Next Article in Journal
Stochastic Models of Particle Trajectories in Turbulent Atmosphere Flows by Using the LANGEVIN Equations
Previous Article in Journal
Lyophilized Extract of Hibiscus sabdariffa L. Induces Cytotoxicity in Breast Cancer Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

Effect of Pulse Consumption on Obesity and the Metagenome †

1
Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA
2
Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523, USA
*
Author to whom correspondence should be addressed.
Presented at the 1st International Electronic Conference on Nutrients—Nutritional and Microbiota Effects on Chronic Disease, 2–15 November 2020; Available online: https://iecn2020.sciforum.net/.
Proceedings 2020, 61(1), 23; https://doi.org/10.3390/IECN2020-07009
Published: 30 October 2020

Abstract

:
Grain legumes, which are commonly referred to as pulses, are staple foods in many parts of the world, but are infrequently consumed in most economically developed countries where the obesity pandemic is prominent. However, even in low pulse consuming countries such as the United States, there are sub-groups of individuals who consume large amounts of pulses. Systematic reviews of population studies indicate that pulse consumers have a lower risk for developing obesity. To determine whether these population-based findings could be modeled in preclinical systems in which such findings can be deconstructed, we used rat and mouse models of dietary induced obesity and reported that lipid accumulation was inhibited. In this study, we examined the relationship between inhibition of fat accumulation and changes in the gut associated microbiome in male C57BL/6 mice fed either a high fat diet with casein as the protein source or that diet formulation in which one of four pulses (chickpea, common bean, dry pea, or lentil) was substituted to provide 70% dietary protein with the remainder provided by casein. The seeds of each pulse were soaked, cooked, and then freeze-dried and milled; the resulting powder was used for diet formulation. Mice were ad libitum fed over the 17-week duration of the feeding trial. Cecal content was obtained at necropsy and immediately snap frozen in liquid nitrogen. Extracted genomic DNA was processed for 16s rRNA sequencing on an Illumina system. Significant differences were observed between each pulse and the high fat control diet in microbial phylogenetic diversity (p < 0.001) and accumulation of lipid in adipose depots (p < 0.01). Differences among pulses were also observed in both metrics. Microbiome alpha and beta diversity metrics, differences in abundance for each detected taxon among treatment groups, and their relationships to changes in lipid accumulation in adipose storage depots are reported.

1. Introduction

The occurrence of obesity, a global pandemic, increases the risk for a number of aging-associated chronic diseases including non-alcoholic fatty liver disease, type-2 diabetes, cardiovascular disease, and certain types of cancer [1]. In these disease states, insulin resistance, chronic inflammation, and oxidation-mediated cell damage are known to contribute to disease pathogenesis [2,3,4,5,6]. Deregulation of interconnected cell signaling pathways among tissues underlies the metabolic dysfunction that is observed. While the importance of environmental effects on gene expression are well-established, there has been a lack of compelling evidence that an individual’s diet exerts effects on the signaling networks that impact disease outcomes including mortality. This has led to increased reliance on prescribed drugs, while limiting the attention given to food-based interventions in chronic disease prevention and control [7]. However, emerging evidence now supports significant impacts of diet that are mediated by its interaction with the gut microbiome that result in reduced risk for chronic diseases [8,9,10,11,12]. As an example of this, recent meta-analyses indicate that consumption of grain legumes, i.e., common bean, chickpea, dry pea, and lentil, also referred to as pulses [13], is associated with improved weight management relative to populations in which the consumption of pulses is low [14,15]. Given the complexity of obesity, we recently sought to determine whether the impact of pulses on body weight observed in prospective clinical studies could be reproduced under the controlled conditions that it is possible to achieve with the use of preclinical rodent models [16]. In polygenic models for obesity in both rats and in mice, pulse consumption was shown to be anti-obesogenic [16,17,18].
It is widely recognized that food components play a major role in establishing and maintaining the gut microbiome. Of the wide array of bioactive constituents in foods, dietary fiber is a key determinant. While there is no requirement for dietary fiber, the level currently recommended (14 g/1000 kcal) is achieved by less than 25% of the US population and the average shortfall exceeds 50% [19]. The problem can be traced to low food quality relative to dietary fiber content; particularly, a lack of grain legumes, i.e., pulse crops in the diet. Our recent publications document that the most commonly consumed pulses, dry bean (Phaseolus vulgaris, L.), chickpea (Cicer arietinum L.), dry pea (Pisum sativum L.), and lentil (Lens culinaris L.) have 2–3 times more fiber per 100 kcal edible portion than other commonly promoted dietary fiber sources, e.g., cereal grains [20,21]. Whether or not these pulses have equivalent effects on the composition and function of the gut microbiome is not known.
The objectives of the work reported herein were: (1) to compare the effects among pulses, i.e., chickpea, common bean, dry pea, and lentil, on the metagenome within the cecum, and (2) to investigate relationships between the effects of these pulses on the microbiome relative to the accumulation of lipid in adipose depots.

2. Materials and Methods

2.1. Experimental Animals and Design

Details of the feeding study have previously been reported [18]. Briefly, NCI C57BL/6NCrl male mice (21–28 days of age) were obtained from Charles River Laboratories NCI (Frederick, MD, USA). Upon arrival, the mice were fed a purified high fat diet. Mice were housed in solid bottomed polycarbonate rodent cages and maintained on a 12 h light/dark cycle at 27.5 ± 2 °C with 30% relative humidity. All mice had ad libitum access to diet and distilled water. All animal studies were performed in accordance with the Colorado State University Institutional Animal Care and Use Committee (protocol 18-7746A). At 5 weeks of age mice were randomized to their treatment groups. Mice were either continued on the high fat (HF) formulation (control diet) or were fed the HF diet formulation to which common bean, chickpea, dry pea, or lentil was added. The formulation of the experimental diets and the rationale for the concentration of pulses has been published [18]. The experimental duration was 17 weeks. At necropsy, inguinal subcutaneous and abdominal visceral adipose tissue were harvested and weighed. Content of the cecum was harvested and snap frozen in liquid nitrogen until it was processed for genomic DNA extraction.

2.2. Microbiota Characterization

Using intestinal specimens collected at necropsy, DNA was extracted using the QIAamp PowerFecal DNA Kit (Qiagen, Germantown, MD, USA). 2 × 250 bp paired-end sequencing libraries of the V4 region of the 16s rRNA gene were constructed by using Earth Microbiome Project protocols as previously published [22]. Functional predictions were made analyzing sequences by PICRUSt2 to infer functional content and visualized using STAMP v2.1.3 [23,24].

2.3. Statistical Analyses

Data were evaluated using ANOVA, PERMANOVA, regression analysis, or multivariate analysis techniques. The Benjamini–Hochberg method was used to adjust p-values to control the false discovery rate. Data analyses were conducted using Systat, version 13.0 (Systat Software, Inc., San Jose, CA, USA), CLC Genomics Workbench version 20.0.4 (Qiagen Bioinformatics, Redwood City, CA, USA) and RStudio version 1.1.456 (RStudio, Boston, MA, USA) running R version 3.6.3 (The R Foundation for Statistical Computing, Vienna, Austria) and SIMCA v15 software (Sartorius Stedim Biotech, Umea, Sweden).

3. Results and Discussion

3.1. Effects of Pulses on Adipose Depot Mass

Previously, we have reported that feeding diets containing 70% of dietary protein from chickpea, common bean, dry pea, or lentil reduced visceral and subcutaneous fat pad mass of male C57BL/6 mice [18]. In the analysis presented in Figure 1, all individual fat pad masses for an animal across treatment groups were subjected to unsupervised principal components analysis. The relative clustering of pulses in terms of their adipose tissue mass in comparison to the positive control (high fat obesogenic diet) and negative control (low fat non obesogenic diet) is shown in the three-dimensional scores plot.
Given that each principal component assigned to an animal is an unbiased measure of its adipose mass, each animal’s first principal component value (PC1) was subjected to ANOVA. The overall p-value was highly significant (p < 0.0009) and post hoc analysis confirmed that all pulses were significantly different from either control, but not different from one another.

3.2. Effect of Pulses on Cecal Microbiome

It is recognized that the composition of microbiome varies throughout the length of the intestinal tract, particularly as it relates to conditions in the gut lumen relative to the growth of facultative and obligate anaerobes which are important groupings of commensal microorganisms. Therefore, we decided to evaluate the impact of pulse consumption on microbial composition of the luminal content of the cecum. In the mouse, the cecum interfaces with the ileum, i.e., the distal segment of the small intestine that plays a central role in metabolic signaling, and the ascending colon with which it abuts and that plays key roles in water reabsorption and short chain fatty acid metabolism. The focus on the cecum is in marked contrast to other reports of the impact of pulses on the fecal microbiome [25,26,27,28], which are translationally important, but may not capture information relevant to metabolic effects of the microbiome in regions of the gut that are anerobic.
Microbial abundance data (level of genus) from the 16s rRNA analyses of DNA extracted from cecal luminal content were subjected to unsupervised principal components analysis in SIMCA in the same way the adipose depot mass data were evaluated, in order to permit an opportunity to compare the clustering of data by diet group. The three-dimensional scores plot is Figure 2.
Using the same strategy as described for adipose depot mass, each animal’s first principal component for microbial abundance was evaluated by ANOVA. The model fit had an R2 of 0.76, overall effect of diet, p < 0.001. Post hoc pairwise comparisons among treatment groups revealed that the unbiased principal component for the positive control (high fat, obesogenic) was significantly different from all other diet groups but differences among pulses and between each pulse and the negative control (low fat, non-obesogenic) were not significant.
Since there were overall effects of diet group on the principle components for adipose mass and microbial abundance (genus level), the relationship between these variables was examined via regression analyses (Figure 3). Overall, these analyses support a relationship between cecal microbial composition and adipose tissue mass, using unbiased measures of both variables. This opens the door to using the loading values from the principal components analyses to identify microorganisms (genus level) that contribute to this relationship.
Another focus of our comparative analyses of the effects of pulses on the microbiome in the luminal content of the cecum employed metrics commonly used in the evaluation of metagenomic data.
Using the taxonomic profiling algorithm in the CLC Genomics Workbench, the impact of pulse consumption on species abundance is shown in Figure 4. Major shifts in percent abundance of microbial species were observed when pulses were compared to either the negative or positive dietary control group. Differences among pulses were also apparent. Of particular note, the changes in Akkermansia muciniphila that were reported by us and determined via qPCR using specific primers [17,18], are consistent with the differences observed in Figure 4. The observed pulse induced increase in A. muciniphila are consistent with health-promoting effects of pulses. Gut levels of A. muciniphila have been reported to be inversely associated with obesity, diabetes, and inflammation [29,30,31,32,33,34]. At the level of phylum, pulses, collectively, induced an increase in bacteria in the phylum Bacteroidetes vs. Firmicutes (p < 0.001 for all). Given the controversial nature of the literature about whether an increase in the Bacteroidetes-to-Firmicutes ratio is consistent with health benefits [35,36,37,38,39,40], the importance of this observation requires further investigation.
We also evaluated the effect of pulse consumption on alpha diversity using CLC Taxonomic Profiler (Figure 5). Unlike other metrics that have been presented thus far, statistically significant differences in alpha diversity were observed. The rank order from lowest to highest was: lentil ≤ dry pea < common bean < chickpea. The lowest alpha diversity was observed for the positive control (high fat, obesogenic). While the commonly held view is that higher phylogenetic diversity is prognostic for a healthy gut, there is no consensus on this point [9]. However, a diverse microbiota gives rise to an immense metabolic potential. The microbiome consists of the genes that the cells constituting the microbiota harbor. A human microbiome collectively contains on the order of 3 million non-redundant genes; whereas, the human genome is comprised of approximately 20,000 genes [41]. Unsurprisingly, the gut microbiota executes essential functions that the body itself is incapable of performing. These functions include promotion of gut maturation, education of the immune system, protection against viral and bacterial pathogens, influence on brain activities and bodily activities.
The effect on beta-diversity was also assessed (Figure 6).
The hierarchical cluster analysis of these data showed that the positive control (high fat, obesogenic diet) was on a branch distinct from all other diet groups.
The metagenomic data were also evaluated for predictions of function using PICRUSt. That output was subject to unsupervised principal components in SIMCA using the same strategy described in the presentation of Figure 1 and Figure 2. The scores plot from that analysis is Figure 7.
The ANOVA of the principle components for each animal from the PCA supported the assessment that there was an overall effect of diet group on predicted functional activity categorized via Kegg defined metabolic pathways. The post hoc analysis indicated that pulses differed from the control groups and among one another. This supports the hypothesis that all pulses are not created equal in terms of microbial populations whose cecal colonization they support.

4. Final Comments

There has been an explosive expansion of the literature on food, the gut microbiome, and human health and disease with major reviews being published regularly as illustrated in these citations [42,43,44]. From this burgeoning literature, the central thesis we advance is that increased granularity in the assessment of pulse-derived chemical exposures in the gut is required to understand their effects on microbial ecology and function. The ultimate effectors of the microbiota on their host are an array of molecules produced due to the transcription activity of the microbiome. Analysis of the effect of pulses on the meta-transcriptome and meta-metabolome are logical next steps in elucidating the role of the microbiome in mediating pulse health effects.

Author Contributions

Conceptualization, H.J.T., T.L.W.; methodology, E.S.N., J.N.M., T.L.W.; formal analysis, T.L.W., E.S.N., J.N.M., H.J.T.; investigation, H.J.T.; data curation, E.S.N., J.N.M., T.L.W.; writing—review and editing, E.S.N., J.N.M., H.J.T., T.L.W.; supervision, E.S.N., J.N.M., H.J.T.; project administration, H.J.T.; funding acquisition, H.J.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Agricultural Research Service, U.S. Department of Agriculture, grant number 3060-21650-001-08S.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Heymsfield, S.B.; Wadden, T.A. Mechanisms, Pathophysiology, and Management of Obesity. N. Engl. J. Med. 2017, 376, 254–266. [Google Scholar] [CrossRef] [PubMed]
  2. Abdelrahim, M.; Morris, E.; Carver, J.; Facchina, S.; White, A.; Verma, A. Liquid chromatographic assay of dityrosine in human cerebrospinal fluid. J. Chromatogr. B Biomed. Sci Appl. 1997, 696, 175–182. [Google Scholar] [CrossRef]
  3. Biddinger, S.B.; Kahn, C.R. From mice to men: insights into the insulin resistance syndromes. Annu. Rev. Physiol 2006, 68, 123–158. [Google Scholar] [CrossRef]
  4. Eyre, H.; Kahn, R.; Robertson, R.M.; ACS/ADA/AHA Collaborative Writing Committee; Clark, N. G.; Doyle, C.; Gansler, T.; Glynn, T.; Hong, Y.; Smith, R.A.; et al. Preventing Cancer, Cardiovascular Disease, and Diabetes: A Common Agenda for the American Cancer Society, the American Diabetes Association, and the American Heart Association. CA: A Cancer J. Clin. 2004, 54, 190–207. [Google Scholar]
  5. Holmes, E.; Wilson, I.D.; Nicholson, J.K. Metabolic phenotyping in health and disease. Cell 2008, 134, 714–717. [Google Scholar] [CrossRef] [PubMed]
  6. Marshall, S. Role of insulin, adipocyte hormones, and nutrient-sensing pathways in regulating fuel metabolism and energy homeostasis: a nutritional perspective of diabetes, obesity, and cancer. Sci. STKE 2006, 2006, re7. [Google Scholar] [CrossRef]
  7. NIH. The Science Behind Healthy Eating Patterns; National Institutes of Health: Wasington, DC, USA, 2020. [Google Scholar]
  8. Health, D.o.H.a.H.S.N.I.o. 2020–2030 Strategic Plan for NIH Nutrition Research. 2020. Available online: https://www.niddk.nih.gov/about-niddk/strategic-plans-reports/strategic-plan-nih-nutrition-research (accessed on 15 October 2020).
  9. Frame, L.A.; Costa, E.; Jackson, S.A. Current Explorations of Nutrition and the Gut Microbiome: A Comprehensive Evaluation of the Review Literature. Nutr. Rev. 2020. [Google Scholar] [CrossRef]
  10. McBurney, M.I.; Davis, C.; Fraser, C.M.; Schneeman, B.O.; Huttenhower, C.; Verbeke, K.; Walter, J.; Latulippe, M.E. Establishing What Constitutes a Healthy Human Gut Microbiome: State of the Science, Regulatory Considerations, and Future Directions. J. Nutr. 2019, 149, 1882–1895. [Google Scholar] [CrossRef]
  11. Bilotta, A.J.; Cong, Y. Gut microbiota metabolite regulation of host defenses at mucosal surfaces: implication in precision medicine. Precis. Clin. Med. 2019, 2, 110–119. [Google Scholar] [CrossRef]
  12. Rodgers, G.P.; Collins, F.S. Precision Nutrition-the Answer to “What to Eat to Stay Healthy”. JAMA 2020. [Google Scholar] [CrossRef]
  13. Organization, F.A.A. Definition and Classification of Commodities: Pulses and Derived Products. Available online: http://www.fao.org/es/faodef/fdef04e.htm (accessed on 20 January 2017).
  14. Viguiliouk, E.; Stewart, S.E.; Jayalath, V.H.; Ng, A.P.; Mirrahimi, A.; de Souza, R.J.; Hanley, A.J.; Bazinet, R.P.; Blanco, M.S.; Leiter, L.A.; et al. Effect of Replacing Animal Protein with Plant Protein on Glycemic Control in Diabetes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2015, 7, 9804–9824. [Google Scholar] [CrossRef] [PubMed]
  15. Viguiliouk, E.; Blanco, M.S.; Kendall, C.W.; Sievenpiper, J.L. Can pulses play a role in improving cardiometabolic health? Evidence from systematic reviews and meta-analyses. Ann. N. Y. Acad. Sci. 2017, 1392, 43–57. [Google Scholar] [CrossRef] [PubMed]
  16. Thompson, H.J.; McGinley, J.N.; Neil, E.S.; Brick, M.A. Beneficial Effects of Common Bean on Adiposity and Lipid Metabolism. Nutrients 2017, 9. [Google Scholar] [CrossRef]
  17. Neil, E.S.; McGinley, J.N.; Fitzgerald, V.K.; Lauck, C.A.; Tabke, J.A.; Streeter-McDonald, M.R.; Yao, L.; Broeckling, C.D.; Weir, T.L.; Foster, M.T.; et al. White Kidney Bean (Phaseolus Vulgaris L.) Consumption Reduces Fat Accumulation in a Polygenic Mouse Model of Obesity. Nutrients 2019, 11, E2780. [Google Scholar] [CrossRef] [PubMed]
  18. McGinley, J.N.; Fitzgerald, V.K.; Neil, E.S.; Omerigic, H.M.; Heuberger, A.L.; Weir, T.L.; McGee, R.; Vandemark, G.; Thompson, H.J. Pulse Crop Effects on Gut Microbial Populations, Intestinal Function, and Adiposity in a Mouse Model of Diet-Induced Obesity. Nutrients 2020, 12. [Google Scholar] [CrossRef] [PubMed]
  19. Thompson, H.J.; Brick, M.A. Perspective: Closing the Dietary Fiber Gap: An Ancient Solution for a 21st Century Problem. Adv. Nutr. 2016, 7, 623–626. [Google Scholar] [CrossRef]
  20. Kleintop, A.E.; Echeverria, D.; Brick, L.A.; Thompson, H.J.; Brick, M.A. Adaptation of the AOAC 2011.25 Integrated Total Dietary Fiber Assay To Determine the Dietary Fiber and Oligosaccharide Content of Dry Edible Beans. J. Agr. Food Chem. 2013, 61, 9719–9726. [Google Scholar] [CrossRef]
  21. Chen, Y.R.; McGee, R.; Vandemark, G.; Brick, M.; Thompson, H.J. Dietary Fiber Analysis of Four Pulses Using AOAC 2011.25: Implications for Human Health. Nutrients 2016, 8. [Google Scholar] [CrossRef]
  22. Lee, D.M.; Battson, M.L.; Jarrell, D.K.; Hou, S.; Ecton, K.E.; Weir, T.L.; Gentile, C.L. SGLT2 inhibition via dapagliflozin improves generalized vascular dysfunction and alters the gut microbiota in type 2 diabetic mice. Cardiovasc. Diabetol. 2018, 17, 62. [Google Scholar] [CrossRef]
  23. Douglas, G.M.; Maffei, V.J.; Zaneveld, J.; Yurgel, S.N.; Brown, J.R.; Taylor, C.M.; Huttenhower, C.; Langille, M.G.I. PICRUSt2: An improved and extensible approach for metagenome inference. bioRxiv 2019, 672295. [Google Scholar] [CrossRef]
  24. Parks, D.H.; Tyson, G.W.; Hugenholtz, P.; Beiko, R.G. STAMP: Statistical analysis of taxonomic and functional profiles. Bioinformatics 2014, 30, 3123–3124. [Google Scholar] [CrossRef] [PubMed]
  25. Monk, J.M.; Lepp, D.; Wu, W.Q.; Graf, D.; McGillis, L.H.; Hussain, A.; Carey, C.; Robinson, L.E.; Liu, R.H.; Asao, R.; et al. Chickpea-supplemented diet alters the gut microbiome and enhances gut barrier integrity in C57Bl/6 male mice. J. Funct. Foods 2017, 38, 663–674. [Google Scholar] [CrossRef]
  26. Monk, J.M.; Lepp, D.; Wu, W.Q.; Pauls, K.P.; Robinson, L.E.; Power, K.A. Navy and black bean supplementation primes the colonic mucosal microenvironment to improve gut health. J. Nutr. Biochem. 2017, 49, 89–100. [Google Scholar] [CrossRef] [PubMed]
  27. Monk, J.M.; Wu, W.Q.; Hutchinson, A.L.; Pauls, P.; Robinson, L.E.; Power, K.A. Navy and black bean supplementation attenuates colitis-associated inflammation and colonic epithelial damage. J. Nutr. Biochem. 2018, 56, 215–223. [Google Scholar] [CrossRef] [PubMed]
  28. Monk, J.M.; Zhang, C.P.; Wu, W.Q.; Zarepoor, L.; Lu, J.T.; Liu, R.H.; Pauls, K.P.; Wood, G.A.; Tsao, R.; Robinson, L.E.; et al. White and dark kidney beans reduce colonic mucosal damage and inflammation in response to dextran sodium sulfate. J. Nutr. Biochem. 2015, 26, 752–760. [Google Scholar] [CrossRef] [PubMed]
  29. Caesar, R.; Tremaroli, V.; Kovatcheva-Datchary, P.; Cani, P.D.; Backhed, F. Crosstalk between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation through TLR Signaling. Cell Metab. 2015, 22, 658–668. [Google Scholar] [CrossRef]
  30. Centre, W.U.A.R. Intestinal bacterium Akkermansia curbs obesity. Available online: https://www.sciencedaily.com/releases/2013/05/130515113744.htm (accessed on 14 September 2019).
  31. Dao, M.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Verger, E.; Rizkalla, S.; Dore, J.; Cani, P.; Clenent, K. Akkermansia Muciniphila and Gut Microbiota Richness are Associated with Improved Metabolic Status after Calorie Restriction. Faseb J. 2015, 29, 601–603. [Google Scholar] [CrossRef]
  32. Derrien, M.; Belzer, C.; de Vos, W.M. Akkermansia muciniphila and its role in regulating host functions. Microb Pathogenesis 2017, 106, 171–181. [Google Scholar] [CrossRef]
  33. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef]
  34. van der Lugt, B.; van Beek, A.A.; Aalvink, S.; Meijer, B.; Sovran, B.; Vermeij, W.P.; Brandt, R.M.C.; de Vos, W.M.; Savelkoul, H.F.J.; Steegenga, W.T.; et al. Akkermansia muciniphila ameliorates the age-related decline in colonic mucus thickness and attenuates immune activation in accelerated aging Ercc1(-/7) mice. Immun. Ageing 2019, 16. [Google Scholar] [CrossRef] [PubMed]
  35. Koliada, A.; Syzenko, G.; Moseiko, V.; Budovska, L.; Puchkov, K.; Perederiy, V.; Gavalko, Y.; Dorofeyev, A.; Romanenko, M.; Tkach, S.; et al. Association between body mass index and Firmicutes/Bacteroidetes ratio in an adult Ukrainian population. BMC Microbiol. 2017, 17, 120. [Google Scholar] [CrossRef] [PubMed]
  36. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef] [PubMed]
  37. Rinninella, E.; Cintoni, M.; Raoul, P.; Lopetuso, L.R.; Scaldaferri, F.; Pulcini, G.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. Food Components and Dietary Habits: Keys for a Healthy Gut Microbiota Composition. Nutrients 2019, 11. [Google Scholar] [CrossRef]
  38. Rinninella, E.; Mele, M.C.; Merendino, N.; Cintoni, M.; Anselmi, G.; Caporossi, A.; Gasbarrini, A.; Minnella, A.M. The Role of Diet, Micronutrients and the Gut Microbiota in Age-Related Macular Degeneration: New Perspectives from the Gut(-)Retina Axis. Nutrients 2018, 10. [Google Scholar] [CrossRef]
  39. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7. [Google Scholar] [CrossRef] [PubMed]
  40. Mariat, D.; Firmesse, O.; Levenez, F.; Guimaraes, V.; Sokol, H.; Dore, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol 2009, 9, 123. [Google Scholar] [CrossRef]
  41. Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef]
  42. Mills, S.; Lane, J.A.; Smith, G.J.; Grimaldi, K.A.; Ross, R.P.; Stanton, C. Precision Nutrition and the Microbiome Part II: Potential Opportunities and Pathways to Commercialisation. Nutrients 2019, 11. [Google Scholar] [CrossRef]
  43. Mills, S.; Stanton, C.; Lane, J.A.; Smith, G.J.; Ross, R.P. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019, 11. [Google Scholar] [CrossRef]
  44. Zinocker, M.K.; Lindseth, I.A. The Western Diet-Microbiome-Host Interaction and Its Role in Metabolic Disease. Nutrients 2018, 10. [Google Scholar] [CrossRef]
Figure 1. Principal components analysis of the effect of feeding pulse diets on adipose tissue mass.
Figure 1. Principal components analysis of the effect of feeding pulse diets on adipose tissue mass.
Proceedings 61 00023 g001
Figure 2. Principal components analysis of the effect of feeding pulse diets on the abundance of microorganism (level of genus).
Figure 2. Principal components analysis of the effect of feeding pulse diets on the abundance of microorganism (level of genus).
Proceedings 61 00023 g002
Figure 3. Regression analyses evaluating the relationship among principal components for adipose depot mass and microbial abundance (cecal luminal content). All regressions were statistically significant (p < 0.01), except for the regression analysis shown in panel D; lower confidence limit: LCL; upper confidence limit: UPL; lower prediction limit: LPL; upper prediction limit: UPL.
Figure 3. Regression analyses evaluating the relationship among principal components for adipose depot mass and microbial abundance (cecal luminal content). All regressions were statistically significant (p < 0.01), except for the regression analysis shown in panel D; lower confidence limit: LCL; upper confidence limit: UPL; lower prediction limit: LPL; upper prediction limit: UPL.
Proceedings 61 00023 g003
Figure 4. Effect of pulse consumption on microbial abundance (species level) using CLC Genomics Taxonomic Profiler.
Figure 4. Effect of pulse consumption on microbial abundance (species level) using CLC Genomics Taxonomic Profiler.
Proceedings 61 00023 g004
Figure 5. Effect of pulse consumption on alpha diversity.
Figure 5. Effect of pulse consumption on alpha diversity.
Proceedings 61 00023 g005
Figure 6. Effect of pulse consumption on beta diversity/.
Figure 6. Effect of pulse consumption on beta diversity/.
Proceedings 61 00023 g006
Figure 7. Effect of pulse consumption predicted function of the microbiome.
Figure 7. Effect of pulse consumption predicted function of the microbiome.
Proceedings 61 00023 g007
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Weir, T.L.; McGinley, J.N.; Neil, E.S.; Thompson, H.J. Effect of Pulse Consumption on Obesity and the Metagenome. Proceedings 2020, 61, 23. https://doi.org/10.3390/IECN2020-07009

AMA Style

Weir TL, McGinley JN, Neil ES, Thompson HJ. Effect of Pulse Consumption on Obesity and the Metagenome. Proceedings. 2020; 61(1):23. https://doi.org/10.3390/IECN2020-07009

Chicago/Turabian Style

Weir, Tiffany L., John N. McGinley, Elizabeth S. Neil, and Henry J. Thompson. 2020. "Effect of Pulse Consumption on Obesity and the Metagenome" Proceedings 61, no. 1: 23. https://doi.org/10.3390/IECN2020-07009

Article Metrics

Back to TopTop