W.J.; Cao, J.; Chen, J.; Chen, J.C.; Bazzano, L.A.; Li, J.X.; et al. Metabolomics Study of Blood Pressure Salt-Sensitivity and Hypertension. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 1681–1692. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, F.H.; Liu, J.; Deng, Q.J.; Qi, Y.; Wang, M.; Wang, Y.; Zhang, X.G.; Zhao, D. Association between Plasma Essential Amino Acids and Atherogenic Lipid Profile in a Chinese Population: A Cross-Sectional Study. Atherosclerosis 2019, 286, 7–13. [Google Scholar] [CrossRef]
  29. Cheng, S.; Rhee, E.P.; Larson, M.G.; Lewis, G.D.; McCabe, E.L.; Shen, D.; Palma, M.J.; Roberts, L.D.; Dejam, A.; Souza, A.L.; et al. Metabolite Profiling Identifies Pathways Associated with Metabolic Risk in Humans. Circulation 2012, 125, 2222–2231. [Google Scholar] [CrossRef] [Green Version]
  30. Ruiz-Canela, M.; Estruch, R.; Corella, D.; Salas-Salvadó, J.; Martínez-González, M.A. Association of Mediterranean Diet with Peripheral Artery Disease: The PREDIMED Randomized Trial. JAMA 2014, 311, 415–417. [Google Scholar] [CrossRef]
  31. Vernon, S.T.; Tang, O.; Kim, T.; Chan, A.S.; Kott, K.A.; Park, J.; Hansen, T.; Koay, Y.C.; Grieve, S.M.; O’sullivan, J.F.; et al. Metabolic Signatures in Coronary Artery Disease: Results from the BioHEART-CT Study. Cells 2021, 10, 980. [Google Scholar] [CrossRef]
  32. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.-I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef] [PubMed]
  33. Lubin, J.H.; Gail, M.H. Biased Selection of Controls for Case-Control Analyses of Cohort Studies. Biometrics 1984, 40, 63–75. [Google Scholar] [CrossRef] [PubMed]
  34. Paynter, N.P.; Balasubramanian, R.; Giulianini, F.; Wang, D.D.; Tinker, L.F.; Gopal, S.; Deik, A.A.; Bullock, K.; Pierce, K.A.; Scott, J.; et al. Metabolic Predictors of Incident Coronary Heart Disease in Women. Circulation 2018, 137, 841–853. [Google Scholar] [CrossRef]
  35. Wang, T.J.; Ngo, D.; Psychogios, N.; Dejam, A.; Larson, M.G.; Vasan, R.S.; Ghorbani, A.; O’Sullivan, J.; Cheng, S.; Rhee, E.P.; et al. 2-Aminoadipic Acid Is a Biomarker for Diabetes Risk. J. Clin. Investig. 2013, 123, 4309–4317. [Google Scholar] [CrossRef] [Green Version]
  36. Elosua, R.; Marrugat, J.; Molina, L.; Pons, S.; Pujol, E. Validation of the Minnesota Leisure Time Physical Activity Questionnaire in Spanish Men. The MARATHOM Investigators. Am. J. Epidemiol. 1994, 139, 1197–1209. [Google Scholar] [CrossRef]
  37. Blom, G. Statistical Estimates and Transformed Beta-Variables; John Wiley & Sons A/S: New York, NY, USA, 1958. [Google Scholar]
  38. Simes, R. An Improved Bonferroni Procedure for Multiple Tests of Significance. Biometrika 1986, 73, 751–754. [Google Scholar] [CrossRef]
Figure 1. Matched odds ratios (95% CI) for the association between baseline threonine (below/above the median) and incident peripheral artery disease (PAD) within each PREDIMED intervention arm. Multivariable model adjusted for the intervention group (Mediterranean diet vs. control group), body mass index (kg/m2), smoking (never, current, former), leisure-time physical activity (metabolic equivalent tasks in min/d), prevalent chronic conditions at baseline (dyslipidemia, hypertension and diabetes), educational level (primary or lower/secondary or higher) and propensity scores predicting randomization to account for small between-group imbalances at baseline. MedDiet: Mediterranean diet.
Figure 1. Matched odds ratios (95% CI) for the association between baseline threonine (below/above the median) and incident peripheral artery disease (PAD) within each PREDIMED intervention arm. Multivariable model adjusted for the intervention group (Mediterranean diet vs. control group), body mass index (kg/m2), smoking (never, current, former), leisure-time physical activity (metabolic equivalent tasks in min/d), prevalent chronic conditions at baseline (dyslipidemia, hypertension and diabetes), educational level (primary or lower/secondary or higher) and propensity scores predicting randomization to account for small between-group imbalances at baseline. MedDiet: Mediterranean diet.
Ijms 24 00270 g001
Table 1. Baseline characteristics of the participants according to the incidence of peripheral artery disease (PAD).
Table 1. Baseline characteristics of the participants according to the incidence of peripheral artery disease (PAD).
Controls (Non-Cases)
(n = 250)
PAD Cases
(n = 167)
Age (years)68.0 (6.8)67.6 (6.8)
Body mass index (kg/m2)29.2 (3.3)29.2 (3.6)
Leisure time physical activity (METs-min/d)285.0 (288)223.0 (221)
Female (%)29.231.1
Hypertension (%)79.277.2
Dyslipidemia (%)65.261.1
Type 2 diabetes (%)52.465.3
Smoking
Never (%)43.235.9
Former (%)20.828.7
Current (%)3635.3
Intervention group
Control (low-fat) (%)30.838.9
MedDiet+EVOO (%)39.627.5
MedDiet+nuts (%)29.633.5
Education
Primary or less (%)73.271.9
Secondary (%)17.622.8
University graduate (%)9.25.39
Mean (standard deviation) is presented unless percentage (%) is specified.
Table 2. Association * between plasma amino acids and incident peripheral artery disease (PAD).
Table 2. Association * between plasma amino acids and incident peripheral artery disease (PAD).
Q1Q2Q3Q4p-Value for Linear TrendPer 1 SDFDR ** Corrected p-Values (Per 1 SD)
GlycineRef.0.78 (0.42–1.45)0.92 (0.49–1.71)0.92 (0.48–1.76)0.8660.93 (0.73–1.18)0.871
AlanineRef.0.70 (0.37–1.35)0.96 (0.49–1.88)0.97 (0.51–1.85)0.8401.02 (0.81–1.29)0.958
ValineRef.0.77 (0.40–1.48)0.50 (0.25–0.99)1.01 (0.54–1.87)0.9481.00 (0.79–1.26)0.991
LeucineRef.1.62 (0.84–3.12)1.28 (0.63–2.61)1.10 (0.56–2.17)0.9181.04 (0.82–1.32)0.945
IsoleucineRef.1.10 (0.57–2.12)1.29 (0.66–2.53)0.84 (0.42–1.70)0.6270.99 (0.79–1.28)0.991
PhenylalanineRef.1.33 (0.68–2.59)1.44 (0.75–2.79)1.76 (0.90–3.44)0.0991.09 (0.87–1.36)0.871
TryptophanRef.1.12 (0.60–2.11)0.65 (0.33–1.29)0.76 (0.38–1.52)0.2640.78 (0.61–0.99)0.315
MethionineRef.0.59 (0.30–1.14)0.60 (0.31–1.16)0.85 (0.45–1.61)0.7090.93 (0.73–1.19)0.871
ProlineRef.1.48 (0.76–2.89)1.28 (0.65–2.54)1.54 (0.78–3.02)0.2861.17 (0.92–1.49)0.697
SerineRef.0.84 (0.46–1.54)0.55 (0.29–1.02)0.41 (0.20–0.84)0.0150.68 (0.53–0.88)0.065
ThreonineRef.0.77 (0.42–1.42)0.41 (0.20–0.84)0.56 (0.29–1.07)0.0260.76 (0.60–0.97)0.264
TyrosineRef.0.90 (0.49–1.64)0.68 (0.35–1.32)1.19 (0.64–2.23)0.7921.02 (0.81–1.29)0.958
AsparagineRef.0.46 (0.23–0.93)0.72 (0.37–1.38)0.77 (0.41–1.44)0.7950.91 (0.72–1.16)0.871
GlutamineRef.0.65 (0.34–1.24)0.70 (0.35–1.39)0.81 (0.42–1.55)0.5560.93 (0.72–1.18)0.871
GlutamateRef.0.41 (0.21–0.79)0.62 (0.34–1.27)0.42 (0.20–0.88)0.0290.78 (0.59–1.03)0.381
HistidineRef.0.74 (0.38–1.45)0.66 (0.34–1.15)1.16 (0.61–2.19)0.6841.10 (0.87–1.40)0.871
LysineRef.0.68 (0.36–1.26)0.84 (0.45–1.55)0.81 (0.44–1.50)0.6250.95 (0.76–1.20)0.945
ArginineRef.1.52 (0.82–2.81)1.22 (0.66–2.26)1.45 (0.74–2.83)0.3871.11 (0.88–1.40)0.871
* Multivariable model adjusted for intervention group, body mass index, smoking, leisure-time physical activity, educational level, dyslipidemia, hypertension and diabetes.** FDR: false discovery rate.
Table 3. Association * between plasma tryptophan–kynurenine/tryptophan–serotonin pathway metabolites and incident peripheral artery disease (PAD).
Table 3. Association * between plasma tryptophan–kynurenine/tryptophan–serotonin pathway metabolites and incident peripheral artery disease (PAD).
Q1Q2Q3Q4p-Value for Linear TrendPer 1 SD
TryptophanRef.1.12 (0.60–2.11)0.65 (0.33–1.29)0.76 (0.38–1.52)0.2640.78 (0.61–0.99)
KynurenineRef.1.16 (0.61–2.24)1.30 (0.65–2.59)1.74 (0.87–3.47)0.1771.23 (0.94–1.59)
Ratio Kyn/Trp **Ref.1.75 (0.87–3.52)1.72 (0.84–3.50)3.11 (1.42–6.82)0.0121.50 (1.14–1.98)
Kynurenic acidRef.0.85 (0.44–1.65)0.83 (0.44–1.58)1.50 (0.80–2.83)0.2841.20 (0.96–1.51)
Hydroxyanthranilic acidRef.0.64 (0.33–1.23)1.27 (0.67–2.40)0.82 (0.38–1.77)0.7790.92 (0.69–1.22)
Quinolinic acidRef.0.76 (0.38–1.50)0.69 (0.35–1.35)1.08 (0.52–2.22)0.9880.90 (0.69–1.18)
SerotoninRef.1.56 (0.77–3.14)1.35 (0.70–2.60)2.30 (1.12–4.73)0.0491.18 (0.91–1.53)
Serotonin/Trp ratioRef.1.53 (0.77–3.02)1.24 (0.62–2.48)1.82 (0.92–3.60)0.1401.22 (0.95–1.57)
* Multivariable model adjusted for the intervention group, body mass index, smoking, leisure-time physical activity, education level, dyslipidemia, hypertension and diabetes. ** Kynurenine/tryptophan ratio.
Table 4. Association * between plasma serine–glycine–threonine pathway metabolites and incident of peripheral artery disease (PAD).
Table 4. Association * between plasma serine–glycine–threonine pathway metabolites and incident of peripheral artery disease (PAD).
Q1Q2Q3Q4p-Value for Linear TrendPer 1 SD
SerineRef.0.84 (0.46–1.54)0.55 (0.29–1.02)0.41 (0.20–0.84)0.0150.67 (0.51–0.86)
GlycineRef.0.78 (0.42–1.45)0.92 (0.49–1.71)0.92 (0.48–1.76)0.8660.93 (0.73–1.18)
ThreonineRef.0.77 (0.42–1.42)0.41 (0.20–0.84)0.56 (0.29–1.07)0.0260.75 (0.59–0.95)
CholineRef.0.89 (0.47–1.68)0.69 (0.36–1.32)1.14 (0.60–2.19)0.8231.02 (0.80–1.29)
BetaineRef.1.04 (0.57–1.91)0.92 (0.49–1.76)0.95 (0.45–2.04)0.8211.03 (0.80–1.32)
DimethylglycineRef.0.74 (0.38–1.42)0.93 (0.48–1.78)0.77 (0.39–1.53)0.5520.96 (0.76–1.21)
* Multivariable model adjusted for the intervention group, body mass index, smoking, leisure-time physical activity, education level, dyslipidemia, hypertension and diabetes.
Table 5. Association between plasma glutamine–glutamate pathway metabolites and incident of peripheral artery disease (PAD). Multivariable model adjusted for the intervention group, body mass index, smoking, leisure-time physical activity, education level, dyslipidemia, hypertension and diabetes.
Table 5. Association between plasma glutamine–glutamate pathway metabolites and incident of peripheral artery disease (PAD). Multivariable model adjusted for the intervention group, body mass index, smoking, leisure-time physical activity, education level, dyslipidemia, hypertension and diabetes.
Q1Q2Q3Q4p-Value for Linear TrendPer 1 SD
GlutamineRef.0.65 (0.34–1.24)0.70 (0.35–1.39)0.81 (0.42–1.55)0.5560.93 (0.73–1.18)
GlutamateRef.0.41 (0.21–0.79)0.62 (0.34–1.15)0.42 (0.20–0.88)0.0290.78 (0.59–1.03)
Gln/Glt ratio *Ref.1.35 (0.68–2.69)1.21 (0.57–2.56)2.03 (0.99–4.15)0.0591.22 (0.92–1.60)
* Multivariable model adjusted for the intervention group, body mass index, smoking, leisure-time physical activity, education level, dyslipidemia, hypertension and diabetes.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Razquin, C.; Ruiz-Canela, M.; Toledo, E.; Clish, C.B.; Guasch-Ferré, M.; García-Gavilán, J.F.; Wittenbecher, C.; Alonso-Gómez, A.; Fitó, M.; Liang, L.; et al. Circulating Amino Acids and Risk of Peripheral Artery Disease in the PREDIMED Trial. Int. J. Mol. Sci. 2023, 24, 270. https://doi.org/10.3390/ijms24010270

AMA Style

Razquin C, Ruiz-Canela M, Toledo E, Clish CB, Guasch-Ferré M, García-Gavilán JF, Wittenbecher C, Alonso-Gómez A, Fitó M, Liang L, et al. Circulating Amino Acids and Risk of Peripheral Artery Disease in the PREDIMED Trial. International Journal of Molecular Sciences. 2023; 24(1):270. https://doi.org/10.3390/ijms24010270

Chicago/Turabian Style

Razquin, Cristina, Miguel Ruiz-Canela, Estefania Toledo, Clary B. Clish, Marta Guasch-Ferré, Jesús F. García-Gavilán, Clemens Wittenbecher, Angel Alonso-Gómez, Montse Fitó, Liming Liang, and et al. 2023. "Circulating Amino Acids and Risk of Peripheral Artery Disease in the PREDIMED Trial" International Journal of Molecular Sciences 24, no. 1: 270. https://doi.org/10.3390/ijms24010270

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop