Grapefruit Juice Flavanones Modulate the Expression of Genes Regulating Inflammation, Cell Interactions and Vascular Function in Peripheral Blood Mononuclear Cells of Postmenopausal Women

Irena Krga, Karla Fabiola Corral-Jara, Nicolas Barber-Chamoux, Claude Dubray, Christine Morand, Dragan Milenkovic, Irena Krga, Karla Fabiola Corral-Jara, Nicolas Barber-Chamoux, Claude Dubray, Christine Morand, Dragan Milenkovic

Abstract

Grapefruit is a rich source of flavanones, phytochemicals suggested excreting vasculoprotective effects. We previously showed that flavanones in grapefruit juice (GFJ) reduced postmenopausal women's pulse-wave velocity (PWV), a measure of arterial stiffness. However, mechanisms of flavanone action in humans are largely unknown. This study aimed to decipher molecular mechanisms of flavanones by multi-omics analysis in PBMCs of volunteers consuming GFJ and flavanone-free control drink for 6 months. Modulated genes and microRNAs (miRNAs) were identified using microarrays. Bioinformatics analyses assessed their functions, interactions and correlations with previously observed changes in PWV. GFJ modified gene and miRNA expressions. Integrated analysis of modulated genes and miRNA-target genes suggests regulation of inflammation, immune response, cell interaction and mobility. Bioinformatics identified putative mediators of the observed nutrigenomic effect (STAT3, NF-κB) and molecular docking demonstrated potential binding of flavanone metabolites to transcription factors and cell-signaling proteins. We also observed 34 significant correlations between changes in gene expression and PWV. Moreover, global gene expression was negatively correlated with gene expression profiles in arterial stiffness and hypertension. This study revealed molecular mechanisms underlying vasculoprotective effects of flavanones, including interactions with transcription factors and gene and miRNA expression changes that inversely correlate with gene expression profiles associated with cardiovascular risk factors.

Clinical trial registration: [ClinicalTrials.gov], identifier [NCT01272167].

Keywords: bioinformatics; flavanone; genomics; naringin; vascular function.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Krga, Corral-Jara, Barber-Chamoux, Dubray, Morand and Milenkovic.

Figures

FIGURE 1
FIGURE 1
A flow-chart describing steps implemented in the genomic and bioinformatic analysis.
FIGURE 2
FIGURE 2
A 2D principal component analysis plot of the genomic profiles of PBMCs from volunteers after consumption of flavanones in GFJ and same volunteers after control drink consumption.
FIGURE 3
FIGURE 3
Functional ontology and network analyses of differentially expressed genes. (A) Network of over-represented gene ontologies of identified differentially expressed genes identified using ShinyGO tool. Two nodes are connected if they share 20% or more genes. Darker nodes are more significantly enriched gene sets. Bigger nodes represent larger gene sets. Thicker edges represent more overlapped genes. (B) Pie chart of significantly over-represented functional gene networks identified using text-mining approach from Metacore. Pie chart represent functional groups of the identified networks, with section proportional to the number of networks in each group. Gene networks of each functional group are represented together with the number of genes in each network. *p < 0.05, **p < 0.01, and ****p < 0.0001.
FIGURE 4
FIGURE 4
Functional pathway analysis of differentially expressed genes presented as a histogram of significantly over-represented pathways and networks of genes present in pathways of functional groups. Yellow bars depict p-values of the identified significant pathways, and orange dots the number of genes in each pathway. Histogram data obtained using BioCarta (†), Kyoto Encyclopedia of Genes and Genomes (*) and NCI Pathways (#) databases through GeneTrail2. Networks of genes present in pathways of functional groups obtained using Cytoscape.
FIGURE 5
FIGURE 5
In silico docking analysis of interactions between naringin metabolites and potential transcription factors and cell signaling proteins. (A) Naringenin 4’-O-glucuronide to STAT3; (B) naringenin-7-O-glucuronide to STAT3; (C) naringenin-7-O-glucuronide to Jak2; (D) naringenin 4’-O-glucuronide to ERK1(MAPK3); (E) naringenin-7-O-glucuronide to Akt.
FIGURE 6
FIGURE 6
The effects of flavanones in GFJ on miRNA expression in PBMCs with a functional analysis of differentially modulated miRNAs. (A) Fold-changes for the identified differentially expressed miRNAs following the consumption of GFJ; (B) Network presentation of miRNAs and potential target genes; (C) Functional pathway analysis of miRNAs target genes. The orange histogram bars depict p-values of the identified significant pathways, and brown dots the number of genes in each pathway. Histogram data obtained using BioCarta (†), Kyoto Encyclopedia of Genes and Genomes (*) and NCI Pathways (#) databases through GeneTrail2.
FIGURE 7
FIGURE 7
Integration analysis of differentially expressed protein-coding genes and miRNAs. (A) Network of the identified putative transcription factors, differentially expressed miRNAs, differentially expressed genes and the identified putative gene targets of differentially expressed miRNAs; (B) Comparison of pathways identified from differentially expressed genes and target genes of differentially expressed miRNAs. The size of each rectangle is proportional to the number of genes in the pathway. Black letters: pathways identified from differentially expressed protein-coding genes; white letters: pathways from differentially expressed miRNAs; red letters: pathways in common for both protein-coding and miRNA genes.
FIGURE 8
FIGURE 8
Pearson correlation analysis between modulation in the expression of genes and changes in pulse wave velocity of the same volunteers. (A) Correlation plot indicating positive and negative correlations, (B) correlation plot between pulse wave velocity and VCL; (C) correlation plot between pulse wave velocity and RAET1K; (D) correlation plot between pulse-wave velocity and PLEKHG4B; (E) correlation plot between pulse wave velocity and SMPD2. All p-values <0.05.
FIGURE 9
FIGURE 9
Correlations between global gene expression profile from volunteers who consumed grapefruit juice and gene expression profile associated with an increase in (A) arterial stiffness and (B) hypertension and arterial stiffness obtained from Gene Expression Omnibus (GEO) repository.
FIGURE 10
FIGURE 10
Schematic presentation of genomic modifications induced by consumption of flavanones in grapefruit juice and its impact on vascular health.

References

    1. Mink PJ, Scrafford CG, Barraj LM, Harnack L, Hong CP, Nettleton JA, Jacobs DR. Flavonoid intake and cardiovascular disease mortality: a prospective study in postmenopausal women. Am J Clin Nutr. (2007) 85:895–909. 10.1093/ajcn/85.3.895
    1. Landberg R, Sun Q, Rimm EB, Cassidy A, Scalbert A, Mantzoros CS, et al. Selected dietary flavonoids are associated with markers of inflammation and endothelial dysfunction in U.S. Women. J Nutr. (2011) 141:618–25. 10.3945/jn.110.133843
    1. Dow CA, Going SB, Chow HHS, Patil BS, Thomson CA. The effects of daily consumption of grapefruit on body weight, lipids, and blood pressure in healthy, overweight adults. Metabolism. (2012) 61:1026–35. 10.1016/j.metabol.2011.12.004
    1. Fujioka K, Greenway F, Sheard J, Ying Y. The effects of grapefruit on weight and insulin resistance: relationship to the metabolic syndrome. J Med Food. (2006) 9:49–54. 10.1089/jmf.2006.9.49
    1. Cristóbal-Luna JM, Álvarez-González I, Madrigal-Bujaidar E, Chamorro-Cevallos G. Grapefruit and its biomedical, antigenotoxic and chemopreventive properties. Food Chem Toxicol. (2018) 112:224–34. 10.1016/j.fct.2017.12.038
    1. Gattuso G, Barreca D, Gargiulli C, Leuzzi U, Caristi C. Flavonoid composition of citrus juices. Molecules. (2007) 12:1641–73. 10.3390/12081641
    1. Cassidy A, Rimm EB, O’Reilly ÉJ, Logroscino G, Kay C, Chiuve SE, et al. Dietary flavonoids and risk of stroke in women. Stroke. (2012) 43:946–51. 10.1161/STROKEAHA.111.637835
    1. Knekt P, Kumpulainen J, Järvinen R, Rissanen H, Heliövaara M, Reunanen A, et al. Flavonoid intake and risk of chronic diseases. Am J Clin Nutr. (2002) 76:560–8. 10.1093/ajcn/76.3.560
    1. Chang SC, Cassidy A, Willett WC, Rimm EB, O’Reilly EJ, Okereke OI. Dietary flavonoid intake and risk of incident depression in midlife and older women. Am J Clin Nutr. (2016) 104:704–14. 10.3945/ajcn.115.124545
    1. Barreca D, Gattuso G, Bellocco E, Calderaro A, Trombetta D, Smeriglio A, et al. Flavanones: citrus phytochemical with health-promoting properties. BioFactors. (2017) 43:495–506. 10.1002/biof.1363
    1. Chanet A, Milenkovic D, Manach C, Mazur A, Morand C. Citrus flavanones: what is their role in cardiovascular protection? J Agric Food Chem. (2012) 60:8809–22. 10.1021/jf300669s
    1. Pontifex MG, Malik MMAH, Connell E, Müller M, Vauzour D. Citrus polyphenols in brain health and disease: current perspectives. Front Neurosci. (2021) 15:115. 10.3389/fnins.2021.640648
    1. Jung UJ, Kim HJ, Lee JS, Lee MK, Kim HO, Park EJ, et al. Naringin supplementation lowers plasma lipids and enhances erythrocyte antioxidant enzyme activities in hypercholesterolemic subjects. Clin Nutr. (2003) 22:561–8. 10.1016/S0261-5614(03)00059-1
    1. Habauzit V, Verny MA, Milenkovic D, Barber-Chamoux N, Mazur A, Dubray C, et al. Flavanones protect from arterial stiffness in postmenopausal women consuming grapefruit juice for 6 MO: a randomized, controlled, crossover trial. Am J Clin Nutr. (2015) 102:66–74. 10.3945/ajcn.114.104646
    1. Chanet A, Milenkovic D, Deval C, Potier M, Constans J, Mazur A, et al. Naringin, the major grapefruit flavonoid, specifically affects atherosclerosis development in diet-induced hypercholesterolemia in mice. J Nutr Biochem. (2012) 23:469–77. 10.1016/j.jnutbio.2011.02.001
    1. Alam MA, Subhan N, Rahman MM, Uddin SJ, Reza HM, Sarker SD. Effect of citrus flavonoids, naringin and naringenin, on metabolic syndrome and their mechanisms of action. Adv Nutr. (2014) 5:404–17. 10.3945/an.113.005603
    1. Heidary Moghaddam R, Samimi Z, Moradi SZ, Little PJ, Xu S, Farzaei MH. Naringenin and naringin in cardiovascular disease prevention: a preclinical review. Eur J Pharmacol. (2020) 887:173535. 10.1016/j.ejphar.2020.173535
    1. Chanet A, Milenkovic D, Claude S, Maier JAM, Kamran Khan M, Rakotomanomana N, et al. Flavanone metabolites decrease monocyte adhesion to TNF-α-activated endothelial cells by modulating expression of atherosclerosis-related genes. Br J Nutr. (2013) 110:587–98. 10.1017/S0007114512005454
    1. Milenkovic D, Jude B, Morand C. miRNA as molecular target of polyphenols underlying their biological effects. Free Radic Biol Med. (2013) 64:40–51. 10.1016/j.freeradbiomed.2013.05.046
    1. Milenkovic D, Deval C, Gouranton E, Landrier JF, Scalbert A, Morand C, et al. Modulation of miRNA expression by dietary polyphenols in APOE deficient mice: a new mechanism of the action of polyphenols. PLoS One. (2012) 7:e29837. 10.1371/journal.pone.0029837
    1. Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, et al. The conduct of in vitro and in vivo drug-drug interaction studies: a PHRMA perspective. J Clin Pharmacol. (2003) 43:443–69. 10.1177/0091270003252519
    1. Stöckel D, Kehl T, Trampert P, Schneider L, Backes C, Ludwig N, et al. Multi-omics enrichment analysis using the genetrail2 web service. Bioinformatics. (2016) 32:1502–8. 10.1093/bioinformatics/btv770
    1. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. (2019) 47:D607–13. 10.1093/nar/gky1131
    1. Licursi V, Conte F, Fiscon G, Paci P. MIENTURNET: an interactive web tool for microRNA-target enrichment and network-based analysis. BMC Bioinformatics. (2019) 20:545. 10.1186/s12859-019-3105-x
    1. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, et al. ClueGO: a cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. (2009) 25:1091–3. 10.1093/bioinformatics/btp101
    1. Milenkovic D, Deval C, Dubray C, Mazur A, Morand C. Hesperidin displays relevant role in the nutrigenomic effect of orange juice on blood leukocytes in human volunteers: a randomized controlled cross-over study. PLoS One. (2011) 6:e26669. 10.1371/journal.pone.0026669
    1. Laura de la Garza A, Etxeberria U, Palacios-Ortega S, Haslberger AG, Aumueller E, Milagro FI, et al. Modulation of hyperglycemia and TNFα-mediated inflammation by helichrysum and grapefruit extracts in diabetic db/db mice. Food Funct. (2014) 5:2120–8. 10.1039/C4FO00154K
    1. Gamboa-Gómez C, Salgado LM, González-Gallardo A, Ramos-Gómez M, Loarca-Piña G, Reynoso-Camacho R. Consumption of Ocimum sanctum L. and Citrus paradisi infusions modulates lipid metabolism and insulin resistance in obese rats. Food Funct. (2014) 5:927–35. 10.1039/C3FO60604J
    1. Liu Y, Su WW, Wang S, Li PB. Naringin inhibits chemokine production in an LPS-induced RAW 264.7 macrophage cell line. Mol Med Rep. (2012) 6:1343–50. 10.3892/mmr.2012.1072
    1. Leray V, Freuchet B, Le Bloc’h J, Jeusette I, Torre C, Nguyen P. Effect of citrus polyphenol- and curcumin-supplemented diet on inflammatory state in obese cats. Br J Nutr. (2011) 106:S198–201. 10.1017/S0007114511002492
    1. Saenz J, Santa-María C, Reyes-Quiroz ME, Geniz I, Jiménez J, Sobrino F, et al. Grapefruit flavonoid naringenin regulates the expression of LXRα in THP-1 macrophages by modulating AMP-activated protein kinase. Mol Pharm. (2018) 15:1735–45. 10.1021/acs.molpharmaceut.7b00797
    1. Carlsson AC, Ruge T, Kjøller E, Hilden J, Kolmos HJ, Sajadieh A, et al. 10-year associations between tumor necrosis factor receptors 1 and 2 and cardiovascular events in patients with stable coronary heart disease: a claricor (effect of clarithromycin on mortality and morbidity in patients with ischemic heart disease) trial S. J Am Heart Assoc. (2018) 7:e008299. 10.1161/JAHA.117.008299
    1. Wingrove JA, Daniels SE, Sehnert AJ, Tingley W, Elashoff MR, Rosenberg S, et al. Correlation of peripheral-blood gene expression with the extent of coronary artery stenosis. Circ Cardiovasc Genet. (2008) 1:31–8. 10.1161/CIRCGENETICS.108.782730
    1. Kim HL, Lee JP, An JN, Kim JH, Lim WH, Seo JB, et al. Soluble tumor necrosis factor receptors and arterial stiffness in patients with coronary atherosclerosis. Am J Hypertens. (2017) 30:313–8. 10.1093/ajh/hpw134
    1. Martynowicz H, Janus A, Nowacki D, Mazur G. The role of chemokines in hypertension. Adv Clin Exp Med. (2014) 23:319–25. 10.17219/acem/37123
    1. Wan W, Murphy PM. Regulation of atherogenesis by chemokines and chemokine receptors. Arch Immunol Ther Exp (Warsz). (2013) 61:1–14. 10.1007/s00005-012-0202-1
    1. Yilma AN, Singh SR, Morici L, Dennis VA. Flavonoid naringenin: a potential immunomodulator for Chlamydia trachomatis inflammation. Mediators Inflamm. (2013) 2013:1–13. 10.1155/2013/102457
    1. Gökaslan S, Özer Gökaslan Ç, Demirel E, Çelik S. Role of aortic stiffness and inflammation in the etiology of young-onset hypertension. Turkish J Med Sci. (2019) 49:1748–53. 10.3906/sag-1908-137
    1. Gu L, Okada Y, Clinton SK, Gerard C, Sukhova GK, Libby P, et al. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor–deficient mice. Mol Cell. (1998) 2:275–81. 10.1016/S1097-2765(00)80139-2
    1. Ishibashi M, Hiasa K, Zhao Q, Inoue S, Ohtani K, Kitamoto S, et al. Critical role of monocyte chemoattractant protein-1 receptor CCR2 on monocytes in hypertension-induced vascular inflammation and remodeling. Circ Res. (2004) 94:1203–10. 10.1161/01.RES.0000126924.23467.A3
    1. Lee CH, Jeong TS, Choi YK, Hyun BH, Oh GT, Kim EH, et al. Anti-atherogenic effect of citrus flavonoids, naringin and naringenin, associated with hepatic ACAT and aortic VCAM-1 and MCP-1 in high cholesterol-fed rabbits. Biochem Biophys Res Commun. (2001) 284:681–8. 10.1006/bbrc.2001.5001
    1. Muller WA, Weigl SA, Deng X, Phillips DM. PECAM-1 is required for transendothelial migration of leukocytes. J Exp Med. (1993) 178:449–60. 10.1084/jem.178.2.449
    1. Pan Y, Yu C, Huang J, Rong Y, Chen J, Chen M. Bioinformatics analysis of vascular RNA-seq data revealed hub genes and pathways in a novel tibetan minipig atherosclerosis model induced by a high fat/cholesterol diet. Lipids Health Dis. (2020) 19:54. 10.1186/s12944-020-01222-w
    1. Nageh MF, Sandberg ET, Marotti KR, Lin AH, Melchior EP, Bullard DC, et al. Deficiency of inflammatory cell adhesion molecules protects against atherosclerosis in mice. Arterioscler Thromb Vasc Biol. (1997) 17:1517–20. 10.1161/01.ATV.17.8.1517
    1. Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res. (2020) 89:1619–26. 10.1038/s41390-020-01177-9
    1. Jaipersad AS, Lip GYH, Silverman S, Shantsila E. The role of monocytes in angiogenesis and atherosclerosis. J Am Coll Cardiol. (2014) 63:1–11. 10.1016/j.jacc.2013.09.019
    1. Spiering D, Hodgson L. Dynamics of the rho-family small GTPases in actin regulation and motility. Cell Adhes Migr. (2011) 5:170–80. 10.4161/cam.5.2.14403
    1. Soriano O, Alcón-Pérez M, Vicente-Manzanares M, Castellano E. The crossroads between RAS and RHO signaling pathways in cellular transformation, motility and contraction. Genes (Basel). (2021) 12:819. 10.3390/genes12060819
    1. Zhao Y, Qian Y, Sun Z, Shen X, Cai Y, Li L, et al. Role of PI3K in the progression and regression of atherosclerosis. Front Pharmacol. (2021) 12:632378. 10.3389/fphar.2021.632378
    1. Szelag M, Piaszyk-Borychowska A, Plens-Galaska M, Wesoly J, Bluyssen HAR. Targeted inhibition of STATs and IRFs as a potential treatment strategy in cardiovascular disease. Oncotarget. (2016) 7:48788–812. 10.18632/oncotarget.9195
    1. Loperena R, Van Beusecum JP, Itani HA, Engel N, Laroumanie F, Xiao L, et al. Hypertension and increased endothelial mechanical stretch promote monocyte differentiation and activation: roles of STAT3, interleukin 6 and hydrogen peroxide. Cardiovasc Res. (2018) 114:1547–63. 10.1093/cvr/cvy112
    1. Kopaliani I, Martin M, Zatschler B, Müller B, Deussen A. Whey peptide isoleucine-tryptophan inhibits expression and activity of matrix metalloproteinase-2 in rat aorta. Peptides. (2016) 82:52–9. 10.1016/j.peptides.2016.05.009
    1. Peng X, Haldar S, Deshpande S, Irani K, Kass DA. Wall stiffness suppresses Akt/eNOS and cytoprotection in pulse-perfused endothelium. Hypertension. (2003) 41:378–81. 10.1161/01.HYP.0000049624.99844.3D
    1. Zeng P, Yang J, Liu L, Yang X, Yao Z, Ma C, et al. ERK1/2 inhibition reduces vascular calcification by activating miR-126-3p-DKK1/LRP6 pathway. Theranostics. (2021) 11:1129–46. 10.7150/thno.49771
    1. Zhao C, Zhao C, Zhao H. Defective insulin receptor signaling in patients with gestational diabetes is related to dysregulated miR-140 which can be improved by naringenin. Int J Biochem Cell Biol. (2020) 128:105824. 10.1016/j.biocel.2020.105824
    1. Fan W, Shi R, Guan M, Chen P, Wu H, Su W, et al. The effects of naringenin on miRNA-mRNA profiles in HEPARG cells. Int J Mol Sci. (2021) 22:2292. 10.3390/ijms22052292
    1. Jamaluddin MS, Weakley SM, Zhang L, Kougias P, Lin PH, Yao Q, et al. miRNAs: roles and clinical applications in vascular disease. Expert Rev Mol Diagn. (2011) 11:79–89. 10.1586/erm.10.103
    1. Wang F, Long G, Zhao C, Li H, Chaugai S, Wang Y, et al. Atherosclerosis-related circulating miRNAs as novel and sensitive predictors for acute myocardial infarction. PLoS One. (2014) 9:e105734. 10.1371/journal.pone.0105734
    1. Mandolini C, Santovito D, Marcantonio P, Buttitta F, Bucci M, Ucchino S, et al. Identification of microRNAs 758 and 33b as potential modulators of ABCA1 expression in human atherosclerotic plaques. Nutr Metab Cardiovasc Dis. (2015) 25:202–9. 10.1016/j.numecd.2014.09.005
    1. Zhang W, Chang G, Cao L, Ding G. Dysregulation of serum miR-361-5p serves as a biomarker to predict disease onset and short-term prognosis in acute coronary syndrome patients. BMC Cardiovasc Disord. (2021) 21:74. 10.1186/s12872-021-01891-0
    1. Simionescu N, Niculescu LS, Sanda GM, Margina D, Sima AV. Analysis of circulating microRNAs that are specifically increased in hyperlipidemic and/or hyperglycemic sera. Mol Biol Rep. (2014) 41:5765–73. 10.1007/s11033-014-3449-2
    1. Tiedt S, Prestel M, Malik R, Schieferdecker N, Duering M, Kautzky V, et al. RNA-Seq identifies circulating miR-125a-5p, miR-125b-5p, and miR-143-3p as potential biomarkers for acute ischemic stroke. Circ Res. (2017) 121:970–80. 10.1161/CIRCRESAHA.117.311572
    1. Rafiei A, Ferns GA, Ahmadi R, Khaledifar A, Rahimzadeh-Fallah T, Mohmmad-Rezaei M, et al. Expression levels of miR-27a, miR-329, ABCA1, and ABCG1 genes in peripheral blood mononuclear cells and their correlation with serum levels of oxidative stress and hs-CRP in the patients with coronary artery disease. IUBMB Life. (2021) 73:223–37. 10.1002/iub.2421
    1. Welten SMJ, Bastiaansen AJNM, de Jong RCM, de Vries MR, Peters EAB, Boonstra MC, et al. Inhibition of 14q32 MicroRNAs miR-329, miR-487b, miR-494, and miR-495 increases neovascularization and blood flow recovery after ischemia. Circ Res. (2014) 115:696–708. 10.1161/CIRCRESAHA.114.304747
    1. van Ingen E, Foks AC, Kröner MJ, Kuiper J, Quax PHA, Bot I, et al. Antisense oligonucleotide inhibition of MicroRNA-494 halts atherosclerotic plaque progression and promotes plaque stabilization. Mol Ther Nucleic Acids. (2019) 18:638–49. 10.1016/j.omtn.2019.09.021
    1. Li Q, Liu J, Jia Y, Li T, Zhang M. miR-623 suppresses cell proliferation, migration and invasion through direct inhibition of XRCC5 in breast cancer. Aging (Albany NY). (2020) 12:10246–58. 10.18632/aging.103182
    1. Valkonen VP, Tuomainen TP, Laaksonen R. DDAH gene and cardiovascular risk. Vasc Med. (2005) 10:S45–8. 10.1191/1358863x05vm600oa
    1. Evangelou E, Warren HR, Mosen-Ansorena D, Mifsud B, Pazoki R, Gao H, et al. Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat Genet. (2018) 50:1412–25. 10.1038/s41588-018-0205-x
    1. He L, Kernogitski Y, Kulminskaya I, Loika Y, Arbeev KG, Loiko E, et al. Corrigendum: pleiotropic meta-analyses of longitudinal studies discover novel genetic variants associated with age-related diseases. Front Genet. (2018) 8:226. 10.3389/fgene.2017.00226
    1. Li L, Lyall GK, Martinez-Blazquez JA, Vallejo FA, Tomas-Barberan F, Birch KM, et al. Blood orange juice consumption increases flow-mediated dilation in adults with overweight and obesity: a randomized controlled trial. J Nutr. (2020) 150:2287–94. 10.1093/jn/nxaa158

Source: PubMed

3
Abonner