Skip to main content

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

Metagenomic and metabolomic remodeling in nonagenarians and centenarians and its association with genetic and socioeconomic factors

An Author Correction to this article was published on 27 June 2022

This article has been updated

Abstract

A better understanding of the biological and environmental variables that contribute to exceptional longevity has the potential to inform the treatment of geriatric diseases and help achieve healthy aging. Here, we compared the gut microbiome and blood metabolome of extremely long-lived individuals (94–105 years old) to that of their children (50–79 years old) in 116 Han Chinese families. We found extensive metagenomic and metabolomic remodeling in advanced age and observed a generational divergence in the correlations with socioeconomic factors. An analysis of quantitative trait loci revealed that genetic associations with metagenomic and metabolomic features were largely generation-specific, but we also found 131 plasma metabolic quantitative trait loci associations that were cross-generational with the genetic variants concentrated in six loci. These included associations between FADS1/2 and arachidonate, PTPA and succinylcarnitine and FLVCR1 and choline. Our characterization of the extensive metagenomic and metabolomic remodeling that occurs in people reaching extreme ages may offer new targets for aging-related interventions.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Analysis of gut microbiota composition and structure.
Fig. 2: Analysis of gut microbial functions.
Fig. 3: Analysis of fecal metabolites.
Fig. 4: Analysis of plasma metabolites.
Fig. 5: Analysis of QTL of metagenomic and metabolomic phenotypes.
Fig. 6: Associations between socioeconomic, lifestyle and medical factors and metagenomic or metabolomic phenotypes.
Fig. 7: Significance of shared genotype–phenotype connections in two generations of longevous families.

Data availability

Data supporting the findings of this study are available in Supplementary Tables. The metagenomic sequencing and exon sequencing data have been deposited in the National Center for Biotechnology Information Sequence Read Archive database under accession numbers PRJNA613947 and PRJNA790003. The stool and plasma metabolomic raw data of the primary and validation Qidong cohorts are in Supplementary Tables 1012.

Code availability

All other analyses were performed using custom codes written in bash (v.4.2) or R (v.4.0.2). These codes are available in GitHub at https://github.com/RYWCY/Longevous.git.

Change history

References

  1. Chang, A. Y., Skirbekk, V. F., Tyrovolas, S., Kassebaum, N. J. & Dieleman, J. L. Measuring population ageing: an analysis of the Global Burden of Disease Study 2017. Lancet Public Health 4, e159–e167 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Passarino, G., De Rango, F. & Montesanto, A. Human longevity: genetics or lifestyle? It takes two to tango. Immun. Ageing 13, 12 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Govindaraju, D., Atzmon, G. & Barzilai, N. Genetics, lifestyle and longevity: lessons from centenarians. Appl. Transl. Genom. 4, 23–32 (2015).

    PubMed  PubMed Central  Google Scholar 

  4. Willcox, B. J. et al. FOXO3A genotype is strongly associated with human longevity. Proc. Natl Acad. Sci. USA 105, 13987–13992 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schachter, F. et al. Genetic associations with human longevity at the APOE and ACE loci. Nat. Genet. 6, 29–32 (1994).

    Article  CAS  PubMed  Google Scholar 

  6. Deelen, J. et al. A meta-analysis of genome-wide association studies identifies multiple longevity genes. Nat. Commun. 10, 3669 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Gonzalez-Covarrubias, V. et al. Lipidomics of familial longevity. Aging Cell 12, 426–434 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Kong, F. et al. Gut microbiota signatures of longevity. Curr. Biol. 26, R832–R833 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Biagi, E. et al. Gut microbiota and extreme longevity. Curr. Biol. 26, 1480–1485 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Kaplanis, J. et al. Quantitative analysis of population-scale family trees with millions of relatives. Science 360, 171–175 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Herskind, A. M. et al. The heritability of human longevity: a population-based study of 2872 Danish twin pairs born 1870–1900. Hum. Genet. 97, 319–323 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. Partridge, L., Deelen, J. & Slagboom, P. E. Facing up to the global challenges of ageing. Nature 561, 45–56 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. van den Berg, N. et al. Longevity defined as top 10% survivors and beyond is transmitted as a quantitative genetic trait. Nat. Commun. 10, 35 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  14. McClellan, J. & King, M. C. Genetic heterogeneity in human disease. Cell 141, 210–217 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. He, Y. et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 24, 1532–1535 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. v, B. H. J. et al. Genetic influence on human lifespan and longevity. Hum. Genet. 119, 312–321 (2006).

    Article  Google Scholar 

  17. Wu, L. et al. A cross-sectional study of compositional and functional profiles of gut microbiota in Sardinian centenarians. mSystems 4, e00325–00319 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Pasolli, E. et al. Accessible, curated metagenomic data through ExperimentHub. Nat. Methods 14, 1023–1024 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rampelli, S. et al. Shotgun metagenomics of gut microbiota in humans with up to extreme longevity and the increasing role of xenobiotic degradation. mSystems https://doi.org/10.1128/mSystems.00124-20 (2020).

  20. Xie, H. et al. Shotgun metagenomics of 250 adult twins reveals genetic and environmental impacts on the gut microbiome. Cell Syst. 3, 572–584 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gupta, V. K. et al. A predictive index for health status using species-level gut microbiome profiling. Nat. Commun. 11, 4635 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kanehisa, M., Sato, Y., Kawashima, M., Furumichi, M. & Tanabe, M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 44, D457–D462 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Morton, J. T. et al. Learning representations of microbe-metabolite interactions. Nat. Methods 16, 1306–1314 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. den Besten, G. et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 54, 2325–2340 (2013).

    Article  Google Scholar 

  25. Heinken, A. et al. Systematic assessment of secondary bile acid metabolism in gut microbes reveals distinct metabolic capabilities in inflammatory bowel disease. Microbiome 7, 75 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Connors, J. et al. The relationship between fecal bile acids and microbiome community structure in pediatric Crohn’s disease. ISME J. 14, 702–713 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Wang, K. et al. Parabacteroides distasonis alleviates obesity and metabolic dysfunctions via production of succinate and secondary bile acids. Cell Rep. 26, 222–235 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Loomba, R. et al. The commensal microbe Veillonella as a marker for response to an FGF19 analog in NASH. Hepatology 73, 126–143 (2021).

  30. Orentreich, N., Brind, J. L., Rizer, R. L. & Vogelman, J. H. Age changes and sex differences in serum dehydroepiandrosterone sulfate concentrations throughout adulthood. J. Clin. Endocrinol. Metab. 59, 551–555 (1984).

    Article  CAS  PubMed  Google Scholar 

  31. Pan, X., Wu, X., Kaminga, A. C., Wen, S. W. & Liu, A. Dehydroepiandrosterone and dehydroepiandrosterone sulfate in Alzheimer’s disease: a systematic review and meta-analysis. Front. Aging Neurosci. https://doi.org/10.3389/fnagi.2019.00061 (2019).

  32. Berr, C., Lafont, S., Debuire, B., Dartigues, J.-F. & Baulieu, E.-E. Relationships of dehydroepiandrosterone sulfate in the elderly with functional, psychological, and mental status, and short-term mortality: a French community-based study. PNAS https://doi.org/10.1073/pnas.93.23.13410 (1996).

  33. Koh, A. et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell 175, 947–961 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Chaleckis, R., Murakami, I., Takada, J., Kondoh, H. & Yanagida, M. Individual variability in human blood metabolites identifies age-related differences. Proc. Natl Acad. Sci. USA 113, 4252–4259 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Banerjee, S. et al. Loss of the PTCH1 tumor suppressor defines a new subset of plexiform fibromyxoma. J. Translat. Med. 17, 246 (2019).

    Article  Google Scholar 

  36. Parker, B. J., Wearsch, P. A., Veloo, A. C. M. & Rodriguez-Palacios, A. The genus Alistipes: gut bacteria with emerging implications to inflammation, cancer, and mental health. Front. Immunol. https://doi.org/10.3389/fimmu.2020.00906 (2020).

  37. Louis, P. & Flint, H. J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 294, 1–8 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Kurilshikov, A. et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat. Genet. 53, 156–165 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bonder, M. J. et al. The effect of host genetics on the gut microbiome. Nat. Genet. 48, 1407–1412 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Glaser, C., Heinrich, J. & Koletzko, B. Role of FADS1 and FADS2 polymorphisms in polyunsaturated fatty acid metabolism. Metab. Clin. Exp. 59, 993–999 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Chen, H. Y. et al. Association of FADS1/2 locus variants and polyunsaturated fatty acids with aortic stenosis. JAMA Cardiol. 5, 694–702 (2020).

    Article  PubMed  Google Scholar 

  42. Peters, M. J. et al. The transcriptional landscape of age in human peripheral blood. Nat. Commun. 6, 8570–8570 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Rothschild, D. et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210–215 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Morgan, X. C. et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 13, R79 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Teague, C. et al. Ethyl glucoside in human urine following dietary exposure: detection by 1H NMR spectroscopy as a result of metabonomic screening of humans. Analyst 129, 259–264 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Falony, G. et al. Population-level analysis of gut microbiome variation. Science 352, 560–564 (2016).

    Article  CAS  PubMed  Google Scholar 

  47. Bar, N. et al. A reference map of potential determinants for the human serum metabolome. Nature 588, 135–140 (2020).

    Article  PubMed  Google Scholar 

  48. Wang, X. et al. Genetic markers of type 2 diabetes: progress in genome-wide association studies and clinical application for risk prediction. J. Diabetes 8, 24–35 (2016).

    Article  PubMed  Google Scholar 

  49. Van Cauwenberghe, C., Van Broeckhoven, C. & Sleegers, K. The genetic landscape of Alzheimer disease: clinical implications and perspectives. Genet. Med. 18, 421–430 (2016).

    Article  PubMed  Google Scholar 

  50. Banin Hirata, B. K. et al. Molecular markers for breast cancer: prediction on tumor behavior. Dis. Markers 2014, 513158 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Uno, M. & Nishida, E. Lifespan-regulating genes in C. elegans. NPJ Aging Mech. Dis. 2, 16010 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Perls, T. T. et al. Life-long sustained mortality advantage of siblings of centenarians. Proc. Natl Acad. Sci. USA 99, 8442–8447 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kaminski, J. et al. High-specificity targeted functional profiling in microbial communities with ShortBRED. PLoS Comput. Biol. 11, e1004557 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Jia, B. et al. CARD 2017: expansion and model-centric curation of the comprehensive antibiotic resistance database. Nucleic Acids Res. 45, D566–D573 (2017).

    Article  CAS  PubMed  Google Scholar 

  55. Franzosa, E. A. et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 15, 962–968 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Stanish, W. M. & Taylor, N. Estimation of the intraclass correlation coefficient for the analysis of covariance model. Am. Stat. 37, 221–224 (1983).

    Google Scholar 

Download references

Acknowledgements

This work was supported by funding from the National Natural Science Foundation of China grant nos. 81730102 (H.L.Q.), 81230057 (H.L.Q.) and 31970111 (N.Q.). We thank B. Zhou, P. He, Z. Cai, T. Liu, Y. Li and Y. Luan at Realbio Genomics Institute for sequencing and data analysis, Q. Yang at Shanghai Tenth Hospital for sample collections and J. Zhang, Y. Fang, J. Zhou and M. Liang at Realbio Genomics Institute for graphic assistance. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

H.L.Q. conceptualized and led the project. H.L.Q., Q. X. and N.Q. designed the study. C.Y.W. led data analyses. Q.X., C.Y.W. and Q.Z. wrote the manuscript. M.V.C. and N.S. critiqued the original manuscript and provided constructive suggestions. S.D.E. contributed to study design. Q.Z., J.Q.L., J.Z., F.Y. and J.F. organized a collection of stool and blood samples, questionnaire survey and documentation of metadata. L.S.H., L.L.D., X.H.Z., Y.H.Z., Y.F.Z., L.L., Y.Q.L., Y.S.C., Y.S., J.W., Z.C. and C.S.F. assisted sample collection, questionnaire survey, documentation of clinical measures and compilation of data. R.Y.G., F.Y. and L.S.H. organized metabolomic analyses. X.X., M.M.B. and X.C. performed data compilation and analyses. All authors read the manuscript and offered valuable suggestions.

Corresponding authors

Correspondence to Nan Qin or Huanlong Qin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Dario Riccardo Valenzano and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Differential gut microbiota species between the advanced age group from Qidong longevous cohort and unrelated CMD healthy controls age-matched with the offspring.

The unrelated healthy controls were 589 healthy controls at ages of 50–75 years (matched with the offspring group) from 26 metagenomic projects deposited in CuratedMetagenomicData (CMD). The boxes represent the interquartile range (IQR), from the first and third quartiles and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers.

Source data

Extended Data Fig. 2 Prevalence distributions of all species of Longevous Gut Microbiota Signature (LGMS) in several advanced age groups and non-advanced age groups.

The advanced age groups included that from Qidong, China and Sardinia, Italy, whereas the non-advanced age groups included the offspring group of Qidong, China, non-advanced age group of Sardinia, Italy (< 80 YO), and 589 CMD healthy controls at ages of 50–75 years.

Source data

Extended Data Fig. 3 Analysis of gut microbiome functions.

a, Comparison of types and RPKM (reads per kilobase per million reads) values of antibiotic resistance genes. b, Differential antibiotic resistance genes between the advanced age (N=116) and offspring (N=232) groups (Wilcoxon rank-sum test, FDR < 0.05). Data are presented as mean values ± SD, indicated by error bars). c and d, The 10 most abundant antibiotic resistance targets (c) and types (d) across advanced age (orange) and offspring (blue) samples. e and f, Highly differential KOs between the two generations (Wilcoxon rank-sum test, FDR < 5 × 10-5) that have (e) or have no (f) Metacyc pathway annotations. Top genus-level contributors to differential KOs in 6 major Metacyc pathways are shown. g, Age-stable KOs between the two generations (Wilcoxon rank-sum test, FDR > 0.1 and ICC < 0). The boxes (a, c and d) represent the interquartile range (IQR), from the first and third quartiles, and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers.

Source data

Extended Data Fig. 4 The most differential KOs between the advanced age (N=116) and offspring (N=232) groups (FDR < 5 × 10-5 and fold change > 2) and their taxonomic contributors.

Fold change is indicated at the top of each scatter plot. The boxes represent the interquartile range (IQR), from the first and third quartiles, and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers.

Source data

Extended Data Fig. 5 Sex-adjusted analysis of androgenic steroids.

FDR and fold change values of each metabolite are marked at its left. The boxes represent the interquartile range (IQR), from the first and third quartiles, and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers.

Source data

Extended Data Fig. 6 Analyses of plasma metabolome.

a, Spearman association analysis between MetaCyc pathways of gut microbiome and plasma metabolites. Rows show MetaCyc pathways and columns show plasma metabolites. Asterisks denote spearman correlation test results (*FDR < 5 × 10-4, **FDR < 1 × 10-4). b, Associations between plasma levels of dehydroepiandrosterone sulfate (DHEA-S; its decline in elderly people is linked to Alzheimer’s disease) or biochemically related epiandrosterone sulfate and microbiome pathways. c, Analysis of two inflammatory cytokines in the Qidong validation cohort (33 pairs of nonagenarians (N=33) and their offspring (N=33)). The boxes represent the interquartile range (IQR), from the first and third quartiles, and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers.

Source data

Extended Data Fig. 7 Analysis of quantitative trait loci (QTL) of plasma metabolites.

Genomic regions accommodating a cluster of associations with a circulating metabolite that are shared by the two generations (a) or found only in the offspring (b). The associations between 3-aminoisobutarate and 5p13.2 were largely offspring-specific, although there were 3 pairs of cross-generational QTLs. FDR value and fold change of the relevant metabolite are marked at the upper left of each box plot (top row or single row, primary Qidong cohort; bottom row; validation Qidong cohort).

Source data

Extended Data Fig. 8 Associations between socioeconomic, lifestyle or medical factors and metagenomic or metabolomic phenotypes.

a, Heatmaps of MaAsLin-based associations between socioeconomic, lifestyle or medical factors and gut microbes, gut Metacyc pathways, plasma metabolites or stool metabolites. Significant associations were marked by + (FDR < 0.1) and * (FDR < 0.05). b, Examples of associations between a metagenomic/metabolic features and a demographic/socioeconomic factors.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, Q., Wu, C., Zhu, Q. et al. Metagenomic and metabolomic remodeling in nonagenarians and centenarians and its association with genetic and socioeconomic factors. Nat Aging 2, 438–452 (2022). https://doi.org/10.1038/s43587-022-00193-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-022-00193-0

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research