Next Article in Journal
Antimicrobial Resistance in Bacteria from Meat and Meat Products: A One Health Perspective
Next Article in Special Issue
Prebiotics, Probiotics, and Synbiotics—A Research Hotspot for Pediatric Obesity
Previous Article in Journal
The Role of the Pancreatic Stone Protein in Predicting Intra-Abdominal Infection-Related Complications: A Prospective Observational Single-Center Cohort Study
Previous Article in Special Issue
Illuminating the Genomic Landscape of Lactiplantibacillus plantarum PU3—A Novel Probiotic Strain Isolated from Human Breast Milk, Explored through Nanopore Sequencing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effects of Lactobacillus johnsonii on Diseases and Its Potential Applications

1
Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
2
State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, China
3
CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200000, China
*
Authors to whom correspondence should be addressed.
Microorganisms 2023, 11(10), 2580; https://doi.org/10.3390/microorganisms11102580
Submission received: 13 July 2023 / Revised: 29 September 2023 / Accepted: 12 October 2023 / Published: 17 October 2023
(This article belongs to the Special Issue Beneficial Microorganisms and Antimicrobials)

Abstract

:
Lactobacillus johnsonii has been used as a probiotic for decades to treat a wide range of illnesses, and has been found to have specific advantages in the treatment of a number of ailments. We reviewed the potential therapeutic effects and mechanisms of L. johnsonii in various diseases based on PubMed and the Web of Science databases. We obtained the information of 149 L. johnsonii from NCBI (as of 14 February 2023), and reviewed their comprehensive metadata, including information about the plasmids they contain. This review provides a basic characterization of different L. johnsonii and some of their potential therapeutic properties for various ailments. Although the mechanisms are not fully understood yet, it is hoped that they may provide some evidence for future studies. Furthermore, the antibiotic resistance of the various strains of L. johnsonii is not clear, and more complete and in-depth studies are needed. In summary, L. johnsonii presents significant research potential for the treatment or prevention of disease; however, more proof is required to justify its therapeutic application. An additional study on the antibiotic resistance genes it contains is also needed to reduce the antimicrobial resistance dissemination.

1. Introduction

Probiotics are correctly defined as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” [1]. Lactobacillus johnsonii, as one of the typical intestinal probiotics, is widely distributed in the gastrointestinal tracts (GITs) of several hosts, including humans, mice, dogs, poultry, pigs, and honeybees [2,3,4], and has a long history of application in the food and fermented feed industries [5,6]. With the rapid development of science and technology, L. johnsonii has also been recognized as having important applications in many fields such as biology, agriculture, animal husbandry [7], and medicine [8]. Many studies have been conducted on L. johnsonii to explore its specific function and mechanism in different diseases, such as colitis, diarrhea, liver disease, and so on. In animal models and human models, researchers have conducted relevant studies, and it should be noted that a large number of studies show that L. johnsonii exhibits the following beneficial abilities: anti-inflammatory, immunomodulatory, intestinal microflora balance, and intestinal barrier protection. Moreover, L. johnsonii co-evolved with different animals at the species or strain level [9,10], which provides a reasonable basis for speculating on its relationship with health benefits. Lactobacilli represent the types of microorganisms to which the mammalian immune systems have learned not to respond, and this is considered a potential driver for the evolution of the human immune system [9]. According to a substantial body of literature, L. johnsonii has been shown to play a crucial role in modulating the host immune system, by altering macrophage [11], T-cell, and Th2 cytokine levels [12,13] and regulating dendritic cell (DC) function [14].
As of 14 February 2023, we retrieved a total of 1313 results in the Web of Science using “Lactobacillus johnsonii” as a keyword, including 1194 papers, 180 reviews, 46 clinical trials, etc. Although there were studies on the beneficial effects of L. johnsonii on certain diseases, we found that there was no review that comprehensively summarized the potential beneficial effects of L. johnsonii on different diseases to date, and the role L. johnsonii plays in disease treatment is unclear. This review summarizes the potential beneficial effects of different strains of L. johnsonii in a variety of common diseases involving various parts of the body and is useful for other researchers to quickly understand the field and to conduct more refined studies.

2. Comprehensive Characteristics of Identified L. johnsonii

We searched the National Center for Biotechnology Information (NCBI) and overviewed information tables for the L. johnsonii (as of 14 February 2023) (Table S1). From Table S1, it is easy to find that these strains were identified through shotgun metagenomic sequencing from host samples, while others were isolated from the host samples. The strains isolated from the hosts came from different body parts of different hosts in different countries, including the human intestine, mouse forestomach, and pig intestine. Based on the available information, we can know that the earliest strains were collected in 1964. However, the culture conditions required for many strains were not described in detail.
Plasmids, as genetic units in the bacterial cytoplasm independent of chromosomes, facilitate bacterial growth. We searched the name of the strain in NCBI, utilizing “Nucleotide” as the search database. In the record page, we were able to access the strain-related plasmid information, which contains the name, description, sequence, and other relevant details of the plasmid, and we summarized the relevant content to obtain the information in Table 1. We found that 9 of these 149 strains contained plasmids, including DC22.2 which contained four plasmids.
High rates of antibiotic resistance were found in multiple Lactobacill species [15,16,17,18], including L. johnsonii [17]. Among them, tet(W/N/W) are the most widely distributed ARGs in L. johnsonii [18]. However, through an extensive literature search, we found that not much research has been done on the antibiotic resistance of L. johnsonii, which means that more research is needed to characterize the antibiotic resistance of L. johnsonii and to investigate the mechanisms of resistance and the possibility of transmission.
Table 1. Summary of plasmid prevalence in L. johnsonii.
Table 1. Summary of plasmid prevalence in L. johnsonii.
StrainBioSampleSize (Kb)RepliconsCDSRelease Date
FI9785SAMEA22724873.55 p9785S:NC_012552.1/AY862141.1 [19]2April 2009
FI9785SAMEA227248726.27 p9785L:NC_013505.1/FN357112.1 [20]26November 2009
BS15SAMN0463127745.84LJBSp1:NZ_CP016630.1/CP016630.143August 2016
UMNLJ22SAMN0457314627.88 pUMNLJ22_1:NZ_CP021705.1/CP021705.134June 2017
UMNLJ22SAMN0457314624.93 pUMNLJ22_2:NZ_CP021706.1/CP021706.124June 2017
UMNLJ21SAMN0457314521.52 pUMNLJ21_1:NZ_CP021701.1/CP021701.122June 2017
UMNLJ21SAMN0457314515.25 pUMNLJ21_2:NZ_CP021702.1/CP021702.120June 2017
pf01SAMN0246959726.46 pLJPF01L:CP024782.1 [21]0November 2017
pf01SAMN0246959714.24 pLJPF01S:CP024783.1 [21]0November 2017
LL8SAMN1326652177.56unnamed:NZ_CM019125.1/CM019125.173December 2019
DC22.2SAMN113719667.65 pLjDC22.2_1:NZ_CP039262.1/CP039262.110January 2020
DC22.2SAMN113719665.75 pLjDC22.2_2:NZ_CP039263.1/CP039263.13January 2020
DC22.2SAMN113719667.08 pLjDC22.1_3:NZ_CP039264.1/CP039264.14January 2020
DC22.2SAMN1137196613.77 pLjDC22.2_4:NZ_CP039265.1/CP039265.16January 2020
G2ASAMN11618738130.11 unnamed1:NZ_CP040855.1/CP040855.1154March 2020
G2ASAMN11618738108.72 unnamed2:NZ_CP040856.1/CP040856.1103March 2020
GHZ10aSAMN1613161413.65 unnamed1:NZ_CP062069.1/CP062069.115October 2020
GHZ10aSAMN1613161415.79 unnamed2:NZ_CP062070.1/CP062070.118October 2020

3. Effects of L. johnsonii on Different Diseases

Probiotics may affect the host through a variety of mechanisms, including enhancing the barrier effect of the intestinal epithelium [22,23,24]; regulating immune function [25,26]; producing organic acids [27], such as the production of oleic acid to play an anti-inflammatory role [28]; interacting with intestinal flora [29]; and interacting with the host through the cell surface structure [30]. Not all mechanisms have been confirmed in humans, nor do they exist in every probiotic strain [31]. The results of previous research we have collected indicate that the common mechanism of action of L. johnsonii in different diseases may include regulating immune function, interacting with intestinal flora, and improving barrier function (Figure 1). Table 2 summarizes some relevant studies and results in detail.

4. The Common Mechanism of L. johnsonii in Different Diseases

4.1. Respiratory Insults

Respiratory syncytial virus (RSV) infects nearly all infants by 2 years of age and is the leading cause of bronchiolitis in children worldwide [66]. Kei E. Fujimura et al. provided evidence that L. johnsonii supplementation significantly reduced the RSV-induced pulmonary responses [38], via immunomodulatory metabolites and altered immune function [14]. Their further study demonstrated that Lactobacillus modulation of the maternal microbiome enhanced airway protection against RSV in neonates. Their evidence was prenatal supplementation with L. johnsonii, which decreased inflammatory metabolites in maternal plasma and breastmilk, and offspring plasma, and resulted in a consistent gut microbiome in mothers and their offspring [12]. The experimental results of Chung-Ming Chen et al. showed that intranasal L. johnsonii administration improved lung development in hyperoxia-exposed neonatal mice [67].

4.2. Gastrointestinal Disease

L. johnsonii NCC 533 (first designed La1) (CNCM I-1225) (Nestlé, Switzerland), isolated from human intestinal microbiome, has been well characterized with regard to its potential antimicrobial effects against the major gastric and enteric bacterial pathogens and rotavirus [68]. Helicobacter pylori infections, colitis, Escherichia coli-induced diarrhea, and subclinical necrotizing colitis in farms were all possible results of L. johnsonii (Figure 2).
L. johnsonii La1 has been shown to exert an anti-inflammatory effect in many double-blind, placebo-controlled clinical trials as a drinkable, whey-based La1 culture supernatant [41], as acidified milk containing live La1 cells (LC-1) [39], or as a probiotic-containing dietary product [40,42] to H. pylori-positive asymptomatic volunteers. Dionyssios N. Sgouras et al. observed that a pronounced anti-inflammatory effect was exerted by La1 in particular on H. pylori-associated neutrophilic and lymphocytic infiltration [43], and a similar effect was found in L. johnsonii MH-68 [45]. In addition to the anti-inflammatory effect mentioned, there are other mechanisms that play a role. Some in vitro results suggest that GroEL proteins from La1 and other lactic acid bacteria might play a role in gastrointestinal homeostasis due to their ability to bind to components of the gastrointestinal mucosa and to aggregate H. pylori [69]. L. johnsonii La1 can also produce bacteriocins, which have inhibitory activity against the human gastric pathogen H. pylori [70], and its antibacterial activity was due to the production of lactic acid and (an) unknown inhibitory substance(s) [71]. However, it would seem highly unlikely that an actively secreted bacteriocin produced by La1 would retain activity, given the abundance of proteolytic activity present in the gastric epithelium [46]. L. johnsonii No. 1088, a novel strain that was isolated from the gastric juice of a healthy Japanese male volunteer, can inhibit the growth of H. pylori and suppress gastric acid secretion [44]. The role of such probiotic strains in the complex regulation of proinflammatory signal strength during early infection and other aspects need to be further identified.
Colitis refers to inflammatory lesions of the colon that occur for various reasons, as a broad concept, which can be subdivided into many categories, and it is a common intestinal disease. The main clinical manifestations are diarrhea, abdominal pain, mucus, and pus and blood stool, etc. Ding-Jia-Cheng Jia et al. uncovered that the abundance of L. johnsonii was lessened in colitis and identified that L. johnsonii relieved experimental colitis [49], drawing the same conclusions as Yunchang Zhang et al. [51]. Rogatien Charlet et al. also provided evidence that the mixed gavage of L. johnsonii and B. thetaiotaomicron alleviated acute colitis induced in mice [52]. In addition, L. johnsonii plays a role in the treatment of different E. coli-induced diarrhea, including enteroinvasive E. coli [48] and enterohemorrhagic E. coli [47], by modulating gut microbiota. In addition to E. coli-induced diarrhea, Keyuan Chen et al. demonstrated that L. johnsonii L531 helps to prevent Salmonella typhimurium-induced diarrhea in mice [72].
In the poultry industry, necrotic enteritis (NE), an enteric bacterial disease, significantly impacts the attempts to increase global poultry production, whereas the more prevalent subclinical form of NE (SNE) is usually difficult to detect, thereby causing considerable economic and profitability losses [73]. Hesong Wang et al. demonstrated in a previous study that feed supplementation with L. johnsonii BS15 may prevent the SNE-caused decrease in the growth performance of broilers [53]. The potential mechanisms include enhancing intestinal immunity and blood parameters related to immunity [54], decreasing fat deposition via adjusting the ratio of Firmicutes/Bacteroidetes in the gut [55], and influencing both lipid synthesis and catabolism in the liver [56]. RNA sequencing of gene expression extracted from liver samples also supported this mechanism [57]. In addition to adding L. johnsonii through feed, vaginal injection of L. johnsonii can modulate the mucosal barrier function and fallopian tube microbiota of laying hens, which may improve egg biosecurity [74].

4.3. Mental Health

The causes of mental health problems are complex. In recent years, many researchers have offered new insights into mental health problems from the perspective of the gut microbiome [75]. The association between the gut environment, host behavior, and potential psychobiotics/probiotics has been extensively investigated presently [76]. Studies have shown that L. johnsonii is a potentially beneficial bacterium that can improve memory impairment and modulate metabolism-related disorders through the brain–gut axis (Figure 3). The hippocampus is considered a crucial brain region in memory ability [77]; therefore, much of the research has focused on inducing hippocampus-related memory dysfunction in animal models and using this as a premise to identify potential psychobiotics or probiotics. In a mouse model of colitis, treatment with L. johnsonii restored the disturbed gut microbiome composition, lowered the gut microbiome, and attenuated memory impairment and colitis [78]. Ning Sun et al. demonstrated that L. johnsonii BS15 can prevent memory dysfunction induced by chronic high-fluorine intake through modulating the intestinal environment and improving gut development [59], and Jinge Xin et al. came to a similar conclusion [58]. Hesong Wang et al. concluded that L. johnsonii BS15 pretreatment enhanced intestinal health and prevented hippocampus-related memory dysfunction [30,31]. All of these indicate the psychoactive effects of L. johnsonii BS15 on positively influencing the brain–gut axis. In the description of mechanisms on how L. johnsonii BS15 yields positive psychiatric effects in psychopathology through the brain–gut axis, they all mentioned the intestinal barrier protective effects of this potential psychobiotic. The present results show that L. johnsonii BS15 pretreatment can reduce levels of TNF-α, IFN-γ, and IL-1β in the small intestines of mice. This result indicates the ability of L. johnsonii BS15 to protect the intestines from inflammation (development, digestive enzyme activities, and anti-inflammatory level). The results show that L. johnsonii BS15 can inhibit proinflammatory cytokines (TNF-α, IFN-γ, and IL-1β) or increase anti-inflammatory cytokines (IL-4 and IL-10) to maintain intestinal integrity [26,27].

4.4. Obesity

In the above reference to SNE, it was noted that L. johnsonii can decrease fat deposition in broiler chickens. This suggests to us that it may also have some beneficial effects on obesity. For rats on a high-fat diet, non-viable L. johnsonii JNU3402 (NV-LJ3402) [63], L. johnsonii N6.2, and blueberry phytophenols [62] can help correct diet-induced dyslipidemia. Another strain, L. johnsonii BFE6154, was also proved to protect against diet-induced hypercholesterolemia through the regulation of cholesterol metabolism in the intestine and liver [64]. In another species, Shaziling pigs, Jie Ma et al. found similar results, namely that L. johnsonii could promote lipid deposition and metabolism [79]. As obesity is a possible risk factor for diabetes, there are also some studies that have targeted diabetes, and found that a multi-strain probiotic supplement including L. johnsonii MH-104 [37], L. johnsonii MH-68 [36], and L. johnsonii N6.2 [33] can reduce diabetes in rats by reducing inflammation and other aspects [34,35,80,81].

4.5. Liver Diseases

There has been a rise in the prevalence of nonalcoholic fatty liver disease (NAFLD) and its more advanced stage, nonalcoholic steatohepatitis (NASH), and this rising disease prevalence will cause an increase in the number of patients with cirrhosis and end-stage liver disease [82]. Insulin resistance, mitochondrial dysfunction, and oxidative stress may all play a role in the disease’s pathogenesis [36,37]. Furthermore, NAFLD can be characterized by inflammation, hepatic steatosis, and hepatocyte apoptosis [83]. Jinge Xin et al. suggested that the treatment with L. johnsonii BS15 may prevent diet-induced NAFLD through adjusting gut flora; improving mitochondrial dysfunction; and reducing gut permeability, serum levels of LPS and IR, and inflammation [65]. Another research study of host glycolipid metabolism noted that L. johnsonii NCC 533 can increase the level of GSH in the serum of mice, boost mitochondrial morphology and function in the liver, reduce hepatic lipids, and improve systemic glucose metabolism [84].
The study on the protecting mechanism of Inonotus hispidus against acute alcoholic liver injury additionally mentioned its ability to upregulate L. johnsonii abundance to safeguard mice from acute alcoholic liver injury [85]. A similar potential mechanism of action was additionally seen in the BaWeiBaiDuSan (BWBDS) protection against sepsis-induced liver injury (SILI) in mice [11].
In addition to the above diseases, there are several studies targeting the treatment of other diseases. L. johnsonii NCC533 (La1) has recently been shown to protect against atopic dermatitis in mice if introduced during the weaning period [32]. L. johnsonii 6084 alleviated sepsis-induced organ injury by modulating gut microbiota. Vazquez-Munoz et al. discovered L. johnsonii had excellent probiotic properties and can prevent or treat mucocutaneous candidiasis [86], especially vulvovaginal candidiasis [87], and that L. johnsonii UBLJ01 is a potential candidate for vaginal probiotics [88]. L. johnsonii has also be found to delay osteoarthritis progression [89] and alleviate the development of acute myocardial infarction [90].

5. Conclusions and Perspectives

Our summary of recent studies on the effects of L. johnsonii on different diseases shows that L. johnsonii acts via a variety of means, including the modulation of immune function, interaction with resident microbiota, interfacing with the host, and improving gut barrier integrity. Although multiple mechanisms are probably co-expressed in a single probiotic, the generation of any given mechanism will depend on many factors, including the physiological state of the host, etc. Despite the complexity of the gastrointestinal tract microbiome, the presence or absence of specific bacterial species can dramatically alter the adaptive immune environment and intestinal environment, such as the different strains of L. johnsonii mentioned above. As various links between the intestinal microbiome and other organs, termed such as the “gut-lung axis” [91], have been proposed in recent years, more attention has begun to be focused on the deeper mechanisms of disease, and a number of researchers have suggested the impact of environmental exposures on the gastrointestinal microbiome, which in turn has an impact on host immunity and thus on the development of host diseases. This has led to greater attention to microbes as an intermediate factor when considering disease.
Bacterial antimicrobial resistance (AMR) to antibiotics has dramatically increased over the past few years due to the overuse of antibiotics, among other factors, and has already reached a level that poses a significant risk to future patients [92,93]. To combat the growing problem of AMR, there is a major dearth of research and development of new antibiotics; therefore, people must turn to other alternative medicines, such as probiotic-related products and their metabolites. Furthermore, if there are numerous pertinent research studies on the potential benefits of probiotics on a particular disease, the majority of them used rat models, which are insufficient to capture the complex variety of pathogenic changes that occur throughout the progression of human diseases. To fully understand the specific potential mechanism between probiotics, intestinal microecology, and disease, in-depth research is still needed, and more large-scale clinical trials are needed to evaluate the efficacy of probiotics in the treatment of disease and the safety of probiotics in the human body. From the above situation, we emphasize that L. johnsonii has broad application prospects in different diseases; however, we also need to consider some of these issues. First of all, since the host specificity of L. johnsonii is only known at the strain level, a question to ponder is whether animal and in vitro experiments can be extrapolated to humans themselves, and deeper mechanisms need to be studied with more human samples. In addition, we found that some researchers in experimental studies used live bacteria, but some researchers used dead bacteria. This may be due to the different culture conditions suitable for different strains, and the inability of some to survive in vitro for long periods of time. Therefore, it is equally important to study the active substances of dead functional probiotics for disease prevention. Indeed, several studies have been conducted on the safety of various strains of L. johnsonii [94,95,96], but some studies have not been conducted on toxicity in animal models, and more strains and further studies should be conducted. In addition to the application of L. johnsonii alone, a study last year added weight to the possible role of probiotics and functional materials in the treatment of disease [97]. Finally, it is worth noting that our summary results showed that not much research has been done on the antibiotic resistance of L. johnsonii, and the mechanisms of its production and transmission have been poorly studied; therefore, more detailed and in-depth studies may be needed.
In conclusion, L. johnsonii still has a bright future in research, especially in the areas of using both living and dead bacteria, as well as combining different biological materials, to treat or prevent disease. Nevertheless, more clinical research is required to pinpoint the precise process in various hosts and convert the findings into practical applications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms11102580/s1, Table S1: Metadata of Lactobacillus johnsonii.

Author Contributions

Z.Z. performed the literature review and drafted the manuscript. Z.L., L.Z. (Lei Zhang) and G.Z. contributed to the revision and editing of the manuscript. Z.Z., L.Z. (Lanlan Zhao), J.W. and Y.P. collected the literature and reviewed the text. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by the National Natural Science Foundation of China (Grant No. 82172320), China Postdoctoral Science Foundation (Grant No. 2022M711915), Shandong Provincial Natural Science Foundation (Grant No. ZR2022QC169), TaiShan Industrial Experts Program tscy20190612, and Shandong University Outstanding Young Scholars Program.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert Consensus Document. The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
  2. Kim, S.-Y.; Adachi, Y. Biological and genetic classification of canine intestinal lactic acid bacteria and bifidobacteria. Microbiol. Immunol. 2007, 51, 919–928. [Google Scholar] [CrossRef] [PubMed]
  3. Carina Audisio, M.; Torres, M.J.; Sabaté, D.C.; Ibarguren, C.; Apella, M.C. Properties of different lactic acid bacteria isolated from Apis mellifera L. Bee-gut. Microbiol. Res. 2011, 166, 1–13. [Google Scholar] [CrossRef] [PubMed]
  4. Duar, R.M.; Lin, X.B.; Zheng, J.; Martino, M.E.; Grenier, T.; Pérez-Muñoz, M.E.; Leulier, F.; Gänzle, M.; Walter, J. Lifestyles in Transition: Evolution and Natural History of the Genus Lactobacillus. FEMS Microbiol. Rev. 2017, 41, S27–S48. [Google Scholar] [CrossRef] [PubMed]
  5. Yan, W.; Sun, C.; Yuan, J.; Yang, N. Gut metagenomic analysis reveals prominent roles of Lactobacillus and cecal microbiota in chicken feed efficiency. Sci. Rep. 2017, 7, 45308. [Google Scholar] [CrossRef] [PubMed]
  6. Rhee, S.J.; Lee, J.-E.; Lee, C.-H. Importance of lactic acid bacteria in Asian fermented foods. Microb. Cell Fact. 2011, 10 (Suppl. S1), S5. [Google Scholar] [CrossRef]
  7. Andrada, E.; Mechoud, M.A.; Abeijón-Mukdsi, M.C.; Chagra Dib, E.P.; Cerviño, S.; Perez Chaia, A.; Medina, R.B. Ferulic acid esterase producing Lactobacillus johnsonii from goat feces as corn silage inoculants. Microorganisms 2022, 10, 1732. [Google Scholar] [CrossRef]
  8. Mager, L.F.; Burkhard, R.; Pett, N.; Cooke, N.C.A.; Brown, K.; Ramay, H.; Paik, S.; Stagg, J.; Groves, R.A.; Gallo, M.; et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 2020, 369, 1481–1489. [Google Scholar] [CrossRef]
  9. O’Callaghan, J.; O’Toole, P.W. Lactobacillus: Host-microbe relationships. Curr. Top. Microbiol. Immunol. 2013, 358, 119–154. [Google Scholar] [CrossRef]
  10. Buhnik-Rosenblau, K.; Matsko-Efimov, V.; Jung, M.; Shin, H.; Danin-Poleg, Y.; Kashi, Y. Indication for co-evolution of Lactobacillus johnsonii with its hosts. BMC Microbiol. 2012, 12, 149. [Google Scholar] [CrossRef]
  11. Fan, X.; Mai, C.; Zuo, L.; Huang, J.; Xie, C.; Jiang, Z.; Li, R.; Yao, X.; Fan, X.; Wu, Q.; et al. Herbal formula BaWeiBaiDuSan alleviates polymicrobial sepsis-induced liver injury via increasing the gut microbiota Lactobacillus johnsonii and regulating macrophage anti-inflammatory activity in mice. Acta Pharm. Sin. B 2023, 13, 1164–1179. [Google Scholar] [CrossRef]
  12. Fonseca, W.; Malinczak, C.-A.; Fujimura, K.; Li, D.; McCauley, K.; Li, J.; Best, S.K.K.; Zhu, D.; Rasky, A.J.; Johnson, C.C.; et al. Maternal gut microbiome regulates immunity to RSV infection in offspring. J. Exp. Med. 2021, 218, e20210235. [Google Scholar] [CrossRef]
  13. Mu, Q.; Zhang, H.; Liao, X.; Lin, K.; Liu, H.; Edwards, M.R.; Ahmed, S.A.; Yuan, R.; Li, L.; Cecere, T.E.; et al. Control of lupus nephritis by changes of gut microbiota. Microbiome 2017, 5, 73. [Google Scholar] [CrossRef] [PubMed]
  14. Fonseca, W.; Lucey, K.; Jang, S.; Fujimura, K.E.; Rasky, A.; Ting, H.-A.; Petersen, J.; Johnson, C.C.; Boushey, H.A.; Zoratti, E.; et al. Lactobacillus johnsonii supplementation attenuates respiratory viral infection via metabolic reprogramming and immune cell modulation. Mucosal Immunol. 2017, 10, 1569–1580. [Google Scholar] [CrossRef] [PubMed]
  15. Sharma, P.; Tomar, S.K.; Sangwan, V.; Goswami, P.; Singh, R. Antibiotic resistance of Lactobacillus sp. isolated from commercial probiotic preparations. J. Food Saf. 2016, 36, 38–51. [Google Scholar] [CrossRef]
  16. Guo, H.; Pan, L.; Li, L.; Lu, J.; Kwok, L.; Menghe, B.; Zhang, H.; Zhang, W. Characterization of antibiotic resistance genes from Lactobacillus isolated from traditional dairy products. J. Food Sci. 2017, 82, 724–730. [Google Scholar] [CrossRef] [PubMed]
  17. Dec, M.; Nowaczek, A.; Stępień-Pyśniak, D.; Wawrzykowski, J.; Urban-Chmiel, R. Identification and antibiotic susceptibility of Lactobacilli isolated from turkeys. BMC Microbiol. 2018, 18, 168. [Google Scholar] [CrossRef] [PubMed]
  18. Ma, Q.; Pei, Z.; Fang, Z.; Wang, H.; Zhu, J.; Lee, Y.; Zhang, H.; Zhao, J.; Lu, W.; Chen, W. Evaluation of tetracycline resistance and determination of the tentative microbiological cutoff values in lactic acid bacterial species. Microorganisms 2021, 9, 2128. [Google Scholar] [CrossRef]
  19. Horn, N.; Wegmann, U.; Narbad, A.; Gasson, M.J. Characterisation of a novel plasmid P9785S from Lactobacillus johnsonii FI9785. Plasmid 2005, 54, 176–183. [Google Scholar] [CrossRef]
  20. Wegmann, U.; Overweg, K.; Horn, N.; Goesmann, A.; Narbad, A.; Gasson, M.J.; Shearman, C. Complete genome sequence of Lactobacillus johnsonii FI9785, a competitive exclusion agent against pathogens in poultry. J. Bacteriol. 2009, 191, 7142–7143. [Google Scholar] [CrossRef]
  21. Lee, J.H.; Chae, J.P.; Lee, J.Y.; Lim, J.-S.; Kim, G.-B.; Ham, J.-S.; Chun, J.; Kang, D.-K. Genome sequence of Lactobacillus johnsonii PF01, isolated from piglet feces. J. Bacteriol. 2011, 193, 5030–5031. [Google Scholar] [CrossRef] [PubMed]
  22. Lyu, M.; Bai, Y.; Orihara, K.; Miyanaga, K.; Yamamoto, N. GAPDH released from Lactobacillus johnsonii MG enhances barrier function by upregulating genes associated with tight junctions. Microorganisms 2023, 11, 1393. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, S.; Li, Y.; Chu, B.; Yuan, L.; Liu, N.; Zhu, Y.; Wang, J. Lactobacillus johnsonii L531 alleviates the damage caused by salmonella typhimurium via inhibiting TLR4, NF-ΚB, and NLRP3 inflammasome signaling pathways. Microorganisms 2021, 9, 1983. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, X.; Xia, B.; He, T.; Li, D.; Su, J.-H.; Guo, L.; Wang, J.; Zhu, Y.-H. Oral administration of a select mixture of Lactobacillus and Bacillus alleviates inflammation and maintains mucosal barrier integrity in the ileum of pigs challenged with salmonella infantis. Microorganisms 2019, 7, 135. [Google Scholar] [CrossRef]
  25. Frei, R.; Akdis, M.; O’Mahony, L. Prebiotics, probiotics, synbiotics, and the immune system: Experimental data and clinical evidence. Curr. Opin. Gastroenterol. 2015, 31, 153–158. [Google Scholar] [CrossRef]
  26. van Baarlen, P.; Wells, J.M.; Kleerebezem, M. Regulation of intestinal homeostasis and immunity with probiotic lactobacilli. Trends Immunol. 2013, 34, 208–215. [Google Scholar] [CrossRef]
  27. Salazar, N.; Ruas-Madiedo, P.; Kolida, S.; Collins, M.; Rastall, R.; Gibson, G.; de Los Reyes-Gavilán, C.G. Exopolysaccharides produced by Bifidobacterium longum IPLA E44 and Bifidobacterium animalis subsp. Lactis IPLA R1 modify the composition and metabolic activity of human faecal microbiota in PH-controlled batch cultures. Int. J. Food Microbiol. 2009, 135, 260–267. [Google Scholar] [CrossRef]
  28. Charlet, R.; Le Danvic, C.; Sendid, B.; Nagnan-Le Meillour, P.; Jawhara, S. Oleic acid and palmitic acid from Bacteroides thetaiotaomicron and Lactobacillus johnsonii exhibit anti-inflammatory and antifungal properties. Microorganisms 2022, 10, 1803. [Google Scholar] [CrossRef]
  29. Ki Cha, B.; Mun Jung, S.; Hwan Choi, C.; Song, I.-D.; Woong Lee, H.; Joon Kim, H.; Hyuk, J.; Kyung Chang, S.; Kim, K.; Chung, W.-S.; et al. The effect of a multispecies probiotic mixture on the symptoms and fecal icrobiota in diarrhea-dominant irritable bowel syndrome: A randomized, double-blind, placebo-controlled trial. J. Clin. Gastroenterol. 2012, 46, 220–227. [Google Scholar] [CrossRef]
  30. Sanders, M.E.; Benson, A.; Lebeer, S.; Merenstein, D.J.; Klaenhammer, T.R. Shared mechanisms among probiotic taxa: Implications for general probiotic claims. Curr. Opin. Biotechnol. 2018, 49, 207–216. [Google Scholar] [CrossRef]
  31. Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef] [PubMed]
  32. Inoue, R.; Otsuka, M.; Nishio, A.; Ushida, K. Primary administration of Lactobacillus johnsonii NCC533 in weaning period suppresses the elevation of proinflammatory cytokines and CD86 gene expressions in skin lesions in NC/Nga mice. FEMS Immunol. Med. Microbiol. 2007, 50, 67–76. [Google Scholar] [CrossRef] [PubMed]
  33. Valladares, R.; Sankar, D.; Li, N.; Williams, E.; Lai, K.-K.; Abdelgeliel, A.S.; Gonzalez, C.F.; Wasserfall, C.H.; Larkin, J.; Schatz, D.; et al. Actobacillus johnsonii N6.2 mitigates the development of type 1 diabetes in BB-DP rats. PLoS ONE 2010, 5, e10507. [Google Scholar] [CrossRef] [PubMed]
  34. Lau, K.; Benitez, P.; Ardissone, A.; Wilson, T.D.; Collins, E.L.; Lorca, G.; Li, N.; Sankar, D.; Wasserfall, C.; Neu, J.; et al. Inhibition of type 1 diabetes correlated to a Lactobacillus johnsonii N6.2-mediated Th17 bias. J. Immunol. 2011, 186, 3538–3546. [Google Scholar] [CrossRef]
  35. Teixeira, L.D.; Kling, D.N.; Lorca, G.L. Lactobacillus johnsonii N6.2 diminishes caspase-1 maturation in the gastrointestinal system of diabetes prone rats. Benef. Microbes 2018, 9, 527–539. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, C.-H.; Yen, H.-R.; Lu, W.-L.; Ho, H.-H.; Lin, W.-Y.; Kuo, Y.-W.; Huang, Y.-Y.; Tsai, S.-Y.; Lin, H.-C. Adjuvant probiotics of Lactobacillus salivarius subsp. Salicinius AP-32, L. Johnsonii MH-68, and Bifidobacterium animalis subsp. Lactis CP-9 attenuate glycemic levels and inflammatory cytokines in patients with type 1 diabetes mellitus. Front. Endocrinol. 2022, 13, 754401. [Google Scholar] [CrossRef]
  37. Hsieh, P.-S.; Ho, H.-H.; Tsao, S.P.; Hsieh, S.-H.; Lin, W.-Y.; Chen, J.-F.; Kuo, Y.-W.; Tsai, S.-Y.; Huang, H.-Y. Multi-strain probiotic supplement attenuates streptozotocin-induced type-2 diabetes by reducing inflammation and β-cell death in rats. PLoS ONE 2021, 16, e0251646. [Google Scholar] [CrossRef]
  38. Fujimura, K.E.; Demoor, T.; Rauch, M.; Faruqi, A.A.; Jang, S.; Johnson, C.C.; Boushey, H.A.; Zoratti, E.; Ownby, D.; Lukacs, N.W.; et al. House dust exposure mediates gut microbiome Lactobacillus enrichment and airway immune defense against allergens and virus infection. Proc. Natl. Acad. Sci. USA 2014, 111, 805–810. [Google Scholar] [CrossRef]
  39. Felley, C.P.; Corthésy-Theulaz, I.; Rivero, J.L.; Sipponen, P.; Kaufmann, M.; Bauerfeind, P.; Wiesel, P.H.; Brassart, D.; Pfeifer, A.; Blum, A.L.; et al. Favourable effect of an acidified milk (LC-1) on Helicobacter pylori gastritis in man. Eur. J. Gastroenterol. Hepatol. 2001, 13, 25–29. [Google Scholar] [CrossRef]
  40. Gotteland, M.; Cruchet, S. Suppressive effect of frequent ingestion of Lactobacillus johnsonii La1 on Helicobacter pylori colonization in asymptomatic volunteers. J. Antimicrob. Chemother. 2003, 51, 1317–1319. [Google Scholar] [CrossRef]
  41. Michetti, P.; Dorta, G.; Wiesel, P.H.; Brassart, D.; Verdu, E.; Herranz, M.; Felley, C.; Porta, N.; Rouvet, M.; Blum, A.L.; et al. Effect of whey-based culture supernatant of Lactobacillus acidophilus (johnsonii) La1 on Helicobacter pylori infection in humans. Digestion 1999, 60, 203–209. [Google Scholar] [CrossRef] [PubMed]
  42. Cruchet, S.; Obregon, M.C.; Salazar, G.; Diaz, E.; Gotteland, M. Effect of the ingestion of a dietary product containing Lactobacillus johnsonii La1 on Helicobacter pylori colonization in children. Nutrition 2003, 19, 716–721. [Google Scholar] [CrossRef] [PubMed]
  43. Sgouras, D.N.; Panayotopoulou, E.G.; Martinez-Gonzalez, B.; Petraki, K.; Michopoulos, S.; Mentis, A. Lactobacillus johnsonii La1 attenuates Helicobacter Pylori-associated gastritis and reduces levels of proinflammatory chemokines in C57BL/6 mice. Clin. Diagn. Lab. Immunol. 2005, 12, 1378–1386. [Google Scholar] [CrossRef] [PubMed]
  44. Aiba, Y.; Nakano, Y.; Koga, Y.; Takahashi, K.; Komatsu, Y. A highly acid-resistant novel strain of Lactobacillus johnsonii No. 1088 has antibacterial activity, including that against Helicobacter Pylori, and inhibits gastrin-mediated acid production in mice. Microbiologyopen 2015, 4, 465–474. [Google Scholar] [CrossRef] [PubMed]
  45. Hsieh, P.-S.; Tsai, Y.-C.; Chen, Y.-C.; Teh, S.-F.; Ou, C.-M.; King, V.A.-E. Eradication of Helicobacter pylori infection by the probiotic strains Lactobacillus johnsonii MH-68 and L. Salivarius ssp. Salicinius AP-32. Helicobacter 2012, 17, 466–477. [Google Scholar] [CrossRef]
  46. Pantoflickova, D.; Corthésy-Theulaz, I.; Dorta, G.; Stolte, M.; Isler, P.; Rochat, F.; Enslen, M.; Blum, A.L. Favourable effect of regular intake of fermented milk containing Lactobacillus johnsonii on Helicobacter pylori associated gastritis. Aliment. Pharmacol. Ther. 2003, 18, 805–813. [Google Scholar] [CrossRef]
  47. Hu, Y.; Zhao, M.; Lu, Z.; Lv, F.; Zhao, H.; Bie, X.L. johnsonii, L. plantarum, and L. rhamnosus alleviated enterohaemorrhagic Escherichia coli-induced diarrhoea in mice by regulating gut microbiota. Microb. Pathog. 2021, 154, 104856. [Google Scholar] [CrossRef]
  48. Wang, Y.; Li, A.; Liu, J.; Mehmood, K.; Wangdui, B.; Shi, H.; Luo, X.; Zhang, H.; Li, J.L. Pseudomesenteroides and L. johnsonii isolated from yaks in Tibet modulate gut microbiota in mice to ameliorate enteroinvasive Escherichia coli-induced diarrhea. Microb. Pathog. 2019, 132, 1–9. [Google Scholar] [CrossRef]
  49. Jia, D.-J.-C.; Wang, Q.-W.; Hu, Y.-Y.; He, J.-M.; Ge, Q.-W.; Qi, Y.-D.; Chen, L.-Y.; Zhang, Y.; Fan, L.-N.; Lin, Y.-F.; et al. Lactobacillus johnsonii alleviates colitis by TLR1/2-STAT3 mediated CD206+ macrophagesIL-10 activation. Gut Microbes 2022, 14, 2145843. [Google Scholar] [CrossRef]
  50. Fukushima, Y.; Miyaguchi, S.; Yamano, T.; Kaburagi, T.; Iino, H.; Ushida, K.; Sato, K. Improvement of nutritional status and incidence of infection in hospitalised, enterally fed elderly by feeding of fermented milk containing probiotic Lactobacillus johnsonii La1 (NCC533). Br. J. Nutr. 2007, 98, 969–977. [Google Scholar] [CrossRef]
  51. Zhang, Y.; Mu, T.; Yang, Y.; Zhang, J.; Ren, F.; Wu, Z. Lactobacillus johnsonii attenuates citrobacter rodentium-induced colitis by regulating inflammatory responses and endoplasmic reticulum stress in mice. J. Nutr. 2021, 151, 3391–3399. [Google Scholar] [CrossRef] [PubMed]
  52. Charlet, R.; Bortolus, C.; Sendid, B.; Jawhara, S. Bacteroides thetaiotaomicron and Lactobacillus johnsonii modulate intestinal inflammation and eliminate fungi via enzymatic hydrolysis of the fungal cell wall. Sci. Rep. 2020, 10, 11510. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, H.; Ni, X.; Qing, X.; Liu, L.; Lai, J.; Khalique, A.; Li, G.; Pan, K.; Jing, B.; Zeng, D. Probiotic enhanced intestinal immunity in broilers against subclinical necrotic enteritis. Front. Immunol. 2017, 8, 1592. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, H.; Ni, X.; Qing, X.; Liu, L.; Xin, J.; Luo, M.; Khalique, A.; Dan, Y.; Pan, K.; Jing, B.; et al. Probiotic Lactobacillus johnsonii BS15 improves blood parameters related to immunity in broilers experimentally infected with subclinical necrotic enteritis. Front. Microbiol. 2018, 9, 49. [Google Scholar] [CrossRef] [PubMed]
  55. Qing, X.; Zeng, D.; Wang, H.; Ni, X.; Liu, L.; Lai, J.; Khalique, A.; Pan, K.; Jing, B. Preventing subclinical necrotic enteritis through Lactobacillus johnsonii BS15 by ameliorating lipid metabolism and intestinal microflora in broiler chickens. AMB Express 2017, 7, 139. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, H.; Ni, X.; Qing, X.; Zeng, D.; Luo, M.; Liu, L.; Li, G.; Pan, K.; Jing, B. Live probiotic Lactobacillus johnsonii BS15 promotes growth performance and lowers fat deposition by improving lipid metabolism, intestinal development, and gut microflora in broilers. Front. Microbiol. 2017, 8, 1073. [Google Scholar] [CrossRef]
  57. Khalique, A.; Zeng, D.; Wang, H.; Qing, X.; Zhou, Y.; Xin, J.; Zeng, Y.; Pan, K.; Shu, G.; Jing, B.; et al. Transcriptome analysis revealed ameliorative effect of probiotic Lactobacillus johnsonii BS15 against subclinical necrotic enteritis induced hepatic inflammation in broilers. Microb. Pathog. 2019, 132, 201–207. [Google Scholar] [CrossRef]
  58. Xin, J.; Wang, H.; Sun, N.; Bughio, S.; Zeng, D.; Li, L.; Wang, Y.; Khalique, A.; Zeng, Y.; Pan, K.; et al. Probiotic alleviate fluoride-induced memory impairment by reconstructing gut microbiota in mice. Ecotoxicol. Environ. Saf. 2021, 215, 112108. [Google Scholar] [CrossRef]
  59. Sun, N.; Ni, X.; Wang, H.; Xin, J.; Zhao, Y.; Pan, K.; Jing, B.; Zeng, D. Probiotic Lactobacillus johnsonii BS15 prevents memory dysfunction induced by chronic high-fluorine intake through modulating intestinal environment and improving gut development. Probiotics Antimicrob. Proteins 2020, 12, 1420–1438. [Google Scholar] [CrossRef]
  60. Wang, H.; He, S.; Xin, J.; Zhang, T.; Sun, N.; Li, L.; Ni, X.; Zeng, D.; Ma, H.; Bai, Y. Psychoactive effects of Lactobacillus johnsonii against restraint stress-induced memory dysfunction in mice through modulating intestinal inflammation and permeability-a study based on the gut-brain axis hypothesis. Front. Pharmacol. 2021, 12, 662148. [Google Scholar] [CrossRef]
  61. Wang, H.; Sun, Y.; Xin, J.; Zhang, T.; Sun, N.; Ni, X.; Zeng, D.; Bai, Y. Lactobacillus johnsonii BS15 prevents psychological stress-induced memory dysfunction in mice by modulating the gut-brain axis. Front. Microbiol. 2020, 11, 1941. [Google Scholar] [CrossRef] [PubMed]
  62. Teixeira, L.D.; Torrez Lamberti, M.F.; DeBose-Scarlett, E.; Bahadiroglu, E.; Garrett, T.J.; Gardner, C.L.; Meyer, J.L.; Lorca, G.L.; Gonzalez, C.F. Lactobacillus johnsonii N6.2 and blueberry phytophenols affect lipidome and gut microbiota composition of rats under high-fat diet. Front. Nutr. 2021, 8, 757256. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, G.; Hong, E.; Oh, S.; Kim, E. Non-viable Lactobacillus johnsonii JNU3402 protects against diet-induced obesity. Foods 2020, 9, 1494. [Google Scholar] [CrossRef] [PubMed]
  64. Yoon, H.; Lee, Y.; Park, H.; Kang, H.-J.; Ji, Y.; Holzapfel, W.H. Lactobacillus johnsonii BFE6154 ameliorates diet-induced hypercholesterolemia. Probiotics Antimicrob. Proteins 2021, 15, 451–459. [Google Scholar] [CrossRef] [PubMed]
  65. Xin, J.; Zeng, D.; Wang, H.; Ni, X.; Yi, D.; Pan, K.; Jing, B. Preventing non-alcoholic fatty liver disease through Lactobacillus johnsonii BS15 by attenuating inflammation and mitochondrial injury and improving gut environment in obese mice. Appl. Microbiol. Biotechnol. 2014, 98, 6817–6829. [Google Scholar] [CrossRef] [PubMed]
  66. Openshaw, P.J.; Dean, G.S.; Culley, F.J. Links between respiratory syncytial virus bronchiolitis and childhood asthma: Clinical and research approaches. Pediatr. Infect. Dis. J. 2003, 22, S58–S64. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, C.-M.; Yang, Y.-C.S.H.; Chou, H.-C.; Lin, S. Intranasal administration of Lactobacillus johnsonii attenuates hyperoxia-induced lung injury by modulating gut microbiota in neonatal mice. J. Biomed. Sci. 2023, 30, 57. [Google Scholar] [CrossRef]
  68. Liévin-Le Moal, V.; Servin, A.L. Anti-infective activities of Lactobacillus strains in the human intestinal microbiota: From probiotics to gastrointestinal anti-infectious biotherapeutic agents. Clin. Microbiol. Rev. 2014, 27, 167–199. [Google Scholar] [CrossRef]
  69. Bergonzelli, G.E.; Granato, D.; Pridmore, R.D.; Marvin-Guy, L.F.; Donnicola, D.; Corthésy-Theulaz, I.E. GroEL of Lactobacillus johnsonii La1 (NCC 533) is cell surface associated: Potential role in interactions with the host and the gastric pathogen Helicobacter pylori. Infect. Immun. 2006, 74, 425–434. [Google Scholar] [CrossRef]
  70. Avonts, L.; De Vuyst, L. Antimicrobial potential of probiotic lactic acid bacteria. Mededelingen (Rijksuniv. Te Gent. Fak. Van Landbouwkd. En Toegepaste Biol. Wet.) 2001, 66, 543–550. [Google Scholar]
  71. Makras, L.; Triantafyllou, V.; Fayol-Messaoudi, D.; Adriany, T.; Zoumpopoulou, G.; Tsakalidou, E.; Servin, A.; De Vuyst, L. Kinetic analysis of the antibacterial activity of probiotic Lactobacilli towards salmonella enterica serovar typhimurium reveals a role for lactic acid and other inhibitory compounds. Res. Microbiol. 2006, 157, 241–247. [Google Scholar] [CrossRef]
  72. Chen, K.; Wang, J.; Guo, L.; Wang, J.; Yang, L.; Hu, T.; Zhao, Y.; Wang, X.; Zhu, Y. Lactobacillus johnsonii L531 ameliorates Salmonella enterica Serovar Typhimurium diarrhea by modulating iron homeostasis and oxidative stress via the IRP2 pathway. Nutrients 2023, 15, 1127. [Google Scholar] [CrossRef]
  73. Skinner, J.T.; Bauer, S.; Young, V.; Pauling, G.; Wilson, J. An economic analysis of the impact of subclinical (mild) necrotic enteritis in broiler chickens. Avian Dis. 2010, 54, 1237–1240. [Google Scholar] [CrossRef]
  74. Nii, T.; Shinkoda, T.; Isobe, N.; Yoshimura, Y. Intravaginal injection of Lactobacillus johnsonii may modulates oviductal microbiota and mucosal barrier function of laying hens. Poult. Sci. 2023, 102, 102699. [Google Scholar] [CrossRef]
  75. Schmidt, C. Thinking from the gut. Nature 2015, 518, S12–S14. [Google Scholar] [CrossRef] [PubMed]
  76. Sarkar, A.; Harty, S.; Lehto, S.M.; Moeller, A.H.; Dinan, T.G.; Dunbar, R.I.M.; Cryan, J.F.; Burnet, P.W.J. The microbiome in psychology and cognitive neuroscience. Trends Cogn. Sci. 2018, 22, 611–636. [Google Scholar] [CrossRef] [PubMed]
  77. Knierim, J.J. The hippocampus. Curr. Biol. 2015, 25, R1116–R1121. [Google Scholar] [CrossRef] [PubMed]
  78. Jang, S.-E.; Lim, S.-M.; Jeong, J.-J.; Jang, H.-M.; Lee, H.-J.; Han, M.J.; Kim, D.-H. Gastrointestinal inflammation by gut microbiota disturbance induces memory impairment in mice. Mucosal Immunol. 2018, 11, 369–379. [Google Scholar] [CrossRef] [PubMed]
  79. Ma, J.; Duan, Y.; Li, R.; Liang, X.; Li, T.; Huang, X.; Yin, Y.; Yin, J. Gut microbial profiles and the role in lipid metabolism in shaziling pigs. Anim. Nutr. 2022, 9, 345–356. [Google Scholar] [CrossRef] [PubMed]
  80. Yamano, T.; Tanida, M.; Niijima, A.; Maeda, K.; Okumura, N.; Fukushima, Y.; Nagai, K. Effects of the probiotic strain Lactobacillus johnsonii strain La1 on autonomic nerves and blood glucose in rats. Life Sci. 2006, 79, 1963–1967. [Google Scholar] [CrossRef] [PubMed]
  81. Kingma, S.D.K.; Li, N.; Sun, F.; Valladares, R.B.; Neu, J.; Lorca, G.L. 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, 1023–1028. [Google Scholar] [CrossRef] [PubMed]
  82. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef] [PubMed]
  83. Powell, E.E.; Wong, V.W.-S.; Rinella, M. Non-alcoholic fatty liver disease. Lancet 2021, 397, 2212–2224. [Google Scholar] [CrossRef] [PubMed]
  84. Rodrigues, R.R.; Gurung, M.; Li, Z.; García-Jaramillo, M.; Greer, R.; Gaulke, C.; Bauchinger, F.; You, H.; Pederson, J.W.; Vasquez-Perez, S.; et al. Transkingdom interactions between Lactobacilli and hepatic mitochondria attenuate western diet-induced diabetes. Nat. Commun. 2021, 12, 101. [Google Scholar] [CrossRef] [PubMed]
  85. Yuan, Y.; Wu, F.; Zhang, F.; Li, X.; Wu, X.; Fu, J. Hepatoenteric protective effect of melanin from Inonotus hispidus on acute alcoholic liver injury in mice. Mol. Nutr. Food Res. 2023, 67, e2200562. [Google Scholar] [CrossRef] [PubMed]
  86. Vazquez-Munoz, R.; Thompson, A.; Russell, J.T.; Sobue, T.; Zhou, Y.; Dongari-Bagtzoglou, A. Insights from the Lactobacillus johnsonii genome suggest the production of metabolites with antibiofilm activity against the pathobiont candida albicans. Front. Microbiol. 2022, 13, 853762. [Google Scholar] [CrossRef]
  87. ElFeky, D.S.; Awad, A.R.; Shamseldeen, A.M.; Mowafy, H.L.; Hosny, S.A. Comparing the therapeutic potentials of Lactobacillus johnsonii vs. Lactobacillus acidophilus against vulvovaginal candidiasis in female rats: An in vivo study. Front. Microbiol. 2023, 14, 1222503. [Google Scholar] [CrossRef]
  88. Ahire, J.J.; Sahoo, S.; Kashikar, M.S.; Heerekar, A.; Lakshmi, S.G.; Madempudi, R.S. In vitro assessment of Lactobacillus crispatus UBLCp01, Lactobacillus gasseri UBLG36, and Lactobacillus johnsonii UBLJ01 as a potential vaginal probiotic candidate. Probiotics Antimicrob. Proteins 2023, 15, 275–286. [Google Scholar] [CrossRef]
  89. Liu, R.; Zhou, Y.; Chen, H.; Xu, H.; Zuo, M.; Chen, B.; Wang, H. Membrane vesicles from Lactobacillus johnsonii delay osteoarthritis progression via modulating macrophage glutamine synthetase/MTORC1 axis. Biomed. Pharmacother. 2023, 165, 115204. [Google Scholar] [CrossRef]
  90. Zhong, X.; Zhao, Y.; Huang, L.; Liu, J.; Wang, K.; Gao, X.; Zhao, X.; Wang, X. Remodeling of the gut microbiome by Lactobacillus johnsonii alleviates the development of acute myocardial infarction. Front. Microbiol. 2023, 14, 1140498. [Google Scholar] [CrossRef]
  91. Dang, A.T.; Marsland, B.J. Microbes, metabolites, and the gut-lung axis. Mucosal Immunol. 2019, 12, 843–850. [Google Scholar] [CrossRef] [PubMed]
  92. Carlet, J.; Jarlier, V.; Harbarth, S.; Voss, A.; Goossens, H.; Pittet, D.; The Participants of the 3rd World Healthcare-Associated Infections Forum. Ready for a World without Antibiotics? The pensières antibiotic resistance call to action. Antimicrob. Resist. Infect. Control 2012, 1, 11. [Google Scholar] [CrossRef] [PubMed]
  93. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Lancet 2022, 399. [CrossRef]
  94. Boucard, A.-S.; Florent, I.; Polack, B.; Langella, P.; Bermúdez-Humarán, L.G. Genome sequence and assessment of safety and potential probiotic traits of Lactobacillus johnsonii CNCM I-4884. Microorganisms 2022, 10, 273. [Google Scholar] [CrossRef] [PubMed]
  95. Adedeji, O.E.; Chae, S.A.; Ban, O.-H.; Bang, W.Y.; Kim, H.; Jeon, H.J.; Chinma, C.E.; Yang, J.; Jung, Y.H. Safety evaluation and anti-inflammatory activity of Lactobacillus johnsonii IDCC 9203 isolated from feces of breast-fed infants. Arch Microbiol 2022, 204, 470. [Google Scholar] [CrossRef] [PubMed]
  96. Gotteland, M.; Cires, M.J.; Carvallo, C.; Vega, N.; Ramirez, M.A.; Morales, P.; Rivas, P.; Astudillo, F.; Navarrete, P.; Dubos, C.; et al. Probiotic screening and safety evaluation of Lactobacillus strains from plants, artisanal goat cheese, human stools, and breast milk. J. Med. Food 2014, 17, 487–495. [Google Scholar] [CrossRef] [PubMed]
  97. Ma, N.; Guo, P.; Chen, J.; Qi, Z.; Liu, C.; Shen, J.; Sun, Y.; Chen, X.; Chen, G.-Q.; Ma, X. Poly-β-hydroxybutyrate alleviated diarrhea and colitis via Lactobacillus johnsonii biofilm-mediated maturation of sulfomucin. Sci. China Life Sci. 2022, 66, 1569–1588. [Google Scholar] [CrossRef]
Figure 1. The common mechanism of L. johnsonii in different diseases. L. johnsonii acts on various parts of the body, including the brain, lungs, liver, stomach, and small intestine, as well as adipose tissue, by modulating immune function, interacting with the intestinal flora, and improving barrier functions. Created with https://www.biorender.com (accessed on 8 March 2023).
Figure 1. The common mechanism of L. johnsonii in different diseases. L. johnsonii acts on various parts of the body, including the brain, lungs, liver, stomach, and small intestine, as well as adipose tissue, by modulating immune function, interacting with the intestinal flora, and improving barrier functions. Created with https://www.biorender.com (accessed on 8 March 2023).
Microorganisms 11 02580 g001
Figure 2. The role played by L. johnsonii for different gastrointestinal diseases. L. johnsonii is beneficial for H. pylori infection, colitis, Escherichia coli-induced diarrhea, and subclinical necrotizing colitis on farms. Created with https://www.biorender.com (accessed on 8 March 2023).
Figure 2. The role played by L. johnsonii for different gastrointestinal diseases. L. johnsonii is beneficial for H. pylori infection, colitis, Escherichia coli-induced diarrhea, and subclinical necrotizing colitis on farms. Created with https://www.biorender.com (accessed on 8 March 2023).
Microorganisms 11 02580 g002
Figure 3. The role played by L. johnsonii through the brain–gut axis. L. johnsonii indirectly prevents hippocampus-related memory dysfunction by affecting normal gut microbes and inhibiting gut inflammatory responses. Created with https://www.biorender.com (accessed on 8 March 2023).
Figure 3. The role played by L. johnsonii through the brain–gut axis. L. johnsonii indirectly prevents hippocampus-related memory dysfunction by affecting normal gut microbes and inhibiting gut inflammatory responses. Created with https://www.biorender.com (accessed on 8 March 2023).
Microorganisms 11 02580 g003
Table 2. L. johnsonii showed improvement in disease in different animal experiments or clinical studies.
Table 2. L. johnsonii showed improvement in disease in different animal experiments or clinical studies.
StrainDiseaseExperimental ModelsDuration of InterventionTreatment ResultsReference
La1 (NCC533)ADAtopic dermatitis NC/Nga miceFrom 20 to 22 days of ageIL-8↓, IL-12↓, IL-23↓, CD86↓[32]
N6.2T1DBB-DP ratsPre-weaning to 1 day old during mother feeding and post-weaning at 21 days oldiNOS↓, IFNγ↓, Cox-2↑, claudin↑, occludin↓[33]
N6.2T1DBB-DP ratsDaily until sacrifice at diabetes onset, or the culmination of the experiment at 140 daysIL-17↑, IL-23↑[34]
N6.2BB-DP ratsAfter weaning to 60 days of agemature caspase-1↓, IL-1β↓[35]
La1HyperglycemiaSTZ-induced diabetes animal model and hyperglycemia model induced by intracranial injection of 2DG2 weeksplasma glucose↓, glucagon levels↓[34]
A multi-strain probiotic supplement including L. johnsonii MH-68T1DPatients with T1D24 weeksfasting blood glucose↓, HbA1c↓, IL-8↓, IL-17↓, MIP-1β↓, RANTES↓, TNF-α↓, TGF-β1↑[36]
A multi-strain probiotic supplement including L. johnsonii MH-68T2DSTZ -induced diabetes animal model8 weeksTNF-α↓, IL-6↓, IL-1β↓, β-cell mass↑[37]
L. johnsoniiAllergic or infectious airway diseaseA similar experimental design as the CRA airway challenge model and RSV infection modelIL-4↓, IL-5↓, IL-13↓, IL-17↓, CD11c/CD11b and CD11c/CD8, as well as CD69 activated CD4 and CD8 T cells↓[38]
L. johnsoniiRSVRSV infection model1 weekIL-4↓, IL-5↓, IL-13↓, IL-6↓, IL-1b↓, TNFα↓, IFNβ↑, DHA↑, AcedoPC↑[14]
L. johnsoniiRSVRSV infection modelIL-4↓, IL-5↓, IL-13↓, IL-17↓, Gob5(mucogenic gene mRNA level) ↓Th2↓, IFN-γ↑[12]
La1H. pylori-associated gastritisHealthy adult volunteers of both genders infected by H. pylori3 weeks[39]
La1H. pylori-associated gastritisHealthy adult volunteers of both genders infected by H. pylori2 weeksδ13CO2 over baseline (DOB)↓[40]
La1H. pylori infectionHealthy adult volunteers of both genders infected by H. pylori2 weeks[41]
La1H. pylori infectionAsymptomatic school children4 weeks[42]
La1H. pylori-associated gastritisH. pylori SS1 strain infection mode in C57BL/6 mice3 monthsanti-H. pylori IgG antibody titers↓[43]
No. 1088H. pylori infectionMale germ-free Balb/c mice2 weeks or 4 weeks[44]
MH-68H. pylori infectionSPF SD male rats4 weeks[45]
L. johnsoniiH. pylori-associated gastritisHealthy adult volunteers of both genders infected by H. pylori16 weeksH. pylori density↓[46]
NJ13Enterohaemorrhagic E. coli-induced diarrhoeaFemale mice[47]
LJ1Enteroinvasive E. coli-induced diarrheaKM mice8-22days[48]
L. johnsoniiUCDSS-induced chronic colitis mice model and human sampleIL10↑, TLR1/2↑, MRC1↑[49]
La1 (NCC533)Completely enterally fed elderly in-patients aged over 70 years12 weeksserum albumin↑, Blood Hb↑, blood phagocytic activity↑, TNF-α↓[50]
L. johnsoniiRodentium-Induced colitisfemale C57BL/6J mice2 weeksCD4, CD8, CD11b, F4/80, TNF-α, IL-1β, IL-6, IL-17A, ssMCP1, Cox2↓[51]
L. johnsonii + B. thetaiotaomicronColitisDSS-induced chronic colitis mice model5 daysIgA↑, IL-1β↓, IL-10↑, TLR9↑, TLR↓, MBL-C↓[52]
BS15SNEBroiler chickens (Cobb 500)days 1-28 or days 29-42CD4↑, CD4/CD8↑, sIgA in ileum↑[53]
BS15SNEBroiler chickens (Cobb 500)28days or 42daysSOD↑, CAT↑, IHR↑, T-AOC↑, IgG↑ and IgA↑ in serum, IFN-γ↑, CD3CD4 percentage↑, CD3CD4/CD3CD8↑[54]
BS15SNEBroiler chickens (Cobb 500)4 weeksALT↓, AST↓, TC↓, HDL-C↑, PPARγ and ATGL↑ in adipose tissue, IGF-1 and EGF↑ in jejunum and ileum, ACC, FAS and SREBP-1c↓ inhepatic expressions, PPARα and CPT-1↑ in hepatic expressions[55]
BS15SNEBroiler chickens (Cobb 500)6 weeksHDL-C↑, TG↓, LDL-C↓, SREBP-1c and FAS↓ in hepatic expressions[56]
BS15SNE induced hepatic inflammationBroiler chickens (Cobb 500)4 weeksFOS↓[57]
BS15Fluoride-induced memory impairmentMale ICR mice98 daysBDNF↑, CREB↑, Bcl-xl↑, Bad↓[58]
BS15Memory dysfunction Induced by chronic high-fluorine intakeMale ICR mice98 daysmRNA levels of Dbn, MAP-2, and SYP↑, T-AOC, and GSH-Px↑ in hippocampuls, sIgA ↓ in the jejunal mucosa, MDA↑, SOD↑, CAT activities↑, GSH↑[59]
BS15Memory dysfunction in mice after RS5C7BL/6J male mice4 weeksthe mRNA expression levels of BDNF, CREB, SCF, c-Fos, and NMDAR↑, DA, 5-HT, and Ach levels↑, the mRNA expression level of IL-4↑, GABA↑, mRNA expression levels of bcl-2 and Bcl-xL↑[60]
BS15Psychological stress-induced memory dysfunctionWAS in ICR male mice4 weeksmRNA-expression levels of tight junction proteins claudin-1, occludin, and ZO-1 in the jejunum and ileum↑, TNF-α↓, IFN-γ↓, and IL-1β↓, mRNA levels of BDNF↑, CREB↑[61]
N6.2 and BBDiet-induced obesityHFD model15 weeksSREBP-1↓, SCAP↓, LCFA in the serum↑[62]
JNU3402Diet-induced obesityHFD model C57BL/6J mice14 weeksACOX↑, CPT1↑, PGC1α↑, PPARγ↑, TG↓, FAS↓, ACC↓, SREBP1c↓, hepatic cholesterol level↓[63]
BFE6154Diet-induced hypercholesterolemiaHFHCD model C57BL/6J mice4 weeksLDL↓, ABCG8↓, NPC1L1↓, ABCG5↑, HDL↑, LDLR↑[64]
BS15NAFLDHFD model ICR mice17 weeksLPS↓, TG↓, LDLC↓, ALT↓, FFA↓, HDLC↓, UCP-2 and cytochrome c↓ in mitochondria, the hepatic expression of Acc 1, Fas, TNFα and PPARγ↓, the hepatic expression of Fiaf↑[65]
2DG: 2-deoxy-D-glucose; ABCG5/8: ATP-binding cassette (ABC) transporters G5 and G8; ACC: acetyl-CoA carboxylase; AcedoPC: 1-docosahexaenoylglycerophosphocholine; ACOX: acetyl-CoA oxidase; AD: atopic dermatitis; ALT: alanine aminotransferase; AST: aspartate transaminase; Bad: Bcl-xL/Bcl-2 associated death promoter; BB: blueberry phytophenols; BB-DP: BioBreeding diabetes-prone; Bcl-xl: B-cell lymphoma-extra large; BDNF: brain-derived neurotrophic factor; CAT: catalase; COX-2: Cyclooxygenase-2; CPT1: carnitine palmitoyltransferase 1; CRA: cockroach allergen; CREB: cAMP response element-binding protein; Dbn: developmentally regulated brain protein; DSS: dextran sulfate sodium; EGF: epidermal growth factor; FOS: proto-oncogene protein; FAS: fatty acid synthase; FFA: free fatty acid; GSH-Px: glutathione peroxidase; Hb: hemoglobin; HbA1c: H glycated hemoglobin; HDL-C: high-density lipoprotein cholesterol; HFD: high-fat diet; HFHCD: high-fat and high-cholesterol diet; IFN-γ: Interferon-gamma; IGF-1: insulin-like growth factors-1; IHR: inhibition of hydroxy radical; iNOS: inducible nitric oxide synthase; LCFA: long chain fatty acids; LDL-C: low-density lipoprotein cholesterol; LDLR: low-density lipoprotein receptor; LPS: lipopolysaccharide; MAP-2: microtubule-associated protein; MCP1: monocyte chemoattractant protein-1; MDA: malondialdehyde; NAFLD: non-alcoholic fatty liver disease; NPC1L1: NiemannPick C1-Like1; PGC1α: peroxisome proliferator-activated receptor gamma coactivator 1-α; PPARγ: Peroxisome proliferator-activated receptor gamma; T1D: Type 1 diabetes; T2D: Type 2 diabetes; T-AOC: total antioxidation capacity; TG: triglyceride; TGF-β1: transforming growth factor-β1; TLR1/2: toll-like receptor1/2; TNF-α: tumor necrosis factor alpha; RS: restraint stress; RSV: respiratory viral infection; SNE: subclinical (mild) necrotic enteritis; SOD: superoxide dismutase; STZ: Streptozotocin; SPF: specific pathogen free; SREBP-1: sterol regulatory element-binding protein 1; SCAP: SREBP cleavage-activating protein; SD: Sprague–Dawley; SYP: synaptophysin; UC: ulcerative colitis; WAS: water-avoidance stress; “↑” and “↓” represent the indicators of increase and decrease in the host after probiotic treatment, respectively.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, Z.; Zhao, L.; Wu, J.; Pan, Y.; Zhao, G.; Li, Z.; Zhang, L. The Effects of Lactobacillus johnsonii on Diseases and Its Potential Applications. Microorganisms 2023, 11, 2580. https://doi.org/10.3390/microorganisms11102580

AMA Style

Zhang Z, Zhao L, Wu J, Pan Y, Zhao G, Li Z, Zhang L. The Effects of Lactobacillus johnsonii on Diseases and Its Potential Applications. Microorganisms. 2023; 11(10):2580. https://doi.org/10.3390/microorganisms11102580

Chicago/Turabian Style

Zhang, Ziyi, Lanlan Zhao, Jiacheng Wu, Yingmiao Pan, Guoping Zhao, Ziyun Li, and Lei Zhang. 2023. "The Effects of Lactobacillus johnsonii on Diseases and Its Potential Applications" Microorganisms 11, no. 10: 2580. https://doi.org/10.3390/microorganisms11102580

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop