Next Article in Journal
In Vitro Anti-Inflammatory and Vasculoprotective Effects of Red Cell Extract from the Black Sea Urchin Arbacia lixula
Next Article in Special Issue
Sex, Nutrition, and NAFLD: Relevance of Environmental Pollution
Previous Article in Journal
Nutrient Patterns and Risk of Osteopenia in Postmenopausal Women
Previous Article in Special Issue
High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Diet and Fertility Status: Relevance in Health and Disease

Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
Nutrients 2023, 15(7), 1669; https://doi.org/10.3390/nu15071669
Submission received: 21 March 2023 / Accepted: 28 March 2023 / Published: 29 March 2023
(This article belongs to the Special Issue Diet and Fertility Status: Relevance in Health and Disease)
The prevalence of obesity and other metabolic disorders is increasing worldwide [1], leading to detrimental effects on human health, including reproductive performance, and thus affecting the quality of life of a large portion of society [2,3]. Increasing evidence suggests that the fertility status of males and females is linked to several metabolic and inflammatory diseases, in addition to dietary habits.
Throughout evolution, mechanisms have developed to sense nutrients, store energy in case of food abundance and adapt physiological responses to nutrient availability, preventing reproduction in nutrient-poor environments; this strict interplay reached its highest degree of complexity in female mammals [4]. Females are responsible for the energy costs of reproduction, and thus regulatory mechanisms which modulate the interplay with metabolic homeostasis have been positively selected and further perfected during evolution to guarantee procreation only in the presence of favorable energy conditions and to tune the overall metabolism to the demanding energy needs of reproduction [4,5].
In the current obesogenic world, this evolutionary strategy has negative effects on overall health and is also impacting fertility, especially in women, who thus lose the metabolic advantages over men guaranteed by female hormones [4,5,6]. In this view, nutrients cannot be merely considered a source of energy, as they exert a bioactive role by acting on several signaling pathways [7].
As summarized in the review by Jurczewska et al. [8], ovulation-dependent female fertility is influenced by several diet-related factors, especially the excessive intake of high-glycemic-index carbohydrates, large amounts of animal protein, saturated fatty acids, and trans fatty acids. It is known that high-fat diet (HFD) regimens negatively impact on several reproductive processes, such as folliculogenesis, oogenesis, and embryo development/implantation, leading to female infertility and transgenerational disorders [9]. Although our currently fragmentary knowledge limits the full comprehension of the underlying mechanisms, current evidence suggests that the negative impact of HFDs on fertility may be due to a direct action on reproductive somatic and germinal cells and/or to an indirect effect mediated by endocrine, metabolic, and immune signaling [9]. The burden of chronic inflammatory diseases, including Crohn’s disease, negatively impact on female reproduction, potentially contributing to irregular menstrual cyclicity, implantation failure, and other negative fertility outcomes [10,11].
In women, subfertility and low success of assisted reproductive technology (ART) have been also associated with low trace elements of copper, selenium, and zinc [12], showing that the proper dietary intake of these elements is essential for reproductive health.
In men, over-exposure to heavy metals such as Cadmium (Cd), which can also occur through the consumption of contaminated water and food, induces structural and functional testicular damage due to oxidative stress, inflammation, and apoptosis [13].
Among the several risk factors for infertility [14], gestational exposure to endocrine disruptors, such as bisphenol A (BPA), in combination with HFD may lead to impaired spermatogenesis in F1 and F2 offspring [15], supporting evidence on the transgenerational inheritance of reproductive disorders as well as of metabolic diseases [16,17,18]. With this in mind, studies aiming to unravel the long-term effects of maternal nutrition [19] may help to further elucidate the relevance and the mechanisms through which gestational nutrients can program metabolic and reproductive functions later in life, as well as in offspring.
Dietary interventions might represent the most promising and invaluable strategy to limit or prevent metabolic and inflammatory complications associated with, or consequent to, subfertility [10,11,20]. Given the adverse effects of inflammation on reproductive processes, anti-inflammatory diets rich in monounsaturated (MUFA) and ω-3 polyunsaturated (PUFA) fatty acids and flavonoids, and poor in red and processed meat, such as the Mediterranean diet, may improve fertility and ART success in women [10,11]. In insulin-resistant PCOS (polycystic ovary syndrome) adolescents with obesity, lifestyle changes and appropriate early dietary interventions aiming to reduce insulin resistance (i.e., diets rich in carbohydrate with low glycemic index, plant protein, MUFA and PUFA, folic acid, vitamin D, antioxidants, and iron) are recommended approaches to restore ovulation and to protect fertility [20].
In males, moderate calorie restriction may have the potential to improve the metabolic profile (i.e., plasma glycemia, glucose-regulating hormones) and reproductive parameters (i.e., plasma testosterone levels, sperm quality, male sexual behavior) [21]. Dietary strategies such as the intake of nutraceuticals, particularly abundant in the Mediterranean diet, can be recommended against Cd-induced testicular injury [13].
Besides fertility, environmentally induced abnormalities in the reproductive cycle should be considered as an important risk factor for female metabolic health. Accordingly, women with impaired or null reproductive cycles (i.e., PCOS women) experience increased incidence of metabolic/inflammatory pathologies, such as non-alcoholic fatty liver disease (NAFLD), metabolic syndrome (MetS), and cardiovascular diseases (CVDs) [4,22]. Nevertheless, more than fertility status per se, the physiological, rhythmic changes of estrogen levels during the reproductive cycle are essential to program and to preserve metabolic homeostasis in females [23,24]. Notably, the lack of the receptor that mediates estrogen action in the liver (hepatic estrogen receptor alpha, ERα) increases female vulnerability to HFD-induced metabolic alterations [25] and an impaired regulation of hepatokines is involved [26]. An alteration of hormonal signaling can have detrimental effects, especially in the female liver, where changes in the transcriptional activity of ERα during the fertile cycle enable the regulation of lipid metabolism according to the energy needs of each reproductive phase [23,24], contributing to sex differences in the regulation of hepatic metabolism [5,27] and in the susceptibility to liver diseases [22,25,28]. The disruption of this hepato–ovarian axis in ovariectomized females leads, indeed, to altered hepatic metabolism [23], which can be, at least in part, rescued by dietary amino acids [29], further demonstrating the potential of nutritional approaches in improving metabolic health when compromised by altered fertility status.
Overall, this Special Issue provides compelling evidence of the relevance of the nutritional status on the mutual relationship between fertility and metabolic homeostasis, as well as between subfertility and metabolic disorders both in males and females. More studies are required to unravel the underlying mechanisms of action and explore the potential of sex-specific nutritional strategies to preserve metabolic homeostasis and fertility in both sexes.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Chew, N.W.S.; Ng, C.H.; Tan, D.J.H.; Kong, G.; Lin, C.; Chin, Y.H.; Lim, W.H.; Huang, D.Q.; Quek, J.; Fu, C.E.; et al. The Global Burden of Metabolic Disease: Data from 2000 to 2019. Cell Metab. 2023, 35, 414–428.e3. [Google Scholar] [CrossRef]
  2. Farquhar, C.M.; Bhattacharya, S.; Repping, S.; Mastenbroek, S.; Kamath, M.S.; Marjoribanks, J.; Boivin, J. Female Subfertility. Nat. Rev. Dis. Primer 2019, 5, 7. [Google Scholar] [CrossRef] [PubMed]
  3. Agarwal, A.; Baskaran, S.; Parekh, N.; Cho, C.-L.; Henkel, R.; Vij, S.; Arafa, M.; Panner Selvam, M.K.; Shah, R. Male Infertility. Lancet Lond. Engl. 2021, 397, 319–333. [Google Scholar] [CrossRef] [PubMed]
  4. Della Torre, S.; Benedusi, V.; Fontana, R.; Maggi, A. Energy Metabolism and Fertility—A Balance Preserved for Female Health. Nat. Rev. Endocrinol. 2014, 10, 13–23. [Google Scholar] [CrossRef] [PubMed]
  5. Della Torre, S.; Maggi, A. Sex Differences: A Resultant of an Evolutionary Pressure? Cell Metab. 2017, 25, 499–505. [Google Scholar] [CrossRef] [Green Version]
  6. Maggi, A.; Della Torre, S. Sex, Metabolism and Health. Mol. Metab. 2018, 15, 3–7. [Google Scholar] [CrossRef] [PubMed]
  7. Fontana, R.; Della Torre, S. The Deep Correlation between Energy Metabolism and Reproduction: A View on the Effects of Nutrition for Women Fertility. Nutrients 2016, 8, 87. [Google Scholar] [CrossRef]
  8. Jurczewska, J.; Szostak-Węgierek, D. The Influence of Diet on Ovulation Disorders in Women—A Narrative Review. Nutrients 2022, 14, 1556. [Google Scholar] [CrossRef]
  9. Di Berardino, C.; Peserico, A.; Capacchietti, G.; Zappacosta, A.; Bernabò, N.; Russo, V.; Mauro, A.; El Khatib, M.; Gonnella, F.; Konstantinidou, F.; et al. High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models. Nutrients 2022, 14, 4341. [Google Scholar] [CrossRef]
  10. Vincenti, A.; Loperfido, F.; De Giuseppe, R.; Manuelli, M.; Bosoni, D.; Righi, A.; Nappi, R.E.; Cena, H. Crohn’s Disease and Female Infertility: Can Nutrition Play a Supporting Role? Nutrients 2022, 14, 2423. [Google Scholar] [CrossRef] [PubMed]
  11. Alesi, S.; Villani, A.; Mantzioris, E.; Takele, W.W.; Cowan, S.; Moran, L.J.; Mousa, A. Anti-Inflammatory Diets in Fertility: An Evidence Review. Nutrients 2022, 14, 3914. [Google Scholar] [CrossRef] [PubMed]
  12. Schmalbrock, L.J.; Weiss, G.; Rijntjes, E.; Reinschissler, N.; Sun, Q.; Schenk, M.; Schomburg, L. Pronounced Trace Element Variation in Follicular Fluids of Subfertile Women Undergoing Assisted Reproduction. Nutrients 2021, 13, 4134. [Google Scholar] [CrossRef]
  13. Marini, H.R.; Micali, A.; Squadrito, G.; Puzzolo, D.; Freni, J.; Antonuccio, P.; Minutoli, L. Nutraceuticals: A New Challenge against Cadmium-Induced Testicular Injury. Nutrients 2022, 14, 663. [Google Scholar] [CrossRef]
  14. Xu, W.; You, Y.; Yu, T.; Li, J. Insights into Modifiable Risk Factors of Infertility: A Mendelian Randomization Study. Nutrients 2022, 14, 4042. [Google Scholar] [CrossRef]
  15. Ho, S.-M.; Rao, R.; Ouyang, B.; Tam, N.N.C.; Schoch, E.; Song, D.; Ying, J.; Leung, Y.-K.; Govindarajah, V.; Tarapore, P. Three-Generation Study of Male Rats Gestationally Exposed to High Butterfat and Bisphenol A: Impaired Spermatogenesis, Penetrance with Reduced Severity. Nutrients 2021, 13, 3636. [Google Scholar] [CrossRef] [PubMed]
  16. Mimouni, N.E.H.; Paiva, I.; Barbotin, A.-L.; Timzoura, F.E.; Plassard, D.; Le Gras, S.; Ternier, G.; Pigny, P.; Catteau-Jonard, S.; Simon, V.; et al. Polycystic Ovary Syndrome Is Transmitted via a Transgenerational Epigenetic Process. Cell Metab. 2021, 33, 513–530.e8. [Google Scholar] [CrossRef] [PubMed]
  17. Franzago, M.; Santurbano, D.; Vitacolonna, E.; Stuppia, L. Genes and Diet in the Prevention of Chronic Diseases in Future Generations. Int. J. Mol. Sci. 2020, 21, 2633. [Google Scholar] [CrossRef]
  18. King, S.E.; Skinner, M.K. Epigenetic Transgenerational Inheritance of Obesity Susceptibility. Trends Endocrinol. Metab. TEM 2020, 31, 478–494. [Google Scholar] [CrossRef]
  19. Mahanani, M.R.; Abderbwih, E.; Wendt, A.S.; Deckert, A.; Antia, K.; Horstick, O.; Dambach, P.; Kohler, S.; Winkler, V. Long-Term Outcomes of in Utero Ramadan Exposure: A Systematic Literature Review. Nutrients 2021, 13, 4511. [Google Scholar] [CrossRef]
  20. Calcaterra, V.; Verduci, E.; Cena, H.; Magenes, V.C.; Todisco, C.F.; Tenuta, E.; Gregorio, C.; De Giuseppe, R.; Bosetti, A.; Di Profio, E.; et al. Polycystic Ovary Syndrome in Insulin-Resistant Adolescents with Obesity: The Role of Nutrition Therapy and Food Supplements as a Strategy to Protect Fertility. Nutrients 2021, 13, 1848. [Google Scholar] [CrossRef]
  21. De Jesús, P.L.; Arenas-Ríos, E.; Ruíz-Ramos, M.; Flores-Alonso, J.C.; Mendoza-Núñez, V.M.; Arrieta-Cruz, I.; Arteaga-Silva, M. Effect of Chronic Moderate Caloric Restriction on the Reproductive Function in Aged Male Wistar Rats. Nutrients 2022, 14, 1256. [Google Scholar] [CrossRef]
  22. Della Torre, S. Non-Alcoholic Fatty Liver Disease as a Canonical Example of Metabolic Inflammatory-Based Liver Disease Showing a Sex-Specific Prevalence: Relevance of Estrogen Signaling. Front. Endocrinol. 2020, 11, 572490. [Google Scholar] [CrossRef]
  23. Villa, A.; Della Torre, S.; Stell, A.; Cook, J.; Brown, M.; Maggi, A. Tetradian Oscillation of Estrogen Receptor Is Necessary to Prevent Liver Lipid Deposition. Proc. Natl. Acad. Sci. USA 2012, 109, 11806–11811. [Google Scholar] [CrossRef] [Green Version]
  24. Della Torre, S.; Mitro, N.; Fontana, R.; Gomaraschi, M.; Favari, E.; Recordati, C.; Lolli, F.; Quagliarini, F.; Meda, C.; Ohlsson, C.; et al. An Essential Role for Liver ERα in Coupling Hepatic Metabolism to the Reproductive Cycle. Cell Rep. 2016, 15, 360–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Meda, C.; Barone, M.; Mitro, N.; Lolli, F.; Pedretti, S.; Caruso, D.; Maggi, A.; Della Torre, S. Hepatic ERα Accounts for Sex Differences in the Ability to Cope with an Excess of Dietary Lipids. Mol. Metab. 2020, 32, 97–108. [Google Scholar] [CrossRef]
  26. Meda, C.; Dolce, A.; Vegeto, E.; Maggi, A.; Della Torre, S. ERα-Dependent Regulation of Adropin Predicts Sex Differences in Liver Homeostasis during High-Fat Diet. Nutrients 2022, 14, 3262. [Google Scholar] [CrossRef] [PubMed]
  27. Della Torre, S.; Mitro, N.; Meda, C.; Lolli, F.; Pedretti, S.; Barcella, M.; Ottobrini, L.; Metzger, D.; Caruso, D.; Maggi, A. Short-Term Fasting Reveals Amino Acid Metabolism as a Major Sex-Discriminating Factor in the Liver. Cell Metab. 2018, 28, 256–267.e5. [Google Scholar] [CrossRef] [Green Version]
  28. Della Torre, S. Beyond the X Factor: Relevance of Sex Hormones in NAFLD Pathophysiology. Cells 2021, 10, 2502. [Google Scholar] [CrossRef]
  29. Della Torre, S.; Benedusi, V.; Pepe, G.; Meda, C.; Rizzi, N.; Uhlenhaut, N.H.; Maggi, A. Dietary Essential Amino Acids Restore Liver Metabolism in Ovariectomized Mice via Hepatic Estrogen Receptor α. Nat. Commun. 2021, 12, 6883. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Della Torre, S. Diet and Fertility Status: Relevance in Health and Disease. Nutrients 2023, 15, 1669. https://doi.org/10.3390/nu15071669

AMA Style

Della Torre S. Diet and Fertility Status: Relevance in Health and Disease. Nutrients. 2023; 15(7):1669. https://doi.org/10.3390/nu15071669

Chicago/Turabian Style

Della Torre, Sara. 2023. "Diet and Fertility Status: Relevance in Health and Disease" Nutrients 15, no. 7: 1669. https://doi.org/10.3390/nu15071669

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

Article Metrics

Back to TopTop