A Microbiota-Directed Food Intervention for Undernourished Children

Robert Y Chen, Ishita Mostafa, Matthew C Hibberd, Subhasish Das, Mustafa Mahfuz, Nurun N Naila, M Munirul Islam, Sayeeda Huq, M Ashraful Alam, Mahabub U Zaman, Arjun S Raman, Daniel Webber, Cyrus Zhou, Vinaik Sundaresan, Kazi Ahsan, Martin F Meier, Michael J Barratt, Tahmeed Ahmed, Jeffrey I Gordon, Robert Y Chen, Ishita Mostafa, Matthew C Hibberd, Subhasish Das, Mustafa Mahfuz, Nurun N Naila, M Munirul Islam, Sayeeda Huq, M Ashraful Alam, Mahabub U Zaman, Arjun S Raman, Daniel Webber, Cyrus Zhou, Vinaik Sundaresan, Kazi Ahsan, Martin F Meier, Michael J Barratt, Tahmeed Ahmed, Jeffrey I Gordon

Abstract

Background: More than 30 million children worldwide have moderate acute malnutrition. Current treatments have limited effectiveness, and much remains unknown about the pathogenesis of this condition. Children with moderate acute malnutrition have perturbed development of their gut microbiota.

Methods: In this study, we provided a microbiota-directed complementary food prototype (MDCF-2) or a ready-to-use supplementary food (RUSF) to 123 slum-dwelling Bangladeshi children with moderate acute malnutrition between the ages of 12 months and 18 months. The supplementation was given twice daily for 3 months, followed by 1 month of monitoring. We obtained weight-for-length, weight-for-age, and length-for-age z scores and mid-upper-arm circumference values at baseline and every 2 weeks during the intervention period and at 4 months. We compared the rate of change of these related phenotypes between baseline and 3 months and between baseline and 4 months. We also measured levels of 4977 proteins in plasma and 209 bacterial taxa in fecal samples.

Results: A total of 118 children (59 in each study group) completed the intervention. The rates of change in the weight-for-length and weight-for-age z scores are consistent with a benefit of MDCF-2 on growth over the course of the study, including the 1-month follow-up. Receipt of MDCF-2 was linked to the magnitude of change in levels of 70 plasma proteins and of 21 associated bacterial taxa that were positively correlated with the weight-for-length z score (P<0.001 for comparisons of both protein and bacterial taxa). These proteins included mediators of bone growth and neurodevelopment.

Conclusions: These findings provide support for MDCF-2 as a dietary supplement for young children with moderate acute malnutrition and provide insight into mechanisms by which this targeted manipulation of microbiota components may be linked to growth. (Supported by the Bill and Melinda Gates Foundation and the National Institutes of Health; ClinicalTrials.gov number, NCT04015999.).

Copyright © 2021 Massachusetts Medical Society.

Figures

Fig. 1:. Study design of a randomized…
Fig. 1:. Study design of a randomized controlled trial of MDCF-2 or RUSF supplementation in children with MAM.
(A) Study design. (B) WLZ during treatment. Best-fit linear regression lines are colored green (MDCF-2) or red (RUSF), and the lighter shaded areas around the lines indicate 95% confidence bands. The β-coefficient of +0.011 represents the weekly change in WLZ in children receiving MDCF-2 compared to those receiving RUSF. The 95% confidence interval is shown in parentheses.
Fig. 2:. Effects of Nutritional Intervention on…
Fig. 2:. Effects of Nutritional Intervention on Plasma Proteins.
(A-C) Schematic depicting the calculation of ‘β-WLZ’ for each participant (panel A), ‘Δprotein abundance’ for each participant (panel B) and the correlation between these two values (panel C). (D) Gene set enrichment analysis (GSEA) of proteins whose abundances were correlated with ponderal growth. The vertical gray line indicates q<0.05. (E-H) Gene Ontology (GO) terms enriched for by the set of WLZ-associated proteins. Shown are the correlation strengths between proteins belonging to a GO term and ponderal growth. Only proteins whose correlations with β-WLZ reached an unadjusted p<0.01 are shown. Proteins are ordered by correlation strength and colored by their p-value (transformed to a −log10 scale so that decreasing values indicate less statistical significance). (I) Differential effects of MDCF-2 and RUSF on WLZ-associated proteins. Proteins are ordered by the log2(fold-change) of the treatment effect of MDCF-2 over RUSF after three months of supplementation. GSEA was used to calculate the enrichment of proteins whose abundances were increased more by MDCF-2 compared to RUSF for the set of 70 proteins that are positively correlated with β-WLZ.
Fig. 3:. Response of the Gut Microbiota…
Fig. 3:. Response of the Gut Microbiota to MDCF-2 and RUSF Supplementation.
(A) Analytical scheme for linear mixed effects modeling of the relationship between WLZ and taxon (ASV) abundance during supplementation. The coefficient β1 represents the change in WLZ for a unit change in the variance-stabilizing, transformed abundance of an ASV. A random effect for participant ID (PID) was included in the model to account for repeated measurements taken from the same individual. Bar graphs indicate β1 (the linear model coefficients) ± SEM for each taxon that was significantly associated with WLZ. ASVs in bold-face were previously identified as ‘ecogroup’ taxa(5), while those with asterisks have previously described associations with weight gain in gnotobiotic mice harboring gut microbial communities obtained from healthy and undernourished children(10). (B)Ratio of 3-month ΔASV (the change in variance-stabilizing transformed ASV counts prior to and after supplementation) between MDCF-2 and RUSF treatment arms. A positive ratio indicates a greater average increase in children treated with MDCF-2. Color scheme: red bars, ASVs with significant positive associations with WLZ; blue bars, ASVs with significant negative associations with WLZ.

References

    1. Black R.E., Allen L.H., Bhutta Z.A., Caufield L.E., de Onis M., Ezzati M., Mathers C., Rivera Maternal J. and child undernutrition: global and regional exposures and health consequences. Lancet. 371, 243–260 (2008).
    1. Black R.E., Victora C.G., Walker S.P., Bhutta Z.A., Christian P., de Onis M., Ezzati M., Grantham-McGregor S., Katz J., Martorell R., Uauy R.. Maternal and child undernutrition and overweight in low-income and middle-income countries. Lancet. 382, 427‐451 (2013).
    1. WHO Multicentre Growth Reference Study Group. WHO Child Growth Standards: Growth velocity based on weight, length and head circumference: Methods and development. Geneva: World Health Organization; (2009).
    1. Subramanian S., Huq S., Yatsunenko T., Haque R., Mahfuz M., Alam M.A., Benezra A., Destefano J., Meier M.F., Muegge B.D., Barratt M.J., VanArendonk L.G., Zhang Q., Province M.A., Petri W.A., Ahmed T., Gordon J.I.. Persistent gut microbiota immaturity in malnourished Bangladeshi children. Nature 510, 417–421 (2014).
    1. Raman A.S., Gehrig J.L., Venkatesh S, Chang H.W., Hibberd M.C., Subramanian S., Kang G., Bessong P.O., Lima A.A.M., Kosek M.N.,Petri W.A., Rodionov D.A., Arzamasov A.A., Leyn S.A., Osterman A.L., Huq S., Mostafa I, Islam M., Mahfuz M., Haque R., Ahmed T., Barratt M.J., Gordon J.I.. A sparse covarying unit that describes healthy and impaired human gut microbiota development. Science 365, eaau4735 (2019).
    1. Dewey K.G.. Reducing stunting by improving maternal, infant and young child nutrition in regions such as South Asia: evidence, challenges and opportunities. Matern Child Nutr. 12, (Suppl 1) 27‐38 (2016).
    1. Goudet S.M., Bogin B.A., Madise N.J., Griffiths P.L.. Nutritional interventions for preventing stunting in children (birth to 59 months) living in urban slums in low- and middle-income countries (LMIC). Cochrane Database of Systematic Reviews, 6, Art. No. CD011695 (2019).
    1. Gehrig J.L., Venkatesh S., Chang H.W., Hibberd M.C., Kung V.L., Cheng J., Chen R.Y., Subramanian S., Cowardin C.A., Meier M., O’Donnell D., Talcott M., Spears L.D., Semenkovich C.C., Henrissat B., Giannone R.J., Hettich R.L., Ilkayeva O., Muehlbauer M., Newgard C.B., Sawyer C., Head R.D., Rodionov D.A., Arzamasov A.A., Leyn S.A., Osterman A.L., Hossain Md I., Islam M., Choudhury N., Sarker S.A., Huq S., Mahnud I., Mostafa I., Mahfuz M., Barratt M.J., Ahmed T., Gordon J.I.. Effects of microbiota-directed foods in gnotobiotic animals and undernourished children. Science 365, eaau4732 (2019).
    1. Roberton T., Carter E.D., Chou V.B., Stegmuller A.R., Jackson B.D., Tam Y. et al. Early estimates of the indirect effects of the COVID-19 pandemic on maternal and child mortality in low-income and middle-income countries: a modelling study. Lancet Global Health DOI:10.1016/S2214-109X(20)30229-1 (2020).
    1. Blanton L.V., Charbonneau M.R., Salih T., Barratt M.J., Venkatesh S., Ilkaveya O., Subramanian S., Manary M.J., Trehan I., Jorgensen J.M., Fan Y.M., Henrissat B., Leyn S.A., Rodionov D.A., Osterman A.L., Maleta K.M., Newgard C.B., Ashorn P.. Dewey K.G., Gordon J.I.. Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children. Science 351, aad3311 (2016)
    1. Mostafa I., Nahar N.N., Islam Md.M., Huq S., Mustafa M., Barratt M.J., Gordon J.I., Ahmed T.. Proof-of-concept study of the efficacy of a microbiota-directed complementary food formulation (MDCF) for treating moderate acute malnutrition. BMC Public Health 20, 242 (2020)
    1. Ritchie M.E., Phipson B., Wu D., Hu Y., Law C.W., Shi W., Smyth G.K.. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research 43 e47 (2015)
    1. Sergushichev A.. An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. bioRxiv (2016).
    1. Gold L., Ayers L.,D., Bertino J., Bock C., Bock A., Brody E.N., Carter J., Dalby A.B., Eaton B.E., Fitzwater T., Flather D., Forbes A., Foreman T., Fowler C., Gawande B., Goss M., Gunn M., Gupta S., Halladay D., Heil J., Heilig J., Hicke B., Husar G., Janjic N., Harvis T., Jennings S., Katilius E., Kenney T.R., Kim N.. Koch T.H., Kraemer S., Kroiss L., Le N., Levine D., Lindsey W., Lollo B., Mayfield W., Mehan M., Mehler R., Nelson S.K., Nelson M., Nieuwlandt D., Nikrad M., Ochsner U., Ostroff R.M., Otis M., Parker T., Pietrasiewicz S., Resnicow D.L., Rohloff J., Sanders G., Sattin S., Schneider D., Singer B., Stanton M., Sterkel A., Stewart A., Stratford S., Vaught J.D., Vrkljan M., Walker J.J., Watrobka M., Waugh S., Weiss A., Wilcox S., Wolfson A., Wolk S., Zhang C., Zichi D.. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS One 5 (2010).
    1. Burger A., Roosenboom J., Hossain M., Weinberg S.M., Hecht J.T., Posey K.L.. Mutant COMP shapes growth and development of skull and facial structures in mice and humans. Mol. Genet. Genomic Med. 1–8 (2020).
    1. Bjarnason R., Andersson B., Kim H.S., Olsson B., Swolin-Eide D., Wickelgren R., Kriström B., Carlsson B., Albertsson-Wikland K., Carlsson L.M.S.. Cartilage oligomeric matrix protein increases in serum after the start of growth hormone treatment in prepubertal children. J. Clin. Endocr. Metab. 89, 5156–5160 (2004).
    1. Chen K., Ng P.Y., Chen R., Hu D., Berry S., Baron R., Gori F., Sfrp4 repression of the Ror2/Jnk cascade in osteoclasts protects cortical bone from excessive endosteal resorption. Proc.. Natl. Acad.Sci. U.S.A. 116, 14138–14143 (2019).
    1. Kim H., Choi Y.J., Lee Y.S., Park S.Y., Baek J.E., Kim H.K., Kim B.J., Lee S.H., Koh J.M.. SLIT3 regulates endochondral ossification by β-catenin suppression in chondrocytes. Biochem. Biophys. Res. Comm. 506, 847–853 (2018).
    1. Diamond M.S., Farzan M.. The broad-spectrum antiviral functions of IFIT and IFITM proteins. Nature Rev. Immunol. 13, 46–57 (2013).
    1. Bernardo B.C., Belluoccio D., Rowley L., Little C.B., Hansen U, Bateman J.F.. Cartilage intermediate layer protein 2 (CILP-2) is expressed in articular and meniscal cartilage and down-regulated in experimental osteoarthritis. J.Biol. Chem. 286, 37758–37767 (2011).
    1. Stenina-adognravi O., Plow E.F.. Thrombospondin-4 in tissue remodeling. Matrix Biol. 75–76, 300–313 (2019).
    1. Sawaya A.L., Martins P., Hoffman D., Roberts S.B.. The Link Between Childhood Undernutrition and Risk of Chronic Diseases in Adulthood : A Case Study of Brazil. Nutr. Rev. 61, 168–175 (2003).
    1. Popkin B.M., Corvalan C., Grummer-Strawn L.M., Dynamics of the double burden of malnutrition and the changing nutrition reality. Lancet 395, 65‐74 (2020).
    1. Wu T., Zhang Q., Wu S., Hu W., Zhou T., Li K., Liu D.. CILP- 2 is a novel secreted protein and associated with insulin resistance. J. Mol. Cell. Biol. 11, 1083–1094 (2019).
    1. Willer C.J., Sanna S., Jackson A.U., Scuteri A., Bonnycastle L.L., Clarke R., Heath S.C., Timpson N.J., Najjar S.S., Stringham H.M., Strait J., Duren W.L., Maschio A., Busonero F., Mulas A., Albai G., Swift A.J., Morken M.A., Narisu N., Mohlke K.L.. Newly identified loci that influence lipid concentrations and risk of coronary artery disease. Nature Genet. 40, 161–169 (2008).
    1. Saxena R., Elbers C.C., Guo Y., Peter I.,Gaunt T.R., Mega J.L., Lanktree M.B., Tare A., Castillo B.A., Li Y.R., Johnson T., Bruinenberg M., Gilbert-Diamond D., Rajagopalan R., Voight B.F., Balasubramanyan A., Barnard J., Braund P.S., Bauer F., Vliet-ostaptchouk J.V. Van. Large-Scale Gene-Centric Meta-Analysis across 39 Studies Identifies Type 2 Diabetes Loci. Am. J. Hum. Genet. 410–425 (2012).
    1. Rzehak P., Covic M., Saffery R., Reischl E., Wahl S., Grote V., Weber M., Xhonneux A., Langhendries J., Ferre N., Escribano J., Verduci E., Riva E., Socha P., DNA-Methylation and Body Composition in Preschool Children : Epigenome-Wide-Analysis in the European Childhood Obesity Project (CHOP) Study. Sci. Rep. 1–13 (2017).
    1. Brix J.M., Krzizek E.C., Hoebaus C., Ludvik B., Schernthaner G., Schernthaner G.H.. Secreted Frizzled-Related Protein 4 (SFRP4) is Elevated in Patients with Diabetes Mellitus. Horm. Metab. Res. 48, 345–348 (2016).
    1. Hoffmann M.M., Werner C., Böhm M., Laufs U., Winkler K.. Association of secreted frizzled-related protein 4 (SFRP4) with type 2 diabetes in patients with stable coronary artery disease. Cardio. Diabetol. 13, 1–8 (2014).
    1. Zierfuss B., Hobaus C., Hertz C.T., Pesau G., Koppensteiner R., Schernthaner G-H.. Thrombospondin-4 indcreases with the severity of peripheral arterial disease and is associated with diabetes. Heart and Vessels. 35, 52–58 (2020)
    1. Conlisk A.J., Barnhart H.X., Martorell R., Grajeda R., Stein A.D.. Maternal and Child Nutritional Supplementation Are Inversely Associated with Fasting Plasma Glucose Concentration in Young Guatemalan Adults. J. Nutr. 134, 890–897 (2004).
    1. Kinra S., Rameshwar Sarma K.V., Ghafoorunissa V. V. Mendu R., Ravikumar R., Mohan V., Wilkinson I.B., Cockcroft J.R. J. R., Smith G.D.. Ben-Shlomo Y., Effect of integration of supplemental nutrition with public health programmes in pregnancy and early childhood on cardiovascular risk in rural Indian adolescents: Long term follow-up of Hyderabad nutrition trial. BMJ. 337, 445–449 (2008).
    1. Abera M., Tesfaye M., Admassu B., Hanlon C., Ritz C., Wibaek R., Michaelsen K.F., Friis H., Wells J.C., Andersen G.S., Girma R., Kaestel P.. Body composition during early infancy and developmental progression from 1 to 5 years of age: The Infant Anthropometry and Body Composition (iABC) cohort study among Ethiopian children. Br. J. Nutr. 119,1263–1273 (2018).
    1. Olsen M.F., Iuel-Brockdorff A.S., Yaméogo C.W., Cichon B., Fabiansen C., Filteau S., Phelan K., Ouédraogo A., Wells J.C., Briend A., Michaelsen K.F., Lauritzen L., Ritz C., Ashorn P., Christensen V.B., Gladstone M., Friss H.. Early development in children with moderate acute malnutrition: A cross-sectional study in Burkina Faso. Mat. Child Nutr. 16, 1–14 (2020).

Source: PubMed

3
Abonnere