Advertisement

Mighty Male Microbes

Both genetic and environmental factors contribute to an individual's susceptibility to autoimmune disease, but the specific environmental influences are not well characterized. Markle et al. (p. 1084, published online 17 January; see the Perspective by Flak et al.) explored how microbial factors, in particular the gut microbiota, influence susceptibility to type 1 diabetes in mice. In the non-obese diabetic (NOD) mouse model of type 1 diabetes, female mice are significantly more susceptible to disease than males; however, this difference was not apparent under germ-free conditions. Transfer of cecal contents from male NOD mice to female NOD mice prior to disease onset protected against pancreatic islet inflammation, autoantibody production, and the development of diabetes and was associated with increased testosterone in female mice. Blocking androgen receptor activity abrogated protection. Thus, the microbiota may be able to regulate sex hormones and influence an individual's susceptibility to autoimmunity.

Abstract

Microbial exposures and sex hormones exert potent effects on autoimmune diseases, many of which are more prevalent in women. We demonstrate that early-life microbial exposures determine sex hormone levels and modify progression to autoimmunity in the nonobese diabetic (NOD) mouse model of type 1 diabetes (T1D). Colonization by commensal microbes elevated serum testosterone and protected NOD males from T1D. Transfer of gut microbiota from adult males to immature females altered the recipient's microbiota, resulting in elevated testosterone and metabolomic changes, reduced islet inflammation and autoantibody production, and robust T1D protection. These effects were dependent on androgen receptor activity. Thus, the commensal microbial community alters sex hormone levels and regulates autoimmune disease fate in individuals with high genetic risk.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Materials and Methods
Figs. S1 to S4
Tables S1 to S6
References (3658)

Resources

File (markle.sm.pdf)
File (markle_1233521_tables2.xls)
File (markle_1233521_tables3.xls)
File (markle_1233521_tables4.xls)

References and Notes

1
Scobie H. M., et al., Anthrax toxin receptor 2-dependent lethal toxin killing in vivo. PLoS Pathog. 2, e111 (2006).
2
Whitacre C. C., Sex differences in autoimmune disease. Nat. Immunol. 2, 777 (2001).
3
Ober C., Loisel D. A., Gilad Y., Sex-specific genetic architecture of human disease. Nat. Rev. Genet. 9, 911 (2008).
4
Bach J. F., The effect of infections on susceptibility to autoimmune and allergic diseases. N. Engl. J. Med. 347, 911 (2002).
5
Makino S., et al., Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu 29, 1 (1980).
6
E. Leiter, M. Atkinson, NOD Mice and Related Strains: Research Applications in Diabetes, AIDS, Cancer and Other Diseases (Landes, Austin, TX, 1998), vol. 2.
7
Serreze D. V., Leiter E. H., Genes and cellular requirements for autoimmune diabetes susceptibility in nonobese diabetic mice. Curr. Dir. Autoimmun. 4, 31 (2001).
8
Miao D., Yu L., Eisenbarth G. S., Role of autoantibodies in type 1 diabetes. Front. Biosci. 12, 1889 (2007).
9
Anderson M. S., Bluestone J. A., The NOD mouse: A model of immune dysregulation. Annu. Rev. Immunol. 23, 447 (2005).
10
Ghosh S., et al., Polygenic control of autoimmune diabetes in nonobese diabetic mice. Nat. Genet. 4, 404 (1993).
11
Pozzilli P., Signore A., Williams A. J., Beales P. E., NOD mouse colonies around the world—recent facts and figures. Immunol. Today 14, 193 (1993).
12
Makino S., Kunimoto K., Muraoka Y., Katagiri K., Effect of castration on the appearance of diabetes in NOD mouse. Jikken Dobutsu 30, 137 (1981).
13
Fox H. S., Androgen treatment prevents diabetes in nonobese diabetic mice. J. Exp. Med. 175, 1409 (1992).
14
Harada M., Kishimoto Y., Makino S., Prevention of overt diabetes and insulitis in NOD mice by a single BCG vaccination. Diabetes Res. Clin. Pract. 8, 85 (1990).
15
Qin H. Y., Sadelain M. W., Hitchon C., Lauzon J., Singh B., Complete Freund's adjuvant-induced T cells prevent the development and adoptive transfer of diabetes in nonobese diabetic mice. J. Immunol. 150, 2072 (1993).
16
Ivakine E. A., et al., The idd4 locus displays sex-specific epistatic effects on type 1 diabetes susceptibility in nonobese diabetic mice. Diabetes 55, 3611 (2006).
17
Ivakine E. A., et al., Sex-specific effect of insulin-dependent diabetes 4 on regulation of diabetes pathogenesis in the nonobese diabetic mouse. J. Immunol. 174, 7129 (2005).
18
See supplementary materials on Science Online.
19
Nicholson J. K., et al., Host-gut microbiota metabolic interactions. Science 336, 1262 (2012).
20
Koal T., Deigner H. P., Challenges in mass spectrometry based targeted metabolomics. Curr. Mol. Med. 10, 216 (2010).
21
Oberbach A., et al., Combined proteomic and metabolomic profiling of serum reveals association of the complement system with obesity and identifies novel markers of body fat mass changes. J. Proteome Res. 10, 4769 (2011).
22
Sadelain M. W., Qin H. Y., Lauzon J., Singh B., Prevention of type I diabetes in NOD mice by adjuvant immunotherapy. Diabetes 39, 583 (1990).
23
Slack E., et al., Innate and adaptive immunity cooperate flexibly to maintain host-microbiota mutualism. Science 325, 617 (2009).
24
Mittelstrass K., et al., Discovery of sexual dimorphisms in metabolic and genetic biomarkers. PLoS Genet. 7, e1002215 (2011).
25
Chai J. G., James E., Dewchand H., Simpson E., Scott D., Transplantation tolerance induced by intranasal administration of HY peptides. Blood 103, 3951 (2004).
26
Koren O., et al., Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 150, 470 (2012).
27
Yatsunenko T., et al., Human gut microbiome viewed across age and geography. Nature 486, 222 (2012).
28
Doran M. F., Pond G. R., Crowson C. S., O'Fallon W. M., Gabriel S. E., Trends in incidence and mortality in rheumatoid arthritis in Rochester, Minnesota, over a forty-year period. Arthritis Rheum. 46, 625 (2002).
29
Weinshenker B. G., Natural history of multiple sclerosis. Ann. Neurol. 36 (suppl.), S6 (1994).
30
Burton P. R., et al.Wellcome Trust Case Control Consortium, Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661 (2007).
31
Patterson C. C., Dahlquist G. G., Gyürüs E., Green A., Soltész G.EURODIAB Study Group, Incidence trends for childhood type 1 diabetes in Europe during 1989-2003 and predicted new cases 2005-20: A multicentre prospective registration study. Lancet 373, 2027 (2009).
32
Brown C. T., et al., Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS ONE 6, e25792 (2011).
33
Frank D. N., Zhu W., Sartor R. B., Li E., Investigating the biological and clinical significance of human dysbioses. Trends Microbiol. 19, 427 (2011).
34
Berer K., et al., Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538 (2011).
35
Wagner B. D., Robertson C. E., Harris J. K., Application of two-part statistics for comparison of sequence variant counts. PLoS ONE 6, e20296 (2011).
36
Smith K., McCoy K. D., Macpherson A. J., Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota. Semin. Immunol. 19, 59 (2007).
37
Dewhirst F. E., et al., Phylogeny of the defined murine microbiota: Altered Schaedler flora. Appl. Environ. Microbiol. 65, 3287 (1999).
38
Frank D. N., BARCRAWL and BARTAB: Software tools for the design and implementation of barcoded primers for highly multiplexed DNA sequencing. BMC Bioinformatics 10, 362 (2009).
39
Frank J. A., et al., Critical evaluation of two primers commonly used for amplification of bacterial 16S rRNA genes. Appl. Environ. Microbiol. 74, 2461 (2008).
40
D. J. Lane, in Nucleic Acid Techniques in Bacterial Systematics, E. Stackebrandt, M. Goodfellow, Eds. (Wiley, New York, 1991), pp. 115–175.
41
Caporaso J. G., et al., Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc. Natl. Acad. Sci. U.S.A. 108 (suppl. 1), 4516 (2011).
42
Frank D. N., et al., The human nasal microbiota and Staphylococcus aureus carriage. PLoS ONE 5, e10598 (2010).
43
Nawrocki E. P., Kolbe D. L., Eddy S. R., Infernal 1.0: Inference of RNA alignments. Bioinformatics 25, 1335 (2009).
44
Haas B. J., et al.Human Microbiome Consortium, Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res. 21, 494 (2011).
45
Li E., et al., Inflammatory bowel diseases phenotype, C. difficile and NOD2 genotype are associated with shifts in human ileum associated microbial composition. PLoS ONE 7, e26284 (2012).
46
Ludwig W., et al., ARB: A software environment for sequence data. Nucleic Acids Res. 32, 1363 (2004).
47
Pruesse E., Peplies J., Glöckner F. O., SINA: Accurate high-throughput multiple sequence alignment of ribosomal RNA genes. Bioinformatics 28, 1823 (2012).
48
Pruesse E., et al., SILVA: A comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res. 35, 7188 (2007).
49
A. E. Magurran, Measuring Biological Diversity (Blackwell, Malden, MA, 2004).
50
Ewing B., Green P., Base-calling of automated sequencer traces using phred. II. Error probabilities. Genome Res. 8, 186 (1998).
51
Ewing B., Hillier L., Wendl M. C., Green P., Base-calling of automated sequencer traces using phred. I. Accuracy assessment. Genome Res. 8, 175 (1998).
52
Gordon D., Abajian C., Green P., Consed: A graphical tool for sequence finishing. Genome Res. 8, 195 (1998).
53
Edgar R. C., Haas B. J., Clemente J. C., Quince C., Knight R., UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27, 2194 (2011).
54
Schloss P. D., Gevers D., Westcott S. L., Reducing the effects of PCR amplification and sequencing artifacts on 16S rRNA-based studies. PLoS ONE 6, e27310 (2011).
55
Gaboriau-Routhiau V., et al., The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31, 677 (2009).
56
Atarashi K., et al., Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337 (2011).
57
Fox C. J., Paterson A. D., Mortin-Toth S. M., Danska J. S., Two genetic loci regulate T cell-dependent islet inflammation and drive autoimmune diabetes pathogenesis. Am. J. Hum. Genet. 67, 67 (2000).
58
Yu L., et al., Early expression of antiinsulin autoantibodies of humans and the NOD mouse: Evidence for early determination of subsequent diabetes. Proc. Natl. Acad. Sci. U.S.A. 97, 1701 (2000).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 339 | Issue 6123
1 March 2013

Article versions

You are viewing the most recent version of this article.

Submission history

Received: 3 December 2012
Accepted: 9 January 2013
Published in print: 1 March 2013

Permissions

Request permissions for this article.

Acknowledgments

We thank S. Bashir for assistance with metabolomic data analysis, C. Vogel Kotter for high-throughput sequencing, and E. Simpson and M. Palmert for helpful discussions. The 16S sequence data have been submitted to the National Center for Biotechnology Information short-read archive project, accession no. SRA064044. Flow cytometry was performed in the SickKids–University Health Network Flow Cytometry Facility with funds from the Ontario Institute for Cancer Research, McEwen Centre for Regenerative Medicine, Canada Foundation for Innovation, and SickKids Foundation. Roche 454 sequencing was performed at the Centre for Applied Genomics, Hospital for Sick Children. Illumina sequencing was performed at the University of Colorado School of Medicine. Supported by Canadian Institutes of Health Research (CIHR) grant 64216 and Juvenile Diabetes Research Foundation (JDRF) grant 17-2011-520 (J.S.D.), Genome Canada (administered by Ontario Genomics Institute) (J.S.D. and A.J.M.), JDRF grant 36-2008-926 and the Genaxen Foundation (A.J.M.), a CIHR Banting and Best fellowship (J.G.M.M.), and NIH grant R21HG005964 (D.N.F. and C.E.R.). J.G.M.M. and J.S.D. designed the study, analyzed data, and wrote the manuscript; J.G.M.M., D.N.F., S.M.-T., C.E.R., M.v.B., K.D.M., and A.J.M. performed experiments and analyzed data; L.M.F. and U.R.-K. performed experiments; and all authors contributed to manuscript editing.

Authors

Affiliations

Janet G. M. Markle
Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada.
Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
Daniel N. Frank
Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO 80045, USA.
Steven Mortin-Toth
Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada.
Charles E. Robertson
Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA.
Leah M. Feazel
Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO 80045, USA.
Ulrike Rolle-Kampczyk
Department of Metabolomics, Helmholtz Center for Environmental Research, 04318 Leipzig, Germany.
Martin von Bergen
Department of Metabolomics, Helmholtz Center for Environmental Research, 04318 Leipzig, Germany.
Department of Proteomics, Helmholtz Center for Environmental Research, 04318 Leipzig, Germany.
Department of Biotechnology, Chemistry and Environmental Engineering, Aalborg University, 9000 Aalborg, Denmark.
Kathy D. McCoy
Maurice Müller Laboratories, Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM), University of Bern, 3008 Bern, Switzerland.
Andrew J. Macpherson
Maurice Müller Laboratories, Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM), University of Bern, 3008 Bern, Switzerland.
Jayne S. Danska* [email protected]
Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada.
Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada.

Notes

*
To whom correspondence should be addressed. E-mail: [email protected]

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Sex differences in tissue immunity, Science, 384, 6692, (159-160), (2024)./doi/10.1126/science.ado8542
    Abstract
  2. Sexual dimorphism in skin immunity is mediated by an androgen-ILC2-dendritic cell axis, Science, 384, 6692, (2024)./doi/10.1126/science.adk6200
    Abstract
  3. Emerging Relationship between the Gut Microbiome and Prostate Cancer, The World Journal of Men's Health, 41, (2023).https://doi.org/10.5534/wjmh.220202
    Crossref
  4. Association between the Inflammatory Potential of the Diet and Biological Aging: A Cross-Sectional Analysis of 4510 Adults from the Moli-Sani Study Cohort, Nutrients, 15, 6, (1503), (2023).https://doi.org/10.3390/nu15061503
    Crossref
  5. Sex-Related Effects of Gut Microbiota in Metabolic Syndrome-Related Diabetic Retinopathy, Microorganisms, 11, 2, (447), (2023).https://doi.org/10.3390/microorganisms11020447
    Crossref
  6. Characterization of Non-Obstructive Azoospermia in Men Using Gut Microbial Profiling, Journal of Clinical Medicine, 12, 2, (701), (2023).https://doi.org/10.3390/jcm12020701
    Crossref
  7. Analysis of 16S rRNA Gene Sequence of Nasopharyngeal Exudate Reveals Changes in Key Microbial Communities Associated with Aging, International Journal of Molecular Sciences, 24, 4, (4127), (2023).https://doi.org/10.3390/ijms24044127
    Crossref
  8. Microbiome and Prostate Cancer: A Novel Target for Prevention and Treatment, International Journal of Molecular Sciences, 24, 2, (1511), (2023).https://doi.org/10.3390/ijms24021511
    Crossref
  9. Alterations in Fecal Microbiota Linked to Environment and Sex in Red Deer (Cervus elaphus), Animals, 13, 5, (929), (2023).https://doi.org/10.3390/ani13050929
    Crossref
  10. Moringa oleifera leaf ethanolic extract benefits cashmere goat semen quality via improving rumen microbiota and metabolome, Frontiers in Veterinary Science, 10, (2023).https://doi.org/10.3389/fvets.2023.1049093
    Crossref
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

Purchase this issue in print

Buy a single issue of Science for just $15 USD.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media