Skip to main content

Mendelian randomization analyses support causal relationships between gut microbiome and longevity

Abstract

Background

Gut microbiome plays a significant role in longevity, and dysbiosis is indeed one of the hallmarks of aging. However, the causal relationship between gut microbiota and human longevity or aging remains elusive.

Methods

Our study assessed the causal relationships between gut microbiome and longevity using Mendelian Randomization (MR). Summary statistics for the gut microbiome were obtained from four genome-wide association study (GWAS) meta-analysis of the MiBioGen consortium (N = 18,340), Dutch Microbiome Project (N = 7738), German individuals (N = 8956), and Finland individuals (N = 5959). Summary statistics for Longevity were obtained from Five GWAS meta-analysis, including Human healthspan (N = 300,447), Longevity (N = 36,745), Lifespans (N = 1,012,240), Parental longevity (N = 389,166), and Frailty (one of the primary aging-linked physiological hallmarks, N = 175,226).

Results

Our findings reveal several noteworthy associations, including a negative correlation between Bacteroides massiliensis and longevity, whereas the genus Subdoligranulum and Alistipes, as well as species Alistipes senegalensis and Alistipes shahii, exhibited positive associations with specific longevity traits. Moreover, the microbial pathway of coenzyme A biosynthesis I, pyruvate fermentation to acetate and lactate II, and pentose phosphate pathway exhibited positive associations with two or more traits linked to longevity. Conversely, the TCA cycle VIII (helicobacter) pathway consistently demonstrated a negative correlation with lifespan and parental longevity.

Conclusions

Our findings of this MR study indicated many significant associations between gut microbiome and longevity. These microbial taxa and pathways may potentially play a protective role in promoting longevity or have a suppressive effect on lifespan.

Introduction

The increases in life expectancy were observed globally during the past 50 years [1, 2]. Previous studies have described growing evidence highlighting geographical disparities in life expectancy at county and sub-county levels [3], which may be associated with genetics [4, 5], environmental factors including intestinal microbiota [6] and socioeconomic factors [7]. Dysbiosis is one the twelve hallmarks of aging [8], and it contributes to multiple pathological conditions associated with age-related diseases, such as diabetes, hypertension, and Alzheimer disease [9, 10]. Age-related morbidities affect the quality and quantity of life, but the composition and function and within the intestinal microbiome as we age are not completely understood. Thus, identifying aging patterns within the gut microbiome could have clinical implications for both monitoring and modifying gut health to achieve healthy aging.

Several studies have characterized the gut microbiota in centenarians [7, 11,12,13,14], providing potential insight into gut microbial trajectories associated with aging. For example, Akkermansia was found to be enriched in the gut of centenarians [7, 11,12,13,14], while Bacteroides and Faecalibacterium were relatively depleted in centenarians [7, 13]. However, most observational research cannot infer the causal relationship between gut microbiota and human longevity or aging. Few mechanisms that link longevity- or aging-correlated pathophysiology with specific microbes or functional pathways have been identified [12, 15]. Some microbial pathways that regulate aging were discovered in invertebrates such as Caenorhabditis elegans (C. elegans) [16, 17] and drosophila [18], however, the genetics of aging is more complex in vertebrates and primates (including Homo sapiens) because of their specialized systems [19]. Population-scale human genome and metagenome sequencing projects combined with comprehensive data on lifespan and age-related diseases are opening new avenues to understand the genetics and microbiome involved in human aging.

In recent years, Mendelian randomization (MR) has attracted wide attention by inferring the causal relationship of modifiable exposures on an outcome such as disease status [20]. For example, a MR analysis indicated a potential causal effect of Morganella on major depressive disorder, implying that this bacterium may play a role in the metabolic modulation of health within the brain-gut axis [21]. Another study employed MR to infer causal relationships between the gut microbiome and metabolites, discovering a potential causal role of Eubacterium rectale in decreasing plasma levels of hydrogen sulfite-a toxin that affects cardiovascular function [22]. These studies uncover potential metabolic capabilities of gut microbes that exert an influence on human health. Therefore, this study assessed the causal relationships between the gut microbiome and longevity using MR. Previous microbiome-longevity studies utilizing MR analysis relied on a limited dataset [23, 24], which resulted in findings that were not comprehensive. Here, we have utilized the most comprehensive GWAS summary data, as of the present moment, pertaining to the gut microbiome and longevity. Our findings provide novel clues for understanding the roles of gut microbiota in longevity and aging development, which may lead to the development of microbiome-based therapies and personalized medicine approaches to delay aging and promote longevity.

Methods

Study design

Figure 1 outlines this study’s overall design and data source. Briefly, a two-sample MR analysis was performed to verify causal links between gut microbiota and traits associated with longevity. It takes genetic variation (single nucleotide polymorphisms, SNPs) as the instrumental variables (IVs) to deduce the causal effect of the exposure (gut microbiota) on the outcome (longevity-related traits), thereby effectively circumventing the confounding biases often encountered in traditional epidemiological studies. Three MR assumptions are used for the unbiased estimation of possible causal relationships: (1) IVs are strongly associated with exposure (relevance); (2) IVs are independent of any confounding factors (independence); and (3) IVs affect the outcome only through the exposure (exclusion restriction) [25]. The genome-wide association study (GWAS) data source of longevity-related traits and gut microbiota has also been summarized in Table S1.

Fig. 1
figure 1

The study design of the MR investigation pertaining to the associations between gut microbiota and longevity

GWAS data of human healthspan, longevity, lifespans, parental longevity, and frailty

Five publicly available GWAS datasets of longevity-related traits were used in this study. The GWAS data of human healthspan consists of 300,477 British-ancestry individuals from the UK Biobank (UKB) [26]. The GWAS data of longevity was derived from 36,745 individuals of European ancestry in multiple studies, encompassing 11,262 cases and 25,483 controls, and the cases were individuals who lived to age above the 90th percentile or 99th percentile [5]. The GWAS data of lifespan consists of 1,012,240 European-ancestry individuals, including 512,047 mother and 500,193 father lifespans [27]. The GWAS data of parental longevity was collected for 389,166 UKB participants of European descent with data recorded on parents’ current ages or parents’ ages of death [28]. Pilling et al. identified all common genetic variants associated with longer parental lifespan, including 7 traits (mother's age at death, father's age at death, mother's attained age, father's attained age, combined parental age at death, combined parental attained age, and both parents in top 10%) [28]. Because one of the primary aging-linked physiological hallmarks is the onset of frailty [29], the GWAS data of frailty is also incorporated in our study, which included 175,226 individuals of European descent and used the frailty index (FI) to measure frailty [30].

GWAS data of gut microbiota

Genetic variants associated with the gut microbiome were obtained from four datasets. The first dataset was conducted by MiBioGen consortium, and integrated 16S rRNA gene sequencing profiles and genotyping data from 18,340 participants across 24 cohorts [31]. Most of participants had European ancestry (N = 13,266). Both genetic data and gut microbiota data were incorporated, and association estimates for a total of 211 bacterial taxa were calculated. After excluding 15 taxa of bacteria without specific names (unknown family or genus), gut microbiota was divided into 196 bacterial taxa including 9 phyla, 16 classes, 20 orders, 32 families, and 119 genera. The second dataset was obtained from 7,738 participants of the Dutch Microbiome Project (DMP), and the gut microbiota was identified by shotgun metagenomic sequencing of stool samples [32]. It contained 207 taxa (105 of which are species) and 205 functional pathways that reflect the composition and activity of gut microbiota. The third dataset was obtained from 8956 German individuals by Ruhlemann et al. [33], which carried out a GWAS involving 430 taxa (from phylum to genus, examining the abundance and prevalence in fecal samples) that reflect the composition of gut microbiota. The fourth dataset was a large-scale population-based cohort of 5959 Finland individuals enrolled in the FINRISK 2002 (FR02) cohort, which carried out a GWAS involving 473 taxa (including species level, examining the abundance in stool) that provided insights into the composition of gut microbiota[21].

Selection of instrumental variables (IVs)

We extracted the gut microbiota taxa as exposure data, including 196 bacterial taxa from the MiBioGen consortium, 412 bacterial traits (207 taxa and 205 pathways) from the DMP, 430 taxa from 8956 German individuals, and 473 taxa from 5,959 Finland individuals. To ensure the authenticity and accuracy of the causal relationship between gut microbiota and longevity, the following quality control procedures were implemented to select IVs. Firstly, we selected IVs for each gut bacterial trait by using a loose cutoff of P < 1 × 10–5. Secondly, the independent IVs with the lowest P-value for each trait (r2 < 0.001 and distance = 10,000 kb) were retained to reduce the influence of correlations among SNPs. Thirdly, we calculated the F-statistic to evaluate the strength of the IVs. SNPs with F-statistics < 10 was disregarded to avoid weak IV bias. Fourthly, the screened SNPs were used as IVs to harmonize with summary statistic of longevity and the palindromic SNPs and fuzzy alleles were removed. At last, we removed SNPs with P < 1 × 10–5 of outcome in harmonized data to avoid strong correlations between SNPs and outcome [14].

Statistical analysis

We primarily employed the inverse-variance weighted (IVW) method for our analysis. To assess the heterogeneity among SNPs, we conducted Cochran's Q test. If significant heterogeneity was observed (P < 0.05), we adopted the random-effects model; otherwise, the fixed-effects model was utilized [34]. The IVW method generated effect estimates for each SNP on gut microbiota and the likelihood of longevity, enabling us to compute the Wald estimates. Additionally, we performed sensitivity analyses to evaluate the robustness of our findings. The weighted median method was used to estimate the potential causal effects when IVs violated standard assumptions to provide a reliable estimate [35]. For the reliability of the final analysis results, the following screening criteria were used as filters for robust significant causality: (1) At least the IVW method suggested a significant causal relationship; (2) The direction of MR analysis results (beta value) was consistent among the three methods (IVW, MR-Egger, and weighted median); 3) We apply the maximum likelihood method or simple median method to replicate significant causal relationships, considering our reliance on the IVW method. MR-Egger method was used to detect directional pleiotropy, and intercept of P > 0.05 was deemed to be no horizontal pleiotropy [36]. Besides, the MR pleiotropy residual sum and outlier (MR-PRESSO) method was also applied to test for possible bias from horizontal pleiotropy and outlier variants removal [37]. Furthermore, the leave-one-out test was conducted to confirm that MR estimates were not driven by strong effect SNPs. The results were visually analyzed by forest plots and scatter plots. All MR analyses were performed in the R software (v4.2.3) using “Mendelian Randomization”, “TwoSampleMR”, and “MRPRESSO” packages. We considered suggestive evidence of a potential causal association when P < 0.05.

Results

Influence of the 403 gut bacterial taxa on longevity‑related traits

We first investigate the causal relationships between the gut microbiome and longevity by performing a two-sample MR analysis using GWAS summary data of 403 gut bacterial taxa (196 taxa sourced from MiBioGen consortium, and 207 taxa from DMP) and longevity-associated traits. We observed suggestive evidence for many bacterial taxa to be associated with longevity, and these causal relationships were statistically significant with a P-value of less than 0.05, at least when employing the IVW method (Table S2).

In terms of healthspan, our findings revealed that Intestinimonas (β = 0.033, P = 0.049), Olsenella (β = 0.044, P = 0.002), and Turicibacter (β = 0.045, P = 0.016) were positively correlated with healthspan, while Anaerostipes (β = − 0.051, P = 0.034), Tyzzerella3 (β = − 0.030, P = 0.026), Ruminiclostridium9 (β = − 0.059, P = 0.012), Ruminococcus obeum (β = − 0.045, P = 0.030), Bacteroides xylanisolvens (β = − 0.034, P = 0.020), Bacteroides vulgatus (β = -0.059, P = 0.009), and Bacteroides eggerthii (β = − 0.038, P = 0.004) were negatively correlated with healthspan. In the MR-Egger regression, there was no evidence of directional pleiotropic effects (intercept p-value > 0.05). There is a significant heterogeneity only for Ruminococcus obeum in the Cochran’s Q test (p = 0.031).

For lifespan, Defluviitaleaceae UCG-011(β = 0.038, P = 0.014), Erysipelotrichaceae UCG003 (β = 0.038, P = 0.016), Senegalimassilia (β = 0.052, P = 0.002), Tyzzerella3 (β = 0.036, P = 0.025), Odoribacter (β = 0.031, P = 0.030), Alistipes senegalensis (β = 0.038, P = 0.008), Bacteroides faecis (β = 0.013, P = 0.033), Holdemania unclassified (β = 0.019, P = 0.040), and Bilophila unclassified (β = 0.030, P = 0.045) were positively associated with lifespan. In comparison, Butyricimonas (β = − 0.034, P = 0.016), Lachnospira (β = − 0.073, P = 0.012), Lachnospiraceae UCG-001 (β = -0.037, P = 0.014), Streptococcus salivarius (Wald ratio, β =− 0.040, P = 0.046), and Collinsella aerofaciens (β = − 0.04, P = 0.005) were negatively associated with lifespan. Intercept of MR-Egger regression also showed no potential horizontal pleiotropy. There is a significant heterogeneity only for Tyzzerella3 (Cochran’s Q test, p = 0.009).

In relation to longevity, our findings revealed a positive correlation between Bilophila wadsworthia (β = 0.309, P = 5 × 10⁻4) and Adlercreutzia equolifaciens (β = 0.172, P = 0.022) with individuals who attained a lifespan exceeding the 90th percentile. Lachnospiraceae bacterium 3_1_46FAA was positively associated with longevity (> 90th percentile, β = 0.195, P = 0.014; > 99th percentile, β = 0.328, P = 0.008). On the contrary, Blautia (β = -0.246, P = 0.001), and Escherichia coli (β = -0.131, P = 0.016) were negatively correlated with longevity (> 90th percentile). Akkermansia muciniphila was negatively correlated with longevity (> 99th percentile, β = -0.219, P = 0.030). Bacteroides massiliensis was negatively associated with longevity (> 90th percentile, β = − 0.179, P = 0.006; > 99th percentile, β = − 0.216, P = 0.038). The forest plot and scatter plot about B. massiliensis were presented in Fig. 2 and Figure S1, while the outcomes of the leave-one-out analysis confirmed that the MR estimates were not driven by strong effect SNPs (Figure S2). In addition, Haemophilus parainfluenzae was also negatively associated with longevity (> 90th percentile, β = -0.154, P = 0.011; > 99th percentile, β = − 0.231, P = 0.016).

Fig. 2
figure 2

The forest plot depicts the causal associations between Bacteroides massiliensis and traits associated with human longevity. The slope value equals the β-value calculated using the three methods (IVW, weighted median, and MR Egger), and it signifies the magnitude of the causal effect. A positive slope indicates that exposure is a contributory factor in promoting the outcome, whereas a negative slope suggests the opposite effect. Abbreviations: CI, confidence interval; OR, odds ratio; Nsnp, the number of single nucleotide polymorphisms (SNPs)

For parental longevity, genus Anaerofilum (β = 0.019, P = 0.023) was positively associated with father's age at death, while Collinsella (β = 0.035, P = 0.035), and Eubacterium rectale group (β = 0.046, P = 0.005) were positively associated with mother's age at death. Desulfovibrio (β = 0.031, P = 0.036), and Eubacterium xylanophilum group (β = 0.061, P = 0.001) were positively associated with combined parental age at death. Slackia (β = 0.033, P = 0.002), Enterorhabdus (β = 0.025, P = 0.031), and Lachnospiraceae bacterium 5_1_63FAA (β = 0.013, P = 0.004) increased parental longevity odds of father's attained age, while Erysipelatoclostridium (β = 0.017, P = 0.044), and Eubacterium rectale group (β = 0.041, P = 0.038) increased parental longevity odds of mother's attained age. Meanwhile, Akkermansia muciniphila was positively associated with mother's age at death (β = 0.021, P = 0.033), combined parental age at death (β = 0.025, P = 0.012), and both parents in top 10% (β = 0.012, P = 0.047). Bacteroides fragilis (β = 0.009, P = 0.034) and Coprobacter fastidiosus (β = 0.022, P = 0.006) were causally associated with mother's attained age. Moreover, Eubacterium eligens was positively associated with parental longevity (both parents in top 10%, β = 0.018, P = 0.007), while Eubacterium rectale was positively associated with combined parental age at death (β = 0.030, P = 0.045) in the IVW method. Regarding negative association, Bacteroides (β = − 0.060, P = 0.006), Butyricicoccus (β = -0.025, P = 0.036), Flavonifractor (β = − 0.041, P = 0.036), Lachnospiraceae UCG008 (β = -0.018, P = 0.020), Odoribacter (β = − 0.045, P = 0.043), Tyzzerella3 (β = − 0.015, P = 0.022), Bacteroides dorei (β = − 0.021, P = 0.021), and Eubacterium biforme (β = − 0.012, P = 0.011) were negatively linked to specific traits of parental longevity. Notably, Oxalobacter in the MiBioGen dataset was negatively associated with mother's age at death (β = − 0.019, P = 0.038), but Oxalobacter in the DMP dataset was positively associated with father's attained age (β = 0.011, P = 0.048).

Finally, frailty, which often accompanies aging, was analyzed as well. Genus Bifidobacterium (β = 0.042, P = 0.013), Clostridium innocuum group (β = 0.023, P = 0.036), Eubacterium coprostanoligenes group (β = 0.054, P = 0.003), Flavonifractor (β = 0.023, P = 0.046), and species Ruminococcus torques (β = 0.035, P = 0.032) were positively associated with the frailty index (FI). The positive associations between the Clostridium innocuum group, the Eubacterium coprostanoligenes group, and frailty are consistent with a previous study[38].

Influence of the 903 gut bacterial taxa on longevity‑related traits

The MR results of the significant associations between 903 gut bacterial taxa (430 taxa from 8956 German individuals, and 473 taxa from 5,959 Finland individuals in the FR02 cohort) and longevity-related phenotypes are summarized in Table S3.

For human healthspan, Porphyromonadaceae (β = 0.033, P = 0.001), Atopobiaceae (β = 0.065, P = 0.042), Ruminococcaceae (β = 0.021, P = 0.005), Caloranaerobacter (β = 0.140, P = 0.003), Sutterella (β = 0.035, P = 0.002), Oscillibacter prevalence (β = 0.019, P = 0.036), Bifidobacterium breve (β = 0.060, P = 0.018), Lawsonibacter sp002161175 (β = 0.124, P = 0.037), and Monoglobus pectinilyticus (β = 0.048, P = 0.017) were positively correlated with healthspan. On the contrary, Alphaproteobacteria (β = -0.038, P = 0.007), and Coprococcus (β = 0.124, P = 0.037) were negatively correlated with healthspan.

For lifespans, Clostridium XlVa (β = 0.074, P = 0.007), Oscillibacter abundance (β = 0.049, P = 0.005), Ruminococcus(β = 0.009, P = 0.020), and Faecalibacterium prausnitzii E (β = 0.063, P = 0.024) were positively correlated with lifespans, while Roseburia (β = − 0.042, P = 0.016), Helicobacter (β = − 0.121, P = 0.004), Alistipes (β = − 0.112, P = 0.038), Photobacterium (β =− 0.145, P = 0.024), Desulfovibrio (β = − 0.022, P = 0.001), Parasutterella prevalence (β = − 0.011, P = 0.049), Oscillibacter prevalence (β = − 0.016, P = 0.031), and Lactococcus lactis (β = -0.050, P = 0.036) were negatively correlated with lifespans. The significant causal relationship between Oscillibacter and longevity-related traits were summarized in Figure S3.

For longevity, Parabacteroides (β = 0.116, P = 0.035), Gordonibacter (β = 0.219, P = 0.030), Sutterella abundance (β = 0.103, P = 0.015), and Alistipes shahii (β = 0.166, P = 0.007) abundance in stool were linked to increased longevity (> 90th percentile). Prevotella sp900317685 (β = − 0.218, P = 0.013) and Blautia A sp900066355 (β = − 0.272, P = 0.026) were negatively correlated with longevity (> 90th percentile). Prevotella (β = − 0.065, P = 0.045), Bacteroides (β = − 0.068, P = 0.004), Escherichia flexneri (β = − 0.341, P = 0.002), and Coprobacillus cateniformis (β = − 0.376, P = 0.007), and Parabacteroides johnsonii (β = − 0.261, P = 0.047) were negatively correlated with longevity (> 99th percentile). Both the Parasutterella prevalence (β = − 0.085, P = 0.035) and Parasutterella abundance (β = − 0.078, P = 0.001) were negatively correlated with longevity (> 90th percentile).

Many of the 430 gut bacterial taxa from 8956 German individuals have been causally associated with parental longevity. Alistipes (β = 0.020, P = 0.009), and Subdoligranulum prevalence (β = 0.011, P = 0.025) were positively correlated with combined parental age at death. Alongside the previously mentioned results, more correlations between Alistipes and longevity were identified (Fig. 3A), especially the two Alistipes species of A. senegalensis and A. shahii (Fig. 3B). The results of the “leave-one-out” test showed that there was no abnormal IV in this analysis affecting the overall results (Figure S4). In addition, the Desulfovibrio abundance was negatively correlated with two parental longevity traits (both parents in top 10%, β = − 0.007, P = 0.038; combined parental age at death, β = − 0.015, P = 0.016), but was positively correlated with another two parental longevity traits (combined parental attained age, β = 0.016, P = 2 × 10–4; father's attained age, β = 0.013, P = 0.001). The Sutterella prevalence was negatively correlated with combined parental age at death (β = − 0.015, P = 0.045). Faecalibacterium abundance was negatively correlated with father’s age at death (β = − 0.021, P = 0.008). More results were summarized in Table S3.

Fig. 3
figure 3

MR results reveal the causal associations between Alistipes and traits associated with human longevity. A Forest plot depicting the associations between Alistipes and longevity-correlated traits. B Scatter plot depicting the associations between Alistipes senegalensis and lifespan, and between Alistipes shahii and longevity (> 90th percentile)

In addition, many of the 473 taxa from 5,959 Finland individuals have been causally associated with parental longevity. For example, Leuconostoc mesenteroide was positively associated with combined parental attained age (β = 0.019, P = 0.046). Prevotella sp000436915 was positively associated with combined parental attained age (β = 0.013, P = 0.048), but was negatively correlated with father's age at death (β = − 0.022, P = 0.005). Bifidobacterium breve was negatively correlated with both parents in top 10% (β = − 0.031, P = 0.002), but was positively correlated with combined parental attained age (β = 0.034, P = 0.010). Bifidobacterium angulatum was negatively correlated with mother's age at death (β = − 0.019, P = 0.043). Dorea phocaeense was negatively correlated with combined parental age at death (β = − 0.050, P = 0.025), but was positively associated with mother's attained age (β = 0.034, P = 0.018) and combined parental attained age (β = 0.035, P = 0.045). Coprobacillus cateniformis abundance was positively associated with father's age at death (β = 0.021, P = 0.036), but was negatively associated with mother's age at death (β = -0.030, P = 0.004). Eubacterium callanderi abundance was positively correlated with parental longevity (father's age at death) (β = 0.050, P = 0.020), but was negatively associated with father's attained age (β = − 0.042, P = 0.010). The abundance of Lactococcus lactis was positively correlated with father's attained age (β = 0.034, P = 0.025), and on the contrary, Clostridium tertium was negatively associated with it (β = − 0.051, P = 0.010). The abundance of Enterococcus faecalis in stool was positively correlated with mother's attained age (β = 0.027, P = 0.045), while Blautia A sp900066355 was negatively linked to it (β = -0.036, P = 0.007). Enorma massiliensis was positively correlated with mother's attained age (β = 0.027, P = 0.013) but was negatively correlated with father's attained age (β = − 0.025, P = 0.022).

The MR results of the association between 903 gut bacterial taxa and frailty indicated that Sutterella prevalence (OTU99_116, β = 0.009, P = 0.009; TestASV_22, β = 0.014, P = 0.002) was positively correlated with FI, while the Sutterella abundance (β = − 0.040, P = 0.004) and Parasutterella abundance (β = -0.008, P = 0.021) were negatively correlated with FI. The abundance of Faecalibacterium (β = 0.033, P = 0.015), and Anaeromassilibacillus sp001305115 (β = 0.047, P = 0.003) were positively correlated with FI. Bacteroides sp002160055 (β = − 0.036, P = 0.017), Bacteroides stercoris (β = − 0.025, P = 0.032), Lawsonibacter sp002161175 (β = − 0.081, P = 0.037), Morganella (β = − 0.038, P = 0.042), and Subdoligranulum abundance (β = − 0.037, P = 0.009) in stool were negatively correlated with FI, at least in the IVW method. To sum up, Subdoligranulum was causally associated with multiple longevity-correlated traits (Fig. 4), and no abnormal IV in this analysis affecting the overall results (Figure S5). Moreover, all the notable associations between Parasutterella and longevity-related traits indicate Parasutterella has a negative impact on lifespan, longevity, and frailty (Figure S6).

Fig. 4
figure 4

The causal associations between Subdoligranulum and traits associated with human longevity. The forest plot A and scatter plot B depicting the association between Subdoligranulum prevalence and Parental longevity (combined parental age at death), and between Subdoligranulum abundance and frailty index, respectively

Influence of the 205 gut functional pathways on longevity‑related traits

The gut microbial taxa potentially regulate longevity through their associated metabolic pathways, prompting us to also analyze the influence of gut functional pathways on longevity‑related traits. Only the gut microbiome from 7,738 participants of the DMP encompassed 205 functional pathways [32], whereas the other three GWAS datasets of gut microbiota exclusively comprised microbial taxa. A total of 65 pathways have been discovered to exhibit significant and robust associations with traits linked to longevity (Table S4). There was no evidence of directional pleiotropic effects (intercept p-value > 0.05), and no significant heterogeneity (Cochran’s Q test, p-value > 0.05).

For human healthspan, microbial pathways of PPGPPMET-PWY: ppGpp biosynthesis (β = 0.037, p = 0.028), and PWY-6507: 4-deoxy-L-threo-hex-4-enopyranuronate degradation (β = 0.017, p = 0.044) showed a positive association, in the IVW method. In contrast, pathways such as PWY-6284: superpathway of unsaturated fatty acids biosynthesis (E. coli) (β =  − 0.049, p = 0.005), HOMOSER-METSYN-PWY: L-methionine biosynthesis I (β =  − 0.051, p = 0.011), DENOVOPURINE2-PWY: superpathway of purine nucleotides de novo biosynthesis II (β =  − 0.042, p = 0.030), PWY-6163: chorismate biosynthesis from 3-dehydroquinate (β =  − 0.043, p = 0.035), and PWY-5913: TCA cycle VI obligate autotrophs (β =  − 0.047, p = 0.004), were negatively correlated with healthspan.

For lifespan, pathways of NONOXIPENT-PWY: pentose phosphate pathway (non-oxidative branch) (β = 0.055, P = 0.002), PWY-6630: superpathway of L-tyrosine biosynthesis (β = 0.023, P = 0.008), PWY-7196: superpathway of pyrimidine ribonucleosides salvage (β = 0.047, P = 0.011), PWY-7209: superpathway of pyrimidine ribonucleosides degradation (β = 0.022, P = 0.032), and PWY0-162: superpathway of pyrimidine ribonucleotides de novo biosynthesis (β = 0.030, P = 0.033) were positively associated. In contrast, UBISYN-PWY: superpathway of ubiquinol 8 biosynthesis (prokaryotic) (β = -0.040, P = 0.001), KDO-NAGLIPASYN-PWY: superpathway of (Kdo)2-lipid A biosynthesis (β = − 0.024, P = 0.006), PWY-7211: superpathway of pyrimidine deoxyribonucleotides de novo biosynthesis (β = -0.032, P = 0.017), PWY_REDCITCYC: TCA cycle VIII (helicobacter) (β = -0.030, P = 0.007), PWY-5918: superpathway of heme biosynthesis from glutamate (β = -0.028, P = 0.022), PWY0-1338: polymyxin resistance (β = − 0.020, P = 0.023), and ENTBACSYN-PWY: enterobactin biosynthesis (β = − 0.039, P = 0.028) demonstrated negative associations with lifespan.

Regarding longevity, the abundances of PWY-6731: starch degradation III was positively associated with age > 90th percentile (β = 0.084, P = 0.020), while PWY-5941: glycogen degradation II (eukaryotic) was positively associated with age > 99th percentile (β = 0.262, P = 0.030). Moreover, THRESYN-PWY: superpathway of L-threonine biosynthesis (β = − 0.338, P = 0.017), and PWY0-1261: anhydromuropeptides recycling (β = − 0.346, P = 0.020) were negatively correlated with longevity (age > 99th percentile).

Regarding parental longevity, 46 associations have been identified. For instance, PWY-7197: pyrimidine deoxyribonucleotide phosphorylation was positively associated with father's attained age in the IVW method (β = 0.019, P = 0.045). PWY-7254: TCA cycle VII (acetate producers) was positively associated with mother's age at death (β = 0.016, P = 0.046). PWY-7456: mannan degradation was positively associated with father’s attained age (β = 0.031, P = 0.001). PWY-6897: thiamin salvage II was positively associated with both parents in top 10% (β = 0.023, P = 0.007). PWY-5088: L-glutamate degradation VIII to propanoate were positively associated with father's attained age (β = 0.015, P = 0.009). GALACTARDEG-PWY: D-galactarate degradation I was positively associated with mother's attained age (β = 0.014, P = 0.021), and combined parental attained age (β = 0.014, P = 0.045). On the contrary, PWY-7323: superpathway of GDP-mannose derived O-antigen building blocks biosynthesis (β = -0.026, P = 0.014) was negatively associated with parental longevity (father’s age at death). PWY-5838: superpathway of menaquinol-8 biosynthesis I (β = − 0.022, P = 0.043), and HOMOSER-METSYN-PWY: L-methionine biosynthesis I (β = − 0.029, P = 0.043) were negatively correlated with mother's age at death. PWY-5667: CDP-diacylglycerol biosynthesis I was negatively correlated with father's attained age (β = − 0.019, P = 0.012). PWY0-1415: superpathway of heme biosynthesis from uroporphyrinogen III (β = − 0.016, P = 0.012), DAPLYSINESYN-PWY: L-lysine biosynthesis I (β = − 0.021, P = 0.017), and ANAEROFRUCAT-PWY: homolactic fermentation (β = − 0.037, P = 0.029) were negatively correlated with combined parental attained age. METHGLYUT-PWY: superpathway of methylglyoxal degradation (β = − 0.008, P = 0.034), and ORNDEG-PWY: superpathway of ornithine degradation (β = − 0.010, P = 0.027) were negatively correlated with both parents in top 10%. Notably, COA-PWY (coenzyme A biosynthesis I) exhibited a positive association with father's age at death (β = 0.035, P = 0.013) and combined parental age at death (β = 0.038, P = 0.023), but exhibited a negative association with combined parental attained age (β = − 0.030, P = 0.002), as illustrated in Fig. 5A. The pathway of P162-PWY: L-glutamate degradation V via hydroxyglutarate was negatively correlated with mother’s age at death (β = − 0.028, P = 0.012) and both parents in top 10% (β = -0.020, P = 0.014), but was positively correlated with mother’s attained age (β = 0.018, P = 0.044) and combined parental attained age (β = 0.019, P = 0.034). PWY-7013: L-1,2-propanediol degradation exhibited a negative correlation with father’s age at death (β = -0.012, P = 0.034) and combined parental age at death (β = − 0.016, P = 0.042), but exhibited a positive correlation with father’s attained age (β = 0.008, P = 0.043) and combined parental attained age (β = 0.009, P = 0.042).

Fig. 5
figure 5

The forest plots reveal the impact of two microbial pathways, COA-PWY and PWY-5100, on traits associated with longevity. A The notable associations between COA-PWY (coenzyme A biosynthesis I) pathway and parental longevity. B The notable associations between PWY-5100 (pyruvate fermentation to acetate and lactate II) and traits associated with lifespan and parental longevity. Four MR methods including IVW, weighted median, simple median, and MR Egger were used. The causal relationships were presented using OR (odds ratio) and 95% CI (confidence interval)

In terms of frailty-correlated pathways, we found PWY-7456: mannan degradation (β = − 0.056, P = 0.008), TRNA-CHARGING-PWY: tRNA charging (β = − 0.032, P = 0.027), POLYAMSYN-PWY: superpathway of polyamine biosynthesis I (β = − 0.062, P = 0.003), and PWY-5101: L-isoleucine biosynthesis II (β = -0.032, P = 0.019) were negatively correlated with frailty index. In comparison, PWY-5920: superpathway of heme biosynthesis from glycine (β = 0.013, P = 0.044), PWY-6630: superpathway of L-tyrosine biosynthesis (β = 0.021, P = 0.049), GLUCONEO-PWY: gluconeogenesis I (β = 0.054, P = 0.009), and GLUCOSE1PMETAB-PWY: glucose and glucose 1-phosphate degradation (β = 0.036, P = 0.026) were positively associated with frailty index.

There are several multiple associations between specific one pathway and longevity traits among different datasets (Table S5). For example, the PWY-5100 (pyruvate fermentation to acetate and lactate II) pathway was positively associated with both lifespan (β = 0.028, P = 0.039) and parental longevity of combined parental age at death (β = 0.032, P = 0.009), but was negatively correlated with father's attained age (β = − 0.022, P = 0.006), which was depicted in Fig. 5B. The NONOXIPEN-PWY: pentose phosphate pathway (non-oxidative branch) exhibited a positive association with lifespan (β = 0.055, P = 0.002), parental longevity of both parents in top 10% (β = 0.026, P = 0.009), and combined parental age at death (β = 0.051, P = 0.006), but exhibited a negative association with mother’s attained age (β = − 0.026, P = 0.017), father’s attained age (β = − 0.024, P = 0.032), and combined parental attained age (β = -0.036, P = 0.001) (Fig. 6A). The PWY-7209: superpathway of pyrimidine ribonucleosides degradation exhibited a positive association with lifespan (β = 0.022, P = 0.032), but exhibited a opposite association with parental longevity of mother's attained age (β = − 0.016, P = 0.040) and combined parental attained age (β = -0.013, P = 0.046). Moreover, the pathway of REDCITCYC: TCA cycle VIII (helicobacter) was consistently negatively associated with lifespan, and three traits of parental longevity (both parents in top 10%, mother's age at death, and combined parental age at death) (Fig. 6B). The of HOMOSER-METSYN-PWY (L-methionine biosynthesis I) was consistently negatively associated with lifespan, and parental longevity of mother’s age at death. The PWY-6507 (4-deoxy-L-threo-hex-4-enopyranuronate degradation) pathway was consistently positively associated with lifespan and parental longevity (both parents in top 10%). In addition, the PWY-7456 (mannan degradation) pathway was positively associated with parental longevity of father's attained age, and negatively associated with FI.

Fig. 6
figure 6

The forest plots reveal the impact of two significant microbial pathways, NONOXIPEN-PWY and REDCITCYC, on multiple traits associated with longevity. A The notable associations between NONOXIPEN-PWY (pentose phosphate pathway, non-oxidative branch) pathway and traits associated with longevity. B The notable associations between REDCITCYC: TCA cycle VIII (helicobacter) pathway and traits associated with longevity. Four MR methods including IVW, weighted median, simple median, and MR Egger were used. The causal relationships were presented using OR (odds ratio) and 95% CI (confidence interval)

Discussion

In this study, we employed a two-sample MR based on GWAS summary data to extensively investigate the potential causal effects between gut microbiota and longevity. To the best of our knowledge, this study used the most comprehensive datasets currently available, including five publicly available GWAS datasets of longevity-related traits and four GWAS datasets of gut microbiota.

Previous research has established a significant correlation between Collinsella and parental longevity [23]. We found Collinsella was positively associated with parental longevity of mother's age at death, but Collinsella aerofaciens was negatively associated with lifespan. Moreover, Liu et al. [24] assessed the causal relationships between human microbiome and longevity by MR analyses in Chinese populations based on GWAS summary statistics of the gut and oral microbiome from the 4D-SZ cohort [39] and longevity from the CLHLS cohort [40], and found genus Oxalobacter and species Lactobacillus amylovorus were positively associated with longevity. Our results indicate that Oxalobacter exhibits a positive correlation with one aspect of parental longevity, specifically the attained age of the father, whereas it demonstrates a negative association with three other characteristics of parental longevity, namely the mother's age at death, the father's age at death, and the combined age at death of both parents. Oxalobacter formigenes, one typical species of Oxalobacter genus, is a key oxalate-degrading bacterium in the mammalian intestinal tract and can reduce the risk of calcium oxalate kidney stone disease [41, 42]. Therefore, Oxalobacter spp may protect humans from kidney stone disease and promote their overall health.

We also observed a negative association between Bacteroides massiliensis and longevity (age > 90th percentile, and > 99th percentile), indicating Bacteroides massiliensis may decrease the likelihood of achieving longevity. A higher relative abundance of B. massiliensis was observed in prostate cancer cases compared to benign controls [43]. A potential mechanism underlying this association may be the possession of glucuronidase genes by B. massiliensis, which, through its glucuronidase deconjugation activity, leads to elevated levels of free estrogens in the bloodstream. These increased estrogens subsequently create apurinic sites within DNA, causing mutations that may stimulate the onset of oncogenesis [44]. Moreover, the Oscillibacter abundance was positively correlated with lifespan and two traits of parental longevity, but was negatively associated with another two traits of parental longevity (mother's attained age, and combined parental attained age). The Alistipes abundance was positively correlated with combined parental age at death. More specific, Alistipes senegalensis was positively associated with lifespan, while Alistipes shahii abundance in stool was linked to increased longevity (age > 90th percentile). Interestingly, an increased relative abundance of fecal Oscillibacter and Alistipes has been causally linked to a decreased triglyceride concentration [39]. Species from the Oscillibacter genus involve cholesterol metabolism and show potential benefits for lipid homeostasis and cardiovascular health [45]. Furthermore, Alistipes may exhibit protective effects against liver cirrhosis and cardiovascular diseases [46]. Therefore, the positive effects of Oscillibacter and Alistipes spp on longevity probably be mediated by lipid homeostasis and cardiovascular health in human subjects. Additionally, the Subdoligranulum prevalence was positively correlated with parental longevity (combined parental age at death), and the Subdoligranulum abundance was negatively associated with frailty. It is noteworthy that the genera Subdoligranulum correlated with slower biological aging [47], potentially attributed to the production of anti-inflammatory short-chain fatty acids. Subdoligranulum was strongly associated with A. muciniphila in an overweight/obese population; however, the supplementation with a Subdoligranulum variabile strain in obese and diabetic mice did not yield beneficial effects [48]. There is a critical need for more extensive animal studies for Subdoligranulum spp, such as in aging-related animal models.

The casual association of some taxa on longevity are inconsistent. For instance, Akkermansia muciniphila was negatively correlated with longevity (age > 99th percentile), but was positively associated with three traits of parental longevity (mother's age at death, combined parental age at death, and both parents in top 10%). In addition, Desulfovibrio in the MiBioGen dataset was positively associated with parental longevity of combined parental age at death. Desulfovibrio in the German dataset was negatively correlated with lifespans and two parental longevity traits (both parents in top 10%, and combined parental age at death), while was positively associated with father's attained age and combined parental attained age. D. piger, a representative species of the Desulfovibrio genus, is identified as one of the gut microbes associated with human aging [49]. Flavonifractor was positively correlated with longevity (age > 99th percentile) and frailty, and was negatively correlated with parental longevity. One species of Flavonifractor genus, Flavonifractor plautii, has been found to protect against elevated arterial stiffness [50], but correlated with colorectal cancer [51, 52]. Consistently, Parasutterella exhibited a negative correlation with both lifespan and longevity (age > 90th percentile). Parasutterella was positively associated with BMI and type 2 diabetes [53], suggesting it may potentially affect metabolic diseases. Moreover, Blautia A sp900066355 was negatively correlated with longevity (age > 90th percentile) and parental longevity of mother's attained age. The mechanisms by which Desulfovibrio spp, Flavonifractor spp, Parasutterella spp, and Blautia A sp900066355 may contribute to disease pathology or healthy aging remains to be investigated.

As for microbial pathways, the abundance of PWY-6731(starch degradation III) was positively associated with longevity of age > 90th percentile. A previous GWAS study indicated that the starch, sucrose and xenobiotic metabolism pathway was highly associated with human longevity in Han Chinese [40]. Species such as Faecalibacterium prausnitzii, Eubacterium rectale, and Roseburia inulinivorans involve in the starch degradation and exert health-regulating effects by producing short-chain fatty acids (SCFAs) [54]. Thus, the starch degradation pathway may have a profound impact on longevity. COA-PWY (coenzyme A biosynthesis I), PWY-5100 (pyruvate fermentation to acetate and lactate II), and NONOXIPEN-PWY (pentose phosphate pathway, non-oxidative branch) exhibited a positive correlation with two or more longevity traits. Previous studies have revealed that acetyl-coenzyme A as a phylogenetically conserved inhibitor of age-associated autophagy and prolongs lifespan [55, 56]. In addition, the Sis2 gene regulated yeast lifespan through the coenzyme A biosynthesis pathway [57], implying a role for this pathway in longevity regulation. Lactate promotes oxidative stress resistance through hormetic ROS signaling [58], while acetate metabolism play a role in the regulation of aging and longevity [59]. The pentose phosphate pathway is crucial for cellular redox balance, and helps to maintain mitochondrial reactive oxygen species homeostasis and to extend lifespan in C. elegans [60]. Researchers also discovered that long‐lived flies exhibit a delayed age-related decline in protein turnover rates and elevated carbon flux into the pentose phosphate pathway [61]. These findings support a causal link between pentose phosphate pathway and lifespan extension. Furthermore, the TCA cycle VIII (helicobacter) pathway was consistently negatively associated with lifespan and three traits of parental longevity. The Helicobacter-specific tricarboxylic acid cycle (TCA cycle VIII) was associated with gastric carcinogenesis [62], indicating a potential harmful mechanism that contributes to the suppression of longevity. These pathways could serve as targets for future therapeutic approaches for aging-related disorders.

These findings have profound implications for our understanding of the causal relationships between the gut microbiome and human longevity. By identifying specific gut microbial taxa and their functional pathways associated with longevity, we gain a more precise picture of which microbial features may influence human lifespan. They not only enhance our understanding of longevity mechanisms but also pave the way for microbiome-targeted interventions to promote healthy aging. For instance, if we increase our intake of dietary fiber to enhance the abundance of starch degradation III pathway or appropriately use medications to eliminate Helicobacter pylori when H. pylori infection has been diagnosed, it is likely to improve our health.

Conclusions

This study reveals numerous causal associations between gut microbiota and longevity. Bacteroides massiliensis and Parasutterella were negatively associated with longevity, while Alistipes and Subdoligranulum exhibited a positive correlation with longevity. Microbial pathways of coenzyme A biosynthesis I, pyruvate fermentation to acetate and lactate II, pentose phosphate pathway, and TCA cycle VIII (helicobacter) were notably associated with longevity in a potentially protective or detrimental manner. More extensive population-based observational studies and longitudinal studies, as well as animal experiments, are needed to elucidate these causal associations and their underlying mechanisms.

Data availability

The datasets analyzed in the current study can be downloaded from the websites that were listed in supplementary Table 1.

References

  1. Partridge L, Deelen J, Slagboom PE. Facing up to the global challenges of ageing. Nature. 2018;561:45–56.

    Article  CAS  PubMed  Google Scholar 

  2. Chen X, Giles J, Yao Y, Yip W, Meng Q, Berkman L, Chen H, Chen X, Feng J, Feng Z, et al. The path to healthy ageing in China: a peking university-lancet commission. Lancet. 2022;400:1967–2006.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Collaborators GUHD. Life expectancy by county, race, and ethnicity in the USA, 2000–19: a systematic analysis of health disparities. Lancet. 2022;400:25–38.

    Article  Google Scholar 

  4. Pilling LC, Atkins JL, Bowman K, Jones SE, Tyrrell J, Beaumont RN, Ruth KS, Tuke MA, Yaghootkar H, Wood AR, et al. Human longevity is influenced by many genetic variants: evidence from 75,000 UK Biobank participants. Aging. 2016;8:547–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Deelen J, Evans DS, Arking DE, Tesi N, Nygaard M, Liu X, Wojczynski MK, Biggs ML, van der Spek A, Atzmon G, et al. A meta-analysis of genome-wide association studies identifies multiple longevity genes. Nat Commun. 2019;10:3669.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Du Y, Gao Y, Zeng B, Fan X, Yang D, Yang M. Effects of anti-aging interventions on intestinal microbiota. Gut Microbes. 2021;13:1994835.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Xu Q, Wu C, Zhu Q, Gao R, Lu J, Valles-Colomer M, Zhu J, Yin F, Huang L, Ding L, et al. Metagenomic and metabolomic remodeling in nonagenarians and centenarians and its association with genetic and socioeconomic factors. Nat Aging. 2022;2:438–52.

    Article  CAS  PubMed  Google Scholar 

  8. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–78.

    Article  CAS  PubMed  Google Scholar 

  9. Haran JP, McCormick BA. Aging, frailty, and the microbiome-how dysbiosis influences human aging and disease. Gastroenterology. 2021;160:507–23.

    Article  CAS  PubMed  Google Scholar 

  10. Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, Zhu D, Koya JB, Wei L, Li J, Chen ZS. Microbiota in health and diseases. Signal Transduct Target Ther. 2022;7:135.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Palmas V, Pisanu S, Madau V, Casula E, Deledda A, Cusano R, Uva P, Loviselli A, Velluzzi F, Manzin A. Gut microbiota markers and dietary habits associated with extreme longevity in healthy Sardinian Centenarians. Nutrients. 2022;14:2436.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sato Y, Atarashi K, Plichta DR, Arai Y, Sasajima S, Kearney SM, Suda W, Takeshita K, Sasaki T, Okamoto S, et al. Novel bile acid biosynthetic pathways are enriched in the microbiome of centenarians. Nature. 2021;599:458–64.

    Article  CAS  PubMed  Google Scholar 

  13. Pang S, Chen X, Lu Z, Meng L, Huang Y, Yu X, Huang L, Ye P, Chen X, Liang J, et al. Longevity of centenarians is reflected by the gut microbiome with youth-associated signatures. Nat Aging. 2023;3:436–49.

    Article  PubMed  Google Scholar 

  14. Chen S, Zhang Z, Liu S, Chen T, Lu Z, Zhao W, Mou X, Liu S. Consistent signatures in the human gut microbiome of longevous populations. Gut Microbes. 2024;16:2393756.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Bodogai M, O’Connell J, Kim K, Kim Y, Moritoh K, Chen C, Gusev F, Vaughan K, Shulzhenko N, Mattison JA, et al. Commensal bacteria contribute to insulin resistance in aging by activating innate B1a cells. Sci Transl Med. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scitranslmed.aat4271.

  16. Shin MG, Lee JW, Han JS, Lee B, Jeong JH, Park SH, Kim JH, Jang S, Park M, Kim SY, et al. Bacteria-derived metabolite, methylglyoxal, modulates the longevity of C. elegans through TORC2/SGK-1/DAF-16 signaling. Proc Natl Acad Sci USA. 2020;117:17142–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Han B, Sivaramakrishnan P, Lin CJ, Neve IAA, He J, Tay LWR, Sowa JN, Sizovs A, Du G, Wang J, et al. Microbial genetic composition tunes host longevity. Cell. 2017;169(1249–1262): e1213.

    Google Scholar 

  18. Xu W, Rustenhoven J, Nelson CA, Dykstra T, Ferreiro A, Papadopoulos Z, Burnham CD, Dantas G, Fremont DH, Kipnis J. A novel immune modulator IM33 mediates a glia-gut-neuronal axis that controls lifespan. Neuron. 2023;111(3244–3254): e3248.

    Google Scholar 

  19. Singh PP, Demmitt BA, Nath RD, Brunet A. The genetics of aging: a vertebrate perspective. Cell. 2019;177:200–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sanderson E, Glymour MM, Holmes MV, Kang H, Morrison J, Munafò MR, Palmer T, Schooling CM, Wallace C, Zhao Q, Davey Smith G. Mendelian randomization. Nat Rev Methods Primers. 2022;2:6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Qin Y, Havulinna AS, Liu Y, Jousilahti P, Ritchie SC, Tokolyi A, Sanders JG, Valsta L, Brozynska M, Zhu Q, et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat Genet. 2022;54:134–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen L, Zhernakova DV, Kurilshikov A, Andreu-Sanchez S, Wang D, Augustijn HE, Vich Vila A, Lifelines Cohort S, Weersma RK, Medema MH, et al. Influence of the microbiome, diet and genetics on inter-individual variation in the human plasma metabolome. Nat Med. 2022;28:2333–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. He D, Liu L, Zhang Z, Yang X, Jia Y, Wen Y, Cheng S, Meng P, Li CE, Zhang H, et al. Association between gut microbiota and longevity: a genetic correlation and mendelian randomization study. BMC Microbiol. 2022;22:302.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Liu X, Zou L, Nie C, Qin Y, Tong X, Wang J, Yang H, Xu X, Jin X, Xiao L, et al. Mendelian randomization analyses reveal causal relationships between the human microbiome and longevity. Sci Rep. 2023;13:5127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Qian H, Gong R, Li Y, Zhu J, Wang L. A genetically informed study reveals modifiable pathways in skin cancer. J Transl Med. 2024;22:916.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Zenin A, Tsepilov Y, Sharapov S, Getmantsev E, Menshikov LI, Fedichev PO, Aulchenko Y. Identification of 12 genetic loci associated with human healthspan. Commun Biol. 2019;2:41.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Timmers PR, Mounier N, Lall K, Fischer K, Ning Z, Feng X, Bretherick AD, Clark DW, Shen X, et al. Genomics of 1 million parent lifespans implicates novel pathways and common diseases and distinguishes survival chances. Elife. 2019;8:e39856.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Pilling LC, Kuo CL, Sicinski K, Tamosauskaite J, Kuchel GA, Harries LW, Herd P, Wallace R, Ferrucci L, Melzer D. Human longevity: 25 genetic loci associated in 389,166 UK biobank participants. Aging. 2017;9:2504–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ghosh TS, Shanahan F, O’Toole PW. The gut microbiome as a modulator of healthy ageing. Nat Rev Gastroenterol Hepatol. 2022;19:565–84.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Atkins JL, Jylhava J, Pedersen NL, Magnusson PK, Lu Y, Wang Y, Hagg S, Melzer D, Williams DM, Pilling LC. A genome-wide association study of the frailty index highlights brain pathways in ageing. Aging Cell. 2021;20: e13459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kurilshikov A, Medina-Gomez C, Bacigalupe R, Radjabzadeh D, Wang J, Demirkan A, Le Roy CI, Raygoza Garay JA, Finnicum CT, Liu X, et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat Genet. 2021;53:156–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lopera-Maya EA, Kurilshikov A, van der Graaf A, Hu S, Andreu-Sanchez S, Chen L, Vila AV, Gacesa R, Sinha T, Collij V, et al. Effect of host genetics on the gut microbiome in 7738 participants of the Dutch microbiome project. Nat Genet. 2022;54:143–51.

    Article  CAS  PubMed  Google Scholar 

  33. Ruhlemann MC, Hermes BM, Bang C, Doms S, Moitinho-Silva L, Thingholm LB, Frost F, Degenhardt F, Wittig M, Kassens J, et al. Genome-wide association study in 8,956 German individuals identifies influence of ABO histo-blood groups on gut microbiome. Nat Genet. 2021;53:147–55.

    Article  PubMed  Google Scholar 

  34. Hemani G, Bowden J, Davey Smith G. Evaluating the potential role of pleiotropy in Mendelian randomization studies. Hum Mol Genet. 2018;27:R195-r208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu B, Lyu L, Zhou W, Song J, Ye D, Mao Y, Chen GB, Sun X. Associations of the circulating levels of cytokines with risk of amyotrophic lateral sclerosis: a Mendelian randomization study. BMC Med. 2023;21:39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhu Z, Zhang F, Hu H, Bakshi A, Robinson MR, Powell JE, Montgomery GW, Goddard ME, Wray NR, Visscher PM, Yang J. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat Genet. 2016;48:481–7.

    Article  CAS  PubMed  Google Scholar 

  37. Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018;50:693–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Cui G, Li S, Ye H, Yang Y, Jia X, Lin M, Chu Y, Feng Y, Wang Z, Shi Z, Zhang X. Gut microbiome and frailty: insight from genetic correlation and mendelian randomization. Gut Microbes. 2023;15:2282795.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Liu X, Tong X, Zou Y, Lin X, Zhao H, Tian L, Jie Z, Wang Q, Zhang Z, Lu H, et al. Mendelian randomization analyses support causal relationships between blood metabolites and the gut microbiome. Nat Genet. 2022;54:52–61.

    Article  CAS  PubMed  Google Scholar 

  40. Zeng Y, Nie C, Min J, Liu X, Li M, Chen H, Xu H, Wang M, Ni T, Li Y, et al. Novel loci and pathways significantly associated with longevity. Sci Rep. 2016;6:21243.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Daniel SL, Moradi L, Paiste H, Wood KD, Assimos DG, Holmes RP, Nazzal L, Hatch M, Knight J. Forty years of oxalobacter formigenes, a gutsy oxalate-degrading specialist. Appl Environ Microbiol. 2021;87: e0054421.

    Article  PubMed  Google Scholar 

  42. Hiremath S, Viswanathan P. Oxalobacter formigenes: a new hope as a live biotherapeutic agent in the management of calcium oxalate renal stones. Anaerobe. 2022;75: 102572.

    Article  CAS  PubMed  Google Scholar 

  43. Golombos DM, Ayangbesan A, O’Malley P, Lewicki P, Barlow L, Barbieri CE, Chan C, DuLong C, Abu-Ali G, Huttenhower C, Scherr DS. The role of gut microbiome in the pathogenesis of prostate cancer: a prospective, pilot study. Urology. 2018;111:122–8.

    Article  PubMed  Google Scholar 

  44. Garbas K, Zapala P, Zapala L, Radziszewski P. The role of microbial factors in prostate cancer development-an up-to-date review. J Clin Med. 2021;10:4772.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li C, Strazar M, Mohamed AMT, Pacheco JA, Walker RL, Lebar T, Zhao S, Lockart J, Dame A, Thurimella K, et al. Gut microbiome and metabolome profiling in Framingham heart study reveals cholesterol-metabolizing bacteria. Cell. 2024;187:1834–52.

    Article  CAS  PubMed  Google Scholar 

  46. Parker BJ, Wearsch PA, Veloo ACM, Rodriguez-Palacios A. The genus alistipes: gut bacteria with emerging implications to inflammation, cancer, and mental health. Front Immunol. 2020;11:906.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Singh S, Giron LB, Shaikh MW, Shankaran S, Engen PA, Bogin ZR, Bambi SA, Goldman AR, Azevedo J, Orgaz L, et al. Distinct intestinal microbial signatures linked to accelerated systemic and intestinal biological aging. Microbiome. 2024;12:31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Van Hul M, Le Roy T, Prifti E, Dao MC, Paquot A, Zucker JD, Delzenne NM, Muccioli G, Clement K, Cani PD. From correlation to causality: the case of Subdoligranulum. Gut Microbes. 2020;12:1–13.

    PubMed  Google Scholar 

  49. Sun L, Li Z, Hu C, Ding J, Zhou Q, Pang G, Wu Z, Yang R, Li S, Li J, et al. Age-dependent changes in the gut microbiota and serum metabolome correlate with renal function and human aging. Aging Cell. 2023;22: e14028.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Luo S, Zhao Y, Zhu S, Liu L, Cheng K, Ye B, Han Y, Fan J, Xia M. Flavonifractor plautii protects against elevated arterial stiffness. Circ Res. 2023;132:167–81.

    Article  CAS  PubMed  Google Scholar 

  51. Yang Y, Du L, Shi D, Kong C, Liu J, Liu G, Li X, Ma Y. Dysbiosis of human gut microbiome in young-onset colorectal cancer. Nat Commun. 2021;12:6757.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gupta A, Dhakan DB, Maji A, Saxena R, Prasoodanan KV, Mahajan S, Pulikkan J, Kurian J, Gomez AM, Scaria J, et al. Association of Flavonifractor plautii, a flavonoid-degrading bacterium, with the gut microbiome of colorectal cancer patients in India. mSystems. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/msystems.00438-19.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Henneke L, Schlicht K, Andreani NA, Hollstein T, Demetrowitsch T, Knappe C, Hartmann K, Jensen-Kroll J, Rohmann N, Pohlschneider D, et al. A dietary carbohydrate - gut Parasutterella - human fatty acid biosynthesis metabolic axis in obesity and type 2 diabetes. Gut Microbes. 2022;14:2057778.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Liu S, Zhao W, Liu X, Cheng L. Metagenomic analysis of the gut microbiome in atherosclerosis patients identify cross-cohort microbial signatures and potential therapeutic target. FASEB J. 2020;34:14166–81.

    Article  CAS  PubMed  Google Scholar 

  55. Schroeder S, Pendl T, Zimmermann A, Eisenberg T, Carmona-Gutierrez D, Ruckenstuhl C, Marino G, Pietrocola F, Harger A, Magnes C, et al. Acetyl-coenzyme A: a metabolic master regulator of autophagy and longevity. Autophagy. 2014;10:1335–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Eisenberg T, Schroeder S, Andryushkova A, Pendl T, Kuttner V, Bhukel A, Marino G, Pietrocola F, Harger A, Zimmermann A, et al. Nucleocytosolic depletion of the energy metabolite acetyl-coenzyme a stimulates autophagy and prolongs lifespan. Cell Metab. 2014;19:431–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Olmez TT, Moreno DF, Liu P, Johnson ZM, McGinnis MM, Tu BP, Hochstrasser M, Acar M. Sis2 regulates yeast replicative lifespan in a dose-dependent manner. Nat Commun. 2023;14:7719.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tauffenberger A, Fiumelli H, Almustafa S, Magistretti PJ. Lactate and pyruvate promote oxidative stress resistance through hormetic ROS signaling. Cell Death Dis. 2019;10:653.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Shimazu T, Hirschey MD, Huang JY, Ho LT, Verdin E. Acetate metabolism and aging: an emerging connection. Mech Ageing Dev. 2010;131:511–6.

    Article  CAS  PubMed  Google Scholar 

  60. Feng X, Wang X, Zhou L, Pang S, Tang H. The impact of glucose on mitochondria and life span is determined by the integrity of proline catabolism in Caenorhabditis elegans. J Biol Chem. 2023;299: 102881.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang L, Davis SS, Borch Jensen M, Rodriguez-Fernandez IA, Apaydin C, Juhasz G, Gibson BW, Schilling B, Ramanathan A, Ghaemmaghami S, Jasper H. JNK modifies neuronal metabolism to promote proteostasis and longevity. Aging Cell. 2019;18: e12849.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Liu C, Ng SK, Ding Y, Lin Y, Liu W, Wong SH, Sung JJ, Yu J. Meta-analysis of mucosal microbiota reveals universal microbial signatures and dysbiosis in gastric carcinogenesis. Oncogene. 2022;41:3599–610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by the National Natural Science Foundation of China (No. 32100039) and Shenzhen Science and Technology Program (No. szbo202215).

Author information

Authors and Affiliations

Authors

Contributions

Sheng Liu contributed to study design. Shu Chen and Wei Chen collected, and analyzed the data. Xudong Wang contributed to the visualization. Shu Chen drafted the manuscript. Sheng Liu revised the manuscript. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Sheng Liu.

Ethics declarations

Ethics approval and concern to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

No conflicts of interest to declare.

Additional information

Publisher's Note

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

Supplementary Information

12967_2024_5823_MOESM1_ESM.pdf

Supplementary material 1: Figure S1. The forest plot depicts the MR effect size for species Bacteroides massiliensis on longevity (age >90th percentile, and age >99th percentile) and frailty.

12967_2024_5823_MOESM2_ESM.pdf

Supplementary material 2: Figure S2. The MR leave-one-out sensitivity analysis for Bacteroides massiliensis on longevity (>90th percentile, and >99th percentile).

12967_2024_5823_MOESM3_ESM.pdf

Supplementary material 3: Figure S3. The forest plot depicts the causal associations between Oscillibacter prevalence, as well as Oscillibacter abundance, and traits associated with human longevity.

12967_2024_5823_MOESM4_ESM.pdf

Supplementary material 4: Figure S4. The MR leave-one-out sensitivity analysis for Alistipes senegalensis on lifespan, and for Alistipes shahii on longevity (>90th percentile).

12967_2024_5823_MOESM5_ESM.pdf

Supplementary material 5: Figure S5. The MR leave-one-out sensitivity analysis for Subdoligranulum prevalence on Parental longevity (combined parental age at death), and for Subdoligranulum abundance on frailty index.

12967_2024_5823_MOESM6_ESM.pdf

Supplementary material 6: Figure S6. The forest plot depicts the causal associations between Parasutterella and longevity-related traits, including frailty. All these associations were negative.

12967_2024_5823_MOESM7_ESM.xlsx

Supplementary material 7: Table S1. Details of the genome-wide association studies and datasets used in our Mendelian Randomization (MR) study.

12967_2024_5823_MOESM8_ESM.xlsx

Supplementary material 8: Table S2. The significant influence of 403 gut bacterial taxa from MiBioGen and DMP datasets on longevity‑related phenotypes.

12967_2024_5823_MOESM9_ESM.xlsx

Supplementary material 9: Table S3. The significant influence of 903 gut bacterial taxa from German and Finland individuals on longevity‑related phenotypes.

12967_2024_5823_MOESM10_ESM.xlsx

Supplementary material 10: Table S4. The significant influence of the 205 microbial pathways from DMP dataset on specific longevity-related phenotypes.

12967_2024_5823_MOESM11_ESM.xlsx

Supplementary material 11: Table S5. The MR results reveal the notable associations between specific one microbial pathway and multiple longevity-related traits among different datasets.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, S., Chen, W., Wang, X. et al. Mendelian randomization analyses support causal relationships between gut microbiome and longevity. J Transl Med 22, 1032 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05823-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05823-2