Skip to main content
  • Research article
  • Open access
  • Published:

Lactobacillus paracasei feeding improves the control of secondary experimental meningococcal infection in flu-infected mice

Abstract

Background

The use of probiotics to improve anti-microbial defence, such as for influenza infections, is increasingly recommended. However, no data are available on the effect of probiotics on flu-associated secondary bacterial infections. There is strong evidence of a spatiotemporal association between influenza virus infection and invasive Neisseria meningitidis. We thus investigated the effect of feeding mice Lactobacillus paracasei CNCM I-1518 in a mouse model of sequential influenza-meningococcal infection.

Methods

We intranasally infected BALB/c mice with a strain of influenza A virus (IAV) H3N2 that was first adapted to mice. Seven days later, a secondary bacterial infection was induced by intranasal administration of bioluminescent N. meningitidis. During the experiment, mice orally received either L. paracasei CNCM I-1518 or PBS as a control. The effect of L. paracasei administration on secondary bacterial infection by N. meningitidis was evaluated.

Results

Oral consumption of L. paracasei CNCM I-1518 reduced the weight loss of infected mice and lowered the bioluminescent signal of infecting meningococci. This improvement was associated with higher recruitment of inflammatory myeloid cells, such as interstitial monocytes and dendritic cells, to the lungs.

Conclusions

Our data highlight the role of the gut-lung axis. L. paracasei CNCM I-1518 may boost the defence against IAV infection and secondary bacterial infection, which should be further studied and validated in clinical trials.

Peer Review reports

Background

Bacterial and viral respiratory infections are responsible for severe morbidity and mortality in children and adults worldwide. Among them, influenza virus is a major source of severe viral respiratory infections in adults, causing annual epidemics that result in significant morbidity and mortality. Flu pandemics during the twentieth century killed more than 100 million people [1]. Flu morbidity is often due to secondary respiratory infections by bacteria, such as Streptococcus pneumoniae and Neisseria meningitidis [2, 3]. Epidemiological studies have clearly shown a spatiotemporal association between influenza A virus (IAV) and invasive N. meningitidis infections [2, 4, 5]. Both infections show a seasonal pattern with most of the cases in winter. Moreover, the winter peaks of invasive meningococcal disease usually follow the peak of influenza-like syndromes [6]. This sequential pattern of flu infection and invasive meningococcal infection has been reproduced in a murine animal model [7]. The mechanisms that enhance bacterial superinfection appear to be multifactorial. Alterations in physical and immunological barriers, as well as changes in the microenvironment, have been shown to contribute to the development of secondary bacterial infections [8]. It has also been suggested that the neuraminidases (NA) of IAV enhance the adhesion process of N. meningitidis to the respiratory epithelium by direct interaction with the sialic-acid containing capsules that constitute a substrate for the NA of IAV. Cleavage of capsular sialic acid at the bacterial surface may unravel sub-capsular meningococcal adhesins and enhance meningococcal adhesion to epithelial cells [6].

Several clinical trials have suggested a positive effect of preventive feeding of probiotics on respiratory infections in children and the elderly [9, 10]. The administration of Lactobacillus species enhances protection against influenza virus infection in mice [11,12,13,14]. We have recently shown that daily consumption of Lactobacillus paracasei CNCM I-1518 allowed better control of IAV infection in a BALB/c mouse model, most likely through the modulation of the immune response in the respiratory system [15]. Here, we aimed to use the murine model of sequential flu-meningococcal infection to explore the effect of feeding mice L. paracasei CNCM I-1518 on the secondary bacterial infection.

Methods

Bacterial and viral strains

Lactobacillus paracasei strain CNCM I-1518 was cultured and prepared using identical conditions as previously described [15]. MRS (Man, Rogosa, Sharpe) broth (DIFCO laboratories Detroit, MI, USA) was used. Bacteria were harvested, washed and prepared in sterile PBS (Fisher Scientific, Graffenstaden, France) at optical density 600Ā nm (OD600nm) of 1 (Spectrometer, Biochrom, France). The bacterial suspension was stored at āˆ’ā€‰80Ā Ā°C in aliquots until use.

Neisseria meningitidis (LNP24198lux), a bioluminescent derivative of strain 24,198 (serogroup C), was cultured on Gonococcal (GC) Base (GCB) agar medium (DIFCO laboratories Detroit, MI, USA) containing the Kellogg supplements [16]. IAV, (A/Scotland/20/74 [H3N2]), adapted to mice, was prepared, stored in aliquots at āˆ’ā€‰80Ā Ā°C, and thawed prior to infection, as previously described [7].

Mice

Six-week old female BALB/c mice were purchased (Janvier, Genest-Saint-Isle, France) and housed for one week prior to the experiment in a biosafety containment facility under the same previously described conditions [15].

Probiotic treatment and viral and secondary bacterial infection

Three experiments were separately conducted. In each experiment, two groups of 10 and nine mice received 200Ā Ī¼l containing 2ā€‰Ć—ā€‰108 colony forming units (CFU)/mouse of L. paracasei CNCM I-1518 or 200Ā Ī¼l PBS (control), respectively, daily by oral gavage. Mice were infected by intranasal infection with IAV on day 0 with 50Ā Ī¼l H3N2 virus (260 plaque forming units, PFU) as previously described [15]. For this purpose, mice were first anesthetized by intraperitoneal injection with a mixture of xylazine and ketamine (MERIAL, Villeurbanne, France). Seven days after IAV infection (day 7), mice were superinfected in the airways with N. meningitidis (107Ā CFU/ mouse in 50Ā Ī¼l of a bacterial suspension at 2ā€‰Ć—ā€‰108 CFU/ml). The infection was performed by the strain LNP24198lux, a serogroup C strain belonging to the hyperinvasive clonal complex ST-11 [17]. The infection was followed by dynamic imaging until 48Ā h after bacterial infection (Fig.Ā 1a) as previously described [18]. Oral gavage with L. paracasei CNCM I-1518 was performed during the entire period of viral and secondary infection.

Fig. 1
figure 1

Effects of L. paracasei consumption on the health status of influenza/N. meningitidis-infected mice. a Schematic representation of the experimental design. b Body-weight loss. c Mortality. d Scores for the appearance of the mice after influenza infection as follows: 3; smooth coat, 2; patches of fur showing piloerection, 1; most of the fur showing piloerection, and 0; mouse appears ā€œpuffyā€. Results are expressed as the meanā€‰Ā±ā€‰SEM for each group (nā€‰=ā€‰50). (*pā€‰<ā€‰0.05, **Pā€‰<ā€‰0.01, ***Pā€‰<ā€‰0.001)

The health status, appearance, and survival of the mice were followed after IAV infection from day 0 to day 16 with identical scoring system as previously described [15, 19]. Survival of the mice was scored and represented as percent survival.

Mice were euthanatized at the end of experiment by injection of a high dose of a mixture of 10Ā mg/Kg Xylazine (Bayer, Puteaux, France) and 5Ā mg/Kg of Ketamine (Merial, Lyon, France). After perfusion with 3Ā ml PBS to wash out blood, the lungs were removed, homogenized, divided into two lots, and the homogenates used to perform the cytokine assay and flow cytometry analysis as previously described [15].

Quantification of N. meningitidis (bioluminescence imaging)

Mice infected intranasally with bioluminescent N. meningitidis were anesthetized with a constant flow of 2.5% isoflurane mixed with oxygen, using an XGI-8 anaesthesia induction chamber (Xenogen Corp.). Bacterial infection images were acquired using an IVIS 100 system (Xenogen Corp., Alameda, CA) according to the instructions of the manufacturer. Analysis and acquisition were performed using Living ImageĀ 4.3.1 software (Xenogen Corp.). Images were acquired using a 1-min as previously described [18]. An uninfected mouse under the same conditions of acquisition was used to subtract the background.

Determination of cytokine levels

Several cytokines (MCP1, IL10, KC, IL12p70, and IL6) were quantified in whole-lung homogenates from mice sacrificed on day 9 (48Ā h after N. meningitidis infection) in one part of the lung homogenates as mentioned above that was cleared by centrifugation at 5000 x g for 10Ā min at 4Ā Ā°C and the supernatants used for cytokine assays using an ELISA kit (R&D Systems, AbcamĀ®), according to the manufacturerā€™s instructions and the data were expressed in pg/ml of of total protein of the lungs as previously described [15].

Antibodies and flow cytometry

The second part of whole-lung homogenates were used to prepare single-cell suspensions were using treatment with collagenase IV (Sigma-Aldrich, St. Quentin Fallavier, France) and ACK (Ammonium-Chloride-Potassium) Lysing Buffer (Fisher Scientific, Graffenstaden, France) as previously described [15, 20]. Analysis was then performed using a LSR Fortessa Flow Cytometer (BD Biosciences, San Jose, CA, USA) and the results analysed with FlowJo software (v10; Tree Star, Ashland OR, USA). The antibodies and the gating strategies used are depicted in the AdditionalĀ fileĀ 1: Figure S1.

Statistical analysis

Data are expressed as the mean sĀ± SEM. Studentā€™s t tests were used to analyse group differences. Values of pā€‰<ā€‰0.05 were considered to be statistically significant.

Results

Effect of L. paracasei CNCM I-1518 on the health status of mice with a secondary bacterial infection

We first assessed the effect of oral administration of L. paracasei CNCM I-1518 on secondary meningococcal infection. Mice were infected intranasally with H3N2 influenza virus at day 0 and superinfected intranasally with N. meningitidis seven days later (Fig. 1a).

After infection with influenza virus, the infected mice rapidly lost weight, but mice fed L. paracasei lost significantly less weight than those given PBS, as measured on days 1, 4, 6, and 7 post-infection (Fig. 1b). The mice also appeared to be ill, according to the clinical score based on the appearance of their fur. The clinical scores for the mice fed L. paracasei were significantly better than those of the PBS-fed mice at days 3, 4, 5, 6, and 7 post-infection (Fig. 1c). Following N. meningitidis infection (on day 7), the weight loss continued in both groups until day 9. However, the weight loss was less in the L. paracasei group and their general health was better (Fig. 1b and c). Moreover, the L. paracasei group showed better survival (90%) at day 9 that the PBS group (75%) (Fig. 1d). Theses data suggest that consumption of L. paracasei before N. meningitidis infection is associated with an improvement in the health of the mice.

Effect of L. paracasei consumption on N. meningitidis respiratory infection

N. meningitidis infection was followed by dynamic bioluminescence imaging. The quantification of bacterial load was performed in IAV-infected mice for both the L. paracasei and PBS groups by following the total photons per second emitted by each mouse 0.5, 3, 6, 24, and 48Ā h after infection (Fig.Ā 2).

Fig. 2
figure 2

Dissemination of N. meningitidis in BALB/c-flu infected mice. Sequential IAV (250 PFU per mouse) and meningococcal infection (107Ā CFU per mouse) were performed by the intranasal route. Bacterial infection was analysed by bioluminescence at the indicated times. Images depict photographs overlaid with colour representations of luminescence intensity, measured in total photons/s and indicated on the scales, in which red is the most intense and blue the least intense. a Ventral views of nine PBS-fed and 10Ā L. paracasei-fed mice. A non-infected mouse was added as a control. b. The luminescence was quantified and expressed as the means Ā± SEM for each category at the indicated times by defining specific representative regions of interest encompassing the entire animal

Dynamic bioluminescence imaging 0.5Ā h after intranasal inoculation showed the bacteria to be present in the upper respiratory tract of infected mice and already starting to reach the lower respiratory pathways and lungs. The infection signal increased 3 and 6Ā h after N. meningitidis infection for both groups of mice (L. paracasei-fed and PBS-fed mice). The infection appeared to become established in the lower respiratory tract and lungs (Fig. 2a), with no significant difference between the two groups of infected mice. The bioluminescent signals decreased after 24 and 48Ā h of meningococcal infection. However, the signals were significantly lower in the L. paracasei fed-mice than the PBS-fed mice after 24Ā h of meningococcal infection. The bioluminescent signal was also lower, although not significantly, in the L. paracasei fed-mice than the PBS-fed mice after 48Ā h of infection (Fig. 2b). These data suggest gavage with L. paracasei was associated with faster clearance of the secondary meningococcal infection.

Effect of L. paracasei consumption on cytokine profiles in the lungs

We next investigated the effect of L. paracasei on the inflammatory status by quantifying the levels of several cytokines (MCP1, IL10, KC, IL12p70, and IL6) in the lungs of mice fed L. paracasei or PBS (Fig.Ā 3) sacrificed on day 9 (48Ā h after N. meningitidis infection). The cytokine levels in the lungs are expressed as the amount of cytokine per mg of total protein of the tested lungs.

Fig. 3
figure 3

Effects of L. paracasei consumption on cytokine profiles relative to those of the control (PBS) group after 48Ā h of secondary meningococcal infection. Results are expressed as the meanā€‰Ā±ā€‰SEM for each group (nā€‰=ā€‰20). Cytokines for which the differences between the two groups were significant are indicated by a star

The levels of the inflammatory cytokines IL6, MCP1, KC, and IL12p70 were higher in the lungs of L. paracasei-fed than PBS-fed mice. However, only the differences in the levels of IL6 and MCP1 reached significance (Fig. 3). In addition, although the levels of the anti-inflammatory cytokine IL10 were higher in the L. paracasei than PBS group, the difference was not significant (Fig. 3).

Effect of L. paracasei consumption on immune-cell recruitment to the lungs

Innate and adaptive immune responses play a key role in controlling infection. We assessed the effect of L. paracasei on immune responses by quantifying total cell counts in the lungs at day 9, 48Ā h after N. meningitidis infection, using trypan blue. The cell counts in the lungs of the mice fed L. paracasei were significantly higher than those of the PBS-fed mice (Fig.Ā 4a).

Fig. 4
figure 4

Effect of L. paracasei consumption on immune-cell recruitment to the lungs relative to that of the control (PBS) group after 48Ā h of secondary meningococcal infection. Results are expressed as the meanā€‰Ā±ā€‰SEM for each group (nā€‰=ā€‰16). Cells for which the differences between the two groups were significant are indicated by a star

Flow cytometry analysis (Fig. 4b) revealed a significantly more dendritic cells, neutrophils, and inflammatory monocytes in the lungs of L. paracasei-fed mice sacrificed at day 9 (48Ā h after N. meningitidis infection) than those of the control group (PBS-fed mice). Although, there were more interstitial macrophages, patrolling monocytes, and eosinophils in the lungs of L. paracasei-fed mice sacrificed at day 9 than in control mice, the difference did not reach significance.

The flow cytometry gating strategies for immune cells are shown in the Additional file 1: Figure S1.

Discussion

Respiratory viral infections are responsible for significant morbidity and mortality in children and adults worldwide. They also predispose individuals to secondary bacterial infections by altering the innate and adaptive immune systems [21]. Enhancing immune function through the consumption of probiotics could thus improve the outcomes of such infections [22]. We have already shown that L. paracasei CNCMI-1518 may exert such benefits by modulating host immune responses during IAV infection in a mouse model [15].

Here, we further show that feeding mice L. paracasei CNCM I-1518 not only improves the health status of mice infected with IAV, but also improves the health status of mice infected with N. meningitis after IAV infection. The reduction in the severity of secondary meningococcal infection may be mainly due to better control of the primary flu infection, allowing better control of the secondary bacterial infection. However, better direct clearance of N. meningitidis may also be possible through higher recruitment of neutrophils, inflammatory monocytes, and DCs (Fig. 4). We previously suggested that L. paracasei consumption induced early recruitment of immune cells, including alveolar macrophages, which may allow better control of the flu infection [15]. A role of alveolar macrophages in the protection against secondary pneumococcal infection after flu was suggested by the association between the early depletion of alveolar macrophages in the lungs and lethal pneumococcal pneumonia in flu-infected mice [23].

Indeed, we have previously reported that mice fed L. paracasei CNCM I-1518 prior to flu infection showed early activation of pro-inflammatory cytokines (IL-1Ī±, IL-1Ī²) and the massive recruitment of immune cells to the lungs (mainly DCs, macrophages, monocytes, and eosinophils) [15]. Such early recruitment may also be responsible for the faster clearance of meningococci 24Ā h after a secondary meningococcal infection. Moreover, we also observed differential modulation of the immune response in the lungs 48Ā h after secondary bacterial infection in mice fed L. paracasei CNCMI-1518 relative to control mice, represented by larger numbers of dendritic cells, inflammatory monocytes, and neutrophils. This may have also contributed to the control of the secondary meningococcal infection. Recent data supports a role for monocytes/macrophages in susceptibility to meningococcal sepsis in a murine model [24]. Similarly, nasal priming with viable Corynebacterium pseudodiphtheriticum 090104 differentially modulated the TLR3-mediated innate antiviral immune response in the respiratory tract of infant mice. This improved resistance to primary infection by the respiratory syncytial virus (RSV) and secondary pneumococcal pneumonia [25]. Oral administration of the probiotic Lactococcus lactis to mice with simultaneous vaccination with pneumococcal protective protein A was shown to reduce lung colonisation by S. pneumoniae [26]. Feeding mice the probiotic Lactobacillus rhamnosus CRL1505 (Lr05) has been reported to decrease the number of S. pneumoniae in the lung and prevent dissemination in the blood. This decrease was associated with increased IL6 and IL10 levels in bronchoalveolar lavages [27], consistent with our results. Mice treated with L. rhamnosus GG showed significantly better seven-day survival than saline-treated control mice in another experimental model of Pseudomonas aeruginosa-induced pneumonia [28]. This model also reveal the gut-lung axis and suggests that probiotic (L. rhamnosus GG) consumption may help to maintain the homeostasis of mucosal barriers [29]. Indeed, the gut microbiota has been reported to improve the outcome of experimental pneumococcal pneumonia in mice, as suggested by increased bacterial dissemination, inflammation, organ damage, and mortality in microbiota-depleted mice [30]. This beneficial effect of the gut microbiota may be due to the enhancement of primary alveolar macrophage function [30].

Conclusions

Our data add a new facet to the impact of probiotic consumption, as they suggest a positive effect by improving recovery from a secondary bacterial infection following an influenzae infection. These observations are highly relevant as primary influenza infection enhances the effects of several types of secondary bacterial infections, such as those by N. meningitidis, staphylococci, pneumococci, streptococci, and Haemophilus influenzae [2, 31,32,33]. Probiotics may help to relieve the burden of such secondary bacterial infections.

Abbreviations

CFU:

colony forming unit

CNCM:

Collection Nationale de Culture de Microorganismes

IAV:

influenza A virus

NA:

neuraminidase

PFU:

plaque forming unit

References

  1. Morens DM, Fauci AS. The 1918 influenza pandemic: insights for the 21st century. J Infect Dis. 2007;195(7):1018ā€“28.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  2. Hubert B, Watier L, Garnerin P, Richardson S. Meningococcal disease and influenza-like syndrome: a new approach to an old question. J Infect Dis. 1992;166(3):542ā€“5.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  3. McCullers JA. Insights into the interaction between influenza virus and pneumococcus. Clin Microbiol Rev. 2006;19(3):571ā€“82.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  4. Abramson JS. The pathogenesis of bacterial infections in infants and children: the role of viruses. Perspect Biol Med. 1988;32(1):63ā€“72.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Moore PS, Hierholzer J, DeWitt W, Gouan K, Djore D, Lippeveld T, Plikaytis B, Broome CV. Respiratory viruses and mycoplasma as cofactors for epidemic group a meningococcal meningitis. JAMA. 1990;264(10):1271ā€“5.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  6. Rameix-Welti MA, Zarantonelli ML, Giorgini D, Ruckly C, Marasescu M, van der Werf S, Alonso JM, Naffakh N, Taha MK. Influenza a virus neuraminidase enhances meningococcal adhesion to epithelial cells through interaction with sialic acid-containing meningococcal capsules. Infect Immun. 2009;77(9):3588ā€“95.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  7. Alonso JM, Guiyoule A, Zarantonelli ML, Ramisse F, Pires R, Antignac A, Deghmane AE, Huerre M, van der Werf S, Taha MK. A model of meningococcal bacteremia after respiratory superinfection in influenza a virus-infected mice. FEMS Microbiol Lett. 2003;222(1):99ā€“106.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  8. Bellinghausen C, Rohde GG, Savelkoul PH, Wouters EF, Stassen FR. Viral-bacterial interactions in the respiratory tract. J Gen Virol. 2016;97(12):3089ā€“102.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  9. Ozen M, Kocabas Sandal G, Dinleyici EC. Probiotics for the prevention of pediatric upper respiratory tract infections: a systematic review. Expert Opin Biol Ther. 2015;15(1):9ā€“20.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  10. Guillemard E, Tondu F, Lacoin F, Schrezenmeir J. Consumption of a fermented dairy product containing the probiotic lactobacillus casei DN-114001 reduces the duration of respiratory infections in the elderly in a randomised controlled trial. Br J Nutr. 2010;103(1):58ā€“68.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  11. Harata G, He F, Hiruta N, Kawase M, Kubota A, Hiramatsu M, Yausi H. Intranasal administration of Lactobacillus rhamnosus GG protects mice from H1N1 influenza virus infection by regulating respiratory immune responses. Lett Appl Microbiol. 2010;50(6):597ā€“602.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  12. Kechaou N, Chain F, Gratadoux JJ, Blugeon S, Bertho N, Chevalier C, Le Goffic R, Courau S, Molimard P, Chatel JM, et al. Identification of one novel candidate probiotic lactobacillus plantarum strain active against influenza virus infection in mice by a large-scale screening. Appl Environ Microbiol. 2013;79(5):1491ā€“9.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  13. Lee YN, Youn HN, Kwon JH, Lee DH, Park JK, Yuk SS, Erdene-Ochir TO, Kim KT, Lee JB, Park SY, et al. Sublingual administration of lactobacillus rhamnosus affects respiratory immune responses and facilitates protection against influenza virus infection in mice. Antivir Res. 2013;98(2):284ā€“90.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  14. Youn HN, Lee DH, Lee YN, Park JK, Yuk SS, Yang SY, Lee HJ, Woo SH, Kim HM, Lee JB, et al. Intranasal administration of live Lactobacillus species facilitates protection against influenza virus infection in mice. Antivir Res. 2012;93(1):138ā€“43.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  15. Belkacem N, Serafini N, Wheeler R, Derrien M, Boucinha L, Couesnon A, Cerf-Bensussan N, Gomperts Boneca I, Di Santo JP, Taha MK, et al. Lactobacillus paracasei feeding improves immune control of influenza infection in mice. PLoS One. 2017;12(9):e0184976.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  16. Kellogg DS Jr, Peacock WL Jr, Deacon WE, Brown L, Pirkle DI. Neisseria gonorrhoeae. I. Virulence genetically linked to clonal variation. J Bacteriol. 1963;85:1274ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  17. Guiddir T, Deghmane AE, Giorgini D, Taha MK. Lipocalin 2 in cerebrospinal fluid as a marker of acute bacterial meningitis. BMC Infect Dis. 2014;14(1):276.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  18. Szatanik M, Hong E, Ruckly C, Ledroit M, Giorgini D, Jopek K, Nicola MA, Deghmane AE, Taha MK. Experimental meningococcal sepsis in congenic transgenic mice expressing human transferrin. PLoS One. 2011;6(7):e22210.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  19. Shrum B, Anantha RV, Xu SX, Donnelly M, Haeryfar SM, McCormick JK, Mele T. A robust scoring system to evaluate sepsis severity in an animal model. BMC Res Notes. 2014;7:233.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  20. Klein Wolterink RG, Kleinjan A, van Nimwegen M, Bergen I, de Bruijn M, Levani Y, Hendriks RW. Pulmonary innate lymphoid cells are major producers of IL-5 and IL-13 in murine models of allergic asthma. Eur J Immunol. 2012;42(5):1106ā€“16.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  21. Smith CM, Sandrini S, Datta S, Freestone P, Shafeeq S, Radhakrishnan P, Williams G, Glenn SM, Kuipers OP, Hirst RA, et al. Respiratory syncytial virus increases the virulence of Streptococcus pneumoniae by binding to penicillin binding protein 1a. A new paradigm in respiratory infection. Am J Respir Crit Care Med. 2014;190(2):196ā€“207.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  22. Tada A, Zelaya H, Clua P, Salva S, Alvarez S, Kitazawa H, Villena J. Immunobiotic Lactobacillus strains reduce small intestinal injury induced by intraepithelial lymphocytes after toll-like receptor 3 activation. Inflamm Res. 2016;65(10):771ā€“83.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  23. Ghoneim HE, Thomas PG, McCullers JA. Depletion of alveolar macrophages during influenza infection facilitates bacterial superinfections. J Immunol. 2013;191(3):1250ā€“9.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  24. Wang X, Zhang D, Sjolinder M, Wan Y, Sjolinder H. CD46 accelerates macrophage-mediated host susceptibility to meningococcal sepsis in a murine model. Eur J Immunol. 2017;47(1):119ā€“30.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  25. Kanmani P, Clua P, Vizoso-Pinto MG, Rodriguez C, Alvarez S, Melnikov V, Takahashi H, Kitazawa H, Villena J. Respiratory commensal Bacteria Corynebacterium pseudodiphtheriticum improves resistance of infant mice to respiratory syncytial virus and Streptococcus pneumoniae superinfection. Front Microbiol. 2017;8:1613.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  26. Vintini E, Villena J, Alvarez S, Medina M. Administration of a probiotic associated with nasal vaccination with inactivated Lactococcus lactis-PppA induces effective protection against pneumoccocal infection in young mice. Clin Exp Immunol. 2010;159(3):351ā€“62.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  27. Salva S, Villena J, Alvarez S. Immunomodulatory activity of Lactobacillus rhamnosus strains isolated from goat milk: impact on intestinal and respiratory infections. Int J Food Microbiol. 2010;141(1ā€“2):82ā€“9.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  28. Khailova L, Baird CH, Rush AA, McNamee EN, Wischmeyer PE. Lactobacillus rhamnosus GG improves outcome in experimental pseudomonas aeruginosa pneumonia: potential role of regulatory T cells. Shock. 2013;40(6):496ā€“503.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  29. Khailova L, Baird CH, Rush AA, Barnes C, Wischmeyer PE. Lactobacillus rhamnosus GG treatment improves intestinal permeability and modulates inflammatory response and homeostasis of spleen and colon in experimental model of Pseudomonas aeruginosa pneumonia. Clin Nutr. 2016;36(6):1549ā€“57.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  30. Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa E Melo F, Roelofs JJ, de Boer JD, Hoogendijk AJ, de Beer R, de Vos A, Belzer C et al: The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut 2016;65(4):575ā€“583.

  31. Jakab GJ, Warr GA, Knight ME. Pulmonary and systemic defenses against challenge with Staphylococcus aureus in mice with pneumonia due to influenza a virus. J Infect Dis. 1979;140(1):105ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  32. Mahmud MI, Jennings R, Potter CW. The infant rat as a model for assessment of the attenuation of human influenza viruses. J Med Microbiol. 1979;12(1):43ā€“54.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  33. McCullers JA, Rehg JE. Lethal synergism between influenza virus and Streptococcus pneumoniae: characterization of a mouse model and the role of platelet-activating factor receptor. J Infect Dis. 2002;186(3):341ā€“50.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

Download references

Funding

The study was supported by the grants CI IMMUNOBIOTIC 1307014/00 IRT.

BAP301 and Danone 30000221, as well as the Institut Pasteur. Danone Nutricia Research provided the Lactobacilli strains and performed the microbiota analysis. The funders had no role in study design, the decision to publish, or preparation of the manuscript.

Availability of data and materials

The data that support the findings of this study are available on request from the corresponding author [MKT].

Author information

Authors and Affiliations

Authors

Contributions

MKT directed the work with the contribution of NB and RBS. NB and MKT performed the experiments and all authors participated in the data analysis. MKT drafted the manuscript and all authors read, edited, and approved the manuscript.

Corresponding author

Correspondence to Muhamed-Kkeir Taha.

Ethics declarations

Ethics approval

This study was carried out in strict accordance with European Union Directive 2010/63/EU (and its revision, 86/609/EEC) on the protection of animals used for scientific purposes. Our laboratory has the administrative authorization for animal experimentation (Number 2013ā€“0109; Protocol 99ā€“174) and the protocol was approved by the Institut Pasteur Review Board, which is part of the Regional Ethics Committee for Animal Experimentation of the Paris Region (Permit Number: 75ā€“1554). All invasive procedures were performed under anaesthesia and all possible efforts were made to minimize animal suffering.

Competing interests

R.B-S is an employee of Danone Nutricia Research. NB and MKT have no competing interests.

Publisherā€™s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional file

Additional file 1:

Figure S1. The figure describes the flow cytometry analysis of immune cells in the lungs: The used antibodies and the gating strategies. (PDF 71 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Belkacem, N., Bourdet-Sicard, R. & Taha, MK. Lactobacillus paracasei feeding improves the control of secondary experimental meningococcal infection in flu-infected mice. BMC Infect Dis 18, 167 (2018). https://doi.org/10.1186/s12879-018-3086-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12879-018-3086-9

Keywords