The therapeutic efficacy of Bifidobacterium animalis subsp. lactis BB-12® in infant colic: A randomised, double blind, placebo-controlled trial

Rita Nocerino, Francesca De Filippis, Gaetano Cecere, Antonio Marino, Maria Micillo, Carmen Di Scala, Carmen de Caro, Antonio Calignano, Cristina Bruno, Lorella Paparo, Anna M Iannicelli, Linda Cosenza, Ylenia Maddalena, Giusy Della Gatta, Serena Coppola, Laura Carucci, Danilo Ercolini, Roberto Berni Canani, Rita Nocerino, Francesca De Filippis, Gaetano Cecere, Antonio Marino, Maria Micillo, Carmen Di Scala, Carmen de Caro, Antonio Calignano, Cristina Bruno, Lorella Paparo, Anna M Iannicelli, Linda Cosenza, Ylenia Maddalena, Giusy Della Gatta, Serena Coppola, Laura Carucci, Danilo Ercolini, Roberto Berni Canani

Abstract

Background: The pathogenesis of infant colic is poorly defined. Gut microbiota seems to be involved, supporting the potential therapeutic role of probiotics.

Aims: To assess the rate of infants with a reduction of ≥50% of mean daily crying duration after 28 days of intervention with the probiotic Bifidobacterium animalis subsp. lactis BB-12® (BB-12). Secondary outcomes were daily number of crying episodes, sleeping time, number of bowel movements and stool consistency.

Methods: Randomized controlled trial (RCT) on otherwise healthy exclusively breastfed infants with infant colic randomly allocated to receive BB-12 (1 × 109 CFU/day) or placebo for 28 days. Gut microbiota structure and butyrate, beta-defensin-2 (HBD-2), cathelicidin (LL-37), secretory IgA (sIgA) and faecal calprotectin levels were assessed.

Results: Eighty infants were randomised, 40/group. The rate of infants with reduction of ≥50% of mean daily crying duration was higher in infants treated with BB-12, starting from the end of 2nd week. No infant relapsed when treatment was stopped. The mean number of crying episodes decreased in both groups, but with a higher effect in BB-12 group (-4.7 ± 3.4 vs -2.3 ± 2.2, P < 0.05). Mean daily stool frequency decreased in both groups but the effect was significantly higher in the BB-12 group; stool consistency was similar between the two groups. An increase in Bifidobacterium abundance (with significant correlation with crying time reduction), butyrate and HBD-2, LL-37, sIgA levels associated with a decrease in faecal calprotectin level were observed in the BB-12 group.

Conclusions: Supplementation with BB-12 is effective in managing infant colic. The effect could derive from immune and non-immune mechanisms associated with a modulation of gut microbiota structure and function.

© 2019 The Authors. Alimentary Pharmacology & Therapeutics Published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Panel (A). The design of the study. Panel (B). The flow of subjects during the phases of the study
Figure 2
Figure 2
Panel 1. The results of the main study outcome (ITT analysis): the rate of infants with reduction of ≥50% of duration of crying after 28 days of treatment. Eighty percent of the BB‐12 group and 32.5% of the placebo group showed a ≥50% reduction in crying duration after 28 days of treatment. The between‐group difference was significantly in favour of BB‐12 and the asterisk indicates a significant difference (* = BB‐12 vs placebo, P < 0.0001). Panel 2. The mean number of crying episodes during the week before treatment (V0‐V1, blue bars) and during the last week of treatment (V4‐V5, light blue bars) in infants enrolled in the two study groups. Values are expressed as mean and SD and symbols indicate a significant difference (* = BB‐12 V0‐V1 vs BB‐12 V4‐V5, P < 0.05; ** = BB‐12 V4‐V5 vs Placebo V4‐V5, P < 0.05; ° = Placebo V0‐V1 vs Placebo V4‐V5, P < 0.05)
Figure 3
Figure 3
Panel 1. The values of innate and acquired immunity biomarkers, calprotectin and butyrate faecal levels at baseline during the week before treatment (V0‐V1, blue bars) and during the last week of treatment (V4‐V5, light blue bars) in the two study groups. Panel A: human β‐defensin 2; panel B: cathelecidin (LL‐37); panel C: secretory IgA; panel D: butyrate; panel E: calprotectin. Values are expressed as mean and SD and asterisks indicate a significant difference (* = P < 0.05). Panel 2. The k‐means clustering (k = 2) of subjects based on the variation (V5‐V1) of the crying time, beta‐defensin 2, LL‐37, sIgA, butyrate, faecal calprotectin levels after 28 days of treatment. Cluster 1 (yellow dot) included 10% of infants enrolled in the BB‐12 group and 67% of infants enrolled in the placebo group. Whereas, Cluster 2 (blue dot) included 90% and 33% of BB‐12 and placebo subjects respectively. Panel 3. The boxplots showing the variation (V5‐V1) of crying time (in minutes), beta‐defensin 2, LL‐37, sIgA, butyrate and faecal calprotectin levels in subjects classified in Cluster 1 or 2. Boxes represent the interquartile range (IQR) between the first and third quartiles, and the line inside represents the median (2nd quartile). Whiskers denote the lowest and the highest values within 1.5 × IQR from the first and third quartiles respectively. Asterisks indicate a significant difference as obtained by pairwise Wilcoxon test (P < 0.05)
Figure 4
Figure 4
Linear regression of the duration of crying in minutes (V5‐V1) as a function of the abundance of Bifidobacterium (V5‐V1) (P < 0.05)

References

    1. Wolke D, Bilgin A, Samara M. Systematic review and meta‐analysis: fussing and crying durations and prevalence of colic in infants. J Pediatr. 2017;185:e4.
    1. James‐Roberts IS. In Barr RG, St James‐Roberts I, Keefe MR, eds. New Evidence on Unexplained Early Infant Crying: Its Origins, Nature and Management. Skillman, New Jersey: Johnson & Johnson Pediatric Institute; 2001:5‐24.
    1. Johnson JD, Cocker K, Chang E. Infantile colic: recognition and treatment. Am Fam Physician. 2015;92:577‐582.
    1. Zeevenhooven J, Browne PD, L'Hoir MP, de Weerth C, Benninga MA. Infant colic: mechanisms and management. Nat Rev Gastroenterol Hepatol. 2018;15:479‐496.
    1. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest. 2015;125:926‐938.
    1. Indrio F, Dargenio VN, Giordano P, Francavilla R. Preventing and treating colic. Adv Exp Med Biol. 2019;1125:49‐56. 10.1007/5584_2018_315
    1. Jungersen M, Wind A, Johansen E, Christensen JE, Stuer‐Lauridsen B, Eskesen D. The science behind the probiotic strain bifidobacterium animalis subsp. lactis BB‐12®. Microorganisms. 2014;2:92‐110.
    1. Xinias I, Analitis A, Mavroudi A, et al. Innovative dietary intervention answers to baby colic. Pediatr Gastroenterol Hepatol Nutr. 2017;20:100‐106.
    1. Bellaïche M, Levy M, Jung C. Treatments for infant colic. J Pediatr Gastroenterol Nutr. 2013;57:S27‐S30.
    1. Hyman PE, Milla PJ, Benninga MA, Davidson GP, Fleisher DF, Taminiau J. Childhood functional gastrointestinal disorders: neonate/toddler. Gastroenterology. 2006;130:1519‐1526.
    1. Heaton KW, Radvan J, Cripps H, Mountford RA, Braddon FE, Hughes AO. Defecation frequency and timing, and stool form in the general population: a prospective study. Gut. 1992;33:818‐824.
    1. Nocerino R, Paparo L, Terrin G, et al. Cow's milk and rice fermented with Lactobacillus paracasei CBA L74 prevent infectious diseases in children: a randomized controlled trial. Clin Nutr. 2017;36:118‐125.
    1. Berni Canani R, Terrin G, Rapacciuolo L, et al. Faecal calprotectin as reliable non‐invasive marker to assess the severity of mucosal inflammation in children with inflammatory bowel disease. Dig Liver Dis. 2008;40:547‐553.
    1. Berni Canani R, Sangwan N, Stefka AT, et al. Lactobacillus rhamnosus GG‐supplemented formula expands butyrate‐producing bacterial strains in food allergic infants. ISME J. 2016;10:742‐750.
    1. Berni Canani R, De Filippis F, Nocerino R, et al. Specific signatures of the gut microbiota and increased levels of butyrate in children treated with fermented cow's milk containing heat‐killed Lactobacillus paracasei CBA L74. Appl Environ Microbiol. 2017;83:e01206‐e1217.
    1. Magoč T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics. 2011;27:2957‐2963.
    1. Schmieder R, Edwards R. Quality control and preprocessing of metagenomic datasets. Bioinformatics. 2011;27:863‐864.
    1. Caporaso JG, Kuczynski J, Stombaugh J, et al. QIIME allows analysis of high‐throughput community sequencing data. Nat Methods. 2010;7:335‐336.
    1. Szajewska H, Gyrczuk E, Horvath A. Lactobacillus reuteri DSM 17938 for the management of infantile colic in breastfed infants: a randomized, double‐blind, placebo‐controlled trial. J Pediatr. 2013;162:257‐262.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B Stat Methodol. 1995;57:289‐300.
    1. Sung V, Cabana MD, D'Amico F, et al. Lactobacillus reuteri DSM 17938 for managing infant colic: protocol for an individual participant data meta‐analysis. BMJ Open. 2014;4:e006475.
    1. Sung V, D'Amico F, Cabana MD, et al. Lactobacillus reuteri to treat infant colic: a meta‐analysis. Pediatrics. 2018;141:e20171811 10.1542/peds.2017-1811.
    1. Dryl R, Szajewska H. Probiotics for management of infantile colic: a systematic review of randomized controlled trials. Arch Med Sci. 2018;14:1137‐1143. 10.5114/aoms.2017.66055.
    1. Gordon M, Biagioli E, Sorrenti M, et al. Dietary modifications for infantile colic. Cochrane Database Syst Rev. 2018;10:CD011029.
    1. Pitkala KH, Strandberg TE, Finne Soveri UH, Ouwehand AC, Poussa T, Salminen S. Fermented cereal with specific bifidobacteria normalizes bowel movements in elderly nursing home residents. A randomized, controlled trial. J Nutr Health Aging. 2007;11:305‐311.
    1. Uchida K, Akashi K, Kusunoki I, et al. Effect of fermented milk containing Bifidobacterium lactis BB‐12® on stool frequency, defecation, fecal microbiota and safety of excessive ingestion in healthy female students. J Nutr Food. 2005;8:39‐51.
    1. Matsumoto M, Imai T, Hironaka T, Kume H, Watanabe M, Benno Y. Effect of yoghurt with Bifidobacterium lactis LKM512 in improving fecal microflora and defecation of healthy volunteers. Intest Microbiol Mag. 2001;14:97‐102.
    1. Kolaček S, Hojsak I, Berni Canani R, et al. Commercial probiotic products: a call for improved quality control. A Position Paper by the ESPGHAN Working Group for Probiotics and Prebiotics. ESPGHAN Working Group for Probiotics and Prebiotics. J Pediatr Gastroenterol Nutr. 2017;65:117‐124.
    1. Lopez P, Gueimonde M, Margolles A, Suarez A. Distinct Bifidobacterium strains drive different immune responses in vitro. Int J Food Microbiol. 2010;138:157‐165.
    1. Latvala S, Pietilä TE, Veckman V, et al. Potentially probiotic bacteria induce efficient maturation but differential cytokine production in human monocyte‐derived dendritic cells. World J Gastroenterol. 2008;14:5570‐5581.
    1. Savino F, Quartieri A, De Marco A, et al. Comparison of formula‐fed infants with and without colic revealed significant differences in total bacteria, Enterobacteriaceae and faecal ammonia. Acta Paediatr. 2017;106:573‐578.
    1. Savino F, Cordisco L, Tarasco V, Calabrese R, Palumeri E, Matteuzzi D. Molecular identification of coliform bacteria from colicky breastfed infants. Acta Paediatr. 2009;98:1582‐1588.
    1. de Weerth C, Fuentes S, Puylaert P, de Vos WM. Intestinal microbiota of infants with colic: development and specific signatures. Pediatrics. 2013;131:e550‐e558.
    1. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol. 2019;. 10.1038/s41575-019-0173-3.
    1. Berni Canani R, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol. 2011;17:1519‐1528.
    1. Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF. The neuropharmacology of butyrate: the bread and butter of the microbiota‐gut‐brain axis? Neurochem Int. 2016;99:110‐132. 10.1016/j.neuint.2016.06.011.
    1. Dinan TG, Cryan JF. The microbiome‐gut‐brain axis in health and disease. Gastroenterol Clin North Am. 2017;46:77‐89. 10.1016/j.gtc.2016.09.007.
    1. Bienenstock J, Kunze W, Forsythe P. Microbiota and the gut‐brain axis. Nutr Rev. 2015;73:28‐31.
    1. van de Wouw M, Boehme M, Lyte JM, et al. Short‐chain fatty acids: microbial metabolites that alleviate stress‐induced brain‐gut axis alterations. J Physiol. 2018;596:4923‐4944.
    1. Ejtahed HS, Hasani‐Ranjbar S. Neuromodulatory effect of microbiome on gut‐brain axis; new target for obesity drugs. J Diabetes Metab Disord. 2019;18:263‐265.
    1. Russo R, Cristiano C, Avagliano C, et al. Gut‐brain axis: Role of lipids in the regulation of inflammation, pain and CNS diseases. Curr Med Chem. 2018;25:3930‐3952.
    1. Mittal R, Debs LH, Patel AP, et al. Butyrate, neuroepigenetics and the gut microbiome: can a high fiber diet improve brain health? J Cell Physiol. 2017;232:2359‐2372.
    1. Bourassa MW, Alim I, Bultman SJ, Ratan RR. Butyrate, neuroepigenetics and the gut microbiome: can a high fiber diet improve brain health? Neurosci Lett. 2016;20:56‐63.
    1. Gill PA, van Zelm MC, Muir JG, Gibson PR. Review article: short chain fatty acids as potential therapeutic agents in human gastrointestinaland inflammatory disorders. Aliment Pharmacol Ther. 2018;48:15‐34.
    1. Russo R, De Caro C, Avagliano C, et al. Sodium butyrate and its synthetic amide derivative modulate nociceptive behaviors in mice. Pharmacol Res. 2016;103:279‐291.
    1. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short‐chain fatty acids in microbiota‐gut‐brain communication. Nat Rev Gastroenterol Hepatol. 2019;16:461‐478.
    1. Rhoads JM, Fatheree NY, Norori J, et al. Altered fecal microflora and increased fecal calprotectin in infants with colic. J Pediatr. 2009;155:e1.
    1. Iacovou M, Craig SS, Yelland GW, Barrett JS, Gibson PR, Muir JG. Randomised clinical trial: reducing the intake of dietary FODMAPs of breastfeeding mothers is associated with a greater improvement of the symptoms of infantile colic than for a typical diet. Aliment Pharmacol Ther. 2018;48:1061‐1073.

Source: PubMed

3
S'abonner