Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children

Microbiota and infant development Malnutrition in children is a persistent challenge that is not always remedied by improvements in nutrition. This is because a characteristic community of gut microbes seems to mediate some of the pathology. Human gut microbes can be transplanted effectively into germ-free mice to recapitulate their associated phenotypes. Using this model, Blanton et al. found that the microbiota of healthy children relieved the harmful effects on growth caused by the microbiota of malnourished children. In infant mammals, chronic undernutrition results in growth hormone resistance and stunting. In mice, Schwarzer et al. showed that strains of Lactobacillus plantarum in the gut microbiota sustained growth hormone activity via signaling pathways in the liver, thus overcoming growth hormone resistance. Together these studies reveal that specific beneficial microbes could potentially be exploited to resolve undernutrition syndromes. Science, this issue p. 10.1126/science.aad3311, p. 854 Microbes from healthy children protect mice from the detrimental effects of the microbiota of malnourished infants. INTRODUCTION As we come to appreciate how our microbial communities (microbiota) assemble following birth, there is an opportunity to determine how this facet of our developmental biology relates to the healthy or impaired growth of infants and children. Childhood undernutrition is a devastating global health problem whose long-term sequelae, including stunting, neurodevelopmental abnormalities, and immune dysfunction, remain largely refractory to current therapeutic interventions. RATIONALE To test the hypothesis that perturbations in the normal development of the gut microbiota are causally related to undernutrition, we first applied random forests (RF), a machine learning method, to bacterial 16S ribosomal RNA data sets generated from fecal samples that were collected serially from healthy Malawian infants and children during their first 3 postnatal years. Age-discriminatory bacterial taxa were identified with distinctive time-dependent changes in their relative abundances; they were used to construct a sparse RF-derived model describing a program of normal postnatal gut microbiota development that is shared across biologically unrelated individuals. A metric based on this model (microbiota-for-age Z-score) was used to define the state of development (maturation) of fecal microbiota from infants and children with varying degrees of undernutrition. Fecal samples obtained from 6- and 18-month-old children with healthy growth patterns or with varying degrees of undernutrition were transplanted into young germ-free mice that were fed a representative Malawian diet. The recipient animals’ rate of lean body mass gain was characterized by serial quantitative magnetic resonance, their metabolic phenotypes were determined by targeted mass spectrometry, and their femoral bone morphologic features were delineated by microcomputed tomography. RESULTS Undernourished children in the Malawian birth cohort that we studied have immature gut microbiota. Unlike microbiota from healthy children, immature microbiota transmit impaired growth, altered bone morphology, and metabolic abnormalities in the muscle, liver, and brain to recipient gnotobiotic mice. The representation of several age-discriminatory taxa in the transplanted microbiota harbored by recipient animals correlated with their growth rates. Microbiota from 6-month-old infants produced greater effects on growth than did microbiota from 18-month-old children, although in each age bin, the growth effects produced by a healthy donor’s community were greater than those produced by an undernourished donor’s community. Cohousing coprophagic mice shortly after they received microbiota from healthy or severely stunted and underweight 6-month-old infants resulted in the invasion of age- and growth-discriminatory taxa from the former into the latter microbiota in the recipient animals, with associated prevention of growth impairments. Introducing cultured members from this group of invasive species ameliorated growth and metabolic abnormalities in recipients of microbiota from undernourished donors. CONCLUSION These preclinical findings provide evidence that gut microbiota immaturity is causally related to childhood undernutrition. The age- and growth-discriminatory taxa that we identified should help direct studies of the effects of host and environmental factors on gut microbial community development, and they represent therapeutic targets for repairing or preventing gut microbiota immaturity. Preclinical evidence that gut microbiota immaturity is causally related to childhood undernutrition. (A) A model of normal gut microbial community development in Malawian infants and children, based on the relative abundances of 25 bacterial taxa that provide a microbial signature defining the “age,” or state of maturation, of an individual’s (fecal) microbiota. (Hierarchical clusterings of operational taxonomic units are indicated on the left.) (B) Fecal samples from healthy (H) or stunted and underweight (Un) infants and children were transplanted into separate groups of young germ-free mice that were fed a Malawian diet. The immature microbiota of Un donors transmitted impaired growth phenotypes to the mice. (C) Evidence that a subset of age-discriminatory taxa are also growth-discriminatory. Cohousing mice shortly after they received microbiota from 6-month-old healthy or undernourished donors resulted in the invasion of taxa from the healthy donor’s microbiota (HCH) into the undernourished donor’s microbiota (UnCH) among recipient animals and prevented growth impairments. Adding cultured invasive growth-discriminatory taxa directly to the Un donor’s microbiota (Un+) improved growth. Undernourished children exhibit impaired development of their gut microbiota. Transplanting microbiota from 6- and 18-month-old healthy or undernourished Malawian donors into young germ-free mice that were fed a Malawian diet revealed that immature microbiota from undernourished infants and children transmit impaired growth phenotypes. The representation of several age-discriminatory taxa in recipient animals correlated with lean body mass gain; liver, muscle, and brain metabolism; and bone morphology. Mice were cohoused shortly after receiving microbiota from healthy or severely stunted and underweight infants; age- and growth-discriminatory taxa from the microbiota of the former were able to invade that of the latter, which prevented growth impairments in recipient animals. Adding two invasive species, Ruminococcus gnavus and Clostridium symbiosum, to the microbiota from undernourished donors also ameliorated growth and metabolic abnormalities in recipient animals. These results provide evidence that microbiota immaturity is causally related to undernutrition and reveal potential therapeutic targets and agents.

[1]  Katherine H. Huang,et al.  Detection of low-abundance bacterial strains in metagenomic datasets by eigengenome partitioning , 2015, Nature Biotechnology.

[2]  J. Berkley,et al.  Environmental Enteric Dysfunction: An Overview , 2015, Food and nutrition bulletin.

[3]  Tanya Yatsunenko,et al.  Functional characterization of IgA-targeted bacterial taxa from undernourished Malawian children that produce diet-dependent enteropathy , 2015, Science Translational Medicine.

[4]  Y. Cheung,et al.  The impact of lipid-based nutrient supplement provision to pregnant women on newborn size in rural Malawi : a randomized controlled trial 1 – 4 , 2015 .

[5]  Mark A. Miller,et al.  Modeling environmental influences on child growth in the MAL-ED cohort study: opportunities and challenges. , 2014, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[6]  I. Rosenberg,et al.  Environmental enteric dysfunction: pathogenesis, diagnosis, and clinical consequences. , 2014, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[7]  Torsten Seemann,et al.  Prokka: rapid prokaryotic genome annotation , 2014, Bioinform..

[8]  Qunyuan Zhang,et al.  Persistent Gut Microbiota Immaturity in Malnourished Bangladeshi Children , 2014, Nature.

[9]  A. Schacht,et al.  Effects of malnutrition on children's immunity to bacterial antigens in Northern Senegal. , 2014, The American journal of tropical medicine and hygiene.

[10]  G. Fitzmaurice,et al.  Impaired IQ and academic skills in adults who experienced moderate to severe infantile malnutrition: A 40-year study , 2014, Nutritional neuroscience.

[11]  Fangfang Xia,et al.  The SEED and the Rapid Annotation of microbial genomes using Subsystems Technology (RAST) , 2013, Nucleic Acids Res..

[12]  Adam Godzik,et al.  Polysaccharides utilization in human gut bacterium Bacteroides thetaiotaomicron: comparative genomics reconstruction of metabolic and regulatory networks , 2013, BMC Genomics.

[13]  William J. Riehl,et al.  RegPrecise 3.0 – A resource for genome-scale exploration of transcriptional regulation in bacteria , 2013, BMC Genomics.

[14]  K. Dewey The Challenge of Meeting Nutrient Needs of Infants and Young Children during the Period of Complementary Feeding: An Evolutionary Perspective , 2013, The Journal of nutrition.

[15]  J. Clemente,et al.  Gut Microbiota from Twins Discordant for Obesity Modulate Metabolism in Mice , 2013, Science.

[16]  Karsten Zengler,et al.  Transcriptional regulation of the carbohydrate utilization network in Thermotoga maritima , 2013, Front. Microbiol..

[17]  R. Martorell,et al.  Maternal and child undernutrition and overweight in low-income and middle-income countries , 2013, The Lancet.

[18]  Mark A. Miller,et al.  Fecal Markers of Intestinal Inflammation and Permeability Associated with the Subsequent Acquisition of Linear Growth Deficits in Infants , 2013, The American journal of tropical medicine and hygiene.

[19]  L. Ursell,et al.  Gut Microbiomes of Malawian Twin Pairs Discordant for Kwashiorkor , 2013, Science.

[20]  V. Tremaroli,et al.  The gut microbiota regulates bone mass in mice , 2012, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[21]  Peter Williams,et al.  IMG: the integrated microbial genomes database and comparative analysis system , 2011, Nucleic Acids Res..

[22]  S. Salzberg,et al.  FLASH: fast length adjustment of short reads to improve genome assemblies , 2011, Bioinform..

[23]  Rob Knight,et al.  Bayesian community-wide culture-independent microbial source tracking , 2011, Nature Methods.

[24]  J. Faith,et al.  Extensive personal human gut microbiota culture collections characterized and manipulated in gnotobiotic mice , 2011, Proceedings of the National Academy of Sciences.

[25]  Robert C. Edgar,et al.  Search and clustering orders of magnitude faster than BLAST , 2010, Bioinform..

[26]  R. Overbeek,et al.  Genomic encyclopedia of sugar utilization pathways in the Shewanella genus , 2010, BMC Genomics.

[27]  C. Hoppel,et al.  Novel isolation procedure for short-, medium-, and long-chain acyl-coenzyme A esters from tissue. , 2008, Analytical biochemistry.

[28]  Elias Chaibub Neto,et al.  Genetic Networks of Liver Metabolism Revealed by Integration of Metabolic and Transcriptional Profiling , 2008, PLoS genetics.

[29]  K. Dewey,et al.  Systematic review of the efficacy and effectiveness of complementary feeding interventions in developing countries. , 2008, Maternal & child nutrition.

[30]  Linda Richter,et al.  Maternal and child undernutrition: consequences for adult health and human capital , 2008, The Lancet.

[31]  Dmitry A Rodionov,et al.  Comparative genomic reconstruction of transcriptional regulatory networks in bacteria. , 2007, Chemical reviews.

[32]  J. Tiedje,et al.  Naïve Bayesian Classifier for Rapid Assignment of rRNA Sequences into the New Bacterial Taxonomy , 2007, Applied and Environmental Microbiology.

[33]  A. Alavi,et al.  Opportunities and Challenges , 1998, In Vitro Diagnostic Industry in China.

[34]  P. Mahadevan,et al.  An overview , 2007, Journal of Biosciences.

[35]  M. Jensen,et al.  Compensatory responses to pyruvate carboxylase suppression in islet beta-cells. Preservation of glucose-stimulated insulin secretion. , 2006, The Journal of biological chemistry.

[36]  A. Sanabria,et al.  Randomized controlled trial. , 2005, World journal of surgery.

[37]  A. Merrill,et al.  Sphingolipidomics: high-throughput, structure-specific, and quantitative analysis of sphingolipids by liquid chromatography tandem mass spectrometry. , 2005, Methods.

[38]  T. Pieber,et al.  LC/MS/MS method for quantitative determination of long-chain fatty acyl-CoAs. , 2005, Analytical chemistry.

[39]  T. Wetter,et al.  Using the miraEST assembler for reliable and automated mRNA transcript assembly and SNP detection in sequenced ESTs. , 2004, Genome research.

[40]  C. Victora,et al.  The WHO Multicentre Growth Reference Study: planning, study design, and methodology. , 2004, Food and nutrition bulletin.

[41]  David Millington,et al.  Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-animal insulin resistance , 2004, Nature Medicine.

[42]  Reynaldo Martorell,et al.  Measurement and standardization protocols for anthropometry used in the construction of a new international growth reference. , 2004, Food and nutrition bulletin.

[43]  D. Briggs,et al.  An Evolutionary Perspective , 2004, J. Decis. Syst..

[44]  Leo Breiman,et al.  Random Forests , 2001, Machine Learning.

[45]  S. Rapoport,et al.  Isolation and quantitation of long-chain acyl-coenzyme A esters in brain tissue by solid-phase extraction. , 1994, Analytical biochemistry.