Skip to main content
Precision Clinical Medicine logoLink to Precision Clinical Medicine
. 2023 May 24;6(2):pbad010. doi: 10.1093/pcmedi/pbad010

Investigating association between gut microbiota and sarcopenia-related traits: a Mendelian randomization study

Jiaxi Zhao 1, Rui Liang 2, Quhong Song 3, Shiyu Song 4, Jirong Yue 5,, Chenkai Wu 6
PMCID: PMC10263384  PMID: 37324750

Abstract

Background

Observational studies have indicated a potential link between gut microbiota and sarcopenia. However, the underlying mechanisms and a causal relationship have not been established. Thus, the objective of this study is to examine the possible causal association between gut microbiota and sarcopenia-related traits, including low hand-grip strength and appendicular lean mass (ALM), to shed light on the gut–muscle axis.

Methods

To investigate the potential impact of gut microbiota on low hand-grip strength and ALM, we utilized a two-sample Mendelian randomization (MR) approach. Summary statistics were obtained from genome-wide association studies of gut microbiota, low hand-grip strength, and ALM. The primary MR analysis employed the random-effects inverse-variance weighted (IVW) method. To assess the robustness, we conducted sensitivity analyses using the MR pleiotropy residual sum and outlier (MR-PRESSO) test to detect and correct for horizontal pleiotropy, as well as the MR-Egger intercept test and leave-one-out analysis.

Results

Alcaligenaceae, Family XIII, and Paraprevotella were positively associated with the risk of low hand-grip strength (P-values < 0.05). Streptococcaceae were negatively associated with low hand-grip strength (P-values < 0.05). Eight bacterial taxa (Actinomycetales, Actinomycetaceae, Bacteroidaceae, Porphyromonadaceae, Prevotellaceae, Bacteroides, Marvinbryantia, and Phascolarctobacterium) were associated with a higher risk of ALM (P-values < 0.05). Eubacterium fissicatena group was negatively associated with ALM (P-values < 0.05).

Conclusion

We found several gut microbiota components causally associated with sarcopenia-related traits. Our findings provided insights into novel strategies for the prevention and treatment of sarcopenia through the regulation of the gut microbiota, contributing to a better understanding of the gut–muscle axis.

Keywords: gut microbiota, sarcopenia, low hand-grip strength, appendicular lean mass, Mendelian randomization, gut–muscle axis

Introduction

The human intestine harbours a diverse community of gut microbiota that can be influenced by various factors such as diet, environment, and genetics.1 Previous studies have demonstrated the crucial role of gut microbiota in the development of immune, metabolic, psychiatric, and inflammatory diseases.2–5 Sarcopenia, characterized by low muscle mass and weak muscle strength, is a common skeletal muscle syndrome that often accompanies ageing and several chronic conditions,6 which leads to impaired mobility, increased morbidity and mortality,7–9 and substantially elevated healthcare expenditures for patients.10 Although the gut–muscle axis, which links gut health to muscle and physical function, has been proposed by several independent research groups,11–13 the association between gut microbiota and sarcopenia remains debatable. A better understanding of the role of gut microbiota in the management of sarcopenia could pave the way for more viable treatments.

The gut–muscle axis has been shown to play a crucial role in regulating age-related muscle health in animal models and human studies. However, the underlying mechanisms remain poorly understood, and the findings have been inconsistent across studies. Observational studies have reported that gut microbiota can impact muscle mass and function through various factors such as inflammation, immunity, substance metabolism, endocrine function, and insulin sensitivity.14 By regulating the composition and metabolism of proteins and amino acids, gut microbiota can influence muscle synthesis.15,16 Tryptophan, for instance, may stimulate the production of insulin-like growth factor-1 in muscle cells, promoting the expression of myofibril-related genes.17 Conversely, a protein-rich diet has been shown to shift bacterial metabolism to the degradation and fermentation of amino acids.18 A healthy gut microbiota can improve intestinal barrier function and act as an immunomodulator. However, short-chain fatty acids (SCFAs), which are produced by certain gut microbiota, have been linked to increased chronic inflammation, which in turn can lead to sarcopenia.19 Nonetheless, observational studies are subject to methodological challenges, such as confounding bias and reverse causation, and methods that can infer causation are needed to identify the association between gut microbiota and sarcopenia.

Mendelian randomization (MR) is a valuable approach for establishing potential relationships that overcomes the methodological issues commonly present in observational studies by using genetic variations as instrumental variables (IVs) for exposures.20 The goal of this study was to uncover the potential causal association between gut microbiota and sarcopenia, which would lead to a better understanding of the gut–muscle axis.

Material and methods

Study design

To establish potential causal relationships between gut microbiota and sarcopenia-related traits, namely low hand-grip strength and appendicular lean mass (ALM), we employed a two-sample MR analysis, which provides stronger associations by minimizing many of the major biases present in traditional epidemiologic observational studies. A flow chart of the study design is shown in Fig. 1.

Figure 1.

Figure 1.

Flowchart for the study of the association between gut microbiota and sarcopenia-related traits. LD: linkage disequilibrium; SNP: single nucleotide polymorphism.

Data sources and instruments

Gut microbiota

We selected single-nucleotide polymorphisms (SNPs) associated with the gut microbiota as IVs for our study. The summary-level data for these SNPs were obtained from the MiBioGen consortium, which analyzed 16S rRNA gene sequencing data from 18 340 participants of various ethnicities and nationalities (72.3% of the population were European, with the remainder including individuals from Middle-Eastern, East Asian, American Hispanic/Latin, and African American populations).21

To select the most appropriate SNPs, we established a P-value threshold of <10−5.21,22 All SNPs need to be independently associated with the microbiota (linkage disequilibrium [LD], r2 < 0.001, clumping distance = 10 000 kb). Following the removal of 15 unknown bacterial traits, we identified a total of 2 559 SNPs associated with 196 bacterial traits, including 9 phyla, 16 classes, 20 orders, 32 families, and 119 genera. An overview of genome-wide association studies (GWAS) datasets for gut microbiota is shown in supplementary Table 1, see online supplementary material. IVs are listed in Supplementary Table 2, see online supplementary material. To avoid potential pleiotropy, we also used the PhenoScanner V2 (http://www.phenoscanner.medschl.cam.ac.uk/) to exclude IVs associated with confounding or risk factors for sarcopenia, such as older age, low socioeconomic status, low physical activity, and poor diet.23 We submitted SNPs strongly linked to gut microbiota for retrieval on the website. SNPs associated with risk factors for sarcopenia, including older age, low socioeconomic status, low physical activity, and poor diet, were excluded (using a P-value threshold of <10−5). Finally, we discovered no SNPs that were significantly associated with the aforementioned four variables. As a result, 2 559 SNPs were available for further MR analysis. All the confounders and excluded SNPs are listed in supplementary Table 3, see online supplementary material.

Sarcopenia-related traits

For each outcome variable, we have chosen the most recent and largest GWAS studies available. Summary-level data for low hand-grip strength were extracted from a large meta-analysis GWAS including 256 523 participants of European ancestry24 (ebi-a-GCST90007526). The cutoff level of low hand-grip strength was male <30 kg, female <20 kg. Summary statistics of ALM were retrieved from a GWAS, in which 450 243 participants of the UK Biobank Study were included25 (ebi-a-GCST90000025). An overview of GWAS datasets for low hand-grip strength and ALM is shown in supplementary Table 1.

Instrument strength

To rule out the influence of weak IVs bias in association effect estimation, we tested the strength of IVs with an F statistic > 10. The following equation was used to compute the F statistic: F = (R2/1−R2) (nk−1/k), where R2 is the variance interpreted by IVs (per gut microbiota), and n is the sample size.26 The minor allele frequency (MAF) and effect estimates (β) were used to calculate R2: R2 = 2MAF (1−MAF) (β/SD)2. SD is the standard deviation. The F values of all variables in the present study were >10.

Statistical analysis

Our study utilized two-sample MR analysis as the primary statistical technique to investigate the potential causal association between gut microbiota and low hand-grip strength and ALM. To obtain a robust and reliable result, we combined several statistical methods. The inverse-variance weighted (IVW) method was used as the primary approach, which has stable and balanced pleiotropic effects. In addition, we employed the MR-Egger regression, weighted median, and MR pleiotropy residual sum and outlier (MR-PRESSO) tests as complementary methods to IVW, to estimate the relationship between exposures and outcomes under various conditions.27,28 When the two-sided P-value was <0.05, statistical significance was considered.

The MR estimates are presented as odds ratios (ORs) and corresponding 95% confidence intervals (CIs). The TwoSampleMR package (version 0.5.6) in R (v4.2.2) was used for the statistical analyses.29

Sensitivity analysis

In our study, we utilized several methods to ensure the robustness and reliability of our findings. To detect any potential heterogeneity, we employed Cochran's Q statistic and visualized funnel plots with a significance threshold of P-value < 0.05. Then we utilized the MR-PRESSO test to detect and remove outliers, and retested the results. Additionally, to detect horizontal pleiotropy, we used the MR-Egger intercept test, with a significance threshold of P-value < 0.05. Results are displayed using scatter plots.

We also performed leave-one-out analyzes where we repeated the IVW analysis, removing one exposure-related SNP at a time, to assess the robustness of our findings. By combining these methods, we aimed to ensure the validity and reliability of our findings in determining the potential causal association between gut microbiota and the risk of low hand-grip strength and ALM.

Results

Gut microbiota and low hand-grip strength

Four bacterial taxa were found to be statistically associated with the risk of low hand-grip strength using the IVW method (Fig. 2). Alcaligenaceae, Family XIII, and Paraprevotella were found to have significant positive causal effects on low hand-grip strength (OR: 1.167, 95% CI: 1.051–1.297, P = 0.004 for Alcaligenaceae; OR: 1.111, 95% CI: 1.022–1.209, P = 0.013 for Family XIII; OR: 1.088, 95% CI: 1.036–1.142, P = 0.001 for Paraprevotella; Table 1). Streptococcaceae had a negative impact on low hand-grip strength (OR: 0.920, 95% CI: 0.855–0.991, P = 0.027). We did not find evidence of heterogeneity or horizontal pleiotropy based on the funnel, scatter, and forest plots (Supplementary Figs. 1–3). Multiple test corrections for the P-values are shown in supplementary Table 4, see online supplementary material.

Figure 2.

Figure 2.

IVW estimates for association between gut microbiota and sarcopenia-related traits. The color of each block represents the IVW-derived P-value for each MR analysis.

Table 1.

MR estimates of the association between gut microbiota and the risk of low hand-grip strength.

Exposure Outcome No. of IVs Method OR (95% CI) P-valuea Heterogeneity MR Egger
Cochran's Q (P-value) Intercept (P-value)
Alcaligenaceae LHGS 18 IVW 1.167 (1.051, 1.297) 0.004 18.589 (0.069) −0.002 (0.922)
MR Egger 1.197 (0.721, 1.986) 0.502
Weighted median 1.194 (1.053, 1.353) 0.006
Family XIII LHGS 14 IVW 1.111 (1.022, 1.209) 0.013 9.990 (0.441) −0.002 (0.866)
MR Egger 1.140 (0.846, 1.534) 0.411
Weighted median 1.109 (0.990, 1.243) 0.075
Streptococcaceae LHGS 17 IVW 0.920 (0.855, 0.991) 0.027 9.492 (0.735) 0.003 (0.782)
MR Egger 0.883 (0.657, 1.186) 0.425
Weighted median 0.906 (0.819, 1.002) 0.054
Paraprevotella LHGS 13 IVW 1.088 (1.036, 1.142) 0.001 9.180 (0.688) 0.009 (0.337)
MR Egger 1.006 (0.858, 1.180) 0.939
Weighted median 1.073 (1.000, 1.151) 0.049

aBold values indicate P-value < 0.05. LHGS, low hand-grip strength.

Gut microbiota and ALM

There were nine bacterial taxa causally associated with ALM in the IVW method (Fig. 2). Actinomycetales, Actinomycetaceae, Bacteroidaceae, Porphyromonadaceae, Prevotellaceae, Bacteroides, Marvinbryantia, Phascolarctobacterium had positive effects on ALM (OR: 1.029, 95% CI: 1.005–1.053, P = 0.017 for Actinomycetales; OR: 1.029, 95% CI: 1.005–1.052, P = 0.016 for Actinomycetaceae; OR: 1.041, 95% CI: 1.010–1.073, P = 0.008 for Bacteroidaceae; OR: 1.036, 95% CI: 1.002–1.071, P = 0.037 for Porphyromonadaceae; OR: 1.034, 95% CI: 1.007–1.061, P = 0.012 for Prevotellaceae; OR: 1.041, 95% CI: 1.010–1.073, P = 0.008 for Bacteroides; OR: 1.041, 95% CI: 1.000–1.046, P = 0.045 for Marvinbryantia; OR: 1.027, 95% CI: 1.001–1.054, P = 0.041 for Phascolarctobacterium). Eubacterium fissicatena group had a negative effect on ALM (OR: 0.983, 95% CI: 0.967–1.100, P = 0.044) (Table 2). Multiple test corrections for the P-values are shown in supplementary Table 4.

Table 2.

MR estimates of the association between gut microbiota and the risk of ALM.

Exposure Outcome No. of IVs Method OR (95% CI) P-valuea Heterogeneity MR Egger
Cochran's Q (P-valuea) Intercept (P-value)
Actinomycetales ALM 5 IVW 1.029 (1.005, 1.053) 0.017 5.085 (0.279) 0.003 (0.358)
MR Egger 0.999 (0.943, 1.058) 0.972
Weighted median 1.015 (0.987, 1.044) 0.278
Actinomycetaceae ALM 5 IVW 1.029 (1.005, 1.052) 0.016 5.064 (0.281) 0.003 (0.361)
MR Egger 0.999 (0.943, 1.058) 0.978
Weighted median 1.015 (0.988, 1.043) 0.275
Bacteroidaceae ALM 12 IVW 1.041 (1.010, 1.073) 0.008 12.005 (0.151) 0.002 (0.711)
MR Egger 1.008 (0.852, 1.192) 0.927
Weighted median 1.022 (0.987, 1.060) 0.215
Porphyromonadaceae ALM 11 IVW 1.036 (1.002, 1.071) 0.037 13.816 (0.087) 0.005 (0.232)
MR Egger 0.951 (0.832, 1.086) 0.481
Weighted median 1.047 (1.010, 1.086) 0.013
Prevotellaceae ALM 18 IVW 1.034 (1.007, 1.061) 0.012 38.021 (0.001) 0.003 (0.359)
MR Egger 0.990 (0.902, 1.087) 0.838
Weighted median 1.018 (0.994, 1.043) 0.142
Bacteroides ALM 12 IVW 1.041 (1.010, 1.073) 0.008 12.005 (0.151) 0.002 (0.711)
MR Egger 1.008 (0.852, 1.192) 0.927
Weighted median 1.022 (0.987, 1.060) 0.223
Eubacterium fissicatena group ALM 9 IVW 0.983 (0.967, 1.100) 0.044 16.317 (0.038) 0.004 (0.540)
MR Egger 0.954 (0.870, 1.047) 0.353
Weighted median 0.978 (0.961, 1.995) 0.010
Marvinbryantia ALM 12 IVW 1.023 (1.000, 1.046) 0.045 12.223 (0.201) −0.001 (0.769)
MR Egger 1.037 (0.946, 1.137) 0.457
Weighted median 1.033 (1.007, 1.059) 0.013
Phascolarctobacterium ALM 11 IVW 1.027 (1.001, 1.054) 0.041 14.772 (0.064) −0.001 (0.888)
MR Egger 1.037 (0.909, 1.183) 0.603
Weighted median 1.014 (0.984, 1.042) 0.322

aBold values indicate P-value < 0.05.

In the sensitivity analysis, only two bacterial taxa, Prevotellaceae and Eubacterium fissicatena group, showed heterogeneity. We removed the outliers and performed the MR-PRESSO test to correct the heterogeneity. The results showed a significant association (outlier-corrected: P = 0.034 for Prevotellaceae; P = 0.039 for Eubacterium fissicatena group). Detailed information is provided in supplementary Table 5, see online supplementary material. We did not find evidence of horizontal pleiotropy based on the funnel, scatter, and forest plots (supplementary Figs. 4–6, see online supplementary material).

Discussion

This study represents the first comprehensive MR analysis investigating the potential causal relationships between gut microbiota and sarcopenia-related traits. Our systematic evaluation of the genetically predicted causal association between gut microbiota and low hand-grip strength and ALM yielded novel findings indicating that different gut microbiota taxa exert different effects on muscle. These findings are consistent with earlier observational studies highlighting the pathophysiological interactions between gut microbiota and muscle, commonly referred to as the gut–muscle axis.11–13

Our results indicated that Alcaligenaceae, Family XIII, and Paraprevotella had significant positive causal effects on low hand-grip strength, which is negatively correlated with muscle strength. In contrast, Streptococcaceae had a significantly negative effect on low hand-grip strength. Previous studies have linked Paraprevotella to the onset and progression of irritable bowel syndrome (IBS) and associated chronic intestinal inflammation.30 Chronic intestinal inflammation and malabsorption of nutrients can lead to decreased muscle function. However, in patients with chronic kidney disease, Paraprevotella inversely correlates with the inflammatory factors interleukin 10 (IL-10) and IL-4,31 which suggests that the association between Paraprevotella and muscle function may be modulated by underlying disease states. In a previous study, Streptococcaceae dietary supplements were found to help muscles recover from strenuous eccentric exercise, with a corresponding decrease in blood IL-6 levels after 3 weeks of supplementation.32 Overall, our MR-derived associations between gut microbiota and muscle function are largely consistent with previous observational studies.

For another sarcopenia-related trait, Actinomycetales, Actinomycetaceae, Bacteroidaceae, Porphyromonadaceae, Prevotellaceae, Bacteroides, Marvinbryantia, and Phascolarctobacterium were found to have significant positive causal effects on ALM, while Eubacterium fissicatena group were found to have significant negative causal effects on ALM in the present study. Patients with liver cirrhosis combined with muscle wasting had a higher abundance of Bacteroides in their gut than those without muscle wasting.33Bacteroides can utilize both endogenous and exogenous polysaccharides to create materials that might be advantageous to the host. When substances like cellulose enter the colon, Bacteroides ferment and break them down into SCFAs, which are thought to reduce inflammation and aid in the maintenance of skeletal muscle mass. SCFAs appear to improve skeletal muscle function, and some studies suggested that SCFAs supplementation may improve exercise performance or strength.34,35 Pei et al.’s research discovered that Bacteroides could also improve the barrier function of the intestinal mucosa and inhibit the Mystn/ActRIIB/SMAD2 pathway to prevent muscle atrophy.36 In general, Bacteroides can affect muscle function and mass by improving intestinal absorption and energy metabolism. A study transplanted the feces of HF (high-function) and LF (low-function) elderly separately into germ-free mice, and the grip strength of HF-colonized mice increased after a 1-month follow-up, which suggested that these gut microbiota had a beneficial role in maintaining muscle strength in the elderly over 65 years old.37 The gut microbiota Prevotellaceae was present in the HF group in the experiment. Community-dwelling older adults with sarcopenia had a higher concentration of Phascolarctobacterium than the control group.38 Moreover, the reduction of Phascolarctobacterium was associated with increased intestinal permeability, inflammation, and nutritional imbalance. Another study found that the body mass index, hand-grip strength, and mid-upper arm muscle circumference were positively correlated with Phascolarctobacterium.39

However, there were several studies’ results inconsistent with the present study. Eubacterium fissicatena group was significantly positively correlated with appendicular skeletal mass index and grip strength, and significantly negatively correlated with five-time chair standing test time, indicating that the Eubacterium fissicatena group's abundance is positively correlated with muscle mass and muscle function.40 The inconsistency in the results could be attributed to the observational studies' confounding factors. The positive correlation between Porphyromonadaceae and excess visceral adipose tissue (VAT) suggests that it may have an impact on the body's endocrine system.41 Through the endocrine pathway of adipokines, obesity, especially central obesity with increased VAT, increases the likelihood of sarcopenia.14 This condition is known as sarcopenic obesity. Further research into whether Porphyromonadaceae affects muscle is necessary. There is currently no research on the effects of Alcaligenaceae and Family XIII on low hand-grip strength, and the effects of Actinomycetales, Actinomycetaceae, Prevotellaceae, and Marvinbryantia on ALM need to be further explored. Theoretically, low hand-grip strength and ALM should coexist in most cases of sarcopenia patients. However, the gut microbiota related to these two variables in this study did not overlap. This may be due to several reasons: first, different gut microbiota may affect muscle mass or strength by secreting similar factors. Second, there is a certain threshold range in the study, and gut microbiota that reaches this threshold range is determined to have statistically significant effects. However, if the threshold is relaxed, gut microbiota related to low hand-grip strength and ALM may have further overlap. In general, the relationships discovered in this study between gut microbiota taxa and ALM were partially consistent with previous research findings, but there were still some research findings inconsistent or even contradictory. Future research into the mechanism underlying the relationship between specific gut microbiota taxa and muscle function and muscle mass is needed.

Our finding demonstrates the presence of four gut microbiota associated with reduced hand-grip strength, and nine gut microbiota associated with ALM. It is worth noting that other gut microbiota not covered in this study may also play a role in regulating muscle function and mass. These findings provide valuable insights for potential pharmacological interventions in the treatment of sarcopenia.42 A study by Eloe-Fadrosh et al. found that a supplement of Lactobacillus could modify resident microbiota to increase the production of the SCFAs, which could increase the cross-sectional area of muscle fibers and stop intramuscular fat from accumulating.43 Munukka et al. suggested that supplement of Faecalibacterium prausnitzii could improve intestinal integrity, reduce inflammatory responses from the body, and improve insulin sensitivity, which will help prevent sarcopenia.44 Though there have been some human and animal studies about the gut–muscle axis, the mechanism behind it is still unclear. An overview of the gut–muscle axis is shown in Fig. 3.

Figure 3.

Figure 3.

Overview of the gut–muscle axis.

This study has several strengths. First, it is the largest and most comprehensive MR study of the gut microbiota and sarcopenia-related traits. Second, the GWAS data used in this paper to identify IVs has a large sample size and a high level of accuracy. Third, the UK Biobank undergoes extensive genetic and phenotypic data analysis and is strictly supervised and operated to ensure high quality. Finally, several MR analysis methods were used in this study, which allowed us to estimate the potential causal relationship between the gut microbiota and the two sarcopenia-related variables without interference from residual confounding or reverse causality. However, the study still has certain limitations. First, because of the inherent nature of MR analysis, we cannot be sure that our findings were not influenced by weak IVs, even though all genetic tools were strongly associated with exposure (F value > 10). Second, most of the GWAS data for gut microbiota were from Europeans, whereas all the GWAS data for sarcopenia-related traits were from Europeans, which may contain some bias. Third, the 16S rRNA gene sequencing method only describes the gut microbiota from genus to phylum level. Future metagenomics and multi-omics approaches may be able to describe gut microbiota taxa at a finer level, avoiding species-related bias. In order to provide more theoretical support for the mechanism research of the 'gut–muscle' axis, we will expand the sample size in future studies to investigate the relationship between different gut microbiota taxa and sarcopenia-related traits at the gene level.

With a particular emphasis on the impacts of various gut microbiota taxa on muscle, our findings give researchers a platform to investigate the connection between gut microbiota and sarcopenia-related traits. Future studies should investigate the mechanism by which the gut microbiota and muscle are related, as well as novel therapeutic strategies involving the gut microbiota in the management of sarcopenia.

Supplementary Material

pbad010_Supplemental_Files

Acknowledgements

This study was supported by grants from Chinese National Science & Technology Pillar Program (Grant No. 2020YFC2005600), Sichuan Science and Technology Program (Grant No. 2021YFS0136), 1·3·5 project for disciplines of excellence–Clinical Research Incubation Project, West China Hospital, Sichuan University (Grant No. 19HXFH012), National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University (Grant No. Z20191003), and 1.3.5 project for disciplines of excellence, West China Hospital, Sichuan University (Grant No. ZYJC21005). The authors certify that they meet the ethical guidelines for authorship and publication of the clinical nutrition.

Contributor Information

Jiaxi Zhao, General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.

Rui Liang, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.

Quhong Song, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.

Shiyu Song, Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210008, Jiangsu, China.

Jirong Yue, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.

Chenkai Wu, Global Health Research Center Duke Kunshan University, Suzhou 215004, Jiangsu, China.

Author contributions

J.Z. and J.Y. contributed to the conception of the study; J.Z. and C.W. contributed significantly to analysis and manuscript preparation; J.Z. performed the data analyses and wrote the manuscript; R.L., Q.S., and Shiyu Song helped perform the analysis and contributed to constructive discussions.

Conflict of interest

The authors declare that they have no conflict of interest.

References

  • 1. Tamburini S, Shen N, Wu HCet al. The microbiome in early life: Implications for health outcomes. Nat Med. 2016;22:713–22. doi: 10.1038/nm.4142. [DOI] [PubMed] [Google Scholar]
  • 2. Tang WH, Kitai T, Hazen SL. Gut microbiota in cardiovascular health and disease. Circ Res. 2017;120:1183–96. doi: 10.1161/CIRCRESAHA.117.309715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Qin J, Li Y, Cai Zet al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60. doi: 10.1038/nature11450. [DOI] [PubMed] [Google Scholar]
  • 4. Zhuang Z, Yang R, Wang Wet al. Associations between gut microbiota and Alzheimer's disease, major depressive disorder, and schizophrenia. J Neuroinflammation. 2020;17:288. doi: 10.1186/s12974-020-01961-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Ni JJ, Xu Q, Yan SSet al. Gut microbiota and psychiatric disorders: A two-sample mendelian randomization study. Front Microbiol. 2022;12:737197. doi: 10.3389/fmicb.2021.737197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Cruz-Jentoft AJ, Bahat G, Bauer Jet al. Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing. 2019;48:16–31. doi: 10.1093/ageing/afy169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Sayer AA, Cruz-Jentoft A.. Sarcopenia definition, diagnosis and treatment: Consensus is growing. Age Ageing. 2022;51:afac220. doi: 10.1093/ageing/afac220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. De Buyser SL, Petrovic M, Taes YEet al. Validation of the FNIH sarcopenia criteria and SOF frailty index as predictors of long-term mortality in ambulatory older men. Age Ageing. 2016;45:602–8. doi: 10.1093/ageing/afw071. [DOI] [PubMed] [Google Scholar]
  • 9. Cawthon PM, Lui LY, Taylor BCet al. Clinical definitions of sarcopenia and risk of hospitalization in community-dwelling older men: The osteoporotic fractures in men study. J Gerontol A Biol Sci Med Sci. 2017;72:1383–9. doi: 10.1093/gerona/glw327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Antunes AC, Araujo DA, Verissimo MTet al. Sarcopenia and hospitalisation costs in older adults: A cross-sectional study. Nutr Diet. 2017;74:46–50. doi: 10.1111/1747-0080.12287. [DOI] [PubMed] [Google Scholar]
  • 11. de Sire R, Rizzatti G, Ingravalle Fet al. Skeletal muscle-gut axis: Emerging mechanisms of sarcopenia for intestinal and extra intestinal diseases. Minerva Gastroenterol Dietol. 2018;64:351–62. doi: 10.23736/S1121-421X.18.02511-4. [DOI] [PubMed] [Google Scholar]
  • 12. Ni LM, Bowyer R, Steves CJ. Dietary protein and muscle in aging people: The potential role of the gut microbiome. Nutrients. 2018;10:929. doi: 10.3390/nu10070929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ticinesi A, Nouvenne A, Cerundolo Net al. Gut microbiota, muscle mass and function in aging: A focus on physical frailty and sarcopenia. Nutrients. 2019;11:1633. doi: 10.3390/nu11071633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Zhao J, Huang Y, Yu X. A narrative review of gut-muscle axis and sarcopenia: The potential role of gut microbiota. Int J Gen Med. 2021;14:1263–73. doi: 10.2147/IJGM.S301141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Karlund A, Gomez-Gallego C, Turpeinen AMet al. Protein supplements and their relation with nutrition, microbiota composition and health: Is more Protein always better for sportspeople?. Nutrients. 2019;11:829. doi: 10.3390/nu11040829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Lin R, Liu W, Piao Met al. A review of the relationship between the gut microbiota and amino acid metabolism. Amino Acids. 2017;49:2083–90. doi: 10.1007/s00726-017-2493-3. [DOI] [PubMed] [Google Scholar]
  • 17. Dukes A, Davis C, El RMet al. The aromatic amino acid tryptophan stimulates skeletal muscle IGF1/p70s6k/mTor signaling in vivo and the expression of myogenic genes in vitro. Nutrition. 2015;31:1018–24. doi: 10.1016/j.nut.2015.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Picca A, Ponziani FR, Calvani Ret al. Gut microbial, inflammatory and metabolic signatures in older people with physical frailty and sarcopenia: Results from the BIOSPHERE study. Nutrients. 2019;12:65. doi: 10.3390/nu12010065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Ticinesi A, Lauretani F, Tana Cet al. Exercise and immune system as modulators of intestinal microbiome: Implications for the gut-muscle axis hypothesis. Exerc Immunol Rev. 2019;25: 84–95. [PubMed] [Google Scholar]
  • 20. Emdin CA, Khera AV, Kathiresan S. Mendelian randomization. JAMA. 2017;318:1925–6. [DOI] [PubMed] [Google Scholar]
  • 21. Liu B, Ye D, Yang Het al. Two-sample mendelian randomization analysis investigates causal associations between gut microbial genera and inflammatory bowel disease, and specificity causal associations in ulcerative colitis or Crohn's disease. Front Immunol. 2022;13:921546. doi: 10.3389/fimmu.2022.921546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Lv WQ, Lin X, Shen Het al. Human gut microbiome impacts skeletal muscle mass via gut microbial synthesis of the short-chain fatty acid butyrate among healthy menopausal women. J Cachexia Sarcopenia Muscle. 2021;12:1860–70. doi: 10.1002/jcsm.12788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Cruz-Jentoft AJ, Sayer AA. Sarcopenia. Lancet. 2019;393:2636–46. doi: 10.1016/S0140-6736(19)31138-9. [DOI] [PubMed] [Google Scholar]
  • 24. Jones G, Trajanoska K, Santanasto AJet al. Genome-wide meta-analysis of muscle weakness identifies 15 susceptibility loci in older men and women. Nat Commun. 2021;12:654. doi: 10.1038/s41467-021-20918-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Pei YF, Liu YZ, Yang XLet al. The genetic architecture of appendicular lean mass characterized by association analysis in the UK Biobank study. Commun Biol. 2020;3:608. doi: 10.1038/s42003-020-01334-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Palmer TM, Lawlor DA, Harbord RMet al. Using multiple genetic variants as instrumental variables for modifiable risk factors. Stat Methods Med Res. 2012;21:223–42. doi: 10.1177/0962280210394459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Bowden J, Davey SG, Haycock PCet al. Consistent estimation in mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40:304–14. doi: 10.1002/gepi.21965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Burgess S, Thompson SG. Interpreting findings from mendelian randomization using the MR-Egger method. Eur J Epidemiol. 2017;32:377–89. doi: 10.1007/s10654-017-0255-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Hemani G, Zheng J, Elsworth Bet al. The MR-Base platform supports systematic causal inference across the human phenome. Elife. 2018;7:e34408. doi: 10.7554/eLife.34408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Chen YJ, Wu H, Wu SDet al. Parasutterella, in association with irritable bowel syndrome and intestinal chronic inflammation. J Gastroenterol Hepatol. 2018;33:1844–52. doi: 10.1111/jgh.14281. [DOI] [PubMed] [Google Scholar]
  • 31. Li F, Wang M, Wang Jet al. Alterations to the gut microbiota and their correlation with inflammatory factors in chronic kidney disease. Front Cell Infect Microbiol. 2019;9: 206. doi: 10.3389/fcimb.2019.00206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Jager R, Purpura M, Stone JDet al. Probiotic Streptococcus thermophilus FP4 and bifidobacterium breve BR03 supplementation attenuates performance and range-of-motion decrements following muscle damaging exercise. Nutrients. 2016;8:642. doi: 10.3390/nu8100642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Ren X, Hao S, Yang Cet al. Alterations of intestinal microbiota in liver cirrhosis with muscle wasting. Nutrition. 2021;83: 111081. doi: 10.1016/j.nut.2020.111081. [DOI] [PubMed] [Google Scholar]
  • 34. Okamoto T, Morino K, Ugi Set al. Microbiome potentiates endurance exercise through intestinal acetate production. Am J Physiol Endocrinol Metab. 2019;316:E956–66. doi: 10.1152/ajpendo.00510.2018 [DOI] [PubMed] [Google Scholar]
  • 35. Scheiman J, Luber JM, Chavkin TAet al. Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat Med. 2019;25:1104–9. doi: 10.1038/s41591-019-0485-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Pei T, Zhu D, Yang Set al. Bacteroides plebeius improves muscle wasting in chronic kidney disease by modulating the gut-renal muscle axis. J Cell Mol Med. 2022;26:6066–78. doi: 10.1111/jcmm.17626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Fielding RA, Reeves AR, Jasuja Ret al. Muscle strength is increased in mice that are colonized with microbiota from high-functioning older adults. Exp Gerontol. 2019;127: 110722. doi: 10.1016/j.exger.2019.110722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Lee YA, Song SW, Jung SYet al. Sarcopenia in community-dwelling older adults is associated with the diversity and composition of the gut microbiota. Exp Gerontol. 2022;167: 111927. doi: 10.1016/j.exger.2022.111927. [DOI] [PubMed] [Google Scholar]
  • 39. Hu J, Zhong X, Liu Yet al. Correlation between intestinal flora disruption and protein-energy wasting in patients with end-stage renal disease. BMC Nephrol. 2022;23:130. doi: 10.1186/s12882-022-02762-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Kang L, Li P, Wang Det al. Alterations in intestinal microbiota diversity, composition, and function in patients with sarcopenia. Sci Rep. 2021;11:4628. doi: 10.1038/s41598-021-84031-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Tavella T, Rampelli S, Guidarelli Get al. Elevated gut microbiome abundance of christensenellaceae, porphyromonadaceae and Rikenellaceae is associated with reduced visceral adipose tissue and healthier metabolic profile in Italian elderly. Gut Microbes. 2021;13:1–19. doi: 10.1080/19490976.2021.1880221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Liu X, Yue J. Precision intervention for sarcopenia. Precis Clin Med. 2022;5:c13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Eloe-Fadrosh EA, Brady A, Crabtree Jet al. Functional dynamics of the gut microbiome in elderly people during probiotic consumption. mBio. 2015;6:e00231–15. doi: 10.1128/mBio.00231-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Munukka E, Rintala A, Toivonen Ret al. Faecalibacterium prausnitzii treatment improves hepatic health and reduces adipose tissue inflammation in high-fat fed mice. ISME J. 2017;11:1667–79. doi: 10.1038/ismej.2017.24. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

pbad010_Supplemental_Files

Articles from Precision Clinical Medicine are provided here courtesy of Oxford University Press

RESOURCES