Introduction

In the last 45 years (1973–2018), global semen quality has decreased by more than 62%1. Globally, the number of people with obesity and overweight are on the rise2. Body mass index (BMI) and the quality of semen were negatively correlated in population surveys3,4. Animal experiments also confirmed that a high-fat diet (HFD) induced testicular injury and spermatogenesis impairment in mice5,6. Paternal Origins of Health and Disease (POHaD) theory manifests that paternal exposure to adverse factors leads to the occurrence and development of adult offspring with chronic diseases7,8. Studies showed that paternal obesity impaired the structure of testicular seminiferous tubules and reduced the number of epididymal sperm in offspring9,10. Based on the above studies, paternal obesity may lead to spermatogenesis disorders in offspring. Recent study found that parental obesity intergenerationally induced reproductive damages in offspring11. Nevertheless, the intergenerational effect of multigenerational paternal obesity on the susceptibility to spermatogenesis disorders in offspring and its mechanism remain unknown.

N6-methyladenosine (m6A) is the most common RNA modification, dynamically regulates post-transcriptional processes12. In mammals, the modification of m6A is conferred by demethylases (ALKBH5 and FTO) and methylases, including METTL3 and METTL413. Increasing evidences have presented that homeostasis of m6A modification is essential for spermatogenesis in mammals14,15,16. An earlier study found that the expression of methylases in mouse testes gradually increased from embryo to adult, while the expression of demethylases gradually decreased17. We previously demonstrated that HFD markedly increased m6A modification levels in contemporary mouse testesFull size image

Paternal HFD enhances testicular Wt1 downregulation and spermatogenesis disorder in offspring via increasing METTL3-mediated paternal sperm m6A level

To further investigate the effect of reduced sperm m6A level on paternal HFD-impaired spermatogenesis in offspring, F0 generation male mice were treated with STM2457, a specific inhibitor of METTL3 activity. Results presented that the sperm count was reduced in HFD1D group, and the above effect was significantly restored after STM2457 treatment (Fig. 9a–c). As shown in Fig. 9d–f, the decreased protein and mRNA levels of Wt1 in HFD1D testes were markedly reversed after STM2457 treatment. In addition, STM2457 treatment evidently restored HFD1D-increased the levels of Wt1 site1 m6A and METTL3 protein in offspring testes (Fig. 9g–i). Further studies found that STM2457 treatment markedly attenuated HFD-increased m6A and Mettl3 mRNA levels in paternal sperm (Fig. 9j, k). Furthermore, the decreased sperm level of Wt1 mRNA in HFD group was also reversed after STM2457 treatment (Fig. 9k). The above results collectively demonstrate that paternal HFD aggravates Cd-induced testicular Wt1 downregulation and spermatogenesis disorder in offspring via increasing METTL3-mediated paternal sperm m6A level.

Fig. 9: Paternal HFD enhances testicular Wt1 downregulation and spermatogenesis disorder in offspring via increasing METTL3-mediated paternal sperm m6A level.
figure 9

F0 generation male mice were fed NC or HFD from 5 weeks to 15 weeks old, and treated with STM2457 once a week from 10 weeks to 15 weeks old. F1 generation male mice were exposed to CdCl2 from 5 weeks to 10 weeks old, and named NCD, NCD + STM, HFD1D or HFD1D + STM group, respectively. a Experimental design flowchart. b, c Epididymal sperm counts were measured. n = 10 mice for NCD group; n = 9 mice for NCD + STM group; n = 11 mice for HFD1D group; n = 8 mice for HFD1D + STM group; DOF = 37, F = 6.10, P = 0.0020. d, e Testicular WT1 protein expression was measured by immunoblotting. n = 6 mice, DOF = 23, F = 8.94, P = 0.0006. f The mRNA level of testicular Wt1 was detected. n = 6 mice, DOF = 23, F = 17.81, P < 0.0001. g Testicular Wt1 site1 m6A level was measured by MeRIP-qPCR. n = 6 mice, DOF = 23, F = 10.20, P = 0.0003. h, i The protein level of testicular METTL3 was measured. n = 6 mice, DOF = 23, F = 27.16, P < 0.0001. j Sperm total RNA m6A level was tested. n = 6 mice, DOF = 23, F = 14.33, P < 0.0001. k Sperm Mettl3 and Wt1 mRNA level were detected by RT-qPCR. n = 6 mice, DOF = 24, F = 15.51 and P < 0.0001 for Mettl3; F = 14.90 and P < 0.0001 for Wt1. *P < 0.05; **P < 0.01 vs NCD. #P < 0.05; ##P < 0.01 vs HFD1D. In regard to Fig. 9c, e–g, i–k, statistical significance was evaluated by two-sided one-way ANOVA with post hoc LSD tests. Data are presented as mean ± SEM. Source data are provided with this paper.

Elevated sperm m6A level and decreased sperm concentration is observed in donors who were overweight or obese

The effects of paternal single- or double-generation HFD exposure on sperm and testicular parameters, retinoic acid level, WT1 and METTL3 expression in offspring mice were investigated. Analogously, the gradual reduction in sperm count, the impairment in testicular germ cell development, the inhibition in retinoic acid synthesis, the downregulation in WT1 expression, the upregulation in METTL3 expression and the elevation in m6A level were observed in offspring testes with the increase of HFD generation (Supplementary Figs. 810). To further verify the relationship among sperm m6A level, sperm concentration and BMI, a case-control study was established. As presented in Fig. 10a, b, the BMI of the case group was higher than that of the control group, but the sperm concentration was lower. Figure 10c showed that sperm m6A levels were elevated in the case group compared to the control group. The correlations of sperm m6A level and BMI or sperm concentration were further analyzed. The positive association was observed between sperm m6A level and BMI (r = 0.57, P < 0.01; Fig. 10e). The negative correlation was also observed between sperm m6A level and sperm concentration (r = −0.51, P < 0.01; Fig. 10f). In addition, restricted cubic splines were used to describe the nonlinear correlation among sperm concentration, BMI and sperm m6A level. As shown in Supplementary Figs. 11a–c, a linear relationship was observed between the three sets of data (P for overall <0.001, and P for nonlinearå 0.05). The above results suggest that elevated sperm m6A level and decreased sperm concentration is observed in donors who were overweight or obese.

Fig. 10: Elevated sperm m6A level and decreased sperm concentration is observed in donors who were overweight or obese.
figure 10

After removal of smoking or alcohol drinking donors, 30 pairs of cases with overweight/obesity and corresponding controls were obtained by matching age. a BMI. n = 30 human sperm, t = −11.20, P < 0.0001. Two-tailed t test was used to analyze the differences. b Sperm concentration. n = 30 human sperm, t = 10.50, P < 0.0001. Two-tailed t test was used to analyze the differences. c Sperm m6A level. n = 30 human sperm, t = −2.74, P = 0.0083. Two-tailed t test was used to analyze the differences. d The association between sperm concentration and BMI. e The association between sperm m6A level and BMI. f The association between sperm m6A level and sperm concentration. g Graphical abstract. **P < 0.01. Source data are provided with this paper.