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.

  • Letter
  • Published:

Food craving-like episodes during pregnancy are mediated by accumbal dopaminergic circuits

Abstract

Preparation for motherhood requires a myriad of physiological and behavioural adjustments throughout gestation to provide an adequate environment for proper embryonic development1. Cravings for highly palatable foods are highly prevalent during pregnancy2 and contribute to the maintenance and development of gestational overweight or obesity3. However, the neurobiology underlying the distinct ingestive behaviours that result from craving specific foods remain unknown. Here we show that mice, similarly to humans, experience gestational food craving-like episodes. These episodes are associated with a brain connectivity reorganization that affects key components of the dopaminergic mesolimbic circuitry, which drives motivated appetitive behaviours and facilitates the perception of rewarding stimuli. Pregnancy engages a dynamic modulation of dopaminergic signalling through neurons expressing dopamine D2 receptors in the nucleus accumbens, which directly modulate food craving-like events. Importantly, persistent maternal food craving-like behaviour has long-lasting effects on the offspring, particularly in males, leading to glucose intolerance, increased body weight and increased susceptibility to develop eating disorders and anxiety-like behaviours during adulthood. Our results reveal the cognitively motivated nature of pregnancy food cravings and advocates for moderating emotional eating during gestation to prevent deterioration of the offspring’s neuropsychological and metabolic health.

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: Sweet taste perception and food craving-like behaviour increase during pregnancy.
Fig. 2: Brain functional networks related to reward and emotions are transiently increased during pregnancy.
Fig. 3: Accumbal D2R neurons underlie pregnancy-specific food craving-like behaviour.
Fig. 4: Recurrent maternal highly palatable food craving-like behaviour deteriorates offspring health.

Similar content being viewed by others

Data availability

The data that support the findings of the study are available from the corresponding authors upon reasonable request. Source data are provided with this paper. All other data are available in the main text or the Supplementary Information.

References

  1. Talbot, L. & Maclennan, K. Physiology of pregnancy. Anaesth. Intensive Care Med. 17, 341–345 (2016).

    Article  Google Scholar 

  2. Orloff, N. C. & Hormes, J. M. Pickles and ice cream! Food cravings in pregnancy: hypotheses, preliminary evidence, and directions for future research. Front. Psychol. 5, 1076 (2014).

  3. Orloff, N. C. et al. Food cravings in pregnancy: preliminary evidence for a role in excess gestational weight gain. Appetite 105, 259–265 (2016).

    Article  PubMed  Google Scholar 

  4. Fontaine, E. Food intake and nutrition during pregnancy, lactation and weaning in the dam and offspring. Reprod. Domest. Anim. 47, 326–330 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Hadjieconomou, D. et al. Enteric neurons increase maternal food intake during reproduction. Nature 587, 455–459 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Speakman, J. R. The physiological costs of reproduction in small mammals. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363, 375–398 (2008).

    Article  PubMed  Google Scholar 

  7. Bodden, C., Hannan, A. J. & Reichelt, A. C. Of ‘junk food’ and ‘brain food’: how parental diet influences offspring neurobiology and behaviour. Trends Endocrinol. Metab. https://doi.org/10.1016/j.tem.2021.04.001 (2021).

  8. Schoonejans, J. M. & Ozanne, S. E. Developmental programming by maternal obesity: lessons from animal models. Diabet. Med. 38, e14694 (2021).

  9. Hasebe, K., Kendig, M. D. & Morris, M. J. Mechanisms underlying the cognitive and behavioural effects of maternal obesity. Nutrients 13, 240 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Todman, D. Soranus of Ephesus (AD 98–138) and the Methodist sect. J. Med. Biogr. 16, 51 (2008).

    Article  PubMed  Google Scholar 

  11. Schroeder, M. et al. A methyl-balanced diet prevents CRF-induced prenatal stress-triggered predisposition to binge eating-like phenotype. Cell Metab. 25, 1269–1281 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Hook, E. B. Dietary cravings and aversions during pregnancy. Am. J. Clin. Nutr. 31, 1355–1362 (1978).

    Article  CAS  PubMed  Google Scholar 

  13. Rosenbaum, D. L. & White, K. S. The role of anxiety in binge eating behavior: a critical examination of theory and empirical literature. Health Psychol. Res. 1, e19 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Ferrario, C. R. et al. Homeostasis meets motivation in the battle to control food intake. J. Neurosci. 36, 11469–11481 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Andermann, M. L. & Lowell, B. B. Toward a wiring diagram understanding of appetite control. Neuron 95, 757–778 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Grandjean, J. et al. Common functional networks in the mouse brain revealed by multi-centre resting-state fMRI analysis. NeuroImage 205, 116278 (2020).

    Article  PubMed  Google Scholar 

  17. Beaulieu, J.-M. & Gainetdinov, R. R. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol. Rev. 63, 182–217 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Lindgren, N. et al. Dopamine D2 receptors regulate tyrosine hydroxylase activity and phosphorylation at Ser40 in rat striatum: D2-mediated regulation of TH in striatum. Eur. J. Neurosci. 13, 773–780 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Domingos, A. I. et al. Leptin regulates the reward value of nutrient. Nat. Neurosci. 14, 1562–1568 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Azevedo, E. P. et al. A role of Drd2 hippocampal neurons in context-dependent food intake. Neuron 102, 873–886 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Luo, Y.-J. et al. Nucleus accumbens controls wakefulness by a subpopulation of neurons expressing dopamine D1 receptors. Nat. Commun. 9, 1576 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Jendryka, M. et al. Pharmacokinetic and pharmacodynamic actions of clozapine-N-oxide, clozapine, and compound 21 in DREADD-based chemogenetics in mice. Sci. Rep. 9, 4522 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Allen, J. A. et al. Discovery of arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy. Proc. Natl Acad. Sci. USA 108, 18488–18493 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Urs, N. M. et al. Distinct cortical and striatal actions of a β-arrestin-biased dopamine D2 receptor ligand reveal unique antipsychotic-like properties. Proc. Natl Acad. Sci. USA 113, E8178–E8186 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Guilloux, J.-P., Seney, M., Edgar, N. & Sibille, E. Integrated behavioral z-scoring increases the sensitivity and reliability of behavioral phenotyping in mice: relevance to emotionality and sex. J. Neurosci. Methods 197, 21–31 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Treasure, J., Duarte, T. A. & Schmidt, U. Eating disorders. Lancet 395, 899–911 (2020).

    Article  PubMed  Google Scholar 

  27. Lippert, R. N. et al. Maternal high-fat diet during lactation reprograms the dopaminergic circuitry in mice. J. Clin. Invest. 130, 3761–3776 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Peleg-Raibstein, D. et al. Enhanced sensitivity to drugs of abuse and palatable foods following maternal overnutrition. Transl. Psychiatry 6, e911 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Blau, L. E., Orloff, N. C., Flammer, A., Slatch, C. & Hormes, J. M. Food craving frequency mediates the relationship between emotional eating and excess weight gain in pregnancy. Eat. Behav. 31, 120–124 (2018).

    Article  PubMed  Google Scholar 

  30. Volkow, N. D., Wise, R. A. & Baler, R. The dopamine motive system: implications for drug and food addiction. Nat. Rev. Neurosci. 18, 741–752 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Caravaggio, F. et al. Ventral striatum binding of a dopamine D2/3 receptor agonist but not antagonist predicts normal body mass index. Biol. Psychiatry 77, 196–202 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Eisenstein, S. A. et al. Emotional eating phenotype is associated with central dopamine D2 receptor binding independent of body mass index. Sci. Rep. 5, 11283 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Domingo-Rodriguez, L. et al. A specific prelimbic-nucleus accumbens pathway controls resilience versus vulnerability to food addiction. Nat. Commun. 11, 782 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Seeley, W. W. The salience network: a neural system for perceiving and responding to homeostatic demands. J. Neurosci. 39, 9878–9882 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Naqvi, N. H., Rudrauf, D., Damasio, H. & Bechara, A. Damage to the insula disrupts addiction to cigarette smoking. Science 315, 531–534 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Faas, M. M., Melgert, B. N. & de Vos, P. A brief review on how pregnancy and sex hormones interfere with taste and food intake. Chemosens. Percept. 3, 51–56 (2010).

    Article  PubMed  Google Scholar 

  37. Yoest, K. E., Quigley, J. A. & Becker, J. B. Rapid effects of ovarian hormones in dorsal striatum and nucleus accumbens. Horm. Behav. 104, 119–129 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gong, S. et al. Targeting cre recombinase to specific neuron populations with bacterial artificial chromosome constructs. J. Neurosci. 27, 9817–9823 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Puighermanal, E. et al. Functional and molecular heterogeneity of D2R neurons along dorsal ventral axis in the striatum. Nat. Commun. 11, 1957 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sclafani, A., Zukerman, S. & Ackroff, K. Postoral glucose sensing, not caloric content, determines sugar reward in C57BL/6J mice. Chem. Senses 40, 245–258 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nickerson, L. D., Smith, S. M., Öngür, D. & Beckmann, C. F. Using dual regression to investigate network shape and amplitude in functional connectivity analyses. Front. Neurosci. 11, 115 (2017).

  42. Timper, K. et al. Mild impairment of mitochondrial OXPHOS promotes fatty acid utilization in POMC neurons and improves glucose homeostasis in obesity. Cell Rep. 25, 383–397 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Altarche-Xifro, W. et al. Functional rescue of dopaminergic neuron loss in Parkinson’s disease mice after transplantation of hematopoietic stem and progenitor cells. EBioMedicine 8, 83–95 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Fan, K. et al. Stress-induced metabolic disorder in peripheral CD4+ T cells leads to anxiety-like behavior. Cell 179, 864–879 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Leger, M. et al. Object recognition test in mice. Nat. Protoc. 8, 2531–2537 (2013).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to J. M. Marimon (Universitat de Barcelona) for mouse vasectomy procedures; A. Garcia (IDIBAPS) for assistance with mouse breeding; S. Luquet (Université de Paris), A. Quintana (Universitat Autònoma de Barcelona) and M. Schneeberger (Yale University) for valuable comments on the manuscript. This study was funded by the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 725004) and supported by: ‘la Caixa’ Foundation (ID100010434) under agreement LCF/PR/HR19/52160016 and the CERCA Programme/Generalitat de Catalunya (to M.C.); grant no. PID2019-105136RB-100, Spanish Ministry of Economy and Competitiveness (MINECO) and European Regional Development Fund (ERDF; to A.B.); Marie Skłodowska-Curie Action fellowship (H2020-MSCA-IF) NEUROPREG (grant agreement no. 891247; to R.H-T.); the Spanish Ministry of Science and Innovation, Juan de la Cierva fellowship (IJC2018-037341-I to S.R.); Miguel Servet contract (CP19/00083) from Instituto de Salud Carlos III co-financed by ERDF (to A.O.). This work was carried out in part at the Esther Koplowitz Centre.

Author information

Authors and Affiliations

Authors

Contributions

R.H.-T. and M.C. conceptualized and supervised the study, and acquired project funding. M.C. administered the project. R.H.-T., S.R., M.M.-G., M.P., I.C., A.O., A.G.G.-V., M.T., E.E., L.M.-R., J.A. and A.B. performed experiments and discussed data. E.M.-M. and G.S. performed fMRI data acquisition and analyses. R.H.-T., S.R., E.M.-M., E.V., G.S., L.M.-R., A.B. and M.C. contributed to method development and data interpretation. R.H.-T., E.M.-M., G.S. and M.C. developed the data visualizations. R.H-T. and M.C. wrote the original draft of the paper with editing and reviewing inputs from all authors.

Corresponding authors

Correspondence to Roberta Haddad-Tóvolli or Marc Claret.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review information

Peer review information

Nature Metabolism thanks Susan Ozanne and the other, anonymous, reviewers for their contribution to the peer review of this work. Primary Handling Editor: Ashley Castellanos-Jankiewicz, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 Sweet taste perception, food craving-like behaviour and anxiety-like states during pregnancy.

a, Preference for diverse sucralose concentrations (0.05–2.5 mM). Black arrow indicates the selected concentration in which virgin female mice were unable to distinguish sucralose from water (n = 5 mice per group/ average of 5 measurements per week). b, Preference for diverse sucrose concentrations (25–200 mM). Black arrow indicates the selected concentration in which virgin female mice were unable to distinguish sucrose from water (n = 5 mice per group/ average of 5 measurements per week). c, Daily food intake of chow when pregnant females (during the second and third week of pregnancy) were exposed to the two-bottle paradigm. Water (n = 32), Sucrose (n = 29), Sucralose (n = 31). d, Schematic illustration of HPF “limited access” paradigm. e, Total daily caloric intake of virgin (n = 8), pseudopregnant (n = 22) and pregnant (n = 13) mice. f, Serum progesterone levels of virgin (n = 5), pseudopregnant (n = 15) and pregnant (n = 8) mice throughout the study. g, h, Recorded parameters to assess open field performance in virgin (n = 10), pregnant (n = 12) and after pregnancy (n = 10) mice, including percentage of time spent per zone (g), and total distance travelled (h). i, j, Recorded parameters to assess dark-light box performance in virgin (n = 8), pregnant (n = 12) and after pregnancy (n = 9) mice, including the latency to cross to the light compartment (i) and the time spent in the light compartment (j). Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by one-way ANOVA with Tukey’s multiple comparisons test for c, g, h, i, j and by two-way ANOVA with Tukey’s multiple comparisons test for e, f. When the factors Group, Time and/or the interaction Group:Time were considered significant, results are shown with significant factor or the interaction effect next to it (f). BP: before pregnancy; W1: first week of pregnancy (or concomitant experimental week); W2: second week of pregnancy (or concomitant experimental week); W3: third week of pregnancy (or concomitant experimental week); AP: after pregnancy.

Source data

Extended Data Fig. 2 The architecture of the female mouse brain resting-state network during pregnancy.

Independent component spatial maps for the 13 cortical and subcortical resting-state networks of the pregnant female mouse brain. Virgins (n = 7), Pregnants (n = 8), After Pregnancy (n = 7). Representative axial, sagittal and coronal slices for each component are shown. a, Cingulate/Retrosplenial Cortex. b, Primary Somatosensory Area 1. c, Prefrontal Cortex. d, Right Thalamus. e, Primary Somatosensory Area 2. f, Left Hippocampus. g, Cortico-striatal network (motor cortex and striatum). h, Thalamus. i, Right Hippocampus. j, Primary Somatosensory Area 4. k, Ventral Striatum. l, Temporal association Area. m, Salience Network (insular cortex and midbrain).

Extended Data Fig. 3 Main dopaminergic markers and Th neuronal activity are not altered in the VTA during pregnancy.

a, Gene expression levels of dopaminergic markers, Th and Slc6a3, in the VTA of virgin (n = 5 for Th/6 for Slca3a), pregnant (n = 6) and after pregnancy (n = 4) mice. b, Representative confocal images of TH (red) and Fos (green) expression in the VTA of virgin, pregnant and after pregnancy mice. Nuclei are stained with DAPI (blue). Scale bar: 50 μm. c, Percentage of Fos positive TH neurons in the VTA of virgin (n = 3), pregnant (n = 4) and after pregnancy (n = 3) mice. d, Number of TH positive neurons throughout the VTA (bregma -3.10 to -3.70) of virgin (n = 3), pregnant (n = 3) and after pregnancy (n = 3) mice. Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by one-way ANOVA with Tukey’s multiple comparisons test for a, c, d. VTA: ventral tegmental area; Th: tyrosine hydroxylase; Slc6a3: Solute Carrier Family 6 Member 3 encoding for the dopamine transporter (DAT); Gapdh: Glyceraldehyde 3-phosphate dehydrogenase.

Source data

Extended Data Fig. 4 Dopamine receptors expression in dStri and NAc of female mice.

a, Representative confocal images of Drd1 (white), Drd2 (green) and Fos (red) mRNA expression in the dStri of virgin, pregnant and after pregnancy mice. Nuclei are stained with DAPI (blue). Scale bar: 7.5 μm. b, DStri Drd1 mRNA particles per neuron of virgin (n = 3), pregnant (n = 3) and after pregnancy (n = 3) mice. c, Percentage of Fos positive Drd1 neurons in the dStri of virgin (n = 3), pregnant (n = 3) and after pregnancy (n = 3) mice. d, DStri Drd2 mRNA particles per neuron of virgin (n = 3), pregnant (n = 3) and after pregnancy (n = 3) mice. e, Percentage of Fos positive Drd2 neurons in the dStri of virgin (n = 3), pregnant (n = 3) and after pregnancy (n = 3) mice. f, Representative confocal images of Drd1 (white), Drd2 (green) and Fos (red) mRNA expression in the NAc of virgin, pregnant and after pregnancy mice. Nuclei are stained with DAPI (blue). Scale bar: 7.5 μm. Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Statistical analysis was performed by one-way ANOVA with Tukey’s multiple comparisons test for b, c, d, e. dStri: dorsal striatum; NAc: nucleus accumbens; Drd1: dopamine receptor 1; Drd2: dopamine receptor 2.

Source data

Extended Data Fig. 5 Dopaminergic signalling is not altered in the dStri of pregnant mice.

a, Immunoblot assessing TH phosphorylation levels at residues Ser31 (p-THS31) and Ser40 (p-THS40) in dStri extract from virgin (n = 5), pregnant (n = 5) and after pregnancy (n = 5) mice. b, Immunoblot assessing G protein-dependent and non-canonical β−arrestin-dependent signalling pathways showing its downstream targets, DARPP-32 phosphorylation levels (at residue pThr34; p-D-32T34) and GSK3β phosphorylation levels (at residue pSer9; p-GSK3βS9), respectively, in dStri extract from virgin (n = 5), pregnant (n = 5) and after pregnancy (n = 5) mice. c, Complete immunoblot assessing TH phosphorylation levels at residues Ser31 (p-THS31) and Ser40 (p-THS40) in NAc extract from virgin (n = 5), pregnant (n = 5) and after pregnancy (n = 5) mice. d, Complete immunoblot assessing G protein-dependent and non-canonical β−arrestin-dependent signalling pathways showing its downstream targets, DARPP-32 phosphorylation levels (at residue pThr34; p-D-32T34) and GSK3β phosphorylation levels (at residue pSer9; p-GSK3βS9), respectively, in NAc extract from virgin (n = 5), pregnant (n = 5) and after pregnancy (n = 5) mice. e-f, Neurotransmitter (DA, DOPAC and HVA) levels (e), dopamine turnover (DOPAC to DA ratio) and dopamine storage (HVA to DA ratio) (f) in the dorsal striatum (dStri) of virgins (n = 9) and pregnant (n = 8) females. Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by one-way ANOVA with Tukey’s multiple comparisons test for a, b and by two-way ANOVA with Sidak’s multiple comparisons test for e, f. dStri: dorsal striatum; Th: tyrosine hydroxylase; DA: dopamine; PKA: protein kinase A; DARPP-32: dopamine- and cAMP-regulated phosphoprotein, Mr 32kDa; Gsk3β: glycogen synthase kinase 3 beta; NAc: nucleus accumbens; DOPAC: 3,4-Dihydroxyphenylacetic acid; HVA: homovanillic acid.

Source data

Extended Data Fig. 6 Pharmacological blockage of pan-DR receptors reverts pregnancy-specific increase in HPF consumption.

a, Schematic illustration of experimental strategy (left) and percentage of daily caloric intake (right) consumed, during the 2-hours of HPF access, by female mice treated with saline or pan-DR antagonist flupenthixol before and during the second week of pregnancy (n = 11 mice/group). b, Total daily caloric intake (chow + HPF) of female mice treated with saline or pan-DR antagonist flupenthixol before and during the second week of pregnancy (n = 11 mice/group). c-e, Recorded parameters to assess open field performance in female mice treated with saline and pan-DR antagonist flupenthixol, including global activity (c), time spent immobile (d), and total distance travelled (e) (n = 3 mice/group). Schematic of the open field test highlights center (purple) and corner (cream) zones is shown. Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by two-way ANOVA with Sidak’s multiple comparisons test for a, b, and by unpaired t-test for c, d, e. BP: before pregnancy; W2: second week of pregnancy.

Source data

Extended Data Fig. 7 Chemogenetic inhibition of accumbal, but not dorsal striatal, D2R-neurons reverts pregnancy-specific food craving-like behavior.

a, Schematic illustration of the DREADD strategy used. b-c, Representative images of mCherry reporter in Drd2Cre/+ female mice injected in the dStri (b) or in the NAc (c) with the AAV-hSyn-DIO-hM4Di-mCherry. Scale bar: 25 μm. d-e, Representative images of mCherry (red) and Fos (green) immunostaining of Drd2+/+ and Drd2Cre/+ mice bilaterally injected in the dStri (d) and in the NAc (e) with the AAV-hSyn-DIO-hM4Di-mCherry. Nuclei are stained with DAPI (blue). Scale bar: 25 μm. f, Number of Fos positive cells per section after CNO injection in the NAc and dStri of Drd2+/+ (n = 3) and Drd2Cre/+ (n = 6 for NAc/5 for dStri) mice. g-h, Representative images of mCherry (red), DARPP-32 (green) and Fos (grey) immunostaining of Drd2Cre/+ mice bilaterally injected in the dStri (g) and in the NAc (h) with the AAV-hSyn-DIO-hM4Di-mCherry. Nuclei are stained with DAPI (blue). Scale bar: 25μm. i, Percentage of Fos positive cells after CNO injection in the NAc (n = 3) and dStri (n = 3) of Drd2Cre/+ mice categorized as D2R (mCherry+ and DARPP-32+) or as D1R (only DARPP-32+). j, Schematic of experimental strategy (left) and total daily caloric intake (chow + HPF) of Drd2+/+ (n = 7) and Drd2Cre/+ (n = 5) female mice after chemogenetic inhibition of D2R neuronal activity in the NAc. k-n, Recorded parameters of open field performance in Drd2+/+ (n = 3) and Drd2Cre/+ (n = 5) female mice after NAc D2R chemogenetic inhibition, including global activity (k), total distance travelled (l), percentage of time spent per zone (m), and representative traces (n). o, Schematic illustration of the experimental strategy (left) and total daily caloric intake (chow + HPF) of Drd2+/+ (n = 4) and Drd2Cre/+ (n = 3) female mice after chemogenetic inhibition of D2R neuronal activity in the dStri. p-s, Recorded parameters to assess open field performance in Drd2+/+ (n = 11) and Drd2Cre/+ (n = 14) female mice after dStri D2R chemogenetic inhibition, including global activity (p), total distance travelled during the test (q), percentage of time spent per zone (r), and representative traces (s). Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by unpaired t-test for k, l, p, q, by two-way ANOVA with Sidak’s multiple comparisons test for f, i, j, m, o, r. dStri: dorsal striatum; NAc: nucleus accumbens; CNO: clozapine-n-oxide; BP: before pregnancy; W2: second week of pregnancy.

Source data

Extended Data Fig. 8 Pharmacological treatment with D2R biased ligand UNC9994 reverts pregnancy-specific increase in HPF consumption.

a, Schematic illustration of experimental strategy (left) and percentage of daily caloric intake consumed (right), during the 2-hours of HPF access, by female mice treated with either vehicle or D2R biased ligand UNC9994 before pregnancy (BP) and during the second week of pregnancy (W2) (n = 6 mice/group). b, Total daily caloric intake (chow + HPF) of female mice treated with either vehicle or D2R biased ligand UNC9994 before pregnancy and during the second week of pregnancy (n = 6 mice/group). c-f, Recorded parameters to assess open field performance in female mice treated with either vehicle or D2R biased ligand UNC9994, including global activity (c) (n = 4 for vehicle and 5 for UNC9994) time spent immobile (d) (n = 7 mice/group) total distance travelled (e) (n = 7 mice/group) and percentage of time spent in each compartment (f) (n = 7 mice/group). Schematic of the open field test highlights center (purple) and corner (cream) zones. Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by two-way ANOVA with Sidak’s multiple comparisons test for a, b, and by unpaired t-test for c, d, e and by two-way ANOVA with Sidak’s multiple comparisons test for f. BP: before pregnancy; W2: second week of pregnancy; VEH: vehicle.

Source data

Extended Data Fig. 9 Characterization of maternal physiology under diverse nutritional paradigms.

a, Body weight of females exposed to either ad libitum chow (n = 4), “limited access” paradigm (n = 5) and ad libitum Western diet (n = 6). Females were crossed with Chow-fed males at 12 weeks of age. b, Glucose tolerance test at E14.5-E16.5 pregnant females exposed to either ad libitum chow (n = 4), “limited access” paradigm (n = 5) and ad libitum Western diet (n = 6). c-e, Blood glucose levels (c), plasma insulin (d), and plasma leptin (e) of E14.5-E16.5 females exposed to either ad libitum Chow (n = 4), “limited access” paradigm (n = 5) and ad libitum Western diet (n = 6) after 6 hours of fasting. Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by two-way ANOVA with Tukey’s multiple comparisons test for a and b. Green P values refer to the comparison between limited access group and Western diet-fed group. Yellow P values refer to the comparison between chow-fed group and Western diet-fed group. Statistical analysis was performed by one-way ANOVA with Tukey’s multiple comparisons test for c, d, e. When the factors Diet, Age/Time and/or the interaction Age/Time:Diet were considered significant, results are shown with significant factor or the interaction effect next to it.

Source data

Extended Data Fig. 10 Predisposition to eating disorders and expression of dopaminergic markers in HPF craving-like offspring.

a-b, Total distance travelled during an open field (a) and a NORT (b) paradigm in 12-week-old male (n = 8 mice/group) and female (n = 7 Chow-O and 8 HPF-O) Chow-O (n = 5) and HPF-O (n = 5) mice. c, Schematic illustration of compulsive eating-like (CE) paradigm used to assess eating disorders predisposition in the offspring. d, Total daily caloric intake of Chow-O and HPF-O male (left panel) and female (right panel) mice exposed to a compulsive eating paradigm during adolescence (n = 5 mice per group/average of 3 measurements per week). e-j, Transcript expression of Th (n = 8 Chow-O and 7 HPF-O males/n = 5 Chow-O and 7 HPF-O females) (e) and Slc6a3 (n = 7 Chow-O and HPF-O males/n = 5 Chow-O and 7 HPF-O females) (f) in the ventral tegmental area (VTA); Drd1 (g) Drd2 (h) in the nucleus accumbens (NAc) (n = 7 Chow-O and HPF-O males/n = 5 Chow-O and 8 HPF-O females); Drd1 (n = 8 Chow-O and HPF-O males/n = 6 Chow-O and HPF-O females) (i) Drd2 (n = 8 Chow-O and HPF-O males/n = 5 Chow-O and 6 HPF-O females) (j) in the dorsal striatum (dStri) of 16-week-old male and female offspring from Chow-O (n = 5) and HPF-O (n = 5) mice. Dots in all panels represent individual sample data. Data are expressed as mean ± SEM. Exact P values are shown. Statistical analysis was performed by ANODE for a, b, e, f, g, h, i, j or ANODE followed by Benjamini–Yekutieli adjustment for d. When the factors Sex, Diet, Time and/or the interaction Sex:Diet, Sex:Time, Diet:Time, Sex:Diet:Time were considered significant, results were shown with significant factor or the interaction effect with Tukey’s post-hoc analysis within the same sex group. VTA: ventral tegmental area; NAc: nucleus accumbens; dStri: dorsal striatum; Th: tyrosine hydroxylase; Slc6a3: Solute Carrier Family 6 Member 3 encoding for the dopamine transporter (DAT); Drd1: dopamine receptor 1; Drd2: dopamine receptor 2.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Haddad-Tóvolli, R., Ramírez, S., Muñoz-Moreno, E. et al. Food craving-like episodes during pregnancy are mediated by accumbal dopaminergic circuits. Nat Metab 4, 424–434 (2022). https://doi.org/10.1038/s42255-022-00557-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00557-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing