Lactobacillus johnsonii N6.2 Modulates the Host Immune Responses: A Double-Blind, Randomized Trial in Healthy Adults

Guillermo E Marcial, Amanda L Ford, Michael J Haller, Salvador A Gezan, Natalie A Harrison, Dan Cai, Julie L Meyer, Daniel J Perry, Mark A Atkinson, Clive H Wasserfall, Timothy Garrett, Claudio F Gonzalez, Todd M Brusko, Wendy J Dahl, Graciela L Lorca, Guillermo E Marcial, Amanda L Ford, Michael J Haller, Salvador A Gezan, Natalie A Harrison, Dan Cai, Julie L Meyer, Daniel J Perry, Mark A Atkinson, Clive H Wasserfall, Timothy Garrett, Claudio F Gonzalez, Todd M Brusko, Wendy J Dahl, Graciela L Lorca

Abstract

Lactobacillus johnsonii N6.2 mitigates the onset of type 1 diabetes (T1D) in biobreeding diabetes-prone rats, in part, through changes in kynurenine:tryptophan (K:T) ratios. The goal of this pilot study was to determine the safety, tolerance, and general immunological response of L. johnsonii N6.2 in healthy subjects. A double-blind, randomized clinical trial in 42 healthy individuals with no known risk factors for T1D was undertaken to evaluate subject responses to the consumption of L. johnsonii N6.2. Participants received 1 capsule/day containing 108 colony-forming units of L. johnsonii N6.2 or placebo for 8 weeks. Comprehensive metabolic panel (CMP), leukocyte subpopulations by complete blood count (CBC) and flow cytometry, serum cytokines, and relevant metabolites in the indoleamine-2,3-dioxygenase pathway were assessed. L. johnsonii N6.2 survival and intestinal microbiota was analyzed. Daily and weekly questionnaires were assessed for potential effects of probiotic treatment on general wellness. The administration of L. johnsonii N6.2 did not modify the CMP or CBC of participants suggesting general safety. In fact, L. johnsonii N6.2 administration significantly decreased the occurrence of abdominal pain, indigestion, and cephalic syndromes. As predicted, increased serum tryptophan levels increased resulting in a decreased K:T ratio was observed in the L. johnsonii N6.2 group. Interestingly, immunophenotyping assays revealed that monocytes and natural killer cell numbers were increased significantly after washout (12 weeks). Moreover, an increase of circulating effector Th1 cells (CD45RO+CD183+CD196-) and cytotoxic CD8+ T cells subset was observed in the L. johnsonii N6.2 group. Consumption of L. johnsonii N6.2 is well tolerated in adult control subjects, demonstrates systemic impacts on innate and adaptive immune populations, and results in a decreased K:T ratio. These data provide support for the safety and feasibility of using L. johnsonii N6.2 in prevention trials in subjects at risk for T1D.

Trial registration: This trial was registered at https://ichgcp.net/clinical-trials-registry/NCT02349360" title="See in ClinicalTrials.gov">NCT02349360.

Keywords: Lactobacillus johnsonii; T cell; diabetes type I; gastrointestinal symptom; immunological response; indoleamine-2,3-dioxygenase; microbiome; probiotic.

Figures

Figure 1
Figure 1
Study flow diagram to illustrate the number of subjects that were screened, consented, and randomized. Code 090115-A was used for the placebo, whereas Code 090115-B was used for Lactobacillus johnsonii N6.2.
Figure 2
Figure 2
Determination of total lactic acid bacteria (LAB) and L. johnsonii N6.2 (Ljo) in stool samples. In placebo and Ljo groups, it was determined: (A) total number of LAB (Log CFU/g). Based on the numbers of LAB obtained, three subgroups were defined: (a) high LAB, (b) low to high LAB, and (c) low LAB. (B) Relative change in LAB for Ljo. (C) Relative change in LAB for placebo. (D) The presence of Ljo was confirmed by performing qRT-PCR of the T285_00345 gene and expressed as genomic equivalents. These data were further stratified based on the determination of total LAB numbers for the Ljo (E) and placebo (F) treatment groups. * indicates statistical differences (p < 0.05) between the groups and time points shown in panels (E,F) using analysis of variance. Comparison of the treatment combinations was performed by least significance difference with a significance level of 5%.
Figure 3
Figure 3
Peripheral tryptophan and kynurenine concentration in plasma of healthy subjects. The concentrations of kynurenine (A) and tryptophan (B) were determined by LC-HRMS/HRMS after 8 or 12 weeks in the placebo or L. johnsonii N6.2 (Ljo) treatment groups. Panel (C) is shown the kynurenine:tryptophan (K:T) ratio. The concentration of the metabolites shown has been normalized to the concentration found at time 0 for each subject. The results obtained were further stratified based on the number of LAB present as described in the Section “Results.” (a) High LAB; (b) low to high LAB, and (c) low LAB.
Figure 4
Figure 4
Monocytes and natural killer (NK) cells in healthy subjects. (A) Mononuclear cells (CD3−CD19−) were stained with specific antibodies to define monocytes (CD14+) and NK cells (CD14−) in the placebo and in the L. johnsonii N6.2 (Ljo) groups. (B) Expression of HLA-DR (mfi) in different NK cells subset: CD16−CD56hi, CD16+CD56lo/−, and CD16+CD56hi after 8 weeks of treatment or 12 weeks (4 weeks into the washout). The concentration of cells shown has been normalized to the concentration found at time 0 for each subject.
Figure 5
Figure 5
T cell subset. CD4+(A) or CD8+(B) T cells populations subsets [Naïve, Tem, activated (CD38+HLA-DR+)] were quantified after 8 or 12 weeks of treatment in the placebo and L. johnsonii N6.2 (Ljo) groups. Naïve (CD197+CD45RA+), Tem (CD197−CD45RA−), Tcm (CD197+CD45RA−), and Temra (CD197−CD45RA+) by labeling with specific antibodies (C). The concentration of cells shown has been normalized to the concentration found at time 0 for each subject.
Figure 6
Figure 6
Expression of CD185+ and CD279+ on T cells subset. Expression of CD279+(A) or CD185+(B) on Naïve, Tem, Tcm, and Temra CD4+ T cells. Expression of CD279+(C) or CD185+(D) on Naïve, Tem, Tcm, and Temra CD8+ T cells. The number of cells in each population was evaluated for the placebo (white bars) and the L. johnsonii N6.2 (Ljo, blue bars) group at 8 and 12 weeks. The concentration of cells shown has been normalized to the concentration found at time 0 for each subject.
Figure 7
Figure 7
T effector cells subset (CD3+CD4+CD45RO+). (A) Th1 (CD183+CD196−), Th2 (CD183−CD196−), Th17 (CD183−CD196+), and Th1/Th17 (CD183+CD196+) were labeled with specific antibodies and quantified in the placebo (white bars) and L. johnsonii N6.2 (Ljo, blue bars) group at 8 and 12 weeks of treatment. (B) HLA-DR+ and (C) HLA-DR+CD38+ are shown for the Th1 and Th1/Th17 effector T cells. The concentration of cells shown has been normalized to the concentration found at time 0 for each subject.
Figure 8
Figure 8
Determination of soluble markers. The concentrations of IFNγ, IgA, and TNFα were quantified in the placebo (white bars) and L. johnsonii N6.2 (Ljo, blue bars) group at time 0 and after 8 weeks of treatment or after the washout (12 weeks).

References

    1. Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol (2015) 14:20–32.10.1038/nrmicro3552
    1. Landy J, Walker AW, Li JV, Al-Hassi HO, Ronde E, English NR, et al. Variable alterations of the microbiota, without metabolic or immunological change, following faecal microbiota transplantation in patients with chronic pouchitis. Sci Rep (2015) 5:12955.10.1038/srep12955
    1. Levy M, Blacher E, Elinav E. Microbiome, metabolites and host immunity. Curr Opin Microbiol (2017) 35:8–15.10.1016/j.mib.2016.10.003
    1. Johnson EL, Heaver SL, Walters WA, Ley RE. Microbiome and metabolic disease: revisiting the bacterial phylum Bacteroidetes. J Mol Med (Berl) (2016) 95:1–8.10.1007/s00109-016-1492-2
    1. Abdul-Aziz MA, Cooper A, Weyrich LS. Exploring relationships between host genome and microbiome: new insights from genome-wide association studies. Front Microbiol (2016) 7:1611.10.3389/FMICB.2016.01611
    1. Kim D, Yoo SA, Kim WU. Gut microbiota in autoimmunity: potential for clinical applications. Arch Pharm Res (2016) 39:1565–76.10.1007/s12272-016-0796-7
    1. Paun A, Yau C, Danska JS. Immune recognition and response to the intestinal microbiome in type 1 diabetes. J Autoimmun (2016) 71:10–8.10.1016/j.jaut.2016.02.004
    1. Noble JA, Erlich HA. Genetics of type 1 diabetes. Cold Spring Harb Perspect Med (2012) 2:a007732.10.1101/cshperspect.a007732
    1. Neu J, Lorca G, Kingma SD, Triplett EW. The intestinal microbiome: relationship to type 1 diabetes. Endocrinol Metab Clin North Am (2010) 39:563–71.10.1016/j.ecl.2010.05.008
    1. Brown CT, Davis-Richardson AG, Giongo A, Gano KA, Crabb DB, Mukherjee N, et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One (2011) 6:e25792.10.1371/journal.pone.0025792
    1. de Goffau MC, Luopajarvi K, Knip M, Ilonen J, Ruohtula T, Harkonen T, et al. Fecal microbiota composition differs between children with β cell autoimmunity and those without. Diabetes (2013) 62:1238–44.10.2337/db12-0526
    1. Kostic AD, Gevers D, Siljander H, Vatanen T, Hyötyläinen T, Hämäläinen AM, et al. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe (2015) 17:260–73.10.1016/j.chom.2015.01.001
    1. Dolpady J, Sorini C, Di Pietro C, Cosorich I, Ferrarese R, Saita D, et al. Oral probiotic VSL # 3 prevents autoimmune diabetes by modulating microbiota and promoting indoleamine 2, 3-dioxygenase-enriched tolerogenic intestinal environment. J Diabetes Res (2016) 2016:7569431.10.1155/2016/7569431
    1. Valladares R, Sankar D, Li N, Williams E, Lai KK, Abdelgeliel AS, et al. Lactobacillus johnsonii N6.2 mitigates the development of type 1 diabetes in BB-DP rats. PLoS One (2010) 5:e10507.10.1371/journal.pone.0010507
    1. Lau K, Benitez P, Ardissone A, Wilson TD, Collins EL, Lorca G, et al. Inhibition of type 1 diabetes correlated to a Lactobacillus johnsonii N6.2-mediated Th17 bias. J Immunol (2011) 186:3538–46.10.4049/jimmunol.1001864
    1. Roesch LF, Lorca GL, Casella G, Giongo A, Naranjo A, Pionzio AM, et al. Culture-independent identification of gut bacteria correlated with the onset of diabetes in a rat model. ISME J (2009) 3:536–48.10.1038/ismej.2009.5
    1. Kemgang TS, Kapila S, Shanmugam VP, Kapila R. Cross-talk between probiotic lactobacilli and host immune system. J Appl Microbiol (2014) 117:303–19.10.1111/jam.12521
    1. Sáez-Lara M, Robles-Sanchez C, Ruiz-Ojeda F, Plaza-Diaz J, Gil A. Effects of probiotics and synbiotics on obesity, insulin resistance syndrome, type 2 diabetes and non-alcoholic fatty liver disease: a review of human clinical trials. Int J Mol Sci (2016) 17:928.10.3390/ijms17060928
    1. Opitz CA, Wick W, Steinman L, Platten M. Tryptophan degradation in autoimmune diseases. Cell Mol Life Sci (2007) 64:2542–63.10.1007/s00018-007-7140-9
    1. Ciorba MA. Indoleamine 2,3 dioxygenase in intestinal disease. Curr Opin Gastroenterol (2013) 29:146–52.10.1097/MOG.0b013e32835c9cb3
    1. Yeung AW, Terentis AC, King NJ, Thomas SR. Role of indoleamine 2,3-dioxygenase in health and disease. Clin Sci (Lond) (2015) 129:601–72.10.1042/CS20140392
    1. Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med (1999) 189:1363–72.10.1084/jem.189.9.1363
    1. Mellor AL, Chandler P, Lee GK, Johnson T, Keskin DB, Lee J, et al. Indoleamine 2,3-dioxygenase, immunosuppression and pregnancy. J Reprod Immunol (2002) 57:143–50.10.1016/S0165-0378(02)00040-2
    1. Grozdics E, Berta L, Bajnok A, Veres G, Ilisz I, Klivényi P, et al. B7 costimulation and intracellular indoleamine-2,3-dioxygenase (IDO) expression in peripheral blood of healthy pregnant and non-pregnant women. BMC Pregnancy Childbirth (2014) 14:306.10.1186/1471-2393-14-306
    1. Valladares R, Bojilova L, Potts AH, Cameron E, Gardner C, Lorca G, et al. Lactobacillus johnsonii inhibits indoleamine 2,3-dioxygenase and alters tryptophan metabolite levels in BioBreeding rats. FASEB J (2013) 27:1711–20.10.1096/fj.12-223339
    1. Leonard MT, Valladares RB, Ardissone A, Gonzalez CF, Lorca GL, Triplett EW. Complete genome sequences of Lactobacillus johnsonii strain N6.2 and Lactobacillus reuteri strain TD1. Genome Announc (2014) 2:5–6.10.1128/genomeA.00397-14
    1. Maecker HT. Standardizing immunophenotyping for the human immunology. Nat Rev Immunol (2012) 12:191–200.10.1016/j.micinf.2011.07.011.Innate
    1. He J, Tsai LM, Leong YA, Hu X, Ma CS, Chevalier N, et al. Circulating precursor CCR7loPD-1hi CXCR5+ CD4+ T cells indicate Tfh cell activity and promote antibody responses upon antigen reexposure. Immunity (2013) 39:770–81.10.1016/j.immuni.2013.09.007
    1. Locci M, Havenar-Daughton C, Landais E, Wu J, Kroenke MA, Arlehamn CL, et al. Human circulating PD-1+CXCR3−CXCR5+ memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity (2013) 39:758–69.10.1016/j.immuni.2013.08.031
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J (2012) 6:1621–4.10.1038/ismej.2012.8
    1. Rideout JR, He Y, Navas-Molina JA, Walters WA, Ursell LK, Gibbons SM, et al. Subsampled open-reference clustering creates consistent, comprehensive OTU definitions and scales to billions of sequences. PeerJ (2014) 2:e545.10.7717/peerj.545
    1. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol (2006) 72:5069–72.10.1128/AEM.03006-05
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics (2010) 26:2460–1.10.1093/bioinformatics/btq461
    1. McMurdie PJ, Holmes S. Phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One (2013) 8:e61217.10.1371/journal.pone.0061217
    1. Wickham H. The split-apply-combine strategy for data analysis. J Stat Softw (2011) 40:1–29.10.18637/jss.v040.i01
    1. Dixon P. VEGAN, a package of R functions for community ecology. J Veg Sci (2003) 14:927–30.10.1111/j.1654-1103.2003.tb02228.x
    1. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics (2014) 30:3123–4.10.1093/bioinformatics/btu494
    1. Stender S, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Extreme bilirubin levels as a causal risk factor for symptomatic gallstone disease. JAMA Intern Med (2013) 173:1222.10.1001/jamainternmed.2013.6465
    1. Knip M, Åkerblom HK, Becker D, Dosch M, Dupre J, Fraser W, et al. Hydrolyzed infant formula and early β-cell autoimmunity: a randomized clinical trial. JAMA (2014) 311:2279–87.10.1001/jama.2014.5610
    1. Chase HP, Boulware D, Rodriguez H, Donaldson D, Chritton S, Rafkin-Mervis L, et al. Effect of docosahexaenoic acid supplementation on inflammatory cytokine levels in infants at high genetic risk for type 1 diabetes. Pediatr Diabetes (2015) 16:271–9.10.1111/pedi.12170
    1. Uusitalo U, Liu X, Yang J, Hummel S, Butterworth M, Rewers M, et al. Association of early exposure of probiotics and islet autoimmunity in the TEDDY study. JAMA Pediatr (2016) 170:20–8.10.1001/jamapediatrics.2015.2757
    1. Fuller R. Probiotics in man and animals. J Appl Bacteriol (1989) 66:365–78.10.1111/j.1365-2672.1989.tb05105.x
    1. Ashraf R, Shah NP. Immune system stimulation by probiotic microorganisms. Crit Rev Food Sci Nutr (2014) 54:938–56.10.1080/10408398.2011.619671
    1. Rizzello V, Bonaccorsi I, Dongarrà ML, Fink LN, Ferlazzo G. Role of natural killer and dendritic cell crosstalk in immunomodulation by commensal bacteria probiotics. J Biomed Biotechnol (2011) 2011:1–10.10.1155/2011/473097
    1. Rask C, Adlerberth I, Berggren A, Ahrén IL, Wold AE. Differential effect on cell-mediated immunity in human volunteers after intake of different lactobacilli. Clin Exp Immunol (2013) 172:321–32.10.1111/cei.12055
    1. Van Baarlen P, Troost FJ, Van Hemert S, Van Der Meer C, De Vos WM. Differential NF-kappaB pathways induction by Lactobacillus plantarum in the duodenum of healthy. Proc Natl Acad Sci U S A (2008) 106:2371–6.10.1073/pnas.0809919106
    1. Michalickova D, Minic R, Dikic N, Andjelkovic M, Kostic-Vucicevic M, Stojmenovic T, et al. Lactobacillus helveticus Lafti L10 supplementation reduces respiratory infection duration in a cohort of elite athletes: a randomized, double-blind, placebo-controlled trial. Appl Physiol Nutr Metab (2016) 41:782–9.10.1139/apnm-2015-0541
    1. Sheikhi A, Shakerian M, Giti H, Baghaeifar M, Jafarzadeh A, Ghaed V, et al. Probiotic yogurt culture Bifidobacterium animalis subsp. lactis BB-12 and Lactobacillus acidophilus LA-5 modulate the cytokine secretion by peripheral blood mononuclear cells from patients with ulcerative colitis. Drug Res (Stuttg) (2016) 66:300–5.10.1055/s-0035-1569414
    1. Sanders ME, Akkermans LM, Haller D, Hammerman C, Heimbach J, Hörmannsperger G, et al. Safety assessment of probiotics for human use. Gut Microbes (2010) 1:164–85.10.4161/gmic.1.3.12127
    1. Grover S, Rashmi HM, Srivastava AK, Batish VK. Probiotics for human health – new innovations and emerging trends. Gut Pathog (2012) 4:15.10.1186/1757-4749-4-15
    1. Hanifi A, Culpepper T, Mai V, Anand A, Ford AL, Ukhanova M, et al. Evaluation of Bacillus subtilis R0179 on gastrointestinal viability and general wellness: a randomised, double-blind, placebo-controlled trial in healthy adults. Benef Microbes (2015) 6:19–27.10.3920/BM2014.0031
    1. Kalman DS, Schwartz HI, Alvarez P, Feldman S, Pezzullo JC, Krieger DR. A prospective, randomized, double-blind, placebo-controlled parallel-group dual site trial to evaluate the effects of a Bacillus coagulans-based product on functional intestinal gas symptoms. BMC Gastroenterol (2009) 9:85.10.1186/1471-230X-9-85
    1. Culpepper T, Christman MC, Nieves C, Specht GJ, Rowe CC, Spaiser SJ, et al. Bifidobacterium bifidum R0071 decreases stress-associated diarrhoea-related symptoms and self-reported stress: a secondary analysis of a randomised trial. Benef Microbes (2016) 7:327–36.10.3920/BM2015.0156
    1. Francavilla R, Miniello V, Magistà AM, De Canio A, Bucci N, Gagliardi F, et al. A randomized controlled trial of Lactobacillus GG in children with functional abdominal pain. Pediatrics (2010) 126:e1445–52.10.1542/peds.2010-0467
    1. Gawrońska A, Dziechciarz P, Horvath A, Szajewska H. A randomized double-blind placebo-controlled trial of Lactobacillus GG for abdominal pain disorders in children. Aliment Pharmacol Ther (2007) 25:177–84.10.1111/j.1365-2036.2006.03175.x
    1. Bauserman M, Michail S. The use of Lactobacillus GG in irritable bowel syndrome in children: a double-blind randomized control trial. J Pediatr (2005) 147:197–201.10.1016/j.jpeds.2005.05.015
    1. Eftekhari K, Vahedi Z, Aghdam MK, Diaz DN. A randomized double-blind placebo-controlled trial of Lactobacillus reuteri for chronic functional abdominal pain in children. Iran J Pediatr (2015) 25:e2616.10.5812/ijp.2616
    1. Ducrotté P, Sawant P, Jayanthi V. Clinical trial: Lactobacillus plantarum 299v (DSM 9843) improves symptoms of irritable bowel syndrome. World J Gastroenterol (2012) 18:4012–8.10.3748/wjg.v18.i30.4012
    1. Dapoigny M, Piche T, Ducrotte P, Lunaud B, Cardot JM, Bernalier-Donadille A. Efficacy and safety profile of LCR35 complete freeze-dried culture in irritable bowel syndrome: a randomized, double-blind study. World J Gastroenterol (2012) 18:2067–75.10.3748/wjg.v18.i17.2067
    1. Amirimani B, Nikfam S, Albaji M, Vahedi S, Nasseri-Moghaddam S, Sharafkhah M, et al. Probiotic vs. placebo in irritable bowel syndrome: a randomized controlled trial. Middle East J Dig Dis (2013) 5:98–102.
    1. Larsen CN, Nielsen S, Kaestel P, Brockmann E, Bennedsen M, Christensen HR, et al. Dose-response study of probiotic bacteria Bifidobacterium animalis subsp lactis BB-12 and Lactobacillus paracasei subsp paracasei CRL-341 in healthy young adults. Eur J Clin Nutr (2006) 60:1284–93.10.1038/sj.ejcn.1602450
    1. Pakdaman MN, Udani JK, Molina JP, Shahani M. The effects of the DDS-1 strain of Lactobacillus on symptomatic relief for lactose intolerance – a randomized, crossover clinical trial. Nutr J (2016) 15:56.10.1186/s12937-016-0172-y
    1. Scagnolari C, Corano Scheri G, Selvaggi C, Schietroma I, Najafi Fard S, Mastrangelo A, et al. Probiotics differently affect gut-associated lymphoid tissue indolamine-2,3-dioxygenase mRNA and cerebrospinal fluid neopterin levels in antiretroviral-treated HIV-1 infected patients: a pilot study. Int J Mol Sci (2016) 17:1–10.10.3390/ijms17101639
    1. Young SN. How to increase serotonin in the human brain without drugs. J Psychiatry Neurosci (2007) 32:394–9.10.1111/jhn.12223
    1. Rao AV, Bested AC, Beaulne TM, Katzman MA, Iorio C, Berardi JM, et al. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog (2009) 1:6.10.1186/1757-4749-1-6
    1. Kai S, Goto S, Tahara K, Sasaki A, Kawano K, Kitano S. Inhibition of indoleamine 2,3-dioxygenase suppresses NK cell activity and accelerates tumor growth. J Exp Ther Oncol (2003) 3:336–45.10.1111/j.1533-869X.2003.01108.x
    1. Kapsenberg ML. Dendritic-cell control of pathogen-driven T-cell polarization. Nat Rev Immunol (2003) 3:984–93.10.1038/nri1246
    1. Cross ML, Ganner A, Teilab D, Fray LM. Patterns of cytokine induction by Gram-positive and Gram-negative probiotic bacteria. FEMS Immunol Med Microbiol (2004) 42:173–80.10.1016/j.femsim.2004.04.001
    1. Hessle C, Hanson LÅ, Wold AE. Lactobacilli from human gastrointestinal mucosa are strong stimulators of IL-12 production. Clin Exp Immunol (1999) 116:276–82.10.1046/j.1365-2249.1999.00885.x
    1. Hatcher GE, Lambrecht RS. Augmentation of macrophage phagocytic activity by cell-free extracts of selected lactic acid-producing bacteria. J Dairy Sci (1993) 76:2485–92.10.3168/jds.S0022-0302(93)77583-9
    1. Takagi A, Matsuzaki T, Sato M, Nomoto K, Morotomi M, Yokokura T. Enhancement of natural killer cytotoxicity delayed murine carcinogenesis by a probiotic microorganism. Carcinogenesis (2001) 22:599–605.10.1093/carcin/22.4.599
    1. Piccioli D, Sammicheli C, Tavarini S, Nuti S, Frigimelica E, Manetti AG, et al. Human plasmacytoid dendritic cells are unresponsive to bacterial stimulation and require a novel type of cooperation with myeloid dendritic cells for maturation. Blood (2009) 113:4232–9.10.1182/blood-2008-10-186890
    1. Kingma SD, Li N, Sun F, Valladares RB, Neu J, Lorca GL. Lactobacillus johnsonii N6.2 stimulates the innate immune response through Toll-like receptor 9 in Caco-2 cells and increases intestinal crypt Paneth cell number in biobreeding diabetes-prone rats. J Nutr (2011) 141:1023–8.10.3945/jn.110.135517
    1. Mailliard RB, Son YI, Redlinger R, Coates PT, Giermasz A, Morel PA, et al. Dendritic cells mediate NK cell help for Th1 and CTL responses: two-signal requirement for the induction of NK cell helper function. J Immunol (2003) 171:2366–73.10.4049/jimmunol.171.5.2366
    1. Fujiwara D, Inoue S, Wakabayashi H, Fujii T. The anti-allergic effects of lactic acid bacteria are strain dependent and mediated by effects on both Th1/Th2 cytokine expression and balance. Int Arch Allergy Immunol (2004) 135:205–15.10.1159/000081305
    1. Yi Z, Stunz LL, Lin WW, Bishop GA. TRAF3 regulates homeostasis of CD8+ central memory T cells. PLoS One (2014) 9:e102120.10.1371/journal.pone.0102120
    1. Kaech SM, Cui W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol (2012) 12:749–61.10.1038/nri3307

Source: PubMed

3
Suscribir