Intestinal microbiology shapes population health impacts of diet and lifestyle risk exposures in Torres Strait Islander communities

Fredrick M Mobegi, Lex Ex Leong, Fintan Thompson, Sean M Taylor, Linton R Harriss, Jocelyn M Choo, Steven L Taylor, Steve L Wesselingh, Robyn McDermott, Kerry L Ivey, Geraint B Rogers, Fredrick M Mobegi, Lex Ex Leong, Fintan Thompson, Sean M Taylor, Linton R Harriss, Jocelyn M Choo, Steven L Taylor, Steve L Wesselingh, Robyn McDermott, Kerry L Ivey, Geraint B Rogers

Abstract

Poor diet and lifestyle exposures are implicated in substantial global increases in non-communicable disease burden in low-income, remote, and Indigenous communities. This observational study investigated the contribution of the fecal microbiome to influence host physiology in two Indigenous communities in the Torres Strait Islands: Mer, a remote island where a traditional diet predominates, and Waiben a more accessible island with greater access to takeaway food and alcohol. Counterintuitively, disease markers were more pronounced in Mer residents. However, island-specific differences in disease risk were explained, in part, by microbiome traits. The absence of Alistipes onderdonkii, for example, significantly (p=0.014) moderated island-specific patterns of systolic blood pressure in multivariate-adjusted models. We also report mediatory relationships between traits of the fecal metagenome, disease markers, and risk exposures. Understanding how intestinal microbiome traits influence response to disease risk exposures is critical for the development of strategies that mitigate the growing burden of cardiometabolic disease in these communities.

Trial registration: ClinicalTrials.gov NCT02909218 NCT03789448.

Keywords: cardiometabolic disease; chronic inflammatory disease; epidemiology; global health; gut microbiome; human; indigenous health; infectious disease; metagenomics; microbiology; risk exposures.

Conflict of interest statement

FM, LL, FT, ST, LH, JC, ST, SW, RM, KI, GR No competing interests declared

© 2020, Mobegi et al.

Figures

Figure 1.. Island-specific differences in the species…
Figure 1.. Island-specific differences in the species composition of the microbiome.
(a) Non-metric Multidimensional Scaling of the Bray Curtis similarity resemblance matrix, with ellipses at 95% confidence interval. n = 100 (b) Fold differences in LDA mean proportions of differentially abundant species in Waiben and Mer. Bars were sorted based on the sequential ranking of significance and separated by the two populations. Only taxa significant at a logarithmic LDA score ≥3 and a factorial Kruskal-Wallis test Alpha (α)≤0.05 are shown. n = 100 (c) A phylogenetic tree based on the NCBI taxonomy of differentially abundant species identified using LEfSe. All species designated ‘unclassified’ were not used in generating the tree. Branches/edges are colored according to phylum rank classification and terminal nodes/species labels are colored based on study site overrepresentation. n = 100.
Figure 1—figure supplement 1.. Taxonomic profile of…
Figure 1—figure supplement 1.. Taxonomic profile of the 100 Torres Strait Islander participants included in the study.
The size of the nodes represents the logarithmically scaled relative abundances of taxa. Only clades consisting of at least five markers, with nodes constituting at least 1% relative abundance, are highlighted. Annotations begin from the phylum level.
Figure 2.. Functional pathways of the gut…
Figure 2.. Functional pathways of the gut microbiota that significantly differed in abundance between Mer and Waiben communities.
(a) Pathway abundances at the whole community level were statistically compared between the groups, and the 95% confidence interval of the effect-size for each pathway was determined using a non-parametric bootstrap method. (b) The heatmap represents community pathway abundances stratified into significant contributions from bacterial species to between-group differences. The color scale depicts the magnitude of the difference of stratified pathway abundances between the groups. All statistical comparisons were performed using the Mann-Whitney test and corrected for multiple testing using the false discovery rate method. Statistical significance for all comparisons was determined at p<0.05. Heatmap boxes with an ‘-’ indicated non-significant stratified pathway abundance differences between the groups.
Figure 2—figure supplement 1.. Heatmap representation of…
Figure 2—figure supplement 1.. Heatmap representation of stratified contribution to whole-community pathway abundances.
Bacterial species that significantly contributed to the stratified pathway abundances between the groups are depicted (Mann-Whitney test with false discovery rate correction for multiple testing, p

Figure 3.. Structural equation model describing the…

Figure 3.. Structural equation model describing the pattern of interrelationships between exposures, microbiome, and host…

Figure 3.. Structural equation model describing the pattern of interrelationships between exposures, microbiome, and host inflammation.
A modified path diagram of the final SEM supporting the mediatory effect of the gut microbiota on inflammation. The heatmaps represent direct associations of diet, behavior, and demographics with the gut microbiota and inflammatory biomarkers. Only significant paths of the effect of the gut microbiota on inflammation are shown. Standardized β coefficients are reported. Species Factors 1–6 denote latent variables of the community gut microbiota modeled as exploratory factor analysis (EFA) regression scores of the species relative abundance; Biomarker Factors 1–3 denote latent variables representing community inflammatory biomarkers as grouped using EFA. *p 0.1–0.05, **p 0.05–0.001, ***p

Figure 3—figure supplement 1.. Theoretical models for…

Figure 3—figure supplement 1.. Theoretical models for assessing the structure of associations between the various…

Figure 3—figure supplement 1.. Theoretical models for assessing the structure of associations between the various human exposures, the gut microbiota, and the host’s inflammatory profile.
(a) Final framework (framework 1): The gut microbiome mediates, in part, the relationship between exposure risk factors and host inflammation. (b) Framework 2: The gut microbiome and exposure risk factors both, independently, impact host inflammation. (c) Framework 3: Exposure risk factors are associated with gut microbiome and inflammatory profile, but there is no relationship between microbiome and inflammation. (d) Framework 4: Exposure risk factors are associated with the inflammatory profile, and the inflammatory profile predicts the gut microbiome composition.

Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the…

Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the…

Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15 and mean arterial pressure.
(a) Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15. The total effect (95% confidence interval) for the model was 0.054 (0.023, 0.090), P-value: <0.001. n = 100 (b) Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with mean arterial pressure. The total effect (95% confidence interval) for the model was −0.006 (–1.530, 1.420), P-value: 0.950. n = 100 a ACME (average causal mediation effects); b ADE (average direct effects).

Figure 4—figure supplement 1.. Role of Lachnospiraceae…

Figure 4—figure supplement 1.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation…

Figure 4—figure supplement 1.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15.
(a) Age-adjusted model. The total effect (95% confidence interval) for the model was 0.049 (0.015, 0.086) P-value: 0.004. (b) Age- and island-adjusted model. The total effect (95% confidence interval) for the model was 0.046 (0.008, 0.085) P-value: 0.016. (c) Multivariate-adjusted model. Includes adjustment for age, island, body mass index, gender, cigarette use, and intakes of fruits, vegetables, takeaways, seafood, and alcohol. The total effect (95% confidence interval) for the model was 0.045 (–0.0003, 0.088) P-value: 0.084.

Figure 4—figure supplement 2.. Role of Lachnospiraceae…

Figure 4—figure supplement 2.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation…

Figure 4—figure supplement 2.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation of sugar-sweetened beverage intake with the mean arterial pressure.
(a) Age-adjusted model. The total effect (95% confidence interval) for the model was 0.842 (−0.822, 2.520) P-value: 0.300. (b) Age-and island-adjusted model. The total effect (95% confidence interval) for the model was 0.948 (−0.694, 2.670) P-value: 0.246. (c) Multivariate-adjusted model. Includes adjustment for age, island, body mass index, gender, cigarette use, and intakes of fruits, vegetables, takeaways, seafood, and alcohol. The total effect (95% confidence interval) for the model was 0.295 (−1.730, 2.170) P-value: 0.808.

Figure 4—figure supplement 3.. 16S rRNA-based phylogeny…

Figure 4—figure supplement 3.. 16S rRNA-based phylogeny of Lachnospiraceae.

Lachnospiraceae family-restricted blast search (blast parameters…

Figure 4—figure supplement 3.. 16S rRNA-based phylogeny of Lachnospiraceae.
Lachnospiraceae family-restricted blast search (blast parameters e-value = 1 e-10, Max targets = 250) using Lachnospiraceae bacterium 8_1_57FAA 16 rRNA as a query against the NCBI non-redundant genes database was performed. The resulting top 250 best blast hits were used to construct a Grishin distance tree using the fast-minimum evolution model in NCBI BLAST. Terminal node labels are Taxonomic Name (Sequence ID).

Figure 5.. Systolic blood pressure in the…

Figure 5.. Systolic blood pressure in the context of an interaction between Alistipes onderdonkii and…

Figure 5.. Systolic blood pressure in the context of an interaction between Alistipes onderdonkii and island systolic blood pressure.
(a) Unadjusted model, (b) age-adjusted model, and (c) multivariate-adjusted model which includes the adjustment for age, body mass index, gender, smoking status, and intakes of fruits, vegetables, takeaways, sugar-sweetened beverages, seafood, and alcohol. Only significant P values (p<0.05) are shown n = 100.
All figures (11)
Similar articles
Cited by
References
    1. Anderson I, Robson B, Connolly M, Al-Yaman F, Bjertness E, King A, Tynan M, Madden R, Bang A, Coimbra CEA, Pesantes MA, Amigo H, Andronov S, Armien B, Obando DA, Axelsson P, Bhatti ZS, Bhutta ZA, Bjerregaard P, Bjertness MB, Briceno-Leon R, Broderstad AR, Bustos P, Chongsuvivatwong V, Chu J, Deji, Gouda J, Harikumar R, Htay TT, Htet AS, Izugbara C, Kamaka M, King M, Kodavanti MR, Lara M, Laxmaiah A, Lema C, Taborda AML, Liabsuetrakul T, Lobanov A, Melhus M, Meshram I, Miranda JJ, Mu TT, Nagalla B, Nimmathota A, Popov AI, Poveda AMP, Ram F, Reich H, Santos RV, Sein AA, Shekhar C, Sherpa LY, Skold P, Tano S, Tanywe A, Ugwu C, Ugwu F, Vapattanawong P, Wan X, Welch JR, Yang G, Yang Z, Yap L. Indigenous and tribal peoples' health (The Lancet–Lowitja Institute Global Collaboration): a population study. The Lancet. 2016;388:131–157. doi: 10.1016/S0140-6736(16)00345-7. - DOI - PubMed
    1. Arnold LW, Hoy WE, Sharma SK, Wang Z. The association between HbA1c and cardiovascular disease markers in a remote indigenous australian community with and without diagnosed Diabetes. Journal of Diabetes Research. 2016;2016:1–8. doi: 10.1155/2016/5342304. - DOI - PMC - PubMed
    1. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, Bertalan M, Borruel N, Casellas F, Fernandez L, Gautier L, Hansen T, Hattori M, Hayashi T, Kleerebezem M, Kurokawa K, Leclerc M, Levenez F, Manichanh C, Nielsen HB, Nielsen T, Pons N, Poulain J, Qin J, Sicheritz-Ponten T, Tims S, Torrents D, Ugarte E, Zoetendal EG, Wang J, Guarner F, Pedersen O, de Vos WM, Brunak S, Doré J, Antolín M, Artiguenave F, Blottiere HM, Almeida M, Brechot C, Cara C, Chervaux C, Cultrone A, Delorme C, Denariaz G, Dervyn R, Foerstner KU, Friss C, van de Guchte M, Guedon E, Haimet F, Huber W, van Hylckama-Vlieg J, Jamet A, Juste C, Kaci G, Knol J, Lakhdari O, Layec S, Le Roux K, Maguin E, Mérieux A, Melo Minardi R, M'rini C, Muller J, Oozeer R, Parkhill J, Renault P, Rescigno M, Sanchez N, Sunagawa S, Torrejon A, Turner K, Vandemeulebrouck G, Varela E, Winogradsky Y, Zeller G, Weissenbach J, Ehrlich SD, Bork P, MetaHIT Consortium Enterotypes of the human gut microbiome. Nature. 2011;473:174–180. doi: 10.1038/nature09944. - DOI - PMC - PubMed
    1. Asnicar F, Weingart G, Tickle TL, Huttenhower C, Segata N. Compact graphical representation of phylogenetic data and metadata with GraPhlAn. PeerJ. 2015;3:e1029. doi: 10.7717/peerj.1029. - DOI - PMC - PubMed
    1. Australian Bureau of Statistics National health survey: first results, 2017-18. Canberra: ABS. 2019;1:97
Show all 59 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3.. Structural equation model describing the…
Figure 3.. Structural equation model describing the pattern of interrelationships between exposures, microbiome, and host inflammation.
A modified path diagram of the final SEM supporting the mediatory effect of the gut microbiota on inflammation. The heatmaps represent direct associations of diet, behavior, and demographics with the gut microbiota and inflammatory biomarkers. Only significant paths of the effect of the gut microbiota on inflammation are shown. Standardized β coefficients are reported. Species Factors 1–6 denote latent variables of the community gut microbiota modeled as exploratory factor analysis (EFA) regression scores of the species relative abundance; Biomarker Factors 1–3 denote latent variables representing community inflammatory biomarkers as grouped using EFA. *p 0.1–0.05, **p 0.05–0.001, ***p

Figure 3—figure supplement 1.. Theoretical models for…

Figure 3—figure supplement 1.. Theoretical models for assessing the structure of associations between the various…

Figure 3—figure supplement 1.. Theoretical models for assessing the structure of associations between the various human exposures, the gut microbiota, and the host’s inflammatory profile.
(a) Final framework (framework 1): The gut microbiome mediates, in part, the relationship between exposure risk factors and host inflammation. (b) Framework 2: The gut microbiome and exposure risk factors both, independently, impact host inflammation. (c) Framework 3: Exposure risk factors are associated with gut microbiome and inflammatory profile, but there is no relationship between microbiome and inflammation. (d) Framework 4: Exposure risk factors are associated with the inflammatory profile, and the inflammatory profile predicts the gut microbiome composition.

Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the…

Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the…

Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15 and mean arterial pressure.
(a) Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15. The total effect (95% confidence interval) for the model was 0.054 (0.023, 0.090), P-value: <0.001. n = 100 (b) Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with mean arterial pressure. The total effect (95% confidence interval) for the model was −0.006 (–1.530, 1.420), P-value: 0.950. n = 100 a ACME (average causal mediation effects); b ADE (average direct effects).

Figure 4—figure supplement 1.. Role of Lachnospiraceae…

Figure 4—figure supplement 1.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation…

Figure 4—figure supplement 1.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15.
(a) Age-adjusted model. The total effect (95% confidence interval) for the model was 0.049 (0.015, 0.086) P-value: 0.004. (b) Age- and island-adjusted model. The total effect (95% confidence interval) for the model was 0.046 (0.008, 0.085) P-value: 0.016. (c) Multivariate-adjusted model. Includes adjustment for age, island, body mass index, gender, cigarette use, and intakes of fruits, vegetables, takeaways, seafood, and alcohol. The total effect (95% confidence interval) for the model was 0.045 (–0.0003, 0.088) P-value: 0.084.

Figure 4—figure supplement 2.. Role of Lachnospiraceae…

Figure 4—figure supplement 2.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation…

Figure 4—figure supplement 2.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation of sugar-sweetened beverage intake with the mean arterial pressure.
(a) Age-adjusted model. The total effect (95% confidence interval) for the model was 0.842 (−0.822, 2.520) P-value: 0.300. (b) Age-and island-adjusted model. The total effect (95% confidence interval) for the model was 0.948 (−0.694, 2.670) P-value: 0.246. (c) Multivariate-adjusted model. Includes adjustment for age, island, body mass index, gender, cigarette use, and intakes of fruits, vegetables, takeaways, seafood, and alcohol. The total effect (95% confidence interval) for the model was 0.295 (−1.730, 2.170) P-value: 0.808.

Figure 4—figure supplement 3.. 16S rRNA-based phylogeny…

Figure 4—figure supplement 3.. 16S rRNA-based phylogeny of Lachnospiraceae.

Lachnospiraceae family-restricted blast search (blast parameters…

Figure 4—figure supplement 3.. 16S rRNA-based phylogeny of Lachnospiraceae.
Lachnospiraceae family-restricted blast search (blast parameters e-value = 1 e-10, Max targets = 250) using Lachnospiraceae bacterium 8_1_57FAA 16 rRNA as a query against the NCBI non-redundant genes database was performed. The resulting top 250 best blast hits were used to construct a Grishin distance tree using the fast-minimum evolution model in NCBI BLAST. Terminal node labels are Taxonomic Name (Sequence ID).

Figure 5.. Systolic blood pressure in the…

Figure 5.. Systolic blood pressure in the context of an interaction between Alistipes onderdonkii and…

Figure 5.. Systolic blood pressure in the context of an interaction between Alistipes onderdonkii and island systolic blood pressure.
(a) Unadjusted model, (b) age-adjusted model, and (c) multivariate-adjusted model which includes the adjustment for age, body mass index, gender, smoking status, and intakes of fruits, vegetables, takeaways, sugar-sweetened beverages, seafood, and alcohol. Only significant P values (p<0.05) are shown n = 100.
All figures (11)
Figure 3—figure supplement 1.. Theoretical models for…
Figure 3—figure supplement 1.. Theoretical models for assessing the structure of associations between the various human exposures, the gut microbiota, and the host’s inflammatory profile.
(a) Final framework (framework 1): The gut microbiome mediates, in part, the relationship between exposure risk factors and host inflammation. (b) Framework 2: The gut microbiome and exposure risk factors both, independently, impact host inflammation. (c) Framework 3: Exposure risk factors are associated with gut microbiome and inflammatory profile, but there is no relationship between microbiome and inflammation. (d) Framework 4: Exposure risk factors are associated with the inflammatory profile, and the inflammatory profile predicts the gut microbiome composition.
Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the…
Figure 4.. Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15 and mean arterial pressure.
(a) Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15. The total effect (95% confidence interval) for the model was 0.054 (0.023, 0.090), P-value: <0.001. n = 100 (b) Lachnospiraceae bacterium 8_1_57FAA mediates the unadjusted relation of sugar-sweetened beverage intake with mean arterial pressure. The total effect (95% confidence interval) for the model was −0.006 (–1.530, 1.420), P-value: 0.950. n = 100 a ACME (average causal mediation effects); b ADE (average direct effects).
Figure 4—figure supplement 1.. Role of Lachnospiraceae…
Figure 4—figure supplement 1.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation of sugar-sweetened beverage intake with the serum concentration of Interleukin 15.
(a) Age-adjusted model. The total effect (95% confidence interval) for the model was 0.049 (0.015, 0.086) P-value: 0.004. (b) Age- and island-adjusted model. The total effect (95% confidence interval) for the model was 0.046 (0.008, 0.085) P-value: 0.016. (c) Multivariate-adjusted model. Includes adjustment for age, island, body mass index, gender, cigarette use, and intakes of fruits, vegetables, takeaways, seafood, and alcohol. The total effect (95% confidence interval) for the model was 0.045 (–0.0003, 0.088) P-value: 0.084.
Figure 4—figure supplement 2.. Role of Lachnospiraceae…
Figure 4—figure supplement 2.. Role of Lachnospiraceae bacterium 8 1 57FAA in mediating the relation of sugar-sweetened beverage intake with the mean arterial pressure.
(a) Age-adjusted model. The total effect (95% confidence interval) for the model was 0.842 (−0.822, 2.520) P-value: 0.300. (b) Age-and island-adjusted model. The total effect (95% confidence interval) for the model was 0.948 (−0.694, 2.670) P-value: 0.246. (c) Multivariate-adjusted model. Includes adjustment for age, island, body mass index, gender, cigarette use, and intakes of fruits, vegetables, takeaways, seafood, and alcohol. The total effect (95% confidence interval) for the model was 0.295 (−1.730, 2.170) P-value: 0.808.
Figure 4—figure supplement 3.. 16S rRNA-based phylogeny…
Figure 4—figure supplement 3.. 16S rRNA-based phylogeny of Lachnospiraceae.
Lachnospiraceae family-restricted blast search (blast parameters e-value = 1 e-10, Max targets = 250) using Lachnospiraceae bacterium 8_1_57FAA 16 rRNA as a query against the NCBI non-redundant genes database was performed. The resulting top 250 best blast hits were used to construct a Grishin distance tree using the fast-minimum evolution model in NCBI BLAST. Terminal node labels are Taxonomic Name (Sequence ID).
Figure 5.. Systolic blood pressure in the…
Figure 5.. Systolic blood pressure in the context of an interaction between Alistipes onderdonkii and island systolic blood pressure.
(a) Unadjusted model, (b) age-adjusted model, and (c) multivariate-adjusted model which includes the adjustment for age, body mass index, gender, smoking status, and intakes of fruits, vegetables, takeaways, sugar-sweetened beverages, seafood, and alcohol. Only significant P values (p<0.05) are shown n = 100.

References

    1. Anderson I, Robson B, Connolly M, Al-Yaman F, Bjertness E, King A, Tynan M, Madden R, Bang A, Coimbra CEA, Pesantes MA, Amigo H, Andronov S, Armien B, Obando DA, Axelsson P, Bhatti ZS, Bhutta ZA, Bjerregaard P, Bjertness MB, Briceno-Leon R, Broderstad AR, Bustos P, Chongsuvivatwong V, Chu J, Deji, Gouda J, Harikumar R, Htay TT, Htet AS, Izugbara C, Kamaka M, King M, Kodavanti MR, Lara M, Laxmaiah A, Lema C, Taborda AML, Liabsuetrakul T, Lobanov A, Melhus M, Meshram I, Miranda JJ, Mu TT, Nagalla B, Nimmathota A, Popov AI, Poveda AMP, Ram F, Reich H, Santos RV, Sein AA, Shekhar C, Sherpa LY, Skold P, Tano S, Tanywe A, Ugwu C, Ugwu F, Vapattanawong P, Wan X, Welch JR, Yang G, Yang Z, Yap L. Indigenous and tribal peoples' health (The Lancet–Lowitja Institute Global Collaboration): a population study. The Lancet. 2016;388:131–157. doi: 10.1016/S0140-6736(16)00345-7.
    1. Arnold LW, Hoy WE, Sharma SK, Wang Z. The association between HbA1c and cardiovascular disease markers in a remote indigenous australian community with and without diagnosed Diabetes. Journal of Diabetes Research. 2016;2016:1–8. doi: 10.1155/2016/5342304.
    1. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, Bertalan M, Borruel N, Casellas F, Fernandez L, Gautier L, Hansen T, Hattori M, Hayashi T, Kleerebezem M, Kurokawa K, Leclerc M, Levenez F, Manichanh C, Nielsen HB, Nielsen T, Pons N, Poulain J, Qin J, Sicheritz-Ponten T, Tims S, Torrents D, Ugarte E, Zoetendal EG, Wang J, Guarner F, Pedersen O, de Vos WM, Brunak S, Doré J, Antolín M, Artiguenave F, Blottiere HM, Almeida M, Brechot C, Cara C, Chervaux C, Cultrone A, Delorme C, Denariaz G, Dervyn R, Foerstner KU, Friss C, van de Guchte M, Guedon E, Haimet F, Huber W, van Hylckama-Vlieg J, Jamet A, Juste C, Kaci G, Knol J, Lakhdari O, Layec S, Le Roux K, Maguin E, Mérieux A, Melo Minardi R, M'rini C, Muller J, Oozeer R, Parkhill J, Renault P, Rescigno M, Sanchez N, Sunagawa S, Torrejon A, Turner K, Vandemeulebrouck G, Varela E, Winogradsky Y, Zeller G, Weissenbach J, Ehrlich SD, Bork P, MetaHIT Consortium Enterotypes of the human gut microbiome. Nature. 2011;473:174–180. doi: 10.1038/nature09944.
    1. Asnicar F, Weingart G, Tickle TL, Huttenhower C, Segata N. Compact graphical representation of phylogenetic data and metadata with GraPhlAn. PeerJ. 2015;3:e1029. doi: 10.7717/peerj.1029.
    1. Australian Bureau of Statistics National health survey: first results, 2017-18. Canberra: ABS. 2019;1:97
    1. Australian Institute of Health and Welfare . Australia’s Health 2018: In Brief. Canberra: AIHW; 2018.
    1. Azzopardi PS, Sawyer SM, Carlin JB, Degenhardt L, Brown N, Brown AD, Patton GC. Health and wellbeing of indigenous adolescents in Australia: a systematic synthesis of population data. The Lancet. 2018;391:766–782. doi: 10.1016/S0140-6736(17)32141-4.
    1. Berger M, Taylor S, Harriss L, Campbell S, Thompson F, Jones S, Makrides M, Gibson R, Paul Amminger G, Sarnyai Z, McDermott R. Cross-sectional association of seafood consumption, polyunsaturated fatty acids and depressive symptoms in two torres strait communities. Nutritional Neuroscience. 2020;23:353–362. doi: 10.1080/1028415X.2018.1504429.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for illumina sequence data. Bioinformatics. 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Brooks AW, Priya S, Blekhman R, Bordenstein SR. Gut Microbiota diversity across ethnicities in the united states. PLOS Biology. 2018;16:e2006842. doi: 10.1371/journal.pbio.2006842.
    1. Clemente JC, Manasson J, Scher JU. The role of the gut microbiome in systemic inflammatory disease. BMJ. 2018;360:j5145. doi: 10.1136/bmj.j5145.
    1. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563. doi: 10.1038/nature12820.
    1. Engen PA, Green SJ, Voigt RM, Forsyth CB, Keshavarzian A. The gastrointestinal microbiome: alcohol effects on the composition of intestinal Microbiota. Alcohol Research : Current Reviews. 2015;37:223–236.
    1. Epskamp S, Epskamp MS. MplusAutomation S: Package ‘semPlot. 1.1.2Github. 2017
    1. Franzosa EA, McIver LJ, Rahnavard G, Thompson LR, Schirmer M, Weingart G, Lipson KS, Knight R, Caporaso JG, Segata N, Huttenhower C. Species-level functional profiling of metagenomes and metatranscriptomes. Nature Methods. 2018;15:962–968. doi: 10.1038/s41592-018-0176-y.
    1. Gassasse Z, Smith D, Finer S, Gallo V. Association between urbanisation and type 2 diabetes: an ecological study. BMJ Global Health. 2017;2:e000473. doi: 10.1136/bmjgh-2017-000473.
    1. He Y, Wu W, Zheng HM, Li P, McDonald D, Sheng HF, Chen MX, Chen ZH, Ji GY, Zheng ZD, Mujagond P, Chen XJ, Rong ZH, Chen P, Lyu LY, Wang X, Wu CB, Yu N, Xu YJ, Yin J, Raes J, Knight R, Ma WJ, Zhou HW. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nature Medicine. 2018;24:1532–1535. doi: 10.1038/s41591-018-0164-x.
    1. Hodge AM, Maple-Brown L, Cunningham J, Boyle J, Dunbar T, Weeramanthri T, Shaw J, O'Dea K. Abdominal obesity and other risk factors largely explain the high CRP in indigenous australians relative to the general population, but not gender differences: a cross-sectional study. BMC Public Health. 2010;10:700. doi: 10.1186/1471-2458-10-700.
    1. Hooper D, Coughlan J. Structural equation modelling: guidelines for determining model fit. Electronic Journal of Business Research Methods. 2008;6:53–60.
    1. Hu FB. Globalization of diabetes: the role of diet, lifestyle, and genes. Diabetes Care. 2011;34:1249–1257. doi: 10.2337/dc11-0442.
    1. Hu L‐t, Bentler PM. Cutoff criteria for fit indexes in covariance structure analysis: conventional criteria versus new alternatives. Structural Equation Modeling: A Multidisciplinary Journal. 1999;6:1–55. doi: 10.1080/10705519909540118.
    1. Human Microbiome Project Consortium Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214. doi: 10.1038/nature11234.
    1. Jie Z, Xia H, Zhong SL, Feng Q, Li S, Liang S, Zhong H, Liu Z, Gao Y, Zhao H, Zhang D, Su Z, Fang Z, Lan Z, Li J, Xiao L, Li J, Li R, Li X, Li F, Ren H, Huang Y, Peng Y, Li G, Wen B, Dong B, Chen JY, Geng QS, Zhang ZW, Yang H, Wang J, Wang J, Zhang X, Madsen L, Brix S, Ning G, Xu X, Liu X, Hou Y, Jia H, He K, Kristiansen K. The gut microbiome in atherosclerotic cardiovascular disease. Nature Communications. 2017;8:845. doi: 10.1038/s41467-017-00900-1.
    1. Jost L. Partitioning diversity into independent alpha and beta components. Ecology. 2007;88:2427–2439. doi: 10.1890/06-1736.1.
    1. Kaibe M, Ohishi M, Ito N, Yuan M, Takagi T, Terai M, Tatara Y, Komai N, Rakugi H, Ogihara T. Serum interleukin-15 concentration in patients with essential hypertension. American Journal of Hypertension. 2005;18:1019–1025. doi: 10.1016/j.amjhyper.2005.02.014.
    1. Kameyama K, Itoh K. Intestinal colonization by a Lachnospiraceae bacterium contributes to the development of diabetes in obese mice. Microbes and Environments. 2014;29:427–430. doi: 10.1264/jsme2.ME14054.
    1. Krolopp JE, Thornton SM, Abbott MJ. IL-15 activates the Jak3/STAT3 signaling pathway to mediate glucose uptake in skeletal muscle cells. Frontiers in Physiology. 2016;7:626. doi: 10.3389/fphys.2016.00626.
    1. Levy M, Blacher E, Elinav E. Microbiome, metabolites and host immunity. Current Opinion in Microbiology. 2017;35:8–15. doi: 10.1016/j.mib.2016.10.003.
    1. Lippert K, Kedenko L, Antonielli L, Kedenko I, Gemeier C, Leitner M, Kautzky-Willer A, Paulweber B, Hackl E. Gut Microbiota dysbiosis associated with glucose metabolism disorders and the metabolic syndrome in older adults. Beneficial Microbes. 2017;8:545–556. doi: 10.3920/BM2016.0184.
    1. Liu B, Zhang Y, Wang R, An Y, Gao W, Bai L, Li Y, Zhao S, Fan J, Liu E. Western diet feeding influences gut Microbiota profiles in apoE knockout mice. Lipids in Health and Disease. 2018;17:159. doi: 10.1186/s12944-018-0811-8.
    1. Martin AM, Sun EW, Rogers GB, Keating DJ. The influence of the gut microbiome on host metabolism through the regulation of gut hormone release. Frontiers in Physiology. 2019;10:428. doi: 10.3389/fphys.2019.00428.
    1. Miller G, McDermott R, McCulloch B, Leonard D, Arabena K, Muller R. The well person's Health Check: a population screening program in indigenous communities in north Queensland. Australian Health Review. 2002;25:136–147. doi: 10.1071/AH020136b.
    1. Morgan XC, Kabakchiev B, Waldron L, Tyler AD, Tickle TL, Milgrom R, Stempak JM, Gevers D, Xavier RJ, Silverberg MS, Huttenhower C. Associations between host gene expression, the mucosal microbiome, and clinical outcome in the pelvic pouch of patients with inflammatory bowel disease. Genome Biology. 2015;16:67. doi: 10.1186/s13059-015-0637-x.
    1. Niccoli T, Partridge L. Ageing as a risk factor for disease. Current Biology. 2012;22:R741–R752. doi: 10.1016/j.cub.2012.07.024.
    1. O'Keefe SJ, Li JV, Lahti L, Ou J, Carbonero F, Mohammed K, Posma JM, Kinross J, Wahl E, Ruder E, Vipperla K, Naidoo V, Mtshali L, Tims S, Puylaert PG, DeLany J, Krasinskas A, Benefiel AC, Kaseb HO, Newton K, Nicholson JK, de Vos WM, Gaskins HR, Zoetendal EG. Fat, fibre and Cancer risk in african americans and rural africans. Nature Communications. 2015;6:6342. doi: 10.1038/ncomms7342.
    1. Oksanen J, Blanchet F, Friendly M, Kindt R, Legendre P, McGlinn D, Minchin P, O’Hara R, Simpson G. R package version; 2018.
    1. Popkin BM, Adair LS, Ng SW. Global nutrition transition and the pandemic of obesity in developing countries. Nutrition Reviews. 2012;70:3–21. doi: 10.1111/j.1753-4887.2011.00456.x.
    1. Queensland Health . The Burden of Disease and Injury in Queensland’s Aboriginal and TorresStrait Islander People 2017 (Reference Year 2011) Brisbane: Queensland Health; 2017.
    1. R Development Core Team . Vienna, Austria: R Foundation for Statistical Computing; 2010.
    1. Rosseel Y. Lavaan : an R package for structural equation modeling. Journal of Statistical Software. 2012;48:v048i02. doi: 10.18637/jss.v048.i02.
    1. Rowley K, Walker KZ, Cohen J, Jenkins AJ, O'Neal D, Su Q, Best JD, O'Dea K. Inflammation and vascular endothelial activation in an aboriginal population: relationships to coronary disease risk factors and nutritional markers. Medical Journal of Australia. 2003;178:495–500. doi: 10.5694/j.1326-5377.2003.tb05324.x.
    1. Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, Hammer RE, Williams SC, Crowley J, Yanagisawa M, Gordon JI. Effects of the gut Microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. PNAS. 2008;105:16767–16772. doi: 10.1073/pnas.0808567105.
    1. Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, ter Horst R, Jansen T, Jacobs L, Bonder MJ, Kurilshikov A, Fu J, Joosten LAB, Zhernakova A, Huttenhower C, Wijmenga C, Netea MG, Xavier RJ. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167:1125–1136. doi: 10.1016/j.cell.2016.10.020.
    1. Schutte AE, Shemesh T, Rowley K, Best JD, McDermott R, O'Dea K. The metabolic syndrome and changing relationship between blood pressure and insulin with age, as observed in aboriginal and Torres strait islander peoples. Diabetic Medicine. 2005;22:1589–1597. doi: 10.1111/j.1464-5491.2005.01747.x.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, Huttenhower C. Metagenomic biomarker discovery and explanation. Genome Biology. 2011;12:R60. doi: 10.1186/gb-2011-12-6-r60.
    1. Shannon C. Acculturation: aboriginal and Torres strait islander nutrition. Asia Pacific Journal of Clinical Nutrition. 2002;11:S576–S578. doi: 10.1046/j.0964-7058.2002.00352.x.
    1. Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56–64. doi: 10.1038/nature18846.
    1. Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535:65–74. doi: 10.1038/nature18847.
    1. Truong DT, Franzosa EA, Tickle TL, Scholz M, Weingart G, Pasolli E, Tett A, Huttenhower C, Segata N. MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nature Methods. 2015;12:902–903. doi: 10.1038/nmeth.3589.
    1. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449:804–810. doi: 10.1038/nature06244.
    1. Vangay P, Johnson AJ, Ward TL, Al-Ghalith GA, Shields-Cutler RR, Hillmann BM, Lucas SK, Beura LK, Thompson EA, Till LM, Batres R, Paw B, Pergament SL, Saenyakul P, Xiong M, Kim AD, Kim G, Masopust D, Martens EC, Angkurawaranon C, McGready R, Kashyap PC, Culhane-Pera KA, Knights D. US immigration westernizes the human gut microbiome. Cell. 2018;175:962–972. doi: 10.1016/j.cell.2018.10.029.
    1. Wang S, Li N, Li N, Zou H, Wu M. A comparative analysis of biosynthetic gene clusters in lean and obese humans. BioMed Research International. 2019;2019:1–7. doi: 10.1155/2019/6361320.
    1. Wang Z, Hoy WE. C-reactive protein: an independent predictor of cardiovascular disease in aboriginal australians. Australian and New Zealand Journal of Public Health. 2010;34:S25–S29. doi: 10.1111/j.1753-6405.2010.00548.x.
    1. Weiland G. The enzyme-linked immunosorbent assay (ELISA)-a new serodiagnostic method for the detection of parasitic infections (author's transl) MMW, Munchener Medizinische Wochenschrift. 1978;120:1457–1460.
    1. Wen L, Duffy A. Factors influencing the gut Microbiota, inflammation, and type 2 diabetes. The Journal of Nutrition. 2017;147:1468S–1475. doi: 10.3945/jn.116.240754.
    1. Whittaker RH. Evolution and measurement of species diversity. Taxon. 1972;21:213–251. doi: 10.2307/1218190.
    1. World Health Organization . Global Status Report on Noncommunicable Diseases. WHO; 2014.
    1. Yeates K, Lohfeld L, Sleeth J, Morales F, Rajkotia Y, Ogedegbe O. A global perspective on cardiovascular disease in vulnerable populations. Canadian Journal of Cardiology. 2015;31:1081–1093. doi: 10.1016/j.cjca.2015.06.035.
    1. Zhao Y, Connors C, Wright J, Guthridge S, Bailie R. Estimating chronic disease prevalence among the remote aboriginal population of the northern territory using multiple data sources. Australian and New Zealand Journal of Public Health. 2008;32:307–313. doi: 10.1111/j.1753-6405.2008.00245.x.

Source: PubMed

3
Abonner