Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Animal Models

Nonnutritive sweetener consumption during pregnancy, adiposity, and adipocyte differentiation in offspring: evidence from humans, mice, and cells

Abstract

Background

Obesity often originates in early life, and is linked to excess sugar intake. Nonnutritive sweeteners (NNS) are widely consumed as “healthier” alternatives to sugar, yet recent evidence suggests NNS may adversely influence weight gain and metabolic health. The impact of NNS during critical periods of early development has rarely been studied. We investigated the effect of prenatal NNS exposure on postnatal adiposity and adipocyte development.

Methods

In the CHILD birth cohort (N = 2298), we assessed maternal NNS beverage intake during pregnancy and child body composition at 3 years, controlling for maternal BMI and other potential confounders. To investigate causal mechanisms, we fed NNS to pregnant C57BL6J mice at doses relevant to human consumption (42 mg/kg/day aspartame or 6.3 mg/kg/day sucralose), and assessed offspring until 12 weeks of age for: body weight, adiposity, adipose tissue morphology and gene expression, glucose and insulin tolerance. We also studied the effect of sucralose on lipid accumulation and gene expression in cultured 3T3-L1 pre-adipocyte cells.

Results

In the CHILD cohort, children born to mothers who regularly consumed NNS beverages had elevated body mass index (mean z-score difference +0.23, 95% CI 0.05–0.42 for daily vs. no consumption, adjusted for maternal BMI). In mice, maternal NNS caused elevated body weight, adiposity, and insulin resistance in offspring, especially in males (e.g., 47% and 15% increase in body fat for aspartame and sucralose vs. controls, p < 0.001). In cultured adipocytes, sucralose exposure at early stages of differentiation caused increased lipid accumulation and expression of adipocyte differentiation genes (e.g., C/EBP-α, FABP4, and FASN). These genes were also upregulated in adipose tissue of male mouse offspring born to sucralose-fed dams.

Conclusion

By triangulating evidence from humans, mice, and cultured adipocytes, this study provides new evidence that maternal NNS consumption during pregnancy may program obesity risk in offspring through effects on adiposity and adipocyte differentiation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Maternal consumption of NNS-sweetened beverages and child body mass index (BMI) at 3 years of age in the CHILD cohort.
Fig. 2: Body composition in male and female mouse offspring of dams fed sucrose, aspartame or sucralose during pregnancy and lactation.
Fig. 3: Glucose tolerance and insulin sensitivity in 10-week-old male and female mouse offspring of dams fed sucrose, aspartame or sucralose during pregnancy and lactation.
Fig. 4: Effect of sucralose on 3T3-L1 adipocyte differentiation in vitro.
Fig. 5: Sucralose increases the expression of pro-adipogenic regulators, fat storage, and mobilization genes in 3T3-L1 cells in vitro.
Fig. 6: Sucralose increases the expression of pro-adipogenic regulators and fat storage and mobilization genes in mouse offspring adipose tissue in vivo.

Similar content being viewed by others

References

  1. Abarca-Gomez L, Abdeen ZA, Hamid ZA, Abu-Rmeileh NM, Acosta-Cazares B, Acuin C, et al. Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults. Lancet. 2017;390:2627–42.

    Google Scholar 

  2. Agarwal P, Morriseau TS, Kereliuk SM, Doucette CA, Wicklow BA, Dolinsky VW. Maternal obesity, diabetes during pregnancy and epigenetic mechanisms that influence the developmental origins of cardiometabolic disease in the offspring. Crit Rev Clin Lab Sci. 2018;55:71–101.

    CAS  PubMed  Google Scholar 

  3. Borengasser SJ, Zhong Y, Kang P, Lindsey F, Ronis MJ, Badger TM, et al. Maternal obesity enhances white adipose tissue differentiation and alters genome-scale DNA methylation in male rat offspring. Endocrinology. 2013;154:4113–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Guan H, Arany E, van Beek JP, Chamson-Reig A, Thyssen S, Hill DJ, et al. Adipose tissue gene expression profiling reveals distinct molecular pathways that define visceral adiposity in offspring of maternal protein-restricted rats. Am J Physiol Endocrinol Metab. 2005;288:E663–73.

    CAS  PubMed  Google Scholar 

  5. Liang X, Yang Q, Fu X, Rogers CJ, Wang B, Pan H, et al. Maternal obesity epigenetically alters visceral fat progenitor cell properties in male offspring mice. J Physiol. 2016;594:4453–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Wen J, Hong Q, Wang X, Zhu L, Wu T, Xu P, et al. The effect of maternal vitamin D deficiency during pregnancy on body fat and adipogenesis in rat offspring. Sci Rep. 2018;8:365.

    PubMed  PubMed Central  Google Scholar 

  7. Yang QY, Liang JF, Rogers CJ, Zhao JX, Zhu MJ, Du M. Maternal obesity induces epigenetic modifications to facilitate Zfp423 expression and enhance adipogenic differentiation in fetal mice. Diabetes. 2013;62:3727–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Hu FB. Resolved: there is sufficient scientific evidence that decreasing sugar-sweetened beverage consumption will reduce the prevalence of obesity and obesity-related diseases. Obes Rev. 2013;14:606–19.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Malik VS, Schulze MB, Hu FB. Intake of sugar-sweetened beverages and weight gain: a systematic review. Am J Clin Nutr. 2006;84:274–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Seferidi P, Millett C, Laverty AA. Sweetened beverage intake in association to energy and sugar consumption and cardiometabolic markers in children. Pediatr Obes. 2018;13:195–203.

    CAS  PubMed  Google Scholar 

  11. Vos MB, Kaar JL, Welsh JA, Van Horn LV, Feig DI, Anderson CAM, et al. Added sugars and cardiovascular disease risk in children: a scientific statement from the American Heart Association. Circulation. 2017;135:e1017–34.

    CAS  PubMed  Google Scholar 

  12. Fitch C, Keim KS. Position of the Academy of Nutrition and Dietetics: use of nutritive and nonnutritive sweeteners. J Acad Nutr Diet. 2012;112:739–58.

    PubMed  Google Scholar 

  13. Gardner C, Wylie-Rosett J, Gidding SS, Steffen LM, Johnson RK, Reader D, et al. Nonnutritive sweeteners: current use and health perspectives: a scientific statement from the American Heart Association and the American Diabetes Association. Diabetes Care. 2012;35:1798–808.

    PubMed  PubMed Central  Google Scholar 

  14. Azad MB, Sharma AK, de Souza RJ, Dolinsky VW, Becker AB, Mandhane PJ, et al. Association between artificially sweetened beverage consumption during pregnancy and infant body mass index. JAMA. 2016;170:662–70.

    Google Scholar 

  15. Sylvetsky AC, Figueroa J, Rother KI, Goran MI, Welsh JA. Trends in low-calorie sweetener consumption among pregnant women in the United States. Curr Dev Nutr. 2019;3:nzz004.

    PubMed  PubMed Central  Google Scholar 

  16. Zhu Y, Olsen SF, Mendola P, Halldorsson TI, Rawal S, Hinkle SN, et al. Maternal consumption of artificially sweetened beverages during pregnancy, and offspring growth through 7 years of age: a prospective cohort study. Int J Epidemiol. 2017;46:1499–508.

    PubMed  PubMed Central  Google Scholar 

  17. Azad MB, Abou-Setta AM, Chauhan BF, Rabbani R, Lys J, Copstein L, et al. Nonnutritive sweeteners and cardiometabolic health: a systematic review and meta-analysis of randomized controlled trials and prospective cohort studies. Can Med Assoc J. 2017;189:E929–39.

    Google Scholar 

  18. Pearlman M, Obert J, Casey L. The association between artificial sweeteners and obesity. Curr Gastroenterol Rep. 2017;19:64.

    PubMed  Google Scholar 

  19. Toews I, Lohner S, Kullenberg de Gaudry D, Sommer H, Meerpohl JJ. Association between intake of non-sugar sweeteners and health outcomes: systematic review and meta-analyses of randomised and non-randomised controlled trials and observational studies. BMJ. 2019;364:k4718.

    PubMed  PubMed Central  Google Scholar 

  20. Archibald AJ, Dolinsky VW, Azad MB. Early-life exposure to non-nutritive sweeteners and the developmental origins of childhood obesity: global evidence from human and rodent studies. Nutrients. 2018;10:194.

    PubMed Central  Google Scholar 

  21. Gillman MW, Rifas-Shiman SL, Fernandez-Barres S, Kleinman K, Taveras EM, Oken E. Beverage intake during pregnancy and childhood adiposity. Pediatrics. 2017;140. https://pediatrics.aappublications.org/content/140/2/e20170031.long.

  22. Araujo JR, Martel F, Keating E. Exposure to non-nutritive sweeteners during pregnancy and lactation: impact in programming of metabolic diseases in the progeny later in life. Reprod Toxicol. 2014;49:196–201.

    CAS  PubMed  Google Scholar 

  23. Collison KS, Makhoul NJ, Zaidi MZ, Saleh SM, Andres B, Inglis A, et al. Gender dimorphism in aspartame-induced impairment of spatial cognition and insulin sensitivity. PLoS ONE. 2012;7:e31570.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Olivier-Van Stichelen S, Rother KI, Hanover JA. Maternal exposure to non-nutritive sweeteners impacts progeny’s metabolism and microbiome. Front Microbiol. 2019;10:1360.

    PubMed  PubMed Central  Google Scholar 

  25. von Poser Toigo E, Huffell AP, Mota CS, Bertolini D, Pettenuzzo LF, Dalmaz C. Metabolic and feeding behavior alterations provoked by prenatal exposure to aspartame. Appetite. 2015;87:168–74.

    Google Scholar 

  26. Soffritti M, Belpoggi F, Manservigi M, Tibaldi E, Lauriola M, Falcioni L, et al. Aspartame administered in feed, beginning prenatally through life span, induces cancers of the liver and lung in male Swiss mice. Am J Ind Med. 2010;53:1197–206.

    CAS  PubMed  Google Scholar 

  27. Simon BR, Parlee SD, Learman BS, Mori H, Scheller EL, Cawthorn WP, et al. Artificial sweeteners stimulate adipogenesis and suppress lipolysis independently of sweet taste receptors. J Biol Chem. 2013;288:32475–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Masubuchi Y, Nakagawa Y, Ma J, Sasaki T, Kitamura T, Yamamoto Y, et al. A novel regulatory function of sweet taste-sensing receptor in adipogenic differentiation of 3T3-L1 cells. PLoS ONE. 2013;8:e54500.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Pandurangan M, Park J, Kim E. Aspartame downregulates 3T3-L1 differentiation. In Vitro Cell Dev Biol Anim. 2014;50:851–7.

    CAS  PubMed  Google Scholar 

  30. Subbarao P, Anand SS, Becker AB, Befus AD, Brauer M, Brook JR, et al. The Canadian Healthy Infant Longitudinal Development (CHILD) Study: examining developmental origins of allergy and asthma. Thorax. 2015;70:998–1000.

    PubMed  Google Scholar 

  31. Patterson RE, Kristal AR, Tinker LF, Carter RA, Bolton MP, Agurs-Collins T. Measurement characteristics of the Women’s Health Initiative food frequency questionnaire. Ann Epidemiol. 1999;9:178–87.

    CAS  PubMed  Google Scholar 

  32. Qiu C, Coughlin KB, Frederick IO, Sorensen TK, Williams MA. Dietary fiber intake in early pregnancy and risk of subsequent preeclampsia. Am J Hypertens. 2008;21:903–9.

    CAS  PubMed  Google Scholar 

  33. Maslova E, Strom M, Olsen SF, Halldorsson TI. Consumption of artificially-sweetened soft drinks in pregnancy and risk of child asthma and allergic rhinitis. PLoS ONE. 2013;8:e57261.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Guenther PM, Casavale KO, Reedy J, Kirkpatrick SI, Hiza HA, Kuczynski KJ, et al. Update of the Healthy Eating Index: HEI-2010. J Acad Nutr Diet. 2013;113:569–80.

    PubMed  Google Scholar 

  35. Pereira TJ, Fonseca MA, Campbell KE, Moyce BL, Cole LK, Hatch GM, et al. Maternal obesity characterized by gestational diabetes increases the susceptibility of rat offspring to hepatic steatosis via a disrupted liver metabolome. J Physiol. 2015;593:3181–97.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Dolinsky VW, Gilham D, Hatch GM, Agellon LB, Lehner R, Vance DE. Regulation of triacylglycerol hydrolase expression by dietary fatty acids and peroxisomal proliferator-activated receptors. Biochim Biophys Acta. 2003;1635:20–8.

    CAS  PubMed  Google Scholar 

  37. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3:Research0034.

    PubMed  PubMed Central  Google Scholar 

  38. Guariguata L, Linnenkamp U, Beagley J, Whiting DR, Cho NH. Global estimates of the prevalence of hyperglycaemia in pregnancy. Diabetes Res Clin Pract. 2014;103:176–85.

    CAS  PubMed  Google Scholar 

  39. Moseti D, Regassa A, Kim WK. Molecular regulation of adipogenesis and potential anti-adipogenic bioactive molecules. Int J Mol Sci. 2016;17:E124.

    PubMed  Google Scholar 

  40. Roberts A, Renwick AG, Sims J, Snodin DJ. Sucralose metabolism and pharmacokinetics in man. Food Chem Toxicol. 2000;38 Suppl 2:S31–41.

    CAS  PubMed  Google Scholar 

  41. Nettleton JE, Reimer RA, Shearer J. Reshaping the gut microbiota: Impact of low calorie sweeteners and the link to insulin resistance? Physiol Behav. 2016;164:488–93.

    CAS  PubMed  Google Scholar 

  42. Ruiz-Ojeda FJ, Plaza-Diaz J, Saez-Lara MJ, Gil A. Effects of sweeteners on the gut microbiota: a review of experimental studies and clinical trials. Adv Nutr. 2019;10 Suppl 1:S31–48.

    PubMed  PubMed Central  Google Scholar 

  43. Asnicar F, Manara S, Zolfo M, Truong DT, Scholz M, Armanini F, et al. Studying vertical microbiome transmission from mothers to infants by strain-level metagenomic profiling. mSystems. 2017;2:e00164–16.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Le Doare K, Holder B, Bassett A, Pannaraj PS. Mother’s milk: a purposeful contribution to the development of the infant microbiota and immunity. Front Immunol. 2018;9:361.

    PubMed  PubMed Central  Google Scholar 

  45. Liou AP, Paziuk M, Luevano JM Jr, Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5:178ra41.

    PubMed  PubMed Central  Google Scholar 

  46. Meijnikman AS, Gerdes VE, Nieuwdorp M, Herrema H. Evaluating causality of gut microbiota in obesity and diabetes in humans. Endocr Rev. 2018;39:133–53.

    PubMed  Google Scholar 

  47. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31.

    PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to all the families who took part in the CHILD study, and the whole CHILD team, which includes interviewers, nurses, computer and laboratory technicians, clerical workers, research scientists, volunteers, managers, and receptionists. We also acknowledge the excellent technical work of Mario Fonseca and Bo Xiang (University of Manitoba), critical review by Shirin Moossavi (University of Manitoba), and editorial assistance from John Schellenberg (University of Manitoba). A preprint of this work was posted on bioRxiv (https://doi.org/10.1101/713974).

Funding

The Canadian Institutes of Health Research (CIHR) and the Allergy, Genes and Environment Network of Centers of Excellence (AllerGen NCE) provided core support for the CHILD Study. This research was supported, in part, by the Canada Research Chairs program. MBA holds the Tier 2 Canada Research Chair in the Developmental Origins of Chronic Disease, and is a Canadian Institute for Advanced Research Fellow in the Humans and the Microbiome Program. VWD holds the Allen Rouse-Manitoba Medical Services Foundation Basic Scientist Award. MMT is the recipient of a Research Manitoba/CHRIM studentship. This research was supported by a Children’s Hospital Research Institute of Manitoba Grant, a CIHR Operating Grant #151540, and a CIHR Environments, Genes and Chronic Disease Team Grant #144626. These entities had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the paper.

Author information

Authors and Affiliations

Authors

Contributions

MBA and VWD conceived of the study design, obtained funding for this research, and drafted the paper. MRS, PS, TJM, SET, PJM, and ABB obtained funding for and oversaw recruitment of the CHILD cohort and data collection. AA performed the statistical analysis of clinical data from the CHILD cohort under the supervision of MBA. RJS contributed nutritional expertise. MMT, AH, and KGC performed mouse and cell culture experiments under the supervision of VWD. All authors critically reviewed and approved the paper. MBA had full access to the human data and VWD had full access to the mouse and adipocyte data, and take final responsibility for the decision to submit for publication.

Corresponding authors

Correspondence to Meghan B. Azad or Vernon W. Dolinsky.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Azad, M.B., Archibald, A., Tomczyk, M.M. et al. Nonnutritive sweetener consumption during pregnancy, adiposity, and adipocyte differentiation in offspring: evidence from humans, mice, and cells. Int J Obes 44, 2137–2148 (2020). https://doi.org/10.1038/s41366-020-0575-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41366-020-0575-x

Search

Quick links