Skip to main content

Homocysteine, blood pressure and gene–diet interactions in relation to vascular function measures of black South Africans

Abstract

Background and Aims

We investigated circulating homocysteine (Hcy), a cardiovascular disease (CVD) risk factor, examining its dietary associations to provide personalized nutrition advice. This study addressed the inadequacy of current dietary interventions to ultimately address the disproportionately high incidence of CVD in Black populations. Methods and Results: Cross-sectional analyses of 1,867 Black individuals of the PURE-SA study allowed the identification of dietary intake and cardiovascular measure interactions on three sub-categories: (1) normal blood pressure (BP), hypertension or Hcy-related hypertension (H-type), (2) low, normal or high Hcy concentrations, and (3) Hcy-related genetic combinations. Favorable body composition, but adverse dietary intake and cardiovascular determinants, were observed in higher Hcy categories. H-types, compared to regular hypertensives, had higher alcohol and lower macronutrient and micronutrient consumption. Inverse associations with carotid-radial pulse wave velocity were evident between monounsaturated fatty acid (FA) consumption and H-type hypertension as well as polyunsaturated FA and CBS883/ins68 TT carriers. Energy intake was positively associated with vascular cell adhesion molecule-1 (VCAM-1) in variant CBST883C/ins68 and CBS9276 GG carriers. VCAM-1 was also positively associated with plant protein intake in CBS9276 GG and MTR2756 AA carriers and negatively with total protein intake and CBS9276 GG carriers. Alcohol intake was positively associated with intercellular adhesion molecule-1 in MTR2756 minor allele carriers. Conclusion: Because Hcy gene-diet interactions are evident, personalized nutrition, by adjusting diets based on genetic profiles (e.g., CBS and MTR variations) and dietary interactions (e.g., FAs and proteins), can enhance cardiovascular outcomes by managing Hcy and related hypertension in genetically susceptible individuals.

Introduction

Cardiovascular disease (CVD) is a leading cause of death in South Africa, with one in six deaths attributed to it [1]. High blood pressure (BP) is a primary risk factor for CVD. Sub-Saharan Africa has higher hypertension prevalence and lower BP control rates than other regions [2], with cardiovascular events occurring about 15 years earlier [3]. National surveys in South Africa reported hypertension rates of 38.4% in 2012 (SANHANES) and 48.2% in 2016 (DHS) [4].

Homocysteine (Hcy) is a nonessential amino acid in a vitamin-regulated, one-carbon methylation pathway. Elevated circulating Hcy concentrations – representing hyperhomocysteinemia (HHcy) – not only impact health adversely [5] but are associated with increased all-cause mortality risk, with each 5 µmol/l rise escalating rates by 34% [6]. HHcy is recognized as an independent risk factor for hypertension and CVD [7], inducing arterial wall structural and functional changes thereby influencing arterial stiffness [8]. However, data on HHcy’s effects among the Black South African population are scarce. Determining HHcy’s role in the heightened mortality, uncontrolled hypertension, and CVD in developing countries such as South Africa, also struggling with nutritional issues [9], is critical.

Diet is a modifier of Hcy concentrations. Determinants encompass protein, B vitamins, vegetables, fruits, omega 3 and 6 fatty acids, pulses, nuts, seeds and alcohol consumption, as previously reported [10,11,12]. A diet impacting Hcy through these determinants suggests potential disease-prevention strategies [13, 14]. Whether diet could be a possible means of influencing HHcy – and by extension cardiovascular function and inflammation – should be investigated while also considering genetic factors.

Several single nucleotide polymorphisms (SNPs) in the vitamin-regulated methylation pathway are relevant to Hcy. For our investigation, we included candidate SNPs known to impact this pathway. The methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism (rs1801133) was selected because it is known to reduce enzyme activity, impairing methionine metabolism and leading to elevated Hcy concentrations. In addition, we included the cystathionine β synthase (CBS) T833C/844ins68 (rs5742905) and G9276A (a novel SNP) polymorphisms due to their roles in Hcy clearance. Variants in the CBS gene are critical for the transsulfuration pathway, which helps to lower Hcy concentrations by converting it to cystathionine. The methionine synthase (MTR) A2756G (rs1805087) polymorphism was also analyzed because it influences the remethylation pathway, promoting the conversion of Hcy to methionine and thus affecting Hcy concentrations.

The etiology of hypertension and CVD remains unclear, highlighting the need for research on Hcy-related genes and their interaction with environmental factors, especially diet, in developing these conditions. This focus is crucial, as diet is one of the few easily modifiable factors. The aim of this study was to advance the understanding of how Hcy interacts with CVD risk markers overall but also in respect of the genetic make-up of individuals with the benefit of precision nutrition. To this end, we used arterial stiffness as indicated by carotid-radialis pulse wave velocity (cr-PWV), endothelial activation [indicated by circulating intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1)] and, also, pulse pressure (PP), heart rate (HR), systolic (SBP), and diastolic BP (DBP) as outcome variables when investigating interactions with Hcy subdivisions and manifesting genotypes for MTHFR C677T, MTR A2756G and CBS T833C/844ins68 and G9276A. This work underscores the importance of including Hcy in hypertension screening and suggests personalized dietary advice for managing CVD risk among Black South Africans, considering individual Hcy concentrations, hypertension status and specific Hcy SNPs. This work highlights the potential of personalized dietary advice, based on individual Hcy concentrations and specific genetic profiles. This approach could possibly improve the management of chronic conditions, particularly CVD risk and improve overall health outcomes in populations vulnerable to hypertension and cardiovascular issues.

Materials and methods

Study design and participants

This cross-sectional study utilized baseline data from the South African arm of the Prospective Urban and Rural Epidemiology (PURE–SA) study, part of a multinational cohort across 17 countries investigating risk factors for chronic diseases in transitioning communities [15]. Following an eligibility screening of 6,000 homes, Tswana-speaking South Africans over 35 from specific North West province areas, deemed healthy and not pregnant or lactating, were randomly selected for participation. Volunteers were informed in the language of their choice and required to provide signed informed consent before any data were acquired. Participants could withdraw or refuse any measurement at any stage of the study without consequence. For this investigation, only individuals who had BP and Hcy data were included for analyses (n = 1,867). North-West University’s Health Research Ethics Committee approved the PURE–SA and sub-study protocols (ethics numbers: 04M10 and NWU-00142-18-A1), adhering to the Declaration of Helsinki principles, ensuring voluntary participation, anonymity and confidentiality.

Questionnaires

Data on demographics and lifestyle were collected through interviews using standardized questionnaires by fieldworkers [15]. Food intake was assessed with quantitative food frequency questionnaires (QFFQs), validated against 7-day weighed food records and biomarkers [16], supplemented by food portion photograph books for accurate portion size reporting. QFFQ data were logged using FoodFinder3® [South African Medical Research Council (SAMRC), Tygerberg, 2007] and analyzed for nutrients by the SAMRC. Dietary supplement use was negligible and excluded from micronutrient intake calculations.

Anthropometric measures

Anthropometric measurements, including height (IP 1465, Invicta, London, UK), weight (Precision Health Scale, A&D Company, Tokyo, Japan), hip (HC) and waist circumference (WC) (Lufkin steel tape, Cooper Tools, Apex, NC, USA), were obtained with calibrated instruments following the International Society for the Advancement of Kinanthropometry (ISAK) guidelines [17]. Body mass index (BMI) and waist-to-hip ratio (WHR) were calculated from height, weight (kg/m2), and circumferences, respectively.

Cardiovascular marker measurements

To ensure accuracy under standardized conditions [18], participants were instructed to avoid exercise, smoking, and caffeine use prior to measurements, and to observe a 5-minute rest. An Omron HEM-757 device (Omron Healthcare, Kyoto, Japan) measured brachial SBP, DBP, HR, and calculated PP in seated participants with their right arm at heart level. cr-PWV was assessed in a supine position on the left side using a Complior SP device (Artech-Medical, Pantin, France). Hypertension was diagnosed at BP ≥ 140/90 mmHg or with anti-hypertensive medication use [18, including thiazide diuretics, calcium channel blockers, and renin–angiotensin system inhibitors. H-type hypertension was identified in hypertensive individuals with Hcy levels ≥ 10 µmol/l [19].

Laboratory biochemical measurements

Volunteers fasted overnight before a registered nurse collected blood samples from the antecubital vein before 11:00. Samples were centrifuged at 2,000 × g for 15 min to separate serum, plasma, and buffy coat, then stored at − 80 °C. Total plasma Hcy was measured using fluorescence polarization immunoassay [CV = 4.52%] on an Abbott immunoassay analyzer (AxSYM, Abbott Laboratories, Abbott Park, IL, USA). Sequential multiple analyzers from Konelab 20i (Thermo Scientific, Vantaa, Finland) and Cobas Integra 400 Plus (Roche, Basel, Switzerland) analyzed biomarkers including high-sensitivity C-reactive protein (hsCRP), gamma-glutamyl transferase (GGT) and all blood lipids [high-density lipoprotein cholesterol (HDL-c), total cholesterol (TC) and triglycerides], with low-density lipoprotein cholesterol (LDL-c) calculated via the Friedewald–Levy–Fredrickson formula [20]. Fasting plasma glucose was determined by the SynchronR System (Beckman Coulter Co., Fullerton, CA, USA) hexokinase method, and fasting glycated hemoglobin A1c (HbA1c) by the Bio-Rad Laboratories (Hercules, CA, USA) D-10 system. Sandwich ELISAs (human sICAM-1 and human sVCAM-1 assay, IBL, Hamburg, Germany) measured serum ICAM-1 and VCAM-1 levels. Whole blood tested for HIV with the First Response HIV 1-2-0 test (Premier Medical Corporation Ltd, Daman, India), confirmed by the Pareeshak card test (BHAT Bio-tech, Bengaluru, India) if positive. Participants on antiretroviral medication were not invited to partake, and those unaware of their HIV status were tested on the same day, ensuring unmedicated status in the analyzed variables even among seropositive individuals.

Genomic DNA was extracted from peripheral blood leukocytes using QIAGEN®, Flexigene® DNA kits (QIAGEN® Valencia, CA, USA; catalog number 51 206) and quantified by NanoDrop™ spectrophotometry (ND-1000, Wilmington, DE, USA). Five Hcy-related SNPs—MTHFR C677T (rs1801133), CBS T833C (rs5742905), CBS 844ins68 (indel, no rs#), CBS G9276A (novel SNP, no rs#) and MTR A2756G (rs1805087)—were genotyped using PCR and restriction fragment length polymorphism methods [21]. Sample DNA batches were analyzed with positive and negative controls to prevent misinterpretation from digestion issues or contamination. MTHFR and MTR genotypes were identified by analyzing enzyme-digested PCR fragments via electrophoresis on an ethidium bromide-stained 2% agarose gel. For CBS alterations, both undigested and digested PCR products were examined. Two independent researchers, blinded to the Hcy phenotype, scored the genotypes and merged their results. Discrepancies were resolved by reanalyzing the genotypes. For primers and digestion enzymes used, see Supplementary Table 1.

Statistical analysis

Statistica version 14.0 (TIBCO Software Inc., Tulsa, OK, USA) and R version 4.2.0 (R Foundation for Statistical Computing, Vienna, Austria; 2020) were used for data analysis. Shapiro–Wilk W and Kolmogorov–Smirnov tests assessed normality. Descriptive statistics for non-normal data were presented as medians and interquartile ranges, and for normal data as means ± standard deviations. Normal and partial Spearman rank correlations (adjusting for age, WHR, HDL-c, and GGT) assessed Hcy’s relationship with dietary intake. Mann–Whitney U tests compared dependent continuous variables across three groups (normal BP vs. hypertension, normal BP vs. H-type hypertension, hypertension vs. H-type hypertension). General linear models (GLMs) identified differences among low (< 7.0 µmol/l), normal (7.0–12.0 µmol/l), and high (> 12.0 µmol/l) Hcy categories [22].

To explore associations between CVD markers, dietary components, Hcy categories, BP categories, and Hcy-related genetic polymorphisms, factorial GLMs, adjusted for age, assessed interaction effects. The CBS T833C and CBS 844ins68 SNPs, being in complete linkage, were reported as CBS T833C/844ins68 [21]. Heterozygotes and homozygotes for variant alleles were combined following the dominant genetic mode. Partial Spearman correlations, adjusted for age and added sugar, analyzed relationships between CVD markers, dietary intake, BP sub-groups, Hcy sub-groups, and genotypes using this genetic coding.

For general analyses, P ≤ 0.05 indicated statistical significance. To address multiple testing and false discovery rates, Hochberg and Benjamini’s method was applied. With a false discovery rate of 25%, considering 4 SNPs (CBS 833 SNP linked with 844ins68), subdivisions as 1, dietary intake categories as 5, and cardiovascular markers as 1 due to associations with dietary factors, the significance threshold was set at P < 0.01 [1/((4 + 1)51))(0.25) = 0.01].

Results

Characteristics compared across Hcy sub-groups and correlations with Hcy

The characteristics and differences across Hcy sub-groups are detailed in Supplementary Table 2. Most of the recruits presented with normal Hcy concentrations (56.7%), whereas 18.3% demonstrated low and 25.0% high levels. Age, HDL-c, and GGT increased over rising Hcy sub-groups. Furthermore, certain body composition variables were more favorable (weight, BMI, and WC decreased; HC and WHR increased) over sub-groups with increasing Hcy concentrations, these associations attenuated after adjusting for sex. A quarter of HIV-seroposiive individuals were in the low-Hcy category, whereas a quarter of those without the virus presented with high Hcy, resulting in frequency differences among the Hcy sub-groups. However, Hcy concentrations did not differ between those with or without HIV (P = 0.99). Alcohol intake was greater in the high-Hcy category (17.2 g/d) – more than double that of the low-Hcy category intake (8.14 g/d); this association remained after adjustments. Energy intake, total protein, animal protein, added sugar, and fat intake (including SFAs, MUFAs, PUFAs, omega-6 FAs, omega-3 FAs and cholesterol) were lower in the highest-Hcy category. Fruit and vegetable intake and B vitamins (biotin, pantothenic acid, vitamin B6, and vitamin B12) were also lowest in the high-Hcy group. For biotin and vitamin B6, those presenting with normal and high Hcy and for vitamin B12 those with high Hcy, respectively, did not meet the recommended daily allowances (RDAs) of 30 µg, 1.3–1.5 mg and 2.4 µg, respectively. For pantothenic acid, none of the Hcy sub-groups met the RDA of 5 mg, with the intake values declining over increasing Hcy sub-groups. Even though folate, niacin and riboflavin intake ranges did not differ among the Hcy sub-groups, no sub-group’s intake met the RDA of 400 µg, 1.1–1.3 mg and 14.0–16.0 mg, respectively, and tended to be lower over increasing Hcy sub-groups. Of the cardiovascular measures, SBP, DBP, PP, HR, and cr-PWV differed among the Hcy sub-groups, being higher in the HHcy sub-group with DBP and cr-PWV exceeding normal healthy ranges [18, 23].

Supplementary Table 3 shows correlations between Hcy and various markers. Initially, Hcy positively correlated with age, WHR, HDL-c, GGT, some cardiovascular markers, and alcohol intake (all r ≥ 0.15), and negatively with most dietary intake markers (r ≤ 0.15). In model 2, after adjusting for age, WHR, HDL-c, and GGT, excluding collinear variables, positive correlations remained for age, HDL-c, GGT and added sugar’s percentage of total carbohydrate intake (all r ≥ 0.15). Post-adjustment, only Hcy and HR showed a weak correlation among cardiovascular markers.

Daily dietary intake compared among blood pressure sub-groups

To explore the association between Hcy as well as its dietary determinants and BP as a cardiovascular risk factor, sub-groups for blood pressure (BP) were created including a group for H-type hypertension; differences are noted in Table 1. While there is an age difference between normal BP and regular hypertension groups, it is not present between normal BP and H-type hypertension. Those with regular hypertension also presented with B-vitamin intake ranges meeting the RDAs except for folate and pantothenic acid, whereas those with H-type hypertension did not satisfy the RDAs except for thiamin and vitamin B12. The difference between the hypertension vs. H-type hypertension groups’ energy intake was not attributed to age, weight, GGT, alcohol, added sugar and carbohydrates alone or combined, but due to differences in either total protein or fat intake. Genotype frequency differences among BP subgroups are detailed in another study [24].

Table 1 Daily dietary intake of participants compared among blood pressure sub-groups

Interactions with dietary intake in relation to cardiovascular markers

In Tables 2, 3 and 4 the interactions (P ≤ 0.01) between the BP sub-groups, Hcy sub-groups or Hcy-related SNPs and dietary intake are presented in relation to cardiovascular markers.

Table 2 Blood pressure category interactions with markers of nutritional status in relation to markers of cardiovascular function
Table 3 Hcy categories interactions with markers of nutritional status in relation to markers of cardiovascular function
Table 4 MTHFR, MTR and CBS gene polymorphisms’ interactions with markers of nutritional status in relation to markers of cardiovascular function

BP sub-group interactions

For those within the normal BP range, cholesterol intake had a positive relationship with SBP. Furthermore, dietary vitamin B12 intake had a protective inverse relationship with DBP in those with hypertension. Total protein, as a percentage of energy, linearly associated with PP in hypertensives and H-type hypertensives; animal protein showed a stronger positive correlation with PP in H-type hypertensives than in other BP categories, but inversely with cr-PWV in normal BP individuals. Consumption of SFAs showed negative associations with cr-PWV across all BP subdivisions, with H-type hypertension manifesting a slightly stronger correlation. MUFAs intake also displayed a negative linear association between cr-PWV in those with normal BP and H-type hypertension, and borderline associations with the hypertension groups as well. All correlations with BP sub-groups, except those with MUFAs intake in normotensives, became inconsequential after adjusting for age; after additionally adjusting for added sugar intake, this association became borderline as well.

Hcy sub-group interactions

Those presenting with Hcy concentrations within the normal ranges had a borderline positive association with biotin and a positive association between fruit and vegetable intake in relation to ICAM-1, whereas no association was observed in the other sub-groups. Partial Spearman correlations, after adjusting for age and then age plus added sugar, revealed stronger biotin associations and consistent positive relations with fruit/vegetable intake. Yet, significance vanished after adjusting interactions for added sugar.

Hcy-related polymorphism interactions

Added sugar displayed a nuanced relationship with cr-PWV among MTHFR genotypes, showing a stronger negative association in variant allele carriers compared to wildtype homozygotes at the 677 locus. Adjusting for fruit/vegetable intake and age nullified the significance.

CBS T883C/ins68 TT genotype carriers showed a notable inverse correlation with cr-PWV and PUFAs intake, not altered by age or added sugar, strengthening post-adjustment. In individuals with the TT genotype of CBS T883C/ins68, there was a positive correlation between thiamine intake and cr-PWV, while no significant correlation was observed in individuals with other genotypes After adjusting for added sugar, the interaction and sub-group correlations with thiamine were no longer significant. In those harboring the variant allele of CBS T883C/ins68, there was a positive linear association, even after additional adjustment, between energy consumption and VCAM-1, whereas in those harboring the wildtype allele no association was observed. The homozygote major G allele carriers of the CBS 9276 genotype also had strong positive correlations between VCAM-1 and energy as well as plant protein consumption only after adjusting for age and added sugar intake.

For MTR A2756G homozygote AA genotypes, a negative linear association between cr-PWV and SFAs intake persisted after adjusting for fruit and vegetable intake but disappeared with PUFAs adjustment. Age and added sugar adjustments further attenuated correlations. The minor G allele carriers exhibited a positive association between ICAM-1 and alcohol intake only after adjusting for age and added sugar. Total, plant proteins, vitamin B6, and folate intake positively correlated with VCAM-1 in AA genotypes. For heterozygotes and GG carriers, folate showed the strongest positive correlation with VCAM-1. Post-adjustment for age and added sugar, protein and folate associations persisted, while vitamin B6’s became borderline.

Discussion

In this study we considered diet interactions across three sub-groups: (1) normal BP, hypertension, and Hcy-related hypertension (H-type); (2) low, normal, and high Hcy levels; and (3) various Hcy-related genotypes. We observed that those with HHcy expressed more favorable energy and macronutrient intake and body composition measures than those with normal and low-Hcy concentrations. However, hyperhomocysteinemic individuals reported noticeably higher alcohol and lower fruit and vegetable intake, possibly contributing to their inadequate vitamin status and higher CVD measures. There were more men than women in the HHcy category and, after accounting for sex, the body composition associations disappeared, although differences in alcohol and diet intake remained noteworthy. Moreover, 9 in 10 of those with HHcy also presented with hypertension (H-type) and when compared with the regular hypertension sub-group, H-type hypertensives recorded higher alcohol intake and lower fruit and vegetable consumption, including lower folate and other B vitamins intake, than those with hypertension alone. To complement our investigation, we report below on specific dietary aspects of BP categories, Hcy categories and Hcy-related genetic characteristics that associate with cardiovascular functional measures and CVD risk in a population already at a high risk.

Blood pressure categories, dietary intake and cardiovascular function markers

BP measurement, an affordable, non-invasive tool, helps healthcare professionals detect and manage chronic conditions and is a step that could complement personalized nutrition. Yet, guidelines overlook H-type hypertension, which could significantly impact hypertension management and precision nutrition. We advise everyone, particularly individuals with hypertension, to consume adequate vitamin B12. Research on the direct effects of vitamin B12 intake and BP are scarce [25]; the most plausible link is through the Hcy folate–vitamin B12 dependent and independent remethylation metabolism, in which a reduced vitamin B12 intake results in higher Hcy concentrations and thereby raised hypertension and CVD risk.

We also advise individuals with regular and H-type hypertension to monitor their total protein intake, as it may raise PP, especially advising caution with animal protein for those with H-type, given PP’s significance in CVD risk [26]. The 10-year Zutphen study on Dutch middle-aged men showed no link between PP and dietary protein intake [26]. Conversely, a study of younger (< 30 years) white and Black South Africans revealed that despite lower total and animal protein intake among Blacks, they had higher serum protein levels [27]. The authors suggested this could stem from heightened protein catabolism or enhanced endogenous amino acid biosynthesis, potentially increasing collagen to mitigate early vascular aging risk. Given our older hypertensive and H-type participants, they might experience elevated PP from higher protein consumption. However, in individuals with normal BP, increased animal protein intake seemingly did not adversely affect arterial function.

MUFA consumption was positively associated with arterial function in all BP sub-groups, particularly in normal and H-type hypertension. The negative correlations between SFA intake and cr-PWV may stem from MUFAs and SFAs collinearity, as adjusting for MUFAs intake weakened these associations. Despite mixed evidence on MUFAs and vascular health, beneficial impacts on serum lipids, BP, and E-selectin suggest a lower CVD risk [28]. Further intervention studies are essential to substantiate MUFA intake recommendations and elucidate underlying mechanisms.

Hcy categories, daily dietary intake and markers of cardiovascular function

Hcy measurements are more invasive and elective than recording BP but should be regarded as a necessary step in the approach to personalized nutrition, particularly in cardiovascular health. Incorporating Hcy screening into hypertension guidelines could differentiate between regular and H-type hypertension for targeted treatment.

Biotin, known for immunoregulatory properties affecting proinflammatory cytokine expression, which in turn induces ICAM-1 and VCAM-1 expression [29], showed positive correlations with Hcy in CBS T833C minor allele carriers [10]. However, research to confirm a direct relationship between dietary biotin and ICAM-1 remains to be done. We revealed that the positive associations between ICAM-1 and biotin get stronger when accounting for age more so than sugar in those with normal Hcy concentrations. It seems that the association between ICAM-1 and fruit and vegetable intake in the same Hcy group was unaffected by either. Notably, biotin and fruit/vegetable consumers ingested more added sugar and significance diminished after adjusting for sugar interactions.

Fructose induces ICAM-1 expression by diminishing endothelial nitric oxide and depleting adenosine triphosphate, leading to vascular cell inflammation [30]. Habitually high intake of carbohydrates, simple sugars, and fructose in particular, contributes to insulin resistance, weight gain and increased BP [31]. Some studies speculate that insulin resistance also produces a drop in methionine transmethylation, hepatic Hcy trans-sulphuration and Hcy clearance, resulting in raised concentrations of circulating Hcy [32, 33]. In Black South Africans, higher socio-economic status improves micronutrient intake through better access to fruits, vegetables, pulses, nuts, and seeds [15, 34] but also raises added sugar and SFA consumption, heightening non-communicable disease risks like CVD [35]. We, therefore, advise an adequate intake of fruit and vegetables, but caution against the accompanying consumption of added sugar.

Hcy-related genetic SNPs, daily dietary intake and markers of cardiovascular function and inflammation

Incorporating genomic testing into CVD and hypertension guidelines could enhance personalized nutrition strategies, emphasizing the role of Hcy-related genetic polymorphisms in dietary impacts on cardiovascular health, particularly for H-type hypertension sufferers.

The relationship between sugar, SFAs and fruit and vegetable intake could explain the disappearance of the inverse association between added sugar and cr-PWV in variant allele carriers, which was stronger than in those homozygous for the wildtype allele at the MTHFR 677 locus, after accounting for fruit and vegetable intake. It could also help clarify why, after adjusting for sugar intake, the folate and VCAM-1 relationships disappeared in those carrying the MTR 2756 minor A genotype. The same can be said for positive associations betweenVCAM-1 and MTR 2756 major allele carriers for vitamin B6 intake. Lastly, the intake of thiamine, a precursor of coenzymes in sugar and amino acid catabolism, led to higher cr-PWV in the TT carriers CBS T883C/ins68 with the interactions disappearing after adjusting for added sugar. It seems therefore that an improved micronutrient status should be accompanied by a balanced, prudent diet of lower added sugar and fructose intake.

cr-PWV was inversely associated with lower PUFA intake in those harboring the TT genotype of CBS T883C/ins68. A previous intervention study reported that daily supplementation of 4,000 mg of omega-3 for 12 weeks decreased carotid-femoral PWV in older, but not younger, healthy adults independent of the effects on BP or arterial wave reflections [36]. The mechanism by which omega-3 PUFAs influence endothelial function (PWV) is mediated by their integration into the phospholipids of biological membranes, so affecting their fluidity. This interaction leads omega-3 PUFAs to bring about their beneficial effects such as increased nitric oxide production and reduced expression of proinflammatory mediators [37]. Despite the study population’s unusually low intake of omega-3 PUFAs, the beneficial implications should not be disregarded. A longitudinal PURE-SA study also found an inverse relationship between omega-6 PUFAs, SBP, and DBP [38], suggesting protective effects may highlight omega-6 long-chain products’ role in physiological functions among those with limited omega-3 consumption. The inverse association between SFAs intake and cr-PWV in those harboring MTR 2756AA could possibly be explained by the accompanying PUFAs intake.

Energy consumption was positively associated with VCAM-1 in CBS T883C/ins68 minor allele carriers and CBS 9276 homozygote major allele carriers. However, after adjustment for total carbohydrate intake the interaction disappeared, hinting at a potential link between energy from carbohydrates. Data on the relationship between energy intake and VCAM-1 are scarce. The additional adjustment for total carbohydrate intake that negated the interaction could be explained by the release of VCAM-1 in a dose-dependent manner when energy from carbohydrates increased glucose levels, as illustrated in a previous in vitro experiment using different doses of glucose in human umbilical vein endothelial cells (HUVECs) [39].

Positive linear associations between plant protein intake and VCAM-1 were observed in those harboring homozygote major alleles for CBS 9276 and/or MTR 2756, associations that remained after adjustments. However, an inverse relationship was observed for total protein intake and the MTR 2756 major homozygote AA carriers, suggesting a beneficial relationship. This is supported by previous studies that reported on animal and plant protein intake that decreased VCAM-1 expression in both unstimulated and stimulated HUVECs [40] as well as vascular endothelial cells [41]. The type of protein and concomitant nutrients could contribute to some of the different outcomes observed here.

Recommendations for precision nutrition

International nutrition and hypertension guidelines offer general recommendations to prevent and manage cardiovascular risk [18, 42]. Although most guidelines are proposed for application across the larger population, and do not currently account for Hcy or H-type hypertension, our personalised nutrition approach still supports and fits within the following general guidelines: (1) lowered sugar intake; (2) sufficient intake of fruit and vegetables; (3) moderate intake of animal products; and (4) an adequate intake of healthy fats. We found that added sugar intake should be limited in those with seemingly normal Hcy or those carrying the T allele at MTHFR 677, to avoid possible detrimental associations with vascular activation (ICAM-1) and vascular function (cr-PWV). Reducing energy and sugar intake benefits CBS T883C/ins68 minor allele and CBS 9276 GG homozygote carriers regarding endothelial activation. Our results endorse consuming fruits and vegetables for their vitamin content, highlighting vitamin B12’s role in DBP management and meeting the RDA for those with hypertension. Achieving optimal vitamin status, particularly through fruits and vegetables, supports arterial health. Animal products, high in cholesterol and SFAs, should be consumed sparingly to manage SBP and PP, with a particular focus on animal protein for H-type hypertensives. Carriers of the MTR 2756 AA and CBS 9276 GG genotypes, on the other hand, should be sensible when consuming plant protein – lower rather than higher consumption could be beneficial in terms of vascular-related inflammation and atherosclerosis. Lastly, our results support an adequate intake of healthy fats such as PUFAs and MUFAs. Sufficient consumption of PUFAs is advised for carriers of the TT genotype of CBS T883C/ins68, owing to their positive influence on cr-PWV. We encourage also the intake of MUFAs within the normal ranges for all BP groups to ensure optimal cr-PWV and arterial wall function. Furthermore, dietary management is an alternative treatment avenue that could be explored for those who are resistant to traditional hypertensive drugs.

Limitations and strengths

The study’s large population allowed detection of changes in cardiovascular function and gene–diet interactions, but a larger sample could better account for low-frequency SNPs. Due to its cross-sectional design, only associations were observed without causal inference. Recall bias in dietary assessment via the QFFQ may lead to under- or overreporting. For more precise results, we suggest using biochemical vitamin measures to capture absorption, metabolism and bioavailability, which dietary questionnaires overlook. Future research should conduct dietary supplementation trials by genotype and BP sub-groups to test their effectiveness. Additional cardiovascular markers such as carotid-femoral PWV, a “gold standard” measure for arterial stiffness, and intima media thickness should also be included in future research.

Conclusions

This study enhances our understanding of Hcy-related nutrigenetics and cardiovascular health in Black South Africans, a population where gene–diet interactions with Hcy, BP, and cardiovascular markers are underexplored. Our findings demonstrate that dietary intake is associated with Hcy levels, BP groupings, and specific CVD risk markers, particularly when considering SNPs. We identified significant differences between regular hypertension and H-type hypertension, with the latter group exhibiting higher alcohol intake and lower consumption of macronutrients and micronutrients. If the gene-diet interactions reported here are replicated in further studies, personalized nutrition strategies, informed by genetic profiles and dietary interactions (e.g., fatty acids and proteins), could hold promise for improving cardiovascular outcomes.

Data availability

The data that support the findings of this study are available upon reasonable request and with the permission of the Health Research Ethics Committee of the North-West University and the principal investigator of the PURE-SA-NW study, Prof. Cristian Ricci (cristian.ricci@nwu.ac.za) at the North-West University’s Africa Unit for Transdisciplinary Health Research.

References

  1. Mortality and causes of death in South Africa. 2014. Findings from death notification/Statistics South Africa.

  2. Yuyun MF, Sliwa K, Kengne AP, Mocumbi AO, Bukhman G. Cardiovascular diseases in sub-saharan Africa compared to high-income countries: an epidemiological perspective. Global Heart. 2020;15(1):15. https://doi.org/10.5334/gh.403.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Yoruk A, Boulos PK, Bisognano JD. The state of hypertension in Sub-saharan Africa: review and commentary. Am J Hypertens. 2018;31:387–8. https://doi.org/10.1093/ajh/hpx196.

    Article  PubMed  Google Scholar 

  4. Kandala N-B, Nnanatu CC, Dukhi N, Sewpaul R, Davids A, Reddy SP. Mapping the burden of hypertension in South Africa: a comparative analysis of the national 2012 SANHANES and the 2016 demographic and Health Survey. Int J Environ Res Public Health. 2021;18:5445. https://doi.org/10.3390/ijerph18105445.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Smith AD, Refsum H. Homocysteine–from Disease Biomarker to Disease Prevention. J Intern Med. 2021;90:826–54. https://doi.org/10.1111/joim.13279.

    Article  CAS  Google Scholar 

  6. Fan R, Zhang A, Zhong F. Association between homocysteine levels and all-cause mortality: a dose-response Meta-analysis of prospective studies. Sci Rep. 2017;7:1–9. https://doi.org/10.1038/s41598-017-05205-3.

    Article  CAS  Google Scholar 

  7. Zhong F, Zhuang L, Wang Y, Ma Y. Homocysteine levels and risk of essential hypertension: a Meta-analysis of published epidemiological studies. Clin Exp Hypertens. 2017;39:160–7. https://doi.org/10.1080/10641963.2016.1226888.

    Article  CAS  PubMed  Google Scholar 

  8. Chen L, Wang B, Wang J, Ban Q, Wu H, Song Y, Zhang J, Cao J, Zhou Z, Liu L, Cao T. Association between Serum Total Homocysteine and arterial stiffness in adults: A Community-based study. J Clin Hypertens. 2018;20:686–93. https://doi.org/10.1111/jch.13246.

    Article  CAS  Google Scholar 

  9. Schutte AE, Srinivasapura Venkateshmurthy N, Mohan S, Prabhakaran D. Hypertension in low-and Middle-Income Countries. Circ Res. 2021;128:808–26. https://doi.org/10.1161/CIRCRESAHA.120.318729.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Du Plessis JP, Melse-Boonstra A, Zandberg L, Nienaber-Rousseau C. Gene Interactions Observed with the HDL-C blood lipid, intakes of protein, Sugar and biotin in relation to circulating homocysteine concentrations in a Group of Black South africans. Mol Genet Metab Rep. 2020;22:e100556. https://doi.org/10.1016/j.ymgmr.2019.100556.

    Article  Google Scholar 

  11. Nienaber-Rousseau C. Dietary strategies to treat hyperhomocysteinaemia based on the Biochemistry of Homocysteine: a review. South Afr J Clin Nutr. 2014;27:93–100. https://doi.org/10.1080/16070658.2014.11734495.

    Article  Google Scholar 

  12. Nienaber-Rousseau C, Pisa PT, Venter CS, Ellis SM, Kruger A, Moss SJ, Melse-Boonstra A, Towers GW. Nutritional Genetics: the case of Alcohol and the Mthfr C677t polymorphism in relation to Homocysteine in a Black South African Population. J Nutrigenet Nutrigenomics. 2013;6:61–72. https://doi.org/10.1159/000348839.

    Article  CAS  PubMed  Google Scholar 

  13. Chen L, Li Q, Fang X, Wang X, Min J, Wang F. Dietary intake of Homocysteine metabolism-related B-Vitamins and the risk of stroke: a dose-response Meta-analysis of prospective studies. Adv Nutr. 2020;11:1510–28. https://doi.org/10.1093/advances/nmaa061.

    Article  PubMed  PubMed Central  Google Scholar 

  14. McNulty H, Strain J, Hughes CF, Ward M, Riboflavin. Mthfr genotype and blood pressure: a Personalized Approach to Prevention and Treatment of Hypertension. Mol Aspects Med. 2017;53:2–9. https://doi.org/10.1016/j.mam.2016.10.002.

    Article  CAS  PubMed  Google Scholar 

  15. Teo K, Chow C, Vaz M, Rangarajan S, Yusuf S. The prospective Urban Rural Epidemiology (pure) study: examining the impact of Societal influences on Chronic Noncommunicable diseases in Low-, Middle-, and high-income countries. Am Heart J. 2009;158:1–7. https://doi.org/10.1016/j.ahj.2009.04.019.

    Article  PubMed  Google Scholar 

  16. MacIntyre U, Venter C, Vorster H. A culture-sensitive quantitative food frequency questionnaire used in an African Population: 2. Relative validation by 7-Day weighed records and biomarkers. Public Health Nutr. 2001;4:63–71. https://doi.org/10.1079/PHN200041.

    Article  CAS  PubMed  Google Scholar 

  17. Marfell-Jones M, Stewart A, Olds T. Kinanthropometry Ix. 1st ed. New York, NY, USA: Taylor & Francis; 2006. pp. 11–128.

    Book  Google Scholar 

  18. Unger T, Borghi C, Charchar F, Khan NA, Poulter NR, Prabhakaran D, Ramirez A, Schlaich M, Stergiou GS, Tomaszewski M, Wainford RD. 2020 International Society of Hypertension Global Hypertension Practice Guidelines. Hypertens. 2020;75:1334–57. https://doi.org/10.1161/HYPERTENSIONAHA.120.15026.

    Article  CAS  Google Scholar 

  19. Li J, Jiang S, Zhang Y, Tang G, Wang Y, Mao G, Li Z, Xu X, Wang B, Huo Y. H-type hypertension and risk of stroke in Chinese adults: a prospective, nested case–control study. J Transl Int Med. 2015;3:171–78. https://doi.org/10.1515/jtim-2015-0027.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Friedewald WT, Levy RI, Fredrickson DS. Estimation of the concentration of Low-Density Lipoprotein Cholesterol in plasma, without Use of the Preparative Ultracentrifuge. Clin Chem. 1972;18:499–502. https://doi.org/10.1093/clinchem/18.6.499.

    Article  CAS  PubMed  Google Scholar 

  21. Nienaber-Rousseau C, Ellis SM, Moss SJ, Melse-Boonstra A, Towers GW. Gene–environment and gene–gene interactions of specific Mthfr, Mtr and Cbs Gene Variants in Relation to Homocysteine in Black South africans. Gene. 2013;530:113–8. https://doi.org/10.1016/j.gene.2013.07.065.

    Article  CAS  PubMed  Google Scholar 

  22. Karger AB, Steffen BT, Nomura SO, Guan W, Garg PK, Szklo M, Budoff MJ, Tsai MY. Association between homocysteine and vascular calcification incidence, prevalence, and progression in the MESA cohort. J Am Heart Assoc. 2020, 9, e013934.

  23. Reference Values for Arterial Stiffness’ Collaboration. Determinants of pulse wave velocity in healthy people and in the presence of cardiovascular risk factors:‘establishing normal and reference values’. Eur Heart J. 2010;31:2338–50.

    Article  Google Scholar 

  24. du Plessis JP, Lammertyn L, Schutte AE, Nienaber-Rousseau C. H-Type hypertension among Black South africans and the relationship between Homocysteine, its genetic determinants and estimates of vascular function. J Cardiovasc Dev Dis. 2022;9:447–63. https://doi.org/10.3390/jcdd9120447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chiu HF, Venkatakrishnan K, Golovinskaia O, Wang CK. Impact of micronutrients on hypertension: evidence from clinical trials with a special focus on Meta-Analysis. Nutrients. 2021;13:588–607. https://doi.org/10.3390/nu13020588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mertens E, Tielemans S, Soedamah-Muthu S, Geleijnse J. Pulse pressure trajectories in relation to Cardiovascular Mortality and Dietary protein intake: the Zutphen Study. Proc Nutr Soc. 2015;74:e346. https://doi.org/10.1017/S0029665115003936.

    Article  Google Scholar 

  27. Mels CM, Delles C, Louw R, Schutte AE. Central systolic pressure and a nonessential amino acid Metabolomics Profile: the African prospective study on the early detection and identification of Cardiovascular Disease and Hypertension. J Hypertens. 2019;37:1157–66. https://doi.org/10.1097/HJH.0000000000002040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Du Y, Taylor CG, Zahradka P. Modulation of endothelial cell responses and vascular function by dietary fatty acids. Nutr Rev. 2019;77:614–29. https://doi.org/10.1093/nutrit/nuz026.

    Article  PubMed  Google Scholar 

  29. Rodriguez-Melendez R, Zempleni J. Regulation of Gene expression by Biotin. J Nutr Biochem. 2003;12:680–90. https://doi.org/10.1016/j.jnutbio.2003.07.001.

    Article  CAS  Google Scholar 

  30. Glushakova O, Kosugi T, Roncal C, Mu W, Heinig M, Cirillo P, Sanchez-Lozada LG, Johnson RJ, Nakagawa T. Fructose induces the inflammatory molecule Icam-1 in endothelial cells. J Am Soc Nephrol. 2008;19:1712–20. https://doi.org/10.1681/ASN.2007121304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Basciano H, Federico L, Adeli K, Fructose. Insulin resistance, and metabolic dyslipidemia. Nutr Metab. 2005;2:1–14. https://doi.org/10.1186/1743-7075-2-5.

    Article  CAS  Google Scholar 

  32. Han N, Chae JW, Jeon J, Lee J, Back HM, Song B, Kwon KI, Kim SK, Yun HY. Prediction of methionine and homocysteine levels in Zucker Diabetic Fatty (Zdf) rats as a T2dm animal model after consumption of a methionine-rich Diet. Nutr Metab. 2018;15:14–22. https://doi.org/10.1186/s12986-018-0247-1.

    Article  CAS  Google Scholar 

  33. Tessari P, Cecchet D, Vettore M, Coracina A, Puricelli L, Kiwanuka E. Decreased Homocysteine Trans-Sulfuration in Hypertension with Hyperhomocysteinemia: relationship with insulin resistance. J Clin Endocrinol Metab. 2017;103:56–63. https://doi.org/10.1210/jc.2017-01076.

    Article  Google Scholar 

  34. Vorster H, Kruger A, Venter C, Margetts B, Macintyre U. Cardiovascular Disease Risk factors and Socio-Economic position of africans in Transition: the Thusa Study: Cardiovascular Topics. Cardiovasc J Afr. 2007;18:282–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Casari S, Di Paola M, Banci E, Diallo S, Scarallo L, Renzo S, Gori A, Renzi S, Paci M, de Mast Q, Pecht T. Changing Dietary habits: the impact of urbanization and rising socio-economic status in families from Burkina Faso in Sub-saharan Africa. Nutrients. 2022;14:1782–800. https://doi.org/10.3390/nu14091782.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Monahan KD, Feehan RP, Blaha C, McLaughlin DJ. Effect of Omega-3 polyunsaturated fatty acid supplementation on central arterial stiffness and arterial Wave reflections in Young and older healthy adults. Physiol Rep. 2015;3:e12438. https://doi.org/10.14814/phy2.12438.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zanetti M, Grillo A, Losurdo P, Panizon E, Mearelli F, Cattin L, Barazzoni R, Carretta R. Omega-3 polyunsaturated fatty acids: Structural and Functional effects on the Vascular Wall. BioMed Res Int. 2015;2015:1–15. https://doi.org/10.1155/2015/791978.

    Article  CAS  Google Scholar 

  38. Zec MM, Schutte AE, Ricci C, Baumgartner J, Kruger IM, Smuts CM. Long-chain polyunsaturated fatty acids are Associated with blood pressure and hypertension over 10-Years in black South African adults undergoing nutritional transition. Foods. 2019;8:394–409. https://doi.org/10.3390/foods8090394.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Videm V, Albrigtsen M. Soluble Icam-1 and Vcam‐1 as markers of endothelial activation. Scand J Immunol. 2008;67:523–31. https://doi.org/10.1111/j.1365-3083.2008.02029.x.

    Article  CAS  PubMed  Google Scholar 

  40. Da Silva MS, Bigo C, Barbier O, Rudkowska I. Whey protein hydrolysate and branched-chain amino acids downregulate inflammation-related genes in vascular endothelial cells. Nutr Res. 2017;38:43–51. https://doi.org/10.1016/j.nutres.2017.01.005.

    Article  CAS  PubMed  Google Scholar 

  41. Burris RL, Ng HP, Nagarajan S. Soy protein inhibits inflammation-Induced Vcam-1 and inflammatory cytokine induction by inhibiting the Nf-Κb and akt signaling pathway in apolipoprotein E–Deficient mice. Eur J Nutr. 2014;53:135–48. https://doi.org/10.1007/s00394-013-0509-7.

    Article  CAS  PubMed  Google Scholar 

  42. Siervo M, Lara J, Chowdhury S, Ashor A, Oggioni C, Mathers JC. Effects of the Dietary Approach to stop hypertension (dash) Diet on Cardiovascular Risk factors: a systematic review and Meta-analysis. Br J Nutr. 2015;113:1–15. https://doi.org/10.1017/S0007114514003341.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We appreciate all contributors, participants and supporting staff of the PURE-SA research team; we especially thank Professor Annamarie Kruger (posthumous); all the fieldworkers and the office personnel of the Africa Unit for Transdisciplinary Health Research (AUTHeR) in the Faculty of Health Sciences of NWU; the PURE-International research team, office personnel and leader Dr. Yusuf and the PURE study.

Funding

The research reported here did not receive any specific grant because it was a secondary analysis of previously collected data. A special mention to the South African Medical Research Council (SAMRC) for granting J.P.dP. a scholarship through its Division of Research Capacity Development under the Bongani Mayosi National Health Scholars Programme from funding received from the Public Health Enhancement Fund/South African National Department of Health. Disclosure Statement: The funders had no role in the study design, data collection or analysis, decision to publish, or preparation of the manuscript, whose content is the sole responsibility of the authors and does not necessarily represent the official views of the funding agencies.

Open access funding provided by North-West University.

Author information

Authors and Affiliations

Authors

Contributions

Authors’ Contributions: Conceptualization C.N-R.; methodology, J.P.dP., C.N-R., L.L. and A.E.S; validation, J.P.dP., L.L., A.E.S and C.N-R.; formal analysis, J.P.dP. and C.N-R.; investigation, J.P.dP. and C.N-R.; resources, L.L., A.E.S and C.N-R.; data curation, C.N-R., L.L. and A.E.S.; writing—original draft preparation, J.P.dP. and C.N-R.; writing—review and editing, J.P.dP, L.L., A.E.S and C.N-R.; visualization, J.P.dP. and C.N-R.; supervision, C.N-R. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Cornelie Nienaber-Rousseau.

Ethics declarations

Ethics approval and consent to participate

The Health Research Ethics Committee at North-West University approved the PURE-SA study protocols (ethics numbers: 04M10 and NWU-00142-18-A1) Informed consent was obtained from all subjects involved in the study ensuring voluntary participation as well as the anonymity and confidentiality of the participants and their data.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

du Plessis, J.P., Lammertyn, L., Schutte, A.E. et al. Homocysteine, blood pressure and gene–diet interactions in relation to vascular function measures of black South Africans. Genes Nutr 19, 14 (2024). https://doi.org/10.1186/s12263-024-00751-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12263-024-00751-8

Keywords