802
Views
0
CrossRef citations to date
0
Altmetric
Brief Report

A high fiber diet or supplementation with Lactococcus lactis subspecies cremoris to pregnant mice confers protection against intestinal injury in adult offspring

ORCID Icon, , , , &
Article: 2337317 | Received 07 Jul 2023, Accepted 27 Mar 2024, Published online: 15 Apr 2024

ABSTRACT

The diet during pregnancy, or antenatal diet, influences the offspring’s intestinal health. We previously showed that antenatal butyrate supplementation reduces injury in adult murine offspring with dextran sulfate sodium (DSS)-induced colitis. Potential modulators of butyrate levels in the intestine include a high fiber diet or dietary supplementation with probiotics. To test this, we supplemented the diet of pregnant mice with high fiber, or with the probiotic bacteria Lactococcus lactis subspecies cremoris or Lactobacillus rhamnosus GG. We then induced chronic colitis with DSS in their adult offspring. We demonstrate that a high fiber antenatal diet, or supplementation with Lactococcus lactis subspecies cremoris during pregnancy diminished the injury from DSS-induced colitis in offspring. These data are evidence that antenatal dietary interventions impact offspring gut health and define the antenatal diet as a therapeutic modality to enhance offspring intestinal health.

Introduction

The fetal environment plays a dynamic role in fetal growth and development and influences the offspring’s long-term health through developmental programming.Citation1 Through pre-clinical and clinical studies, our understanding of the influence of the diet during pregnancy (antenatal diet) on the offspring’s health has expanded.Citation2 For example, it was reported in mice that a high fat diet during pregnancy causes an expansion of Firmicutes in the offspring’s gut microbiome and increases the offspring’s susceptibility to neonatal intestinal inflammation.Citation3 Another study in mice reported that a diet rich in the aryl hydrocarbon receptor ligand indole-3-carbinol, prevented the development of necrotizing enterocolitis in neonatal mice.Citation4 Attesting to clinical relevance, several studies in humans have corroborated the importance of the antenatal diet on the offspring’s health.Citation5–9

Recently, our research group evaluated the effect of antenatal butyrate supplementation to pregnant mice on the intestinal health of their offspring.Citation10 Butyrate is a short-chain fatty acid (SCFA) produced by the gut microbiome through the fermentation of undigested dietary fiber.Citation11 Butyrate influences intestinal health through enhancement of the intestinal barrier,Citation12 down regulation of intestinal inflammationCitation13 and regulation of intestinal immune response.Citation12 Supplementation of the antenatal diet with butyrate reduced injury in adult offspring in a model of dextran sodium sulfate (DSS) induced colitis, with associated reduction in pro-inflammatory gene expression.Citation10 Supplementing the antenatal diet with butyrate also resulted in shifts in the adult offspring’s gut microbiome composition with increased prevalence of Bifidobacterium, and increased alpha diversity.Citation10

Based on these findings, we sought to evaluate whether supplementing the antenatal diet to increase endogenous butyrate levels would also reduce injury from colitis in adult offspring. Butyrate production by the microbiome may be increased through a high fiber diet (HFD),Citation14,Citation15 or by dietary supplementation with lactic acid bacteria, where bacteria produce lactate that is subsequently converted to butyrate by gut microbes that harbor a gene coding for butyryl-CoA:acetate CoA-transferase.Citation16,Citation17 Our research group identified the novel probiotic Lactococcus lactis subsp. cremoris (LLC) as being highly efficacious in suppressing colitis-induced gut injury.Citation18,Citation19 Herein, we report that a high fiber antenatal diet, or supplementation of the antenatal diet with LLC lowered colitis disease activity in offspring, and propose that supplementation of the antenatal diet with interventions that enhance butyrate generation may be an effective therapeutic strategy to enhance gut health in offspring.

Methods

Animal studies

Experiments were performed using wild-type C57BL/6 mice (Jackson Laboratories, Bar Harbor, ME). The mice were bred at the Department of Animal Resources facility at Emory University. Animal procedures were approved by the Institutional Animal Care and Use Committee at Emory University.

Bacterial strains and culture preparation

Probiotic bacteria purchased from the American Type Culture Collection (ATCC) (Manasas, VA) included Lactococcus lactis subspecies cremoris ATCC 19,257 (LLC) and Lactobacillus rhamnosus GG ATCC 53,103 (LGG). All media were propagated according to ATCC instructions. Probiotics were suspended in hanks balanced salt solution (HBSS) to a dose of 1 × 10Citation9 colony forming units, which has been reported to be beneficial to murine intestinal health.Citation19,Citation20

Murine diets

The mice received chow ad libitum. Breeding pairs received either a regular diet, a high fiber diet (HFD), LLC or LGG during pregnancy. The regular diet used was 5053-PicoLab® Rodent Diet 20, 5053 6% Fiber (LabDiet Inc, MO). The high fiber diet (HFD) used was isocaloric Modified AIN-93 G Purified Rodent Diet with 30% Fiber (15% wt:wt) with a 30% soluble fiber:70% cellulose ratio (Dyets Inc., PA, Lot #7284–5).

Mating pairs and experimental groups

Breeding pairs of mice (one male and one female) were randomly assigned to a diet which they received during pregnancy. In each experiment, mice originated from the same cage, ensuring that the microbiome composition was homogenous between all groups at the beginning of the experiment. The experimental groups included dietary variables fed to female mice before conception and during pregnancy, including: HFD, daily LLC supplementation via oral gavage, daily LGG supplementation via oral gavage, LLC with HFD, or regular diet (control). The mating pairs which did not receive probiotic supplementation received daily oral gavage of HBSS. Within 24 h of birth, all mating pairs were placed on a regular diet and did not receive any further dietary supplementation. The offspring of all mating pairs were weaned at 3 weeks of age based on their sex and experimental group and received a regular diet. For each experimental variable, two to three breeding pairs of mice generated all the offspring for each specific group.

DSS-induced colitis

Chronic colitis was induced in offspring at 6–8 weeks of age through administration of DSS (Thermo Scientific, Fair Lawn, NJ, Lot # 187742 and Lot # 207466), established previously as a murine model of colitis.Citation20,Citation21 To induce colitis, DSS was dissolved in the drinking water of the mice. The mice received DSS for 2 cycles. First, they received 2% DSS for 7 days, followed by 7 days of recovery with no DSS. Last, they received another 7 days of 2% DSS. The degree of colitis was evaluated by using a disease activity index (DAI) scoring tool, colon length measurement at the time of sacrifice and histomorphological injury scoring. DAI scoring is an established method to measure the degree of colitis in models of DSS colitis; it accounts for stool consistency, presence of blood in the stool and percent weight loss of each mouse.Citation22 DAI scores range from 0 to 12 and the score is proportional to the degree of intestinal injury with a score of ‘0’ being no injury, and a score of ‘12’ indicating severe colitis. DAI scoring began at day 3 of DSS colitis model. Colon length is another measure of colonic injury, as colitis causes contraction and shortening of the colon.Citation23 Thus, colon length is inversely related to the severity of colitis and was measured at sacrifice.

Hematoxylin-eosin staining and scoring of tissue for intestinal injury

Following completion of the chronic colitis model, the murine offspring were sacrificed, and their colon was harvested. Each colon was fixed in 4% formalin, dehydrated, paraffin embedded and then sectioned. The sections underwent deparaffinization, hydration in alcohol gradient, and staining with Hematoxylin and Eosin (H&E) by Emory Winship Pathology Core. Images were scored for the degree of colitis using a 2-tiered scoring system, adapted from an established scoring guideline.Citation24 The histologic activity index (HAI) score evaluates the degree of inflammatory cell infiltration and the integrity of the mucosal architecture. HAI score ranges from 0 to 6 and is proportional to the degree of injury. Inflammatory cell infiltration is considered mild, moderate, or severe based on the degree of inflammatory cells in the colonic tissue. Inflammatory cell infiltration to the level of the mucosa is considered mild (score of 1), to the mucosa and submucosa is considered moderate (score of 2) and transmural inflammatory cell infiltration is considered severe (score of 3). Next, the extent of epithelial injury is determined based on architectural epithelial changes such as focal erosions (score of 1), erosions with focal ulcerations (score of 2) or extended ulcerations with or without granulation tissue or pseudo polyps (score of 3). The HAI score is calculated as a sum of the inflammatory cell infiltrate score (0–3) and epithelial architecture score (0–3). Sections were scored in blinded fashion.

Statistics

Statistical significance was determined by Student’s T test for paired samples and one-way ANOVA for data with more than 2 groups using Prism 8 (GraphPad, San Diego, CA). A p-value of <0.05 was defined as statistically significant. All data are presented as mean ± standard error of the mean (SEM).

Results

A high fiber antenatal diet in mice reduces DSS-induced chronic colitis injury in adult offspring

At 6 weeks of age, offspring mice from dams that received an HFD or control regular diet were subjected to a model of DSS-induced chronic colitis. Health monitoring during the chronic colitis model revealed that offspring from HFD supplemented mothers had significantly lower DAI compared to offspring from control diet fed mothers (). For example, at day 7, the last day of the first cycle of DSS, the mean DAI score for the control group was 5 ± 1.5, whereas the mean DAI for the HFD group was 2.6 ± 0.5 (p < 0.01). At day 21 of the experiment, the final day of the second cycle of DSS, the mean DAI score for the control group was 4.8 ± 1.6, whereas the mean DAI for the HFD group was 2.5 ± 0.7 (p < 0.01). No significant differences in colon lengths were detected between the two groups (p = 0.16) (), but histopathologic analysis revealed antenatal HFD offspring had lower injury scores than control group offspring with HAI scores of 3.1 ± 0.3 for the antenatal HFD offspring and HAI scores of 3.5 ± 0.4 for the control group offspring (p = 0.048) (). In each of our experimental group, we did not detect a sex-specific effect. Together, these data show that an antenatal HFD can elicit enduring cytoprotective effects in the intestine of murine offspring up to 9 weeks of age.

Figure 1. Antenatal high fiber diet reduces injury from DSS colitis in adult murine offspring.

(a) Quantification of intestinal injury through DAI score in 6-week-old C57BL/6 mice subjected to chronic 2% DSS-induced colitis model. Experimental groups include offspring from mothers on a HFD antenatal diet (n = 10), and offspring from mothers on a control antenatal diet (n = 8). DAI score ranges from 0 to 6 and is proportional to the degree of intestinal injury. Student’s t-test. *p < 0.05, **p < 0.01. (b) Colon length of mice described in (a) measured at the time of sacrifice (9 weeks-old). (c) HAI score quantifies colonic injury by a validated scoring tool, the score ranges from 0 to 6 and is proportional to the degree of colonic injury. Student’s t-test. *p < 0.05). (d) Representative H&E images of proximal colonic tissue in mice in antenatal control group. (e) Representative H&E images of proximal colonic tissue in mice in antenatal HFD group. Scale bar is 50μm.
Figure 1. Antenatal high fiber diet reduces injury from DSS colitis in adult murine offspring.

Antenatal supplementation of L. lactis cremoris (LLC) reduces DSS-induced chronic colitis injury in adult offspring

At 6 weeks of age, offspring mice from dams that were supplemented LLC, LGG or control group were subjected to a model of DSS-induced chronic colitis. The offspring from the antenatal LLC supplemented group exhibited significantly lower DAI scores compared to the offspring from the control groups (). At day 7, the mean DAI score for the control group was 5 ± 1.5, whereas mean DAI for the LLC group was 3.6 ± 0.9 (p = 0.04). On day 21 of DSS, the mean DAI score for the control group was 4.8 ± 1.6, versus the mean DAI for the LLC group was 2.6 ± 0.5 (p < 0.01). By contrast, the DAI scores of the offspring in the antenatal LGG group were not significantly different from the DAI scores of offspring in the control group (). Specifically, at day 7, the mean DAI score for the control group was 5 ± 1.5, and the mean DAI for the LGG group was 4.5 ± 2 (p = 0.3). On day 21 of DSS, the mean DAI score for the control group was 4.8 ± 1.6, versus the mean DAI for the LGG group was 4.2 ± 1 (p = 0.2). Control group offspring had significantly longer colons (22.9 ± 1.6 cm) than the offspring from the antenatal LLC (18.9 ± 0.8 cm) and LGG (19 ± 3.1 cm) supplemented groups (p < 0.004 and p < 0.003, respectively) (). Despite lower DAI scores, histopathologic grading (HAI) to assess the intestinal injury did not show a significant difference in the level of injury between the control group and the antenatal LLC group (p = 0.21), or the control group and the antenatal LGG group (p = 0.43) (). However, we did detect lower HAI score in the antenatal LLC supplemented group compared to the LGG group, with a mean HAI score of 3.2 ± 0.3 in the antenatal LLC group and a mean HAI score of 3.6 ± 0.4 in the antenatal LGG group (p = 0.07) (). These data show that supplementation of the antenatal diet with LLC, but not LGG, conferred cytoprotection in the intestine of 9-week-old murine offspring.

Figure 2. Supplementation of the antenatal diet with probiotics impacts the extent of DSS-induced colitis injury in adult offspring.

(a) Quantification of intestinal injury through DAI score on 6-week-old C57BL/6 mice subjected to chronic 2% DSS-induced colitis model. Experimental groups include offspring from mothers whose antenatal diet was supplemented daily with 1 × 109 CFU of Lactococcus lactis subsp. Cremoris (LLC) (n = 5), Lactobacillus rhamnosus GG (LGG) (n = 6), or with equal volume of HBSS (control) (n = 8). DAI score ranges from 0 to 6 and is proportional to the degree of intestinal injury. Student’s t-test. * p < 0.05, ** p < 0.01. (b) Colon length of mice described in (a) measured at the time of sacrifice (9 weeks-old). One-way ANOVA. ** p < 0.01. (c) HAI score quantifies colonic injury by a validated scoring tool, the score ranges from 0 to 6 and is proportional to the degree of colonic injury. One-way ANOVA. (d) Representative H&E images of proximal colonic tissue in mice described in (a). Scale bar is 50μm. (e) Quantification of intestinal injury through DAI score on 6-week-old C57BL/6 mice subjected to chronic 2% DSS-induced colitis model. Experimental groups include offspring from mothers on a HFD antenatal and supplemented daily with LLC (LLC HF) (n = 6), or on a control diet supplemented daily with LLC (n = 18) (LLC). Student’s t-test. (f) Colon length of mice described in (e) measured at the time of sacrifice (9 weeks-old). Student’s t-test. (g) HAI score quantifies colonic injury by a validated scoring tool, the score ranges from 0 to 6 and is proportional to the degree of colonic injury. Student’s t-test. (h) Representative H&E images of proximal colonic tissue in mice described in (f). Scale bar is 50μm.
Figure 2. Supplementation of the antenatal diet with probiotics impacts the extent of DSS-induced colitis injury in adult offspring.

Given the reduction in intestinal injury among adult offspring born to dams with antenatal HFD or LLC supplementation during pregnancy, we evaluated if a combination of LLC supplementation and a HFD during pregnancy would work in synergy to improve gut cytoprotection in offspring. Adult offspring from antenatal HFD with LLC or antenatal LLC supplementation were exposed to DSS-induced chronic colitis. Analysis revealed that there was no further reduction in intestinal injury in offspring whose mothers were fed antenatal HFD and supplemented with LLC, compared to offspring from mothers with antenatal LLC supplementation (). DAI scores were not significantly different between the two groups. On day 7 of DSS, the mean DAI score for the LLC group was 5 ± 1.2, and the mean DAI for the LLC with HFD group was 5.6 ± 2.2 (p = 0.2). On day 21 of DSS, the mean DAI score for the LLC group was 5.9 ± 2.3, versus the mean DAI for the LLC with HFD group was 6.7 ± 2.6 (p = 0.3). There was also no significant difference in colon length or HAI scores between the two groups, with mean HAI scores of 3.7 ± 0.7 in the LLC group and 3.6 ± 0.6 in the HFD with LLC group (p = 0.48).

Discussion

In this report, we investigated the effects of antenatal dietary supplementation with a HFD, or LLC and LGG probiotics on the adult offspring’s response to experimentally induce chronic colitis. This study corroborates previous findings that the antenatal diet influences the intestinal health of the offspring.Citation3,Citation4,Citation10 Given our previous findings that antenatal butyrate supplementation in mice leads to a reduction in gut injury in offspring exposed to models of DSS colitis, we anticipated that antenatal supplementation with HFD would also reduce injury in DSS-colitis among adult offspring, as supplementation with HFD has been reported to increase butyrate production by the microbiome.Citation11 Indeed, our results highlight that a HFD during pregnancy led to a significant reduction in gut injury in adult offspring exposed to DSS colitis, with a significant reduction in colitis by DAI and HAI score. Interestingly, the lowering of DAI and HAI by HFD during pregnancy did not impact colon length, which is often used as a faithful parameter to quantitate the extent of gut DSS-induced injury.

In addition, we found that supplementation of the antenatal diet with LLC was associated with a reduction in intestinal injury in the adult offspring compared to supplementation of the antenatal diet with LGG, evinced by lower DAI scores. We also evaluated whether supplementing the antenatal diet with a HFD and LLC would be more protective to the offspring than LLC alone. However, HFD and LLC did not engender a cumulative cytoprotective effect. We note that the HFD combined with LLC experiment outlined in was done with a different batch of DSS reagent than the experiments outlined in , and therefore DAI scores could not be directly compared. Nevertheless, the data intriguingly demonstrate that the LLC probiotic, but not LGG was effective in conferring protection in offspring.

A description of the mechanism whereby a HFD or dietary supplementation with LLC to pregnant dams transmits beneficial influences to offspring is currently elusive. We speculate that one possible mechanism is via the generation of elevated levels of SCFAs within the pregnant dams. Indeed, a HFDCitation14,Citation15 or dietary supplementation with lactic acid bacteriaCitation16,Citation17 has been reported to increase levels of SCFAs in gut tissue and serum. However, measurement of butyrate concentrations in gestating pups within the uterus at day E19, or in the gut tissue of 2-week-old offspring did not detect any significant differences in SCFAs levels, even in E19 pups or 2-week-old offspring from dams directly supplemented with butyrate during gestation. These observations support the notion that the E19 pups are not directly exposed to elevated butyrate and that the 2-week-old offspring do not themselves generate elevated butyrate levels in their microbiome. These observations may suggest that the elevated cytoprotective states in offspring may be due to microbiome transfer from dams to offspring and that the inherited microbiome contains a selection of beneficial microbes. It is also possible that gestating pups may be subject to epigenetic changes in utero since fetal development constitutes the most active period for epigenetic DNA imprinting. Given butyrate’s activity as an HDAC inhibitor, it is plausible that part of antenatal butyrate’s protection against intestinal injury in offspring is due to inherited epigenetic effects on genes within the offspring’s intestinal epithelium. As of the publication of this manuscript, the mechanism whereby an HFD or dietary supplementation with LLC to pregnant dams transmits beneficial influence to offspring remains elusive, although it is the subject of intense scrutiny within our research group.

Together, these data suggest that antenatal HFD provides the most significant and consistent protection against intestinal injury in adult offspring exposed to DSS-induced chronic colitis. Antenatal LLC also reduced injury in adult offspring; however, this reduction in injury was not as consistent with HFD, with reduction in DAI but not HAI. Interestingly, antenatal LGG did not induce any reduction in intestinal injury in adult offspring. We postulate that these findings are related to the amount of butyrate produced from each of these diets, with enhanced butyrate production in the HFD and LLC diets. Additionally, since the combination of HFD and LLC supplementation did not provide additional cytoprotection, this suggests that there may be a limit to the amount of butyrate that induces protection or the amount of butyrate produced by our gut microbiome. Thus, a high fiber diet or LLC probiotic when consumed during pregnancy may provide therapeutic options to enhance offspring intestinal health.

Conclusion

These data show that supplementing the diet of a pregnant mice with HFD or specific probiotics elicits cytoprotective effects on the intestinal health of adult offspring. The molecular mechanisms whereby the maternal microbiome transmits and endows enduring beneficial cell signal events in the gut of offspring is the focus of intense current research within our group.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

The authors confirm that the data supporting the findings of this study are available within the article, any further data can be requested from the corresponding author.

Additional information

Funding

The work was supported by the Children’s Healthcare of Atlanta [Junior Faculty Focused Award]; Emory University [Junior Faculty Focused Award]; Emory University [K12HD072245]; Eunice Kennedy Shriver National Institute of Child Health and Human Development [K12HD072245].

References

  • Salam RA, Das JK, Bhutta ZA. Impact of intrauterine growth restriction on long-term health. Curr Opin Clin Nutr Metab Care. 2014;17(3):249–8. doi:10.1097/MCO.0000000000000051.
  • Mirpuri J. Evidence for maternal diet-mediated effects on the offspring microbiome and immunity: implications for public health initiatives. Pediatr Res. 2021;89(2):301–306. doi:10.1038/s41390-020-01121-x.
  • Babu ST, Niu X, Raetz M, Savani RC, Hooper LV, Mirpuri J. Maternal high-fat diet results in microbiota-dependent expansion of ILC3s in mice offspring. JCI Insight. 2018;3(19):3. doi:10.1172/jci.insight.99223.
  • Lu P, Yamaguchi Y, Fulton WB, Wang S, Zhou Q, Jia H, Kovler ML, Salazar AG, Sampah M, Prindle T. et al. Maternal aryl hydrocarbon receptor activation protects newborns against necrotizing enterocolitis. Nat Commun. 2021;12(1):1042. doi:10.1038/s41467-021-21356-4.
  • Sausenthaler S, Koletzko S, Schaaf B, Lehmann I, Borte M, Herbarth O, von Berg A, Wichmann H-E, Heinrich J. Maternal diet during pregnancy in relation to eczema and allergic sensitization in the offspring at2 y of age. Am J Clin Nutr. 2007;85(2):530–537. doi:10.1093/ajcn/85.2.530.
  • Calvani M, Alessandri C, Sopo SM, Panetta V, Pingitore G, Tripodi S, Zappalà D, Zicari AM. Consumption of fish, butter and margarine during pregnancy and development of allergic sensitizations in the offspring: role of maternal atopy. Pediatr Allergy Immunol. 2006;17(2):94–102. doi:10.1111/j.1399-3038.2005.00367.x.
  • Romieu I, Torrent M, Garcia-Esteban R, Ferrer C, Ribas-Fito N, Anto JM, Sunyer J. Maternal fish intake during pregnancy and atopy and asthma in infancy. Clin Exp Allergy. 2007;37(4):518–525. doi:10.1111/j.1365-2222.2007.02685.x.
  • Miyake Y, Sasaki S, Tanaka K, Ohfuji S, Hirota Y. Maternal fat consumption during pregnancy and risk of wheeze and eczema in Japanese infants aged 16–24 months: the Osaka maternal and child health study. Thorax. 2009;64(9):815–821. doi:10.1136/thx.2009.115931.
  • Chu DM, Antony KM, Ma J, Prince AL, Showalter L, Moller M, Aagaard KM. The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med. 2016;8(1):77. doi:10.1186/s13073-016-0330-z.
  • Barbian ME, Owens JA, Naudin CR, Denning PW, Patel RM, Jones RM. Butyrate supplementation to pregnant mice elicits cytoprotection against colonic injury in the offspring. Pediatr Res. 2022;92(1):125–134. doi:10.1038/s41390-021-01767-1.
  • Macfarlane GT, Macfarlane S. Bacteria, colonic fermentation, and gastrointestinal health. J AOAC Int. 2012;95(1):50–60. doi:10.5740/jaoacint.SGE_Macfarlane.
  • Goncalves P, Araujo JR, Di Santo JP. A cross-talk between microbiota-derived short-chain fatty acids and the host mucosal immune system regulates intestinal homeostasis and inflammatory bowel disease. Inflamm Bowel Dis. 2018;24(3):558–572. doi:10.1093/ibd/izx029.
  • Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14(10):667–685. doi:10.1038/nri3738.
  • Flint HJ, Duncan SH, Scott KP, Louis P. Links between diet, gut microbiota composition and gut metabolism. Proc Nutr Soc. 2015;74(1):13–22. doi:10.1017/S0029665114001463.
  • Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7(3):189–200. doi:10.1080/19490976.2015.1134082.
  • Duncan SH, Barcenilla A, Stewart CS, Pryde SE, Flint HJ. Acetate utilization and butyryl coenzyme a (CoA): acetate-CoA transferase in butyrate-producing bacteria from the human large intestine. Appl Environ Microbiol. 2002;68(10):5186–5190. doi:10.1128/AEM.68.10.5186-5190.2002.
  • Tyagi AM, Yu M, Darby TM, Vaccaro C, Li JY, Owens JA, Hsu E, Adams J, Weitzmann MN, Jones RM. et al. The microbial metabolite butyrate stimulates bone formation via T regulatory cell-mediated regulation of WNT10B expression. Immunity. 2018;49(6):1116–31 e7. doi:10.1016/j.immuni.2018.10.013.
  • Song AA, LLA I, Lim SHE, Rahim RA. A review on Lactococcus lactis: from food to factory. Microb Cell Fact. 2017;16(1):55. doi:10.1186/s12934-017-0669-x.
  • Naudin CR, Maner-Smith K, Owens JA, Wynn GM, Robinson BS, Matthews JD, Reedy AR, Luo L, Wolfarth AA, Darby TM. et al. Lactococcus lactis subspecies cremoris elicits protection against metabolic changes induced by a Western-Style Diet. Gastroenterology. 2020;159(2):639–51 e5. doi:10.1053/j.gastro.2020.03.010.
  • Darby TM, Owens JA, Saeedi BJ, Luo L, Matthews JD, Robinson BS, Naudin CR, Jones RM. Lactococcus Lactis subsp. cremoris is an efficacious beneficial bacterium that limits tissue injury in the intestine. iScience. 2019;12:356–367. doi:10.1016/j.isci.2019.01.030.
  • Wirtz S, Popp V, Kindermann M, Gerlach K, Weigmann B, Fichtner-Feigl S, Neurath MF. Chemically induced mouse models of acute and chronic intestinal inflammation. Nat Protoc. 2017;12(7):1295–1309. doi:10.1038/nprot.2017.044.
  • Zhang L, Zhang Y, Zhong W, Di C, Lin X, Xia Z. Heme oxygenase-1 ameliorates dextran sulfate sodium-induced acute murine colitis by regulating Th17/Treg cell balance. J Biol Chem. 2014;289(39):26847–26858. doi:10.1074/jbc.M114.590554.
  • Cao H, Liu J, Shen P, Cai J, Han Y, Zhu K, Fu Y, Zhang N, Zhang Z, Cao Y. et al. Protective effect of Naringin on DSS-Induced ulcerative colitis in mice. J Agric Food Chem. 2018;66(50):13133–13140. doi:10.1021/acs.jafc.8b03942.
  • Erben U, Loddenkemper C, Doerfel K, Spieckermann S, Haller D, Heimesaat MM, Zeitz M, Siegmund B, Kühl AA. A guide to histomorphological evaluation of intestinal inflammation in mouse models. Int J Clin Exp Pathol. 2014;7:4557–4576.