Lactobacillus rhamnosus HN001 decreases the severity of necrotizing enterocolitis in neonatal mice and preterm piglets: evidence in mice for a role of TLR9

M Good, CP Sodhi, JA Ozolek… - American Journal …, 2014 - journals.physiology.org
M Good, CP Sodhi, JA Ozolek, RH Buck, KC Goehring, DL Thomas, A Vikram, K Bibby
American Journal of Physiology-Gastrointestinal and Liver …, 2014journals.physiology.org
Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in
premature infants and develops partly from an exaggerated intestinal epithelial immune
response to indigenous microbes. There has been interest in administering probiotic
bacteria to reduce NEC severity, yet concerns exist regarding infection risk. Mechanisms of
probiotic activity in NEC are unknown although activation of the microbial DNA receptor Toll-
like receptor-9 (TLR9) has been postulated. We now hypothesize that the Gram-positive …
Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants and develops partly from an exaggerated intestinal epithelial immune response to indigenous microbes. There has been interest in administering probiotic bacteria to reduce NEC severity, yet concerns exist regarding infection risk. Mechanisms of probiotic activity in NEC are unknown although activation of the microbial DNA receptor Toll-like receptor-9 (TLR9) has been postulated. We now hypothesize that the Gram-positive bacterium Lactobacillus rhamnosus HN001 can attenuate NEC in small and large animal models, that its microbial DNA is sufficient for its protective effects, and that protection requires activation of the Toll-like receptor 9 (TLR9). We now show that oral administration of live or UV-inactivated Lactobacillus rhamnosus HN001 attenuates NEC severity in newborn mice and premature piglets, as manifest by reduced histology score, attenuation of mucosal cytokine response, and improved gross morphology. TLR9 was required for Lactobacillus rhamnosus-mediated protection against NEC in mice, as the selective decrease of TLR9 from the intestinal epithelium reversed its protective effects. Strikingly, DNA of Lactobacillus rhamnosus HN001 reduced the extent of proinflammatory signaling in cultured enterocytes and in samples of resected human ileum ex vivo, suggesting the therapeutic potential of this probiotic in clinical NEC. Taken together, these findings illustrate that Lactobacillus rhamnosus HN001 is an effective probiotic for NEC via activation of the innate immune receptor TLR9 and that Lactobacillus rhamnosus DNA is sufficient for its protective effects, potentially reducing concerns regarding the infectious risk of this novel therapeutic approach.
American Physiological Society