Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-05-31T09:59:38.247Z Has data issue: false hasContentIssue false

Sexual dimorphism in the context of nutrition and health

Published online by Cambridge University Press:  04 September 2023

Matthew G. Pontifex*
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
David Vauzour
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
Michael Muller
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
*
*Corresponding author: Matthew G. Pontifex, email m.pontifex@uea.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Diets and dietary constituents that we consume have a considerable impact on disease risk. Intriguingly these effects may be modulated to some extent by sex. Lack of female representation in nutritional studies as well as a lack of stratification by sex has and continues to limit our understanding of these sex × diet interactions. Here we provide an overview of the current and available literature describing how exposure to certain dietary patterns (Western-style diet, Mediterranean diet, vegetarian/vegan, ketogenic diet) and dietary constituents (dietary fibre, PUFA and plant bioactive) influences disease risk in a sex-specific manner. Interestingly, these sex differences appear to be highly disease-specific. The identification of such sex differences in response to diet stresses the importance of sex stratification in nutritional research.

Type
Conference on ‘Understanding the role of sex and gender in nutrition research’
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

A poor diet substantially increases the risk of developing numerous chronic health conditions including CVD, cancer and diabetes. In 2019, dietary risks were responsible for 7⋅94 million (6⋅47–9⋅76) deaths among adults globally(Reference Vos, Lim and Abbafati1). As such, diet remains a considerably important factor in the mitigation of disease burden, particularly metabolic diseases which are notoriously difficult to treat. Females have been largely underrepresented in scientific research to date. This is certainly true from a nutritional research perspective, in which our current understanding remains heavily male skewed. Despite this there is reason to believe that food components and dietary patterns modulate disease risk in a sex-specific manner(Reference Wirfält, Hedblad and Gullberg2,Reference Chen, Kim and Paye3) . Indeed, sexual dimorphism exists in many organs and body systems, such as the heart, kidney, adipose tissue, immune system and the central nervous system(Reference Shepherd, Cheung and Pang4,Reference Deegan, Nigam and Engel5,Reference Starling6,Reference Bond Simon, Calkin Anna and Drew Brian7) . From a metabolic perspective it is increasingly apparent that sex differences similarly exist(Reference Varghese, Song and Singer8,Reference Maggi and Della Torre9) . The reasons for such differences have not been entirely elucidated; however, sex hormones, X chromosome dosage and the microbiome have been posited as contributing factors(Reference Link, Chen and Arnold10,Reference Santos-Marcos, Mora-Ortiz and Tena-Sempere11) . Involvement of sex hormones (to some degree) is highly probable and signifies a potential dynamic element to these sex effects, evolving throughout the ageing process (particularly for women across the menopause transition). Such metabolic differences will have implications from a nutritional perspective and may in part explain discrepancies in effectiveness of some nutritional interventions to date. It is therefore important for nutritional guidance to account for and adapt to these changes in order to enhance implementation. Fortunately, nutritional research is now being increasingly conducted across both sexes (although with still significant work to do) with various research councils and funding bodies making the inclusion of both sexes a mandatory component of experimental design. This will no doubt aid our understanding of these complex interactions, enabling us to make more informed decisions in relation to these issues.

In the present review we explore how sex differences modulate physiological responses to various dietary patterns/constituents in the context of health, with a particular emphasis on cardiometabolic diseases. Comprehensively (but not exhaustively) reviewing the current evidence we highlight gaps in knowledge and comment on potential opportunities to further develop this important area of research.

Sex differences in cardiometabolic disorders

Cardiometabolic diseases are a group of common but often preventable conditions which span from obesity and type 2 diabetes right through to CVD. Reviewed extensively by Gerdts and Regitz-Zagrosek(Reference Gerdts and Regitz-Zagrosek12), cardiometabolic disease appears to be modulated by sex with sex-specific molecular mechanisms beginning to be uncovered. Type 2 diabetes for instance is an interesting example of this, with women exhibiting a stronger obesity-related diabetes risk than men of whom have greater susceptibility at a lower BMI(Reference Kautzky-Willer, Harreiter and Pacini13). Interestingly, such sex differences appear to differ from country to country, with cultural, lifestyle and socioeconomic factors presumably responsible(Reference Kautzky-Willer, Harreiter and Pacini13). Indeed, diet plays a prominent role in the development of cardiometabolic diseases. Given the emerging evidence implicating sex as a modulator of metabolism, it is probable that physiological responses to diet similarly differ across sex, perhaps contributing in part to the sex differences in cardiometabolic disorders.

Sex differences across dietary patterns

Western-style diet

Although not particularly well-defined, a Western-style dietary pattern generally consists of high intake of refined, energy-dense, nutrient-poor food sources(Reference Odermatt14).

Evaluation of Western-style diet across sex is relatively extensive in preclinical models compared to other dietary patterns/constituents (Table 1). Overall, this evidence appears to suggest that young male rodents have more negative changes in the body composition profile, as well as a higher susceptibility to diet-induced obesity when exposed to a high-fat diet (HFD)(Reference de Souza, Wasinski and Donato15,Reference Maric, Krieger and van der Velden16,Reference Oraha, Enriquez and Herzog17,Reference Dhanraj, van Heerden and Pepper18,Reference Guerra-Cantera, Frago and Collado-Pérez19) . It must however be mentioned that some discrepancies exist and may relate to species and/or diet differences(Reference Maric, Krieger and van der Velden16,Reference Gannon, Robison and Salinero20) . Young female mice appear to have a greater ability to utilise fat in the diet as a source of fuel(Reference Oraha, Enriquez and Herzog17), increase energy expenditure(Reference Maric, Krieger and van der Velden16) and increase AQP7/Aqp7 glycerol channel abundance (regulation influences glycerol release by adipocytes and reduced function is associated with obesity)(Reference Iena, Jul and Vegger21). Additionally, a more favourable immune response is observed in young female rodents exposed to an HFD(Reference Binenbaum, Atamni and Fotakis22,Reference Braga Tibaes, Azarcoya-Barrera and Wollin23) .

Table 1. Preclinical evidence for Western-style diet-related sex differences

FGF21, fibroblast growth factor 21; HFD, high-fat diet; IGF2, insulin-like growth factor 2.

Although a consensus appears to be emerging in young animals, for aged animals the picture is less clear. It appears that the protection from HFD observed in young female mice diminishes with age, with females having greater weight gain and impairment in glucose tolerance compared to males(Reference Guerra-Cantera, Frago and Collado-Pérez19,Reference Salinero, Anderson and Zuloaga24) . This may in part relate to changes in sex hormones. Indeed, ovariectomy of HFD-fed female mice enhanced adipose tissue inflammation leading to moderate changes in metabolism. However, gonadectomised HFD-fed males had improved metabolic outcomes that were associated with increased CD11c+ adipose tissue macrophages and increased proinflammatory cytokines(Reference Varghese, Griffin and Abrishami25). It should be noted that many diets utilised to model high-fat/Western-style diets in rodents (as outlined earlier) are refined (i.e. made from individual purified component rather than whole food). As such they lack many components of a complete control ‘chow’ diet. Indeed, dietary fibre source (e.g. soluble v. insoluble) and even amount are often overlooked in these studies, compromising the validity of these experiments. Morrison et al. utilised a refined diet with matched fibre source/content in their experimentation of low-fat diet v. HFD across both sexes, reporting that the lack of soluble fibre and not fat content primarily drives gut microbiota alterations previously associated with a refined HFD. In contrast to the aforementioned results, they report that male body weight increase is independent of dietary fat. However, when the amount of dietary fibre is comparable in all dietary groups, aged females do still appear to display increased weight gain in response to HFD(Reference Morrison, Jašarević and Howard26). This is in line with recent reports that the prebiotic effects of dietary fibres are sex-specific(Reference Kadyan, Park and Wang27), although the mechanisms responsible for such differences remain to be elucidated.

Sex differences in response to other components of the Western-style dietary pattern (e.g. high fructose, high sugar, low fibre) have been less extensively covered, and the results are generally mixed. Greater metabolic abnormalities have been reported in female animals receiving 10 % fructose supplementation(Reference Roglans, Baena and Sangüesa28). Similarly, a sweet-fat diet (standard laboratory control diet supplemented with sweet cookies, sunflower seeds and lard) resulted in more intense fat accumulation and weight gain in females as a result of suppressed carbohydrate and fat metabolism(Reference Bazhan, Iakovleva and Dubinina29). Furthermore, female mice maintained on a cafeteria diet had more extensive liver steatosis, higher non-alcoholic fatty liver disease scores and elevated triglyceride (TAG) content compared to males, with no difference in body weight gain or adiposity index observed(Reference Gasparin, Carreño and Mewes30). High-sucrose consumption in mice led to more extensive dysregulation of the oxylipin profile (oxidation products of PUFA) in the brains of female mice(Reference Norman, Nuthikattu and Milenkovic31). The mechanistic basis for which remains unclear. Intriguingly, others have reported the complete opposite with males displaying greater weight gain, glucose intolerance and hepatic inflammation on either high-fat, high-sugar or high-fat, high-fructose diets in agreement with the HFD studies outlined earlier(Reference Daly, Saxena and Singh32,Reference De Souza, Barros and De Souza33) .

Ethical consideration and lack of stratification by sex mean that clinical evaluation of Western-style diets across sex is scarce. However, in one such study conducted in young healthy adults 7-d exposure to a high-fat, high-energy diet did not result in any metabolic outcomes in either males or females(Reference Whytock, Shepherd and Cocks34), indicating that young healthy individuals can tolerate acute exposure to a Western-style diet. Consumption of a high-fructose meal however led to increased postprandial hepatic de novo lipogenesis in females only(Reference Low, Cornfield and Charlton35), suggesting that women may be more responsive to higher levels of fructose in the diet. It should however be noted that the opposite has also been reported(Reference Tran, Jacot-Descombes and Lecoultre36) and in line with this, Couchepin et al. observed that healthy young female mice were more resistant to fructose overfeeding compared to their male counterparts(Reference Couchepin, Lê and Bortolotti37).

Observational studies evaluating sex differences are similarly lacking and can be difficult to discern whether differences relate to biological/metabolic effects or merely food preference/portion size. Indeed, in a cross-sectional multi-ethnic study of middle-aged individuals (45–57 years) it was reported that women have a higher diet quality (as assessed by the HEI-2010)(Reference Maskarinec, Namatame and Kang38). Diet quality reduced adiposity across both sexes but intriguingly females displayed a stronger association than men(Reference Maskarinec, Namatame and Kang38). In line with this, Ruiz-Canela et al. reported (in a study population of 55–80-year-olds with CVD risk) that significant differences in BMI relating to consumption of a pro-inflammatory diet were restricted to females, although other indices of general and abdominal obesity were consistent across both sexes(Reference Ruiz-Canela, Zazpe and Shivappa39). Furthermore, in a Japanese cohort, increased SFA intake was associated with increased all-cause mortality in females only(Reference Nagata, Nakamura and Wada40). Similarly, UK Biobank analysis (of 40–69 years) suggested that higher sugar, SFA and dietary fibre intake may subtly modulate all-cause mortality and/or dementia risk to a greater extent in females(Reference McKenzie, Harris and Peters41). In contrast to the aforementioned studies, a cross-sectional study of a Taiwanese population with dyslipidaemia described that greater consumption of a Western-dietary pattern (highest quartile) increased general obesity, central obesity and high body fat regardless of sex(Reference Lin, Hsu and Lee42).

It has been posited that these sex difference may relate to changes in the microbiota. Indeed, in human subjects, a Western-style diet (high-fat/low-fibre) reportedly leads to an altered microbial profile across males and females, with higher levels of Campylobacter, Blautia, Flavonifractor and Erysipelatoclostridium in females(Reference Bailén, Bressa and Martínez-López43). However, a functional understanding of these changes requires further elucidation. HFD feeding in rats induces sex-related alterations in gut microbiome composition and metabolome(Reference Bailén, Bressa and Martínez-López43,Reference Shi, Wei and Xing44,Reference Hases, Stepanauskaite and Birgersson45) which correlate to metabolic measures such as insulin resistance(Reference Hases, Stepanauskaite and Birgersson45). Kim et al. suggest that the microbial impact may be mediated via the pregnane X receptor (a xenobiotic-sensing nuclear receptor) which reportedly primes the gut microbiome towards an obesity-prone microbial configuration in a sex (male) specific manner(Reference Kim, Choi and Dutta46) (Table 2).

Table 2. Human evidence for Western-style diet-related sex differences

Mediterranean diet

A Mediterranean diet (MedDiet) pattern appears to be highly beneficial, with adherence associated with a reduction in all-cause mortality. The MedDiet consists of a proportionally higher intake of unprocessed cereals, legumes, olive oil, fruits, nuts and vegetables, along with moderate consumption of fish, dairy and meat products(Reference Sikalidis, Kelleher and Kristo47).

In contrast to Western diet, preclinical studies investigating MedDiet across sexes are limited. This predominantly relates to the fact that preclinical studies tend to focus on aspects/constituents of the MedDiet rather than MedDiet in its entirety. Some of these constituents such as dietary fibres, lipids (e.g. MUFA and PUFA) and plant bioactives will be discussed in later sections.

Human evidence evaluating MedDiet across sexes is surprisingly limited with many studies failing to provide stratification of results/analysis by sex, despite inclusion of both sexes in the experiment. This is quite a significant issue, presumably relating to a lack of power that needs to be resolved imminently. From the available evidence, studies in younger (24–53 years, premenopausal) adults suggest that the MedDiet confers more favourable changes in glucose/insulin homoeostasis in men than in women(Reference Bédard, Corneau and Lamarche48,Reference Bédard, Riverin and Dodin49) . In line with this, improvements in TAG levels, HDL-cholesterol ratios and waist circumference are more pronounced in men than in women(Reference Leblanc, Bégin and Hudon50). Furthermore, MedDiet adherence leads to a significant decrease in adiponectin concentration in men only(Reference Bédard, Tchernof and Lamarche51), as well as a more favourable redistribution of LDL subclasses from smaller to larger LDL(Reference Bédard, Corneau and Lamarche52). This appears to be independent of circulating NEFA concentrations (believed to be an important factor in insulin resistance). Similar results were reported after 3-year MedDiet adherence in older (~66 years) overweight/obese individuals with metabolic syndrome, in which a reduction in weight, waist circumference, fasting glucose, insulin and TAG were more pronounced in men than in women(Reference Soldevila-Domenech, Pastor and Sala-Vila53). This appears to be consistent with CVD, in which an association between MedDiet adherence appears to be stronger(Reference GómezSánchez, Gómez Sánchez and Patino-Alonso54,Reference Bilenko, Fraser and Vardi55) , although no difference has also been reported(Reference Bédard, Dodin and Corneau56). In contrast to this, 1-year Mediterranean-like diet intervention in elderly healthy subjects led to female-specific (but also country-specific) reduction in epigenetic ageing score(Reference Gensous, Garagnani and Santoro57). Also, from a neurological disease perspective, women appear to have more favourable outcomes in response to MedDiet adherence. Indeed, an inverse association between MedDiet and dementia risk was established among women, but not among men(Reference Andreu-Reinón, Chirlaque and Gavrila58). Similarly, in a cross-sectional analysis adherence to both MedDiet and Mediterranean-dietary approaches to stop hypertension intervention for neurodegenerative delay diet was significantly associated with a higher age of Parkinson's disease onset(Reference Metcalfe-Roach, Yu and Golz59), especially in women. However, for colorectal cancer no disease-modifying effect was observed as a result of MedDiet(Reference Schulpen and van den Brandt60). This was also true for all cancer risk, which despite displaying an inverse association in females only, failed to reach significance after full adjustment of confounding factors(Reference Schulpen and van den Brandt60,Reference Schulpen and van den Brandt61) (Table 3).

Table 3. Human evidence for Mediterranean style diet-related sex differences

ApoA-2, apolipoprotein A2; HDL-C, HDL-cholesterol; LDL-C, LDL-cholesterol; MedDiet, Mediterranean diet; MIND, Mediterranean-dietary approaches to stop hypertension intervention for neurodegenerative delay; PD, Parkinson's disease; sdLDL, small-dense LDL.

Vegetarian/vegan diets

Food constituents derived from animal sources are limited/absent from vegetarian/vegan diets. Despite an abundance of studies investigating such diets in the context of metabolism and disease, sub-analysis by sex is consistently missing. As such, the existence of any sex differences in response to vegetarian diet is not entirely clear. Blood sampling of healthy age-matched vegetarians and non-vegetarians revealed a purportedly beneficial increase in adiponectin levels in female vegetarians, which was not present in males(Reference Vučić Lovrenčić, Gerić and Košuta63). However, no diet-dependent or sex-dependent differences were found in insulin, Homeostatic model assessment for insulin resistance index (HOMA2-IRI), inflammatory and metabolic biomarkers(Reference Vučić Lovrenčić, Gerić and Košuta63). Reviewed extensively by Adams and Sabaté, there is evidence to suggest that the cardio-protective effects of a vegetarian diet may be sex-specific(Reference Adams and Sabaté64). The available evidence suggests that a vegetarian dietary pattern is associated with a reduction in CVD outcomes for vegetarian men relative to omnivorous men, whilst for women this association is less strong/non-existent(Reference Adams and Sabaté64). In line with this a 4-year longitudinal study reported that low intake of vegetables was significantly associated with type 2 diabetes risk in men, but not in women. Although this may relate to the lack of study power (fewer women in the low-intake category), it could also relate to differences in vegetable preference(Reference Whytock, Shepherd and Cocks34). Conversely, Kim et al. did not find any association between plant-based diet and CVD, nor any apparent sex differences within a US population(Reference Kim, Caulfield and Rebholz65). They did however report that those with a high plant-based diet index (i.e. above median) had a 5 % lower risk in all-cause mortality in the overall study population which was not influenced by sex(Reference Kim, Caulfield and Rebholz65). Together there is some evidence to support vegetarian diet-related sex differences, particularly in the context of CVD however further investigation is clearly warranted to gain a greater understanding metabolically and for other diseases.

Ketogenic diet

Ketogenic diets are low in carbohydrate content and high in fat, shifting energy reliance from glucose to ketone bodies. Twenty-five day ketogenic diet adherence in individuals with severe obesity resulted in significantly larger excess body weight loss and a greater reduction in γ-glutamyl transferase in males(Reference D'Abbondanza, Ministrini and Pucci66). This greater benefit in males has been reported by others(Reference Volek, Sharman and Gómez67,Reference Lyngstad, Nymo and Coutinho68) . Interestingly, this difference does not appear to be present when considering post-menopausal females(Reference D'Abbondanza, Ministrini and Pucci66), again emphasising the importance of the menopause (and likely sex hormones) in metabolism and response to diet. In rats maintained on a high-fat, high-sugar-diet, the beneficial effects of ketogenic diet intervention were largely similar across both sexes although these benefits correlated significantly with plasma β-hydroxybutyrate in females only(Reference Sahagun, Bachman and Kinzig69). In the context of pancreatic cancer, strict ketogenic diet in combination with gemcitabine (chemotherapy medication) prolonged survival. Intrudingly, when stratified by sex this result remained significant for males only(Reference Cortez, Rodriguez Lanzi and Hong70).

Sex differences across other dietary constituents

Dietary fibre

Often overlooked as a key contributor in health and disease, dietary fibre is the undigestible part of the plant, typically obtained from wholegrain cereals, fruits and vegetables. European and US guidelines suggest an intake of 30–35 g daily for men and 25–32 g daily for women (discrepancy between males and females relates to fact that many countries calculate recommendation based upon for total energy intake) but actual dietary fibre intake is significantly lower(Reference Barber, Kabisch and Pfeiffer71). In adolescents, increasing dietary fibre to recommendation levels decreased predicted fasting glucose, fasting insulin, Homeostatic model assessment for insulin resistance (HOMA-IR), Systolic blood pressure (Hg SBP), and diastolic blood pressure (Hg DBP) regardless of sex(Reference Dong, Chen and Gutin72). In line with this a cross-sectional analysis found that higher daily dietary fibre consumption was associated with beneficial effects on cholesterol in both males and females(Reference Zhou, Wu and Tang73). Analysis of the European prospective investigation into cancer and nutrition cohort revealed that total dietary fibre was inversely associated with colorectal cancer (Hazard Ratio per 10 g daily increase in fibre 0⋅87, 95 % CI: 0⋅79, 0⋅96) which did not differ by sex(Reference Murphy, Norat and Ferrari74). Similarly, an inverse relationship between dietary fibre and multiple sclerosis has been reported in a case control study with the trends similar across males and females(Reference Ghadirian, Jain and Ducic75). In the context of depression, dietary fibre may be more favourable in females with an inverse association between depression and dietary fibre consumption established in females only(Reference Chrzastek, Guligowska and Piglowska76). Discrepancies in the impact of dietary fibres across sex may relate to changes in the gut microbiota, indeed oligofructose supplementation in mice led to broad changes in faecal community structure (increasing Bacteroidetes at the expense of Lachnospiraceae) in females but not males. How dietary fibre type (e.g. soluble or insoluble) influences metabolism and health outcomes across sex is yet to be explored and represents a major gap in our current knowledge.

PUFA

As alluded to in the Western-diet section of this review, lipid metabolism appears to be sexually dimorphic. Indeed, vast differences in lipid species have been identified across sex, particularly when considering age × sex interaction, with the most prevalent of these differences found across phosphatidylcholine, sphingomyelin and TAG species(Reference Slade, Irvin and Xie77). It is therefore not surprising that specific dietary lipid types e.g. PUFA exert different effects across the sexes. Females have significantly higher peripheral DHA than males(Reference West, Michaelson and Miles78). In rats this higher DHA concentrations is found in the liver, plasma, erythrocytes and heart (53, 75, 36 and 25 % higher, respectively, compared with males) but not the brain(Reference Kitson, Smith and Marks79). This may be linked to higher Δ6-desaturase expression in females relative to males, which appears to be limited to the liver(Reference Kitson, Smith and Marks79). Women show a greater increase in circulating EPA in response to α-linolenic acid consumption(Reference Childs, Kew and Finnegan80). Similarly, EPA and DHA supplementation increases plasma TAG EPA to a greater extent in females(Reference Walker, Browning and Mander81). The source of PUFA (e.g. krill oil v. fish oil) may also alter these sex differences adding further complexity to the interaction(Reference Ghasemifard, Hermon and Turchini82).

Sex × diet interactions may influence brain PUFA concentrations(Reference Chen, Haven and Lecaj83). Higher n-3 PUFA concentrations appear to benefit different cognitive domains in a sex-specific manner(Reference Duchaine, Fiocco and Carmichael84). In mice receiving DHA supplementation, a reduction in anxiety and depressive-like behaviours was observed in male mice only and coincided with sex-specific gut microbiota interactions in response to DHA which correlated with behavioural finding(Reference Davis, Hecht and Jasarevic85). This in contrasts with a report in human subjects which showed n-3 fatty acid intake to be negatively associated with depressive symptoms in only women(Reference Beydoun, Fanelli Kuczmarski and Beydoun86). From a diabetes perspective n-3 PUFA status was inversely associated with diabetes in overweight/obese females but not in males(Reference Abbott, Burrows and Thota87). This is supported by a systematic review and meta-analysis of randomised controlled trials which found that n-3 PUFA intervention improved insulin resistance in women but not in men(Reference Abbott, Burrows and Thota88). Furthermore, PUFA appear to be more protective against hypertriglyceridaemia in females, compared to males(Reference Kaviani, Taylor and Stevenson89). Interestingly, the ability of n-3 PUFA to reduce platelet aggregation (a factor in CVD) is sex-specific. In men, only EPA treatment reduces aggregation, whilst in women, only DHA treatment reduced platelet aggregation(Reference Phang, Lincz and Garg90). Both increased n-3 and n-6 PUFA intake were found to be inversely associated with non-alcoholic fatty liver disease risk, irrespective of sex(Reference Cui, Li and Ren91). There is growing evidence suggesting that oxylipin (bioactive oxidation products of PUFA) production and profile is differentially altered across sexes in response to the intake of various n-3 and n-6 PUFA(Reference Gabbs, Zahradka and Taylor92,Reference Ferdouse, Leng and Winter93,Reference Pauls, Ragheb and Winter94,Reference Leng, Winter and Aukema95) , although this seems to be less extensive in the brain(Reference Ferdouse, Leng and Winter96). As mediators of PUFA, such differences in the oxylipin profile may provide in part some explanation for the varying disease-modifying influences observed across sexes.

Plant bioactives

Sex has been suggested to modulate both the metabolism(Reference Zamora-Ros, Achaintre and Rothwell97) and physiological effects of plant bioactives such as (poly)phenols(Reference Gibney, Milenkovic and Combet98). HPLC-MS/MS analysis of acute doses of grape seed (poly)phenols established clear sex differences in the metabolism and distribution of flavanols throughout the bodies of rats, with quantitative differences found in the plasma and brain(Reference Margalef, Pons and Iglesias-Carres99). In line with this supplementation with an oral formulation of resveratrol, JOTROL™ in 3xTg-AD mice resulted in Alzheimers disease (AD)-related gene expression changes (Adam10, Bace1, Bdnf, Psen1) some of which were brain region-dependent and sex-specific(Reference Dennison, Volmar and Modarresi100). Analysis of the Primary prevention of cardiovascular disease with a mediterranean diet (PREDIMED) study revealed that catechins, proanthocyanidins, hydroxybenzoic acids and lignans were inversely associated with type 2 diabetes, with women displaying stronger inverse associations. Additionally, a cross-sectional analysis of a Korean population reported an inverse association between flavonoid intake and obesity in women, whilst for men a positive association was determined for some subclasses (namely, flavonols, flavanones and anthocyanidins)(Reference Kim, Kim and Jun101). In a randomised double-blind parallel trial, a combination of 548 mg daily of polyphenols and 2 g daily of l-citrulline reduced ambulatory systolic blood pressure in women, but not in men(Reference Vors, Rancourt-Bouchard and Couillard102). Furthermore, a systematic review and meta-analysis described an inverse association between (poly)phenol consumption and gastric cancer. Interestingly, the risk reduction was greater for females, which the authors suggest may be partly explained by the fact that (poly)phenols can regulate female hormones which play a protective role against cancer(Reference Fagundes, Silva and Fernandes103). These differences may relate to impact on the gut microbiota which may be modulated in a sex-specific manner, indeed microbial changes associated with 7,8-dihydroxyflavone predicted body weight changes in females but not in males(Reference Sharma, Wu and Kumaraswamy104). In contrast to the female-specific improvements outlined earlier, the Reasons for geographic and racial differences in stroke (REGARDS) prospective cohort study reported that the inverse association between flavanone intake and ischaemic stroke risk did not differ by sex(Reference Goetz, Judd and Hartman105). Additionally, in mouse models of CVD both blackberry and gallic acid supplementation reduced atherosclerosis in males only(Reference Clark, Centner and Ukhanov106). Consistent with this, nettle extract altered lipid metabolism differently across sexes, with the activation of transcription factors that control lipid metabolism, and subsequent increase in HDL-cholesterol, specific to male mice(Reference Domjanić Drozdek, Odeh and Đikić107).

Conclusions

Despite considerable underreporting, it is apparent from emerging literature that sex differences exist in response to various dietary patterns and components. These differences are not trivial as they likely contribute to sexual dimorphism that similarly exists in the patterns of health and disease. Such discrepancies (and heterogeneity between males and females) may even explain why some promising nutritional interventions fail to show benefits at more advanced stages of experimentation. These interactions are complex and display both disease and region specificity. As such, future nutritional studies should aim to consistently provide comparison across both sexes, either in initial experiment set up or via extended subgroup analysis. This could potentially improve the effectiveness of dietary advice and treatments enabling us to adapt to specific needs of both men and women as we strive towards a more personal/precise nutritional approach.

Conflict of Interest

None.

Authorship

M. G. P., D. V. and M. M. jointly planned, wrote and edited the manuscript.

References

Vos, T, Lim, SS, Abbafati, C et al. (2020) Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the global burden of disease study 2019. Lancet 396, 12041222.CrossRefGoogle Scholar
Wirfält, E, Hedblad, B, Gullberg, B et al. (2001) Food patterns and components of the metabolic syndrome in men and women: a cross-sectional study within the Malmö diet and cancer cohort. Am J Epidemiol 154, 11501159.CrossRefGoogle Scholar
Chen, Y, Kim, M, Paye, S et al. (2022) Sex as a biological variable in nutrition research: from human studies to animal models. Annu Rev Nutr 42, 227250.CrossRefGoogle ScholarPubMed
Shepherd, R, Cheung, AS, Pang, K et al. (2021) Sexual dimorphism in innate immunity: the role of sex hormones and epigenetics. Front Immunol 11.CrossRefGoogle ScholarPubMed
Deegan, DF, Nigam, P & Engel, N (2021) Sexual dimorphism of the heart: genetics, epigenetics, and development. Front Cardiovasc Med 8.CrossRefGoogle ScholarPubMed
Starling, S (2017) The sexually dimorphic kidney. Nat Rev Nephrol 13, 596596.CrossRefGoogle ScholarPubMed
Bond Simon, T, Calkin Anna, C & Drew Brian, G (2021) Sex differences in white adipose tissue expansion: emerging molecular mechanisms. Clin Sci 135, 26912708.CrossRefGoogle ScholarPubMed
Varghese, M, Song, J & Singer, K (2021) Age and sex: impact on adipose tissue metabolism and inflammation. Mech Ageing Dev 199, 111563.CrossRefGoogle ScholarPubMed
Maggi, A & Della Torre, S (2018) Sex, metabolism and health. Mol Metab 15, 37.CrossRefGoogle ScholarPubMed
Link, JC, Chen, X, Arnold, AP et al. (2013) Metabolic impact of sex chromosomes. Adipocyte 2, 7479.CrossRefGoogle ScholarPubMed
Santos-Marcos, JA, Mora-Ortiz, M, Tena-Sempere, M et al. (2023) Interaction between gut microbiota and sex hormones and their relation to sexual dimorphism in metabolic diseases. Biol Sex Differ 14, 4.CrossRefGoogle ScholarPubMed
Gerdts, E & Regitz-Zagrosek, V (2019) Sex differences in cardiometabolic disorders. Nat Med 25, 16571666.CrossRefGoogle ScholarPubMed
Kautzky-Willer, A, Harreiter, J & Pacini, G (2016) Sex and gender differences in risk, pathophysiology and complications of type 2 diabetes mellitus. Endocr Rev 37, 278316.CrossRefGoogle ScholarPubMed
Odermatt, A (2011) The Western-style diet: a major risk factor for impaired kidney function and chronic kidney disease. Am J Physiol-Renal Physiol 301, F919F931.CrossRefGoogle Scholar
de Souza, GO, Wasinski, F & Donato, J (2022) Characterization of the metabolic differences between male and female C57BL/6 mice. Life Sci 301, 120636.CrossRefGoogle ScholarPubMed
Maric, I, Krieger, JP, van der Velden, P et al. (2022) Sex and species differences in the development of diet-induced obesity and metabolic disturbances in rodents. Front Nutr 9, 828522.CrossRefGoogle ScholarPubMed
Oraha, J, Enriquez, RF, Herzog, H et al. (2022) Sex-specific changes in metabolism during the transition from chow to high-fat diet feeding are abolished in response to dieting in C57BL/6J mice. Int J Obes 46, 17491758.CrossRefGoogle ScholarPubMed
Dhanraj, P, van Heerden, MB, Pepper, MS et al. (2021) Sexual dimorphism in changes that occur in tissues, organs and plasma during the early stages of obesity development. Biology (Basel) 10.Google ScholarPubMed
Guerra-Cantera, S, Frago, LM, Collado-Pérez, R et al. (2021) Sex differences in metabolic recuperation after weight loss in high fat diet-induced obese mice. Front Endocrinol 12, 796661.CrossRefGoogle ScholarPubMed
Gannon, OJ, Robison, LS, Salinero, AE et al. (2022) High-fat diet exacerbates cognitive decline in mouse models of Alzheimer's disease and mixed dementia in a sex-dependent manner. J Neuroinflammation 19, 110.CrossRefGoogle Scholar
Iena, FM, Jul, JB, Vegger, JB et al. (2020) Sex-specific effect of high-fat diet on glycerol metabolism in murine adipose tissue and liver. Front Endocrinol 11, 577650.CrossRefGoogle ScholarPubMed
Binenbaum, I, Atamni, HA, Fotakis, G et al. (2020) Container-aided integrative QTL and RNA-seq analysis of collaborative cross mice supports distinct sex-oriented molecular modes of response in obesity. BMC Genomics 21, 761.CrossRefGoogle ScholarPubMed
Braga Tibaes, JR, Azarcoya-Barrera, J, Wollin, B et al. (2022) Sex differences distinctly impact high-fat diet-induced immune dysfunction in Wistar rats. J Nutr 152, 13471357.CrossRefGoogle ScholarPubMed
Salinero, AE, Anderson, BM & Zuloaga, KL (2018) Sex differences in the metabolic effects of diet-induced obesity vary by age of onset. Int J Obes 42, 10881091.CrossRefGoogle ScholarPubMed
Varghese, M, Griffin, C, Abrishami, S et al. (2021) Sex hormones regulate metainflammation in diet-induced obesity in mice. J Biol Chem 297, 101229.CrossRefGoogle ScholarPubMed
Morrison, KE, Jašarević, E, Howard, CD et al. (2020) It's the fiber, not the fat: significant effects of dietary challenge on the gut microbiome. Microbiome 8, 15.CrossRefGoogle Scholar
Kadyan, S, Park, G, Wang, B et al. (2023) Dietary fiber modulates gut microbiome and metabolome in a host sex-specific manner in a murine model of aging. Front Mol Biosci 10.CrossRefGoogle Scholar
Roglans, N, Baena, M, Sangüesa, G et al. (2021) Chronic liquid fructose supplementation does not cause liver tumorigenesis but elicits clear sex differences in the metabolic response in Sprague–Dawley rats. Food Nutr Res 65.CrossRefGoogle Scholar
Bazhan, NM, Iakovleva, TV, Dubinina, AD et al. (2020) Impact of sex on the adaptation of adult mice to long consumption of sweet-fat diet. Vavilovskii Zh Genet Sel 24, 844852.Google Scholar
Gasparin, FRS, Carreño, FO, Mewes, JM et al. (2018) Sex differences in the development of hepatic steatosis in cafeteria diet-induced obesity in young mice. Biochim Biophys Acta Mol Basis Dis 1864, 24952509.CrossRefGoogle ScholarPubMed
Norman, JE, Nuthikattu, S, Milenkovic, D et al. (2022) A high sucrose diet modifies brain oxylipins in a sex-dependent manner. Prostaglandins, Leukotrienes Essent Fatty Acids 186, 102506.CrossRefGoogle Scholar
Daly, CM, Saxena, J, Singh, J et al. (2022) Sex differences in response to a high fat, high sucrose diet in both the gut microbiome and hypothalamic astrocytes and microglia. Nutr Neurosci 25, 321335.CrossRefGoogle ScholarPubMed
De Souza, L, Barros, WM, De Souza, RM et al. (2021) Impact of different fructose concentrations on metabolic and behavioral parameters of male and female mice. Physiol Behav 228, 113187.CrossRefGoogle ScholarPubMed
Whytock, KL, Shepherd, SO, Cocks, M et al. (2021) Young, healthy males and females present cardiometabolic protection against the detrimental effects of a 7-day high-fat high-calorie diet. Eur J Nutr 60, 16051617.CrossRefGoogle ScholarPubMed
Low, WS, Cornfield, T, Charlton, CA et al. (2018) Sex differences in hepatic de novo lipogenesis with acute fructose feeding. Nutrients 10.CrossRefGoogle ScholarPubMed
Tran, C, Jacot-Descombes, D, Lecoultre, V et al. (2010) Sex differences in lipid and glucose kinetics after ingestion of an acute oral fructose load. Br J Nutr 104, 11391147.CrossRefGoogle ScholarPubMed
Couchepin, C, , K-A, Bortolotti, M et al. (2008) Markedly blunted metabolic effects of fructose in healthy young female subjects compared with male subjects. Diabetes Care 31, 12541256.CrossRefGoogle ScholarPubMed
Maskarinec, G, Namatame, LA, Kang, M et al. (2020) Differences in the association of diet quality with body fat distribution between men and women. Eur J Clin Nutr 74, 14341441.CrossRefGoogle ScholarPubMed
Ruiz-Canela, M, Zazpe, I, Shivappa, N et al. (2015) Dietary inflammatory index and anthropometric measures of obesity in a population sample at high cardiovascular risk from the PREDIMED (PREvención con DIeta MEDiterránea) trial. Br J Nutr 113, 984995.CrossRefGoogle Scholar
Nagata, C, Nakamura, K, Wada, K et al. (2012) Total fat intake is associated with decreased mortality in Japanese men but not in women. J Nutr 142, 17131719.CrossRefGoogle Scholar
McKenzie, BL, Harris, K, Peters, SAE et al. (2022) The association of energy and macronutrient intake with all-cause mortality, cardiovascular disease and dementia: findings from 120 963 women and men in the UK Biobank. Br J Nutr 127, 18581867.CrossRefGoogle ScholarPubMed
Lin, LY, Hsu, CY, Lee, HA et al. (2019) Gender difference in the association of dietary patterns and metabolic parameters with obesity in young and middle-aged adults with dyslipidemia and abnormal fasting plasma glucose in Taiwan. Nutr J 18, 75.CrossRefGoogle Scholar
Bailén, M, Bressa, C, Martínez-López, S et al. (2020) Microbiota features associated with a high-fat/low-fiber diet in healthy adults. Front Nutr 7, 583608.CrossRefGoogle ScholarPubMed
Shi, Y, Wei, L, Xing, L et al. (2022) Sex difference is a determinant of gut microbes and their metabolites SCFAs/MCFAs in high fat diet fed rats. Curr Microbiol 79, 347.CrossRefGoogle ScholarPubMed
Hases, L, Stepanauskaite, L, Birgersson, M et al. (2023) High-fat diet and estrogen modulate the gut microbiota in a sex-dependent manner in mice. Commun Biol 6, 20.CrossRefGoogle Scholar
Kim, S, Choi, S, Dutta, M et al. (2021) Pregnane X receptor exacerbates nonalcoholic fatty liver disease accompanied by obesity- and inflammation-prone gut microbiome signature. Biochem Pharmacol 193, 114698.CrossRefGoogle ScholarPubMed
Sikalidis, AK, Kelleher, AH & Kristo, AS (2021) Mediterranean diet. Encyclopedia 1, 371387.CrossRefGoogle Scholar
Bédard, A, Corneau, L, Lamarche, B et al. (2014) Sex-related differences in the effects of the Mediterranean diet on glucose and insulin homeostasis. J Nutr Metab 2014, 424130.CrossRefGoogle ScholarPubMed
Bédard, A, Riverin, M, Dodin, S et al. (2012) Sex differences in the impact of the Mediterranean diet on cardiovascular risk profile. Br J Nutr 108, 14281434.CrossRefGoogle ScholarPubMed
Leblanc, V, Bégin, C, Hudon, AM et al. (2014) Gender differences in the long-term effects of a nutritional intervention program promoting the Mediterranean diet: changes in dietary intakes, eating behaviors, anthropometric and metabolic variables. Nutr J 13, 107.CrossRefGoogle ScholarPubMed
Bédard, A, Tchernof, A, Lamarche, B et al. (2014) Effects of the traditional Mediterranean diet on adiponectin and leptin concentrations in men and premenopausal women: do sex differences exist? Eur J Clin Nutr 68, 561566.CrossRefGoogle ScholarPubMed
Bédard, A, Corneau, L, Lamarche, B et al. (2015) Sex differences in the impact of the Mediterranean diet on LDL particle size distribution and oxidation. Nutrients 7, 37053723.CrossRefGoogle ScholarPubMed
Soldevila-Domenech, N, Pastor, A, Sala-Vila, A et al. (2022) Sex differences in endocannabinoids during 3 years of Mediterranean diet intervention: association with insulin resistance and weight loss in a population with metabolic syndrome. Front Nutr 9.CrossRefGoogle Scholar
GómezSánchez, M, Gómez Sánchez, L, Patino-Alonso, MC et al. (2020) Adherence to the Mediterranean diet in Spanish population and its relationship with early vascular aging according to sex and age: EVA study. Nutrients 12.Google Scholar
Bilenko, N, Fraser, D, Vardi, H et al. (2005) Mediterranean diet and cardiovascular diseases in an Israeli population. Prev Med 40, 299305.CrossRefGoogle Scholar
Bédard, A, Dodin, S, Corneau, L et al. (2014) Impact of the traditional Mediterranean diet on the Framingham risk score and the metabolic syndrome according to sex. Metab Syndr Relat Disord 12, 95101.CrossRefGoogle ScholarPubMed
Gensous, N, Garagnani, P, Santoro, A et al. (2020) One-year Mediterranean diet promotes epigenetic rejuvenation with country- and sex-specific effects: a pilot study from the NU-AGE project. Geroscience 42, 687701.CrossRefGoogle ScholarPubMed
Andreu-Reinón, ME, Chirlaque, MD, Gavrila, D et al. (2021) Mediterranean diet and risk of dementia and Alzheimer's disease in the EPIC-Spain dementia cohort study. Nutrients 13.CrossRefGoogle ScholarPubMed
Metcalfe-Roach, A, Yu, AC, Golz, E et al. (2021) MIND and Mediterranean diets associated with later onset of Parkinson's disease. Mov Disord 36, 977984.CrossRefGoogle ScholarPubMed
Schulpen, M & van den Brandt, PA (2020) Mediterranean diet adherence and risk of colorectal cancer: the prospective Netherlands cohort study. Eur J Epidemiol 35, 2535.CrossRefGoogle ScholarPubMed
Schulpen, M & van den Brandt, PA (2021) Adherence to the Mediterranean diet and overall cancer incidence: the Netherlands cohort study. J Acad Nutr Diet 121, 242252.CrossRefGoogle Scholar
de la Rubia Ortí, JE, García-Pardo, MP, Drehmer, E et al. (2018) Improvement of main cognitive functions in patients with Alzheimer's disease after treatment with coconut oil enriched Mediterranean diet: a pilot study. J Alzheimers Dis 65, 577587.CrossRefGoogle ScholarPubMed
Vučić Lovrenčić, M, Gerić, M, Košuta, I et al. (2020) Sex-specific effects of vegetarian diet on adiponectin levels and insulin sensitivity in healthy non-obese individuals. Nutrition 79–80, 110862.CrossRefGoogle ScholarPubMed
Adams, M & Sabaté, J (2019) Sexual dimorphism in cardiovascular disease risk and risk factors among vegetarians: an exploration of the potential mechanisms. Curr Atheroscler Rep 21, 35.CrossRefGoogle ScholarPubMed
Kim, H, Caulfield, LE & Rebholz, CM (2018) Healthy plant-based diets are associated with lower risk of all-cause mortality in US adults. J Nutr 148, 624631.CrossRefGoogle ScholarPubMed
D'Abbondanza, M, Ministrini, S, Pucci, G et al. (2020) Very low-carbohydrate ketogenic diet for the treatment of severe obesity and associated non-alcoholic fatty liver disease: the role of sex differences. Nutrients 12.Google ScholarPubMed
Volek, JS, Sharman, MJ, Gómez, AL et al. (2004) Comparison of energy-restricted very low-carbohydrate and low-fat diets on weight loss and body composition in overweight men and women. Nutr Metab 1, 13.CrossRefGoogle ScholarPubMed
Lyngstad, A, Nymo, S, Coutinho, SR et al. (2019) Investigating the effect of sex and ketosis on weight-loss-induced changes in appetite. Am J Clin Nutr 109, 15111518.CrossRefGoogle ScholarPubMed
Sahagun, E, Bachman, BB & Kinzig, KP (2021) Sex-specific effects of ketogenic diet after pre-exposure to a high-fat, high-sugar diet in rats. Nutr Metab Cardiovasc Dis 31, 961971.CrossRefGoogle ScholarPubMed
Cortez, NE, Rodriguez Lanzi, C, Hong, BV et al. (2022) A ketogenic diet in combination with gemcitabine increases survival in pancreatic cancer KPC mice. Cancer Res Commun 2, 951965.CrossRefGoogle ScholarPubMed
Barber, TM, Kabisch, S, Pfeiffer, AFH et al. (2020) The health benefits of dietary fibre. Nutrients 12.CrossRefGoogle ScholarPubMed
Dong, Y, Chen, L, Gutin, B et al. (2019) Total, insoluble, and soluble dietary fiber intake and insulin resistance and blood pressure in adolescents. Eur J Clin Nutr 73, 11721178.CrossRefGoogle ScholarPubMed
Zhou, Q, Wu, J, Tang, J et al. (2015) Beneficial effect of higher dietary fiber intake on plasma HDL-C and TC/HDL-C ratio among Chinese rural-to-urban migrant workers. Int J Environ Res Public Health 12, 47264738.CrossRefGoogle ScholarPubMed
Murphy, N, Norat, T, Ferrari, P et al. (2012) Dietary fibre intake and risks of cancers of the colon and rectum in the European prospective investigation into cancer and nutrition (EPIC). PLoS ONE 7, e39361.CrossRefGoogle ScholarPubMed
Ghadirian, P, Jain, M, Ducic, S et al. (1998) Nutritional factors in the aetiology of multiple sclerosis: a case-control study in Montreal, Canada. Int J Epidemiol 27, 845852.CrossRefGoogle ScholarPubMed
Chrzastek, Z, Guligowska, A, Piglowska, M et al. (2022) Association between sucrose and fiber intake and symptoms of depression in older people. Nutr Neurosci 25, 886897.CrossRefGoogle ScholarPubMed
Slade, E, Irvin, MR, Xie, K et al. (2021) Age and sex are associated with the plasma lipidome: findings from the GOLDN study. Lipids Health Dis 20, 30.CrossRefGoogle ScholarPubMed
West, AL, Michaelson, LV, Miles, EA et al. (2021) Lipidomic analysis of plasma from healthy men and women shows phospholipid class and molecular Species differences between sexes. Lipids 56, 229242.CrossRefGoogle ScholarPubMed
Kitson, AP, Smith, TL, Marks, KA et al. (2012) Tissue-specific sex differences in docosahexaenoic acid and Δ6-desaturase in rats fed a standard chow diet. Appl Physiol Nutr Metab 37, 12001211.CrossRefGoogle ScholarPubMed
Childs, CE, Kew, S, Finnegan, YE et al. (2014) Increased dietary α-linolenic acid has sex-specific effects upon eicosapentaenoic acid status in humans: re-examination of data from a randomised, placebo-controlled, parallel study. Nutr J 13, 113.CrossRefGoogle ScholarPubMed
Walker, CG, Browning, LM, Mander, AP et al. (2014) Age and sex differences in the incorporation of EPA and DHA into plasma fractions, cells and adipose tissue in humans. Br J Nutr 111, 679689.CrossRefGoogle ScholarPubMed
Ghasemifard, S, Hermon, K, Turchini, GM et al. (2015) Metabolic fate (absorption, β-oxidation and deposition) of long-chain n-3 fatty acids is affected by sex and by the oil source (krill oil or fish oil) in the rat. Br J Nutr 114, 684692.CrossRefGoogle Scholar
Chen, CT, Haven, S, Lecaj, L et al. (2020) Brain PUFA concentrations are differentially affected by interactions of diet, sex, brain regions, and phospholipid pools in mice. J Nutr 150, 31233132.CrossRefGoogle ScholarPubMed
Duchaine, CS, Fiocco, AJ, Carmichael, PH et al. (2022) Serum ω-3 fatty acids and cognitive domains in community-dwelling older adults from the NuAge study: exploring the associations with other fatty acids and sex. J Nutr 152, 21172124.CrossRefGoogle ScholarPubMed
Davis, DJ, Hecht, PM, Jasarevic, E et al. (2017) Sex-specific effects of docosahexaenoic acid (DHA) on the microbiome and behavior of socially-isolated mice. Brain Behav Immun 59, 3848.CrossRefGoogle ScholarPubMed
Beydoun, MA, Fanelli Kuczmarski, MT, Beydoun, HA et al. (2013) ω-3 fatty acid intakes are inversely related to elevated depressive symptoms among United States women. J Nutr 143, 17431752.CrossRefGoogle ScholarPubMed
Abbott, KA, Burrows, TL, Thota, RN et al. (2020) Association between plasma phospholipid omega-3 polyunsaturated fatty acids and type 2 diabetes is sex dependent: the hunter community study. Clin Nutr 39, 10591066.CrossRefGoogle ScholarPubMed
Abbott, KA, Burrows, TL, Thota, RN et al. (2016) Do ω-3 PUFAs affect insulin resistance in a sex-specific manner? A systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 104, 14701484.CrossRefGoogle Scholar
Kaviani, S, Taylor, CM, Stevenson, JL et al. (2019) A 7-day high-PUFA diet reduces angiopoietin-like protein 3 and 8 responses and postprandial triglyceride levels in healthy females but not males: a randomized control trial. BMC Nutr 5, 1.CrossRefGoogle ScholarPubMed
Phang, M, Lincz, LF & Garg, ML (2013) Eicosapentaenoic and docosahexaenoic acid supplementations reduce platelet aggregation and hemostatic markers differentially in men and women. J Nutr 143, 457463.CrossRefGoogle ScholarPubMed
Cui, J, Li, L, Ren, L et al. (2021) Dietary n-3 and n-6 fatty acid intakes and NAFLD: a cross-sectional study in the United States. Asia Pac J Clin Nutr 30, 8798.Google ScholarPubMed
Gabbs, M, Zahradka, P, Taylor, CG et al. (2021) Time course and sex effects of α-linolenic acid-rich and DHA-rich supplements on human plasma oxylipins: a randomized double-blind crossover trial. J Nutr 151, 513522.CrossRefGoogle ScholarPubMed
Ferdouse, A, Leng, S, Winter, T et al. (2019) Dietary n-6 and n-3 PUFA alter the free oxylipin profile differently in male and female rat hearts. Br J Nutr 122, 252261.CrossRefGoogle ScholarPubMed
Pauls, SD, Ragheb, M, Winter, T et al. (2020) Spleen oxylipin and polyunsaturated fatty acid profiles are altered by dietary source of polyunsaturated fatty acid and by sex. Lipids 55, 261270.CrossRefGoogle ScholarPubMed
Leng, S, Winter, T & Aukema, HM (2017) Dietary LA and sex effects on oxylipin profiles in rat kidney, liver, and serum differ from their effects on PUFAs. J Lipid Res 58, 17021712.CrossRefGoogle ScholarPubMed
Ferdouse, A, Leng, S, Winter, T et al. (2019) The brain oxylipin profile is resistant to modulation by dietary n-6 and n-3 polyunsaturated fatty acids in male and female rats. Lipids 54, 6780.CrossRefGoogle ScholarPubMed
Zamora-Ros, R, Achaintre, D, Rothwell, JA et al. (2016) Urinary excretions of 34 dietary polyphenols and their associations with lifestyle factors in the EPIC cohort study. Sci Rep 6, 26905.CrossRefGoogle ScholarPubMed
Gibney, ER, Milenkovic, D, Combet, E et al. (2019) Factors influencing the cardiometabolic response to (poly)phenols and phytosterols: a review of the COST action POSITIVe activities. Eur J Nutr 58, 3747.CrossRefGoogle ScholarPubMed
Margalef, M, Pons, Z, Iglesias-Carres, L et al. (2016) Gender-related similarities and differences in the body distribution of grape seed flavanols in rats. Mol Nutr Food Res 60, 760772.CrossRefGoogle ScholarPubMed
Dennison, JL, Volmar, CH, Modarresi, F et al. (2022) JOTROL, a novel formulation of resveratrol, shows beneficial effects in the 3xTg-AD mouse model. J Alzheimers Dis 86, 173190.CrossRefGoogle ScholarPubMed
Kim, SA, Kim, J, Jun, S et al. (2020) Association between dietary flavonoid intake and obesity among adults in Korea. Appl Physiol Nutr Metab 45, 203212.CrossRefGoogle ScholarPubMed
Vors, C, Rancourt-Bouchard, M, Couillard, C et al. (2021) Sex may modulate the effects of combined polyphenol extract and L-citrulline supplementation on ambulatory blood pressure in adults with prehypertension: a randomized controlled trial. Nutrients 13.CrossRefGoogle ScholarPubMed
Fagundes, MA, Silva, ARC, Fernandes, GA et al. (2022) Dietary polyphenol intake and gastric cancer: a systematic review and meta-analysis. Cancers (Basel) 14.CrossRefGoogle ScholarPubMed
Sharma, P, Wu, G, Kumaraswamy, D et al. (2021) Sex-dependent effects of 7,8-dihydroxyflavone on metabolic health are associated with alterations in the host gut microbiome. Nutrients 13.CrossRefGoogle ScholarPubMed
Goetz, ME, Judd, SE, Hartman, TJ et al. (2016) Flavanone intake is inversely associated with risk of incident ischemic stroke in the REasons for Geographic and Racial Differences in Stroke (REGARDS) study. J Nutr 146, 22332243.CrossRefGoogle ScholarPubMed
Clark, M, Centner, AM, Ukhanov, V et al. (2022) Gallic acid ameliorates atherosclerosis and vascular senescence and remodels the microbiome in a sex-dependent manner in ApoE(−/−) mice. J Nutr Biochem 110, 109132.CrossRefGoogle Scholar
Domjanić Drozdek, S, Odeh, D, Đikić, D et al. (2022) The effects of nettle extract consumption on liver PPARs, SIRT1, ACOX1 and blood lipid levels in male and female C57Bl6 mice. Nutrients 14.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Preclinical evidence for Western-style diet-related sex differences

Figure 1

Table 2. Human evidence for Western-style diet-related sex differences

Figure 2

Table 3. Human evidence for Mediterranean style diet-related sex differences