Next Article in Journal
Yeast Diversity Associated with the Phylloplane of Corn Plants Cultivated in Thailand
Next Article in Special Issue
Interactions between Kazachstania humilis Yeast Species and Lactic Acid Bacteria in Sourdough
Previous Article in Journal
Reuterin Demonstrates Potent Antimicrobial Activity Against a Broad Panel of Human and Poultry Meat Campylobacter spp. Isolates
Previous Article in Special Issue
Schizosaccharomyces pombe Can Reduce Acetic Acid Produced by Baijiu Spontaneous Fermentation Microbiota
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages In Vitro

by
Luz María Rocha-Ramírez
1,*,
Beatriz Hernández-Ochoa
2,
Saúl Gómez-Manzo
3,
Jaime Marcial-Quino
4,
Noemí Cárdenas-Rodríguez
5,
Sara Centeno-Leija
6 and
Mariano García-Garibay
7
1
Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Secretaría de Salud Dr. Márquez No. 162, Col Doctores, Delegación Cuauhtémoc, Ciudad de México 06720, Mexico
2
Laboratorio de Inmunoquímica y Biología Celular, Hospital Infantil de México Federico Gómez, Secretaría de Salud. Dr. Márquez No. 162, Col Doctores, Delegación Cuauhtémoc, Ciudad de México 06720, Mexico
3
Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaria de Salud, Ciudad de México 04530, Mexico
4
Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico
5
Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico
6
Consejo Nacional Ciencia y Tecnologia (CONACYT) Laboratorio de Agrobiotecnología, Tecnoparque CLQ, Universidad de Colima, Carretera Los Limones-Loma de Juárez, Colima 28629, Mexico
7
Departamento de Ciencias de la Alimentación, Unidad Lerma, Universidad Autónoma Metropolitana, Av. San Rafael Atlixco No. 186. Col Vicentina, Ciudad de México 09340, Mexico
*
Author to whom correspondence should be addressed.
Microorganisms 2020, 8(1), 79; https://doi.org/10.3390/microorganisms8010079
Submission received: 23 November 2019 / Revised: 2 January 2020 / Accepted: 5 January 2020 / Published: 7 January 2020
(This article belongs to the Special Issue Feature Papers in Food Microbiology)

Abstract

:
Most Lactobacillus species have beneficial immunological (“immunoprobiotic”) effects in the host. However, it is unclear how probiotic bacteria regulate immune responses. The present study investigated the effects of heat-killed Lactobacillus casei IMAU60214 on the activity of human monocyte-derived macrophages (MDMs). Human MDMs were treated with heat-killed L. casei at a ratio (bacteria/MDM) of 50:1, 100:1, 250:1, and 500:1, and then evaluated for the following: NO production, by Griess reaction; phagocytosis of FITC-labeled Staphylococcus aureus particles; cytokine secretion profile (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-12p70, IL-10, and transforming growth factor (TGF)-β) by ELISA; and costimulatory molecule (CD80 and CD86) surface expression, by flow cytometry. Heat-killed L. casei IMAU60214 enhanced phagocytosis, NO production, cytokine release, and surface expression of CD80 and CD86 in a dose-dependent manner. All products were previously suppressed by pretreatment with a Toll-like receptor 2 (TLR2)-neutralizing antibody. Overall, our findings suggest that this probiotic strain promotes an M1-like pro-inflammatory phenotype through the TLR2 signaling pathway. These effects on macrophage phenotype help explain the probiotic efficacy of Lactobacillus and provide important information for the selection of therapeutic targets and treatments compatible with the immunological characteristics of this probiotic strain.

Graphical Abstract

1. Introduction

Lactic acid bacteria (LAB) constitute a heterogeneous group of Gram-positive, non-toxigenic facultative anaerobic microorganisms that efficiently produce lactic acid from carbohydrates, which makes them useful as starter cultures for food fermentation [1]. The most known LAB are the Lactobacillus, a genus that comprises a large heterogeneous group of LAB with more than 200 species and 29 subspecies of which more than 20 have been sequenced [1,2]. Among these is the Lactobacillus casei group (LCG), including L. casei and related species L. paracasei and L. rhamnosus [3]. The LCG group are the most extensively studied Lactobacillus species with documented health benefits [3,4] such as enhanced resistance against infection by increasing the secretion of pro-inflammatory cytokines (e.g., tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-12) from various immune cells analyzed on models of in vitro and in vivo investigations [5,6,7,8]. Multiple investigations have reported that treatment with probiotic bacteria increases resistance against pathogens that cause gastrointestinal and respiratory infections [9,10]. Furthermore, probiotic strains (both viable and non-viable), as well as specific cell wall components (e.g., peptidoglycans (PGs), lipoteichoic acids (LTAs), proteins, and exopolysaccharides (EPSs)) have the ability to stimulate the immune response [11,12]. In addition, the regions of these molecules exposed on the surface are considered microbe-associated molecular patterns (MAMPs), which can activate innate immune responses by binding to pattern recognition receptors (PRRs) expressed on immune cells and many other cell types [13,14]. Several reports have shown that the immunostimulant effect of the organism, in response to probiotics, is dependent in part on activation of Toll-like receptors (TLRs) [15,16]. Monocytes and macrophages express high levels of TLRs, mainly TLR2, TLR4, and TLR6, which have been demonstrated to induce cytokine and chemokine production upon stimulation by several probiotic species and strains [17,18,19]. Macrophages also participate in the activation and regulation of the immune response through antitumor activities, antigen presentation, and secretion of both pro-inflammatory cytokines (e.g., TNF-α, IL-1, and IL-6) and anti-inflammatory cytokines (IL-10 and transforming growth factor (TGF)-β) that act to regulate immune homeostasis [20,21,22]. In addition, probiotics act as mediators of inflammation and play an important role in the control of infection of several pathogens [23,24]. Several recent reports have examined the regulation of the macrophage immune response to probiotic strains of Lactobacillus casei [25]. However, the immune effects of individual probiotic strains cannot be generalized as each species of probiotic strain possesses unique functional properties and it is also important to consider that their effects are dependent on the specific host’s conditions [26,27]. Previously, we reported a screening study on some strains of Lactobacillus isolated from different fermented milks. Secretion profiles of inflammatory mediators such as cytokines varied depending on the Lactobacillus species present in the fermented milk [28].
In the present study, we extend the research on the isolate of L. casei IMAU60214, which has not been as extensively studied and is less characterized in comparison to others strains of probiotics, such as L. rhamnosus (LGG) [29]. Therefore, the aim of this study was to evaluate the immunomodulation activity in order to understand its probiotic potential and the role of microbial homeostasis in health and disease.

2. Materials and Methods

2.1. Bacterial Culture and Preparation of Heat-Killed Probiotic Bacteria

Strain L. casei IMAU60214 for this study was kindly provided by Dr. Guerrero from the fermented milk (Universidad Autonoma, Metropolitana, México). The L. casei IMAU60214 strain was grown in MRS broth (de Man, Ragosa, and Sharp Broth; BDL, Franklin Lakes, NJ, USA) at 37 °C for 18–24 h. Bacterial cells were harvested by centrifugation, and washed three times in physiological saline. The number of colony-forming units (CFUs) was determined by cell counting in dilutions of base 10. The concentration was adjusted to 1 × 109 CFU and heat-killed at 85 °C for 10 min.

2.2. Preparation and Treatment of Human Monocyte-Derived Macrophages (MDMs)

Human peripheral blood mononuclear cells were obtained by gradient centrifugation using Lymphoprep (Nycomed, Zurich, Switzerland). Blood sampling, MDM isolation, and subsequent experiments were conducted with prior authorization and consent according to established institutional guidelines. Monocytes from healthy donors were enriched using the Monocyte Separation Kit II (Miltenyi Biotec, Bergisch Gladbach, Germany) and plated at 1 × 105 cells per well in 24-well plates in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) at 37 °C under a 5% CO2 atmosphere. Differentiated macrophages were obtained after seven days in culture. The purity after these cells was evaluated for phenotypic markers, CD14 and HLA-DR molecules (Biosciences, San Diego, CA, USA), determined by flow cytometry. Cells were then stimulated with heat-sensitive L. casei IMAU60214 at a ratio 50:1, 100:1, 250:1, and 500:1 of (bacteria/MDM) as indicated for 24 h. The research procedures in our study were approved by the Ethics Committee in Research (HIM-2014-013) of the Children′s Hospital, México and project code is HIM-2014-013, 1120 SSA (16 December 2014).

2.3. NO Production Assay

Nitric oxide (NO) production was assessed by quantifying the concentration of nitrite compounds (end-products of NO) in supernatants using a colorimetric assay based on the Griess reaction. Supernatants were recovered from untreated and treated MDM cells for 24 h with heat-killed L. casei IMAU60214, lipopolysaccharide (LPS) (1μg/mL) alone, LPS with polymyxin B (PMB) (1μg/mL) alone, the TLR2 agonist Pam3CSK4 (1μg/mL), or the TLR2 agonist with polymyxin B, respectively, and mixed with equal volumes of the Griess reagent (0.1% sulfanilamide, 5% phosphoric acid, and 1% ethylenediamine dihydrochloride). The mixture was incubated for 10 min at room temperature and the optical density measured at 540 nm. Nitrite concentration was obtained by extrapolation to a sodium nitrite standard curve.

2.4. Quantification of Cytokine Release

Cytokine levels (TNF-α, IL-1β, IL-6, IL-12p70, IL-10, and TGF-β) were measured in supernatants of treated and untreated MDMs using specific ELISA kits (BD Bioscience, San Diego, CA, USA) according to the manufacturer’s instructions.

2.5. Flow Cytometric Assay for Surface Marker Expression

Briefly, macrophages were treated with heat-killed L. casei IMAU60214 at ratio 500:1 (bacteria/MDM) at 37 °C under a 5% CO2 atmosphere for 24 h. Cells were harvested, washed twice with a 2 mM PBS/EDTA (pH 7.2) solution for 5 min, harvested, centrifuged in PBS (300× g for 5 min, 4 °C), resuspended in a blocking buffer solution (PBS supplemented with 2% FBS, 5 mM EDTA, human IgG immunoglobulin, and 0.1% sodium azide), and placed on ice for 30 min. Cells were centrifuged (300× g for 5 min), washed twice with PBS, and incubated for 30 min at 4 °C with specific markers in staining buffer (PBS supplemented with 2% FBS, 5 mM EDTA, and 0.1% sodium azide) containing FITC-conjugated anti-human CD80 and PE-conjugated anti-human CD86 (BD Bioscience, San Diego, CA, USA). Subsequently, cells were washed with 250 μL of staining solution and fixed with 2% paraformaldehyde in PBS containing 0.1% sodium azide. Isotype controls were used to estimate non-specific staining. In this study, we used PE-conjugated anti-IgG2a and FITC-conjugated IgG1 (Santa Cruz Biotechnology, Dallas, TX, USA). The proportions of cells expressing each marker as well as the average fluorescence value were acquired using a FACS Calibur flow cytometer (Becton Dickinson, Franklin Lakes, NJ, USA). Data were analyzed using WinMDI 2.9 software (manufacturer, city, state abbreviation, country). Results are expressed as mean fluorescence intensity (MFI) relative to control isotype for four independent experiments.

2.6. Phagocytosis Assay

Macrophages were pretreated with heat-killed L. casei IMAU60214 at a ratio 250:1 of (bacteria/MDM) for 24 h. A suspension containing FITC-labeled Staphylococcus aureus was added at a ratio 10:1 of (bacteria/MDM) and incubated for 1 h. The number of engulfed particles was visualized under a Zeiss Axiocam epifluorescence microscope (Zeiss AG, Oberkochen, Germany). The proportion of phagocytic macrophages and the number of engulfed particles were counted in randomly selected fields containing 60 cells.

2.7. Assays with a Toll-Like Receptor 2 (TLR2)-Neutralizing Antibody

The contributions of TLR2-dependent signaling to macrophage effector functions induced by heat-killed L. casei IMAU60214 at a ratio 500:1 of (bacteria/MDM) were analyzed by repeating these experiments in the presence of a TLR2-neutralizing antibody (TLR2 Bioscience clone TL 2.1, city, state abbreviation, country). Briefly, MDMs were treated with 10 μg/mL neutralizing antibody for 1 h at 37 °C followed by stimulation with probiotic bacteria for 24 hours as indicated.

2.8. Statistical Analysis

Data are presented as a mean ± standard deviation (SD); each experiment was performed with four donors and was performed in duplicate. Differences for nitric oxide and cytokine measurements between experimental groups (treated vs. untreated) were compared by Mann–Whitney U test. A p < 0.05 (two-tailed) was considered for statistical significance. Mean fluorescence intensity differences were shown as histograms.

3. Results

3.1. Purity and Morphological Identification of MDM Cells

CD14 and HLA-DR expression were used as phenotypic markers for MDM cells during the differentiation process. FACS analysis of gated cells showed that 95% ± 2.9% expressed CD14 (Figure 1a) and a similar percentage was observed in the cells positive for HLA-DR (Figure 1b). Additionally, the purity of MDM cell was demonstrated by its morphology (Figure 1c).

3.2. Induction of NO Release from MDMs by Heat-Killed L. casei IMAU60214

The treatment of MDMs with heat-killed L. casei IMAU60214 at a ratio (bacteria/MDM) of 100:1, 250:1, and 500:1 for 24 h induced NO level elevation compared to the control negative (Figure 2). The highest induction was observed with (500:1, 250:1, 100:1) under these experimental conditions. In addition, the stimulatory activity of heat-killed L. casei IMAU60214 also revealed a dose-dependent increase in extracellular nitrate accumulation, a biochemical measure of NO release. Exposure to Escherichia coli 0111:B4 lipopolysaccharide (LPS, 1 μg/mL) induced NO release that was substantially greater than that produced by the probiotic heat-killed bacteria. However, the treatment with polymyxin B (an LPS inhibitor) effectively inhibited the NO production induced by LPS but had no effect on Pamp3CK4-induced NO production, indicating that enhanced NO production by L. casei IMAU60214 was not the result of endotoxin contamination.

3.3. Induction of Cytokine Release from MDMs by Heat-Killed L. casei IMAU60214

The treatment with heat-killed L. casei IMAU60214 dose-dependently induced secretion of TNF-α, IL-1β, IL-6, IL-12p70, IL-10, and TGF-β with substantially greater extracellular accumulation at a ratio (bacteria/MDM) of 250:1 and 500:1 compared to 100:1 and 50:1 (Figure 3). In addition, accumulations of pro-inflammatory cytokines were higher than accumulations of the anti-inflammatory factors IL-10 and TGF-β, suggesting a shift toward pro-inflammatory status.

3.4. Induction of CD80 and CD86 Surface Expression by MDMs In Response to Heat-Killed L. casei IMAU60214

An increase in basal CD80 expression was induced when the MDMs were exposed to L. casei IMAU60214 at a ratio (bacteria/MDM) of 500:1 as compared with control cells without stimulation (Figure 4a). In addition, the treatment also increased surface expression of the costimulatory molecule CD86 compared to baseline and negative control stimulation (Figure 4b). Similar results were observed when the MDMs were stimulated with TLR2 agonist (Pam3CSK4) utilized as positive control of the assay, which induced expression substantially greater than that produced by the probiotic heat-killed bacteria.

3.5. Induction of Macrophage Phagocytic Activity by Heat-Killed L. casei IMAU60214

The effect of L. casei IMAU60214 at ratio (500:1) on macrophage phagocytosis was investigated by uptake of fluorophore-tagged Staphylococcus aureus particles (Figure 5a). Indeed, the number of fluorescent macrophages (containing S. aureus particles) increased and the phagocytic index was significantly higher than control (untreated) MDMs (Figure 5b). Thus, exposure to heat-sensitive L. casei IMAU60214 promotes the phagocytic activity of macrophages (Figure 5c).

3.6. Effects of L. casei IMAU60214 on Macrophages Are Dependent on Activation of Toll-Like Receptor 2

As shown (Figure 6), the pretreatment with neutralizing antibody for 1 h significantly reduced NO release by 70% (Figure 6a), and TNF-α, IL-1β, IL-6, IL-12p70, IL-10, and TGF-β release (by 50–70%) (Figure 6b). Similarly, data were observed on surface expression of CD80 and CD86 (Figure 6c,d).

4. Discussion

The maintenance of gastrointestinal homeostasis is critical for the prevention of inflammatory and metabolic disorders [30]. The positive influences of probiotics on intestinal homeostasis and host health depend mainly on antagonism of undesirable bacteria and maintenance of a “physiological state of inflammation” that contributes to the control of certain infectious and immunological reactions [31,32]. However, unresolved questions concern the scopes of local and systemic effects as well as the molecular pathways responsible for interactions with host cells. The local immunomodulatory contributions of LAB have been demonstrated in several intestinal mucosal epithelial cell lines and mucosal immune cells (e.g., T lymphocytes, dendritic cells, and monocytes-macrophages) [33,34].
Probiotics have been used as alternative therapies in intestinal inflammatory disorders. It is recognized that the stimulatory and inhibitory actions of probiotic bacteria in the gut are dependent on species, cell condition (i.e., whether viable or dead), and the host-specific effects of soluble components [35,36]. However, there is an increasing interest in the use of non-viable bacteria to reduce the risk of microbial translocation and infection and the possibility of application in some immunocompromised patients (i.e., oncology where the administration of live microorganisms may not be appropriate). In this study, we showed that the probiotic strain L. casei IMAU60214 in the non-viable state has immunostimulating activity on human MDMs.
In the present study, we focused on the effects of non-viable bacteria only, because many of the effects obtained from viable cells of probiotics are also obtained from populations of dead cells [37,38]. In addition, the use of dead probiotics as biological response modifiers has several attractive advantages; such products would be very safe and have a long shelf-life. We found that non-viable L. casei IMAU60214 enhanced the production of nitric oxide, a mediator of oxygen-dependent microbicide critical for the control of pathogens [39]. Nitric oxide also plays an important role in innate immunity mediated by macrophages. Release of NO can produce cytotoxic reactive species such as peroxynitrites, which are effective antimicrobials [40,41]. The synthesis of NO by MDMs was dependent on the relative abundance of added probiotic bacteria. Moreover, polymyxin B (an LPS inhibitor) effectively inhibited the NO production induced by LPS but had no effect on Pam3CSK4 -induced NO production, indicating that enhanced NO production by L. casei IMAU60214 was not the result of endotoxin contamination. Similarly, L. casei IMAU60214 also dose-dependently induced the release of both pro- and anti-inflammatory cytokines. TNF-α, IL-1β, IL-6, and IL-12 are inflammatory cytokines that participate in pathogen clearance [42]. TNF-α activates inflammatory cells such as neutrophils, monocytes, macrophages, and dendritic cells, and increases the recruitment of these cells to the site of inflammation. The cytokine TNF-α also promotes the expression of inducible nitric oxide synthase (iNOS), thereby enhancing NO production. Similarly, IL-6 also maintains host immunity as deficiency leads to failure of both innate and adaptive immune responses to viral and bacterial infections [43]. Likewise, IL-12 can activate natural killer cells and stimulate the synthesis of IFN-γ, one of the main activators of macrophages. IL-12 increases the expression of enzymes (NADPH oxidase, iNOS) and also promotes the secretion of cytokines from TH1 cells. In the current study, L. casei IMAU60214 activated the release of all of these cytokines from macrophages. Based on the substantial increase in IL-1β production, we suggest that these probiotic bacteria can effectively activate the entire inflammasome, in accordance with previous studies on other Lactobacillus strains such as L. rhamnosus, demonstrating activation of the inflammasome and suppression of viral infection [44].
On the other hand, L. casei IMAU60214 also induced the secretion of anti-inflammatory cytokines such as IL-10 and TGF-β. IL-10 is one of the most important immunosuppressive cytokines for the maintenance of immune homeostasis [45], whereas TGF-β exerts systemic immunosuppression and inhibits host immunosurveillance [46]. Our current findings of enhanced NO and cytokine production are in accordance with a previous report by Jung et al. [47] demonstrating similar effects of Lactobacillus sakei KO40706 on RAW264.7 macrophages and the enhancement of NO and cytokine secretion (mainly TNF-α and IL-6) in mice immunosuppressed by cyclophosphamide treatment. Similarly, several strains of Lactobacillus have been shown to trigger the release of TNF-α, IL-6, and IL-10 from macrophages and dendritic cells [48]. The efficacy of probiotics to enhance the production of cytokines such as IL-12 and IL-10 is considered a critical attribute for health promotion and possible therapeutic application. In addition, previous research has also demonstrated the capacity of probiotic bacteria to flexibly regulate cytokine production by macrophages and T cells for immune regulation and multifunctional immune activities [49].
In contrast, live and heat-killed L. casei Lbs2 (MTCC5953) significantly decreased LPS-induced production of TNF-α and IL-6, and increased the expression levels of IL-10 and TGF-β in mouse colon tissue [50]. Sun et al. [51] also reported downregulation of LPS-induced TNF-α and IL-6 production in mononuclear cells and human THP-1 monocytes by L. paracasei. These variations in macrophage effector responses may be related to the different host cells or Lactobacillus strains used. In addition to triggering NO and cytokine production by macrophages, L. casei IMAU60214 also enhanced the surface expression of the costimulatory molecules CD80 and CD86. Both CD80 and CD86 are generally expressed at low levels, but expression levels are increased upon stimulation by certain molecules such as LPS or Pam3CSK4. In this study, heat-killed L. casei IMAU60214 had similar effects. Molecules CD80 and CD86 are the secondary signal of costimulation in antigen-presenting cells and promote the differentiation and maturation of these cells. Moreover, they stimulate the activation of TH1 cells. Increased CD86 and CD80 expression suggests M1 differentiation. In accordance with this finding, a recent study found that L. johnsonni NBRC13952 activated IL-1β secretion as well as CD80 expression and that these responses were important for suppression of Aggregatibacter actinomycetemcomitans infection [52].
Other studies have reported rapid upregulation of immune-related genes such as IL-7R, CD80, CD86, NF-kappa B, and CCL4 in THP-1 cells upon stimulation by L. acidophilus L-92 [53]. Similarly, recent studies have provided evidence that certain Lactobacillus species such as L. crispatus can increase expression of costimulatory molecules, including CD40, CD80, and CD86, suggesting differentiation of monocytes into precursors of Langerhans-like cells [54]. In the current study, we also demonstrated that heat-killed L. casei IMAU60214 exposure can enhance phagocytosis of Staphylococcus aureus by MDMs. This is an essential activity of polymorphonuclear cells and monocytes-macrophages in innate immunity. Similar results have been reported using different labeled bacteria as well as Escherichia coli [55]. A previous study also documented a significant increase in phagocytosis after treatment with different strains of Lactobacillus both in vitro and in vivo [56,57,58].
It is recognized that cellular components derived from probiotics interact via PRRs, including NOD-like receptors and the family of TLRs, which can modulate cytokine production [59]. It has been reported that TLR2 is involved in the secretion of both pro- and anti-inflammatory cytokines in response to probiotic strains [60,61]. We demonstrated that the mechanism responsible for macrophage activation by heat-killed probiotic bacteria is TLR2-dependent, blocking antibody treatments and showing that L. casei IMAU60214 acted as a TLR2 agonist in macrophages, in accordance with a recent investigation demonstrating immune regulation through TLR2/TLR6 signaling in response to LAB [62].
Based on these findings, heat-killed L. casei IMAU60214 appears to stimulate immune activity by inducing differentiation of macrophages towards the M1 phenotype, which could have immunoregulatory actions in immunocompromised hosts. Additional studies in vivo are warranted to examine the potential of this probiotic strain for improvement of human health.

5. Conclusions

In conclusion, this study demonstrates that heat-killed Lactobacillus casei IMAU60214 dose-dependently promotes the immune activities of macrophages via TLR2 activation and so may be one of the main routes of activation in early immunity. Likewise, our findings confirm that probiotics in their non-viable condition are potentially beneficial to human health.

Author Contributions

Conceptualization, L.M.R.-R. and M.G.-G.; methodology, L.M.R.-R., B.H.-O., M.G.-G., N.C-R.., S.C.-L., S.G.-M., and J.M.-Q.; investigation, L.M.R.-R., B.H.-O., N.C.-R. M.G.-G., S.C.-L., S.G.-M., and J.M.-Q.; data curation, L.M.R.-R.; writing—original draft preparation, L.M.R.-R., B.H.-O., S.G.-M., and J.M.-Q.; project administration, L.M.R.-R.; funding acquisition, L.M.R.-R. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by grants from the Fondos Federales México (HIM/2014/013 SSA. 1120) and by the E022 Program, National Institute of Pediatrics, Mexico City, Mexico (Recursos Fiscales para la Investigación. J.M.-Q. was supported by INP 024/2017. S.G.-M. was supported by INP 031/2018 and INP 038/2019. N.C.-R. was supported by INP 041/2018). J.M.-Q. was supported by CONACYT grant 259201, Cátedras CONACYT (2184) project number 2057.

Acknowledgments

The technical assistance of Camila Marcial, Ximena Gomez-Gonzalez, and Maria Jose Gomez-Gonzalez is greatly appreciated. Finally, we wish to thank Javier Gallegos Infante (Instituto de Fisiología Celular, UNAM) for assistance with the bibliographic materials. We appreciate the English language editing carried out by M.D. Cindy Karel Arregoitia-Sarabia (MD, Medical Group, Dallas, TX, USA).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Douillard, F.P.; de Vos, W.M. Functional genomics of lactic acid bacteria: From food to health. Microb. Cell Factories 2014, 13, S8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Sun, Z.; Harris, H.M.; McCann, A.; Guo, C.; Argimón, S.; Zhang, W.; Yang, X.; Jeffery, I.B.; Cooney, J.C.; Kagawa, T.F.; et al. Expanding the biotechnology potential of lactobacilli through comparative genomics of 213 strains and associated genera. Nat. Commun. 2015, 6, 8322. [Google Scholar] [CrossRef] [PubMed]
  3. Hill, D.; Sugrue, I.; Tobin, C.; Hill, C.; Stanton, C.; Ross, R.P. The Lactobacillus casei Group: History and Health Related Applications. Front. Microbiol. 2018, 9, 2107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Aureli, P.; Carpuso, L.; Castellazzi, A.M.; Clerici, M.; Giovannini, M.; Morelli, L.; Poli, A.; Pregliasco, F.; Salvini, F.; Zuccotti, G.V. Probiotics and health; An evidence-based review. Pharmacol. Res. 2011, 63, 366–376. [Google Scholar] [CrossRef]
  5. Bezirtzoglou, E.; Stavropoulou, E. Immunology and probiotic impact of the newborn and young children intestinal microflora. Anaerobe 2011, 17, 369–374. [Google Scholar] [CrossRef]
  6. Elmadfa, I.; Klein, P.; Meyer, A.L. Immune-stimulating effects of lactic acid bacteria in vivo and in vitro. Proc. Nutr. 2010, 69, 416–420. [Google Scholar] [CrossRef] [Green Version]
  7. Meyer, A.L.; Elmadfa, I.; Herbacek, I.; Micksche, M. Probiotic, as well as conventional yogurt, can enhance the stimulated production of proinflammatory cytokines. J. Hum. Nutr. Diet. 2007, 20, 590–598. [Google Scholar] [CrossRef]
  8. Karthikeyan, T.; Pravin, M.; Muthusamy, V.S.; Bharathi, R.R.; Lakshmi, B.S. In vitro Investigation of the Immunomodulatory Potencial of Probiotic Lactobacillus casei. Probiotics Antimicrob Proteins 2013, 5, 51–58. [Google Scholar] [CrossRef]
  9. Liévin-Le, M.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] [Green Version]
  10. Villena, J.; Chiba, E.; Tomosada, Y.; Salva, S.; Marranzino, G.; Kitazawa, H.; Alvarez, S. Orally administered Lactobacillus rhamnosus modulates the respiratory immune response triggered by the viral pathogen-associated molecular pattern poly(I:C). BMC Immunol. 2012, 13, 53. [Google Scholar] [CrossRef] [Green Version]
  11. Cai, S.; Bay, B.H.; Lee, Y.K.; Lu, J.; Mahendran, R. Live and lyophilized Lactobacillus species elicit differential immunomodulatory effects on immune cells. FEMS Microbiol. Lett. 2010, 30, 189–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Claes, I.J.; Segers, M.E.; Verhoeven, T.L.; Dusselier, M.; Sels, B.F.; De Keersmaecker, S.C.; Vanderleyden, J.; Lebeer, S. Lipoteichoic acid is an important microbe-associated molecular pattern of Lactobacillus rhamnosus GG. Microb. Cell Factories 2012, 15, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Erbs, G.; Newman, M.A. The role of lipopolysaccharide and peptidoglycan, two glycosylated bacterial microbe-associated molecular patterns (MAMPs), in plant innate immunity. Mol. Plant Pathol. 2012, 13, 95–104. [Google Scholar] [CrossRef] [PubMed]
  14. Lebeer, S.; Vanderleyden, J.; De Keersmaecker, S.C. Host interactions of probiotic bacterial surface molecules: Comparison with commensals and pathogens. Nat. Rev. Microbiol. 2010, 8, 171–184. [Google Scholar] [CrossRef] [PubMed]
  15. Rizzo, A.; Losacco, A.; Carratelli, C.R. Lactobacillus crispatus modulates epithelial cell defense against Candida albicans Toll-like receptors 2 and 4, interleukin 8 and human β-defensins 2 and 3. Immunol. Lett. 2013, 156, 102–109. [Google Scholar] [CrossRef] [PubMed]
  16. Hoang, T.K.; He, B.; Wang, T.; Tran, D.Q.; Rhoads, J.M.; Liu, Y. Protective effect of Lactobacillus reuteri DSM 17938 against experimental necrotizing enterocolitis is mediated by Toll-like receptor 2. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 315, G231–G240. [Google Scholar] [CrossRef] [Green Version]
  17. Ryu, S.H.; Park, J.H.; Choi, S.Y.; Jeon, H.Y.; Park, J.I.; Kim, J.Y.; Ham, S.H.; Choi, Y.K. The Probiotic Lactobacillus Prevents Citrobacter rodentium-Induced Murine Colitis in a TLR2-Dependent Manner. J. Microbiol. Biotechnol. 2016, 26, 1333–1340. [Google Scholar] [CrossRef]
  18. Muzio, M.; Bosisio, D.; Polentarutti, N.; D’amico, G.; Stoppacciaro, A.; Mancinelli, R.; van’t Veer, C.; Penton-Rol, G.; Ruco, L.P.; Allavena, P.; et al. Differential expression and regulation of toll-like receptors (TLR) in human leukocytes: Selective expression of TLR3 in dendritic cells. J. Immunol. 2000, 164, 5998–6004. [Google Scholar]
  19. Jensen, H.; Drømtorp, S.M.; Axelsson, L.; Grimmer, S. Immunomodulation of monocytes by probiotic and selected lactic Acid bacteria. Probiotics Antimicrob. Proteins 2015, 7, 14–23. [Google Scholar] [CrossRef]
  20. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef]
  21. Chomarat, P.; Banchereau, J.; Davoust, J.; Palucka, A.K. IL-6 switches the differentiation of monocytes from dendritic cells to macrophages. Nat. Immunol. 2000, 1, 510–514. [Google Scholar] [CrossRef] [PubMed]
  22. Commins, S.P.; Borish, L.; Steinke, J.W. Immunologic messenger molecules: Cytokines, interferons, and chemokines. J. Allergy Clin. Immunol. 2010, 125, S53–S72. [Google Scholar] [CrossRef] [PubMed]
  23. Wu, Q.; Liu, M.C.; Yang, J.; Wang, J.F.; Zhu, Y.H. Lactobacillus rhamnosus GR-1 Ameliorates Escherichia coli-Induced Inflammation and Cell Damage via Attenuation of ASC-Independent NLRP3 Inflammasome Activation. Appl. Environ. Microbiol. 2015, 82, 1173–1182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lee, Y.T.; Ngo, V.L.; Cho, Y.H.; Ko, E.J.; Hong, S.M.; Kim, K.H.; Jang, J.H.; Oh, J.S.; Park, M.K.; Kim, C.H.; et al. Heat-killed Lactobacillus casei confers broad protection against influenza A virus primary infection and develops heterosubtypic immunity against future secondary infection. Sci. Rep. 2017, 7, 17360. [Google Scholar]
  25. Yasuda, E.; Serata, M.; Sako, T. Suppressive effect on activation of macrophages by Lactobacillus casei strain Shirota genes determining the synthesis of cell wall-associated polysaccharides. Appl. Environ Microbiol. 2008, 74, 4746–4755. [Google Scholar] [CrossRef] [Green Version]
  26. Lee, I.C.; Tomita, S.; Kleerebezem, M.; Bron, P.A. The quest for probiotic effector molecules-unraveling strain specificity at the molecular level. Pharmacol. Res. 2013, 69, 61–74. [Google Scholar] [CrossRef]
  27. Habil, N.; Al-Murrani, W.; Beal, J.; Foey, A.D. Probiotic bacterial strain differentially modulate macrophage cytokine production in a strain dependent and cell subset-specific manner. Benef. Microbes 2011, 2, 4283–4293. [Google Scholar] [CrossRef]
  28. Rocha-Ramírez, L.M.; Pérez-Solano, R.A.; Castañón-Alonso, S.L.; Moreno-Guerrero, S.S.; Ramírez-Pacheco, A.; García-Garibay, M.; Eslava, C. Probiotic Lactobacillus Strains Stimulate the Inflammatory Response and Activate Human Macrophages. J. Immunol. Res. 2017, 2017, 4607491. [Google Scholar] [CrossRef] [Green Version]
  29. Carpuso, L. Thirty Years of lactobacillus rhamnosus GG: A review. J. Clin. Gastroenterol. 2019, 53, S1–S41. [Google Scholar]
  30. Hotamisligil, G.S.; Erbay, E. Nutrient sensing and inflammation in metabolic diseases. Nat. Rev. Immunol. 2008, 8, 923–934. [Google Scholar] [CrossRef] [Green Version]
  31. 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] [PubMed]
  32. Amara, A.A.; Shibl, A. Role of Probiotics in health improvement, infection control and disease treatment and management. Saudi Pharm. J. 2015, 23, 107–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Maldonado-Galdeano, C.; Cazorla, S.I.; Lemme Dumit, J.M.; Vélez, E.; Perdigón, G. Beneficial Effects of Probiotic Consumption on the Immune System. Ann. Nutr. Metab. 2019, 74, 115–124. [Google Scholar] [CrossRef] [PubMed]
  34. Ferreira Dos Santos, T.; Alves Melo, T.; Almeida, M.E.; Passos Rezende, R.; Romano, C.C. Immunomodulatory Effects of Lactobacillus plantarum Lp62 on Intestinal Epithelial and Mononuclear Cells. Biomed Res. Int. 2016, 2016, 8404156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Adams, C.A. The probiotic paradox: Live and dead cells are biological response modifiers. Nutr. Res. Rev. 2010, 23, 37–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rodríguez-Nogales, A.; Algieri, F.; Vezza, T.; Garrido-Mesa, N.; Olivares, M.; Comalada, M.; Riccardi, C.; Utrilla, M.P.; Rodríguez-Cabezas, M.E.; Galvez, J. The viability of Lactobacillus fermentum CECT5716 is not essential to exert intestinal anti-inflammatory properties. Food Funct. 2015, 6, 1176–1184. [Google Scholar] [CrossRef]
  37. Sashihara, T.; Sueki, N.; Ikegami, S. An analysis of the effectiveness of heat-killed lactic acid bacteria in alleviating allergic diseases. J. Dairy Sci. 2006, 89, 2846–2855. [Google Scholar] [CrossRef]
  38. Zhang, L.; Nan, L.; Caicedo, R.; Neu, J. Alive and dead Lactobacillus rhamnosus GG decrease tumor necrosis factor-a -induced interleukin-8 production in Caco-2 cells. J. Nutr. 2005, 135, 1752–1756. [Google Scholar] [CrossRef]
  39. Bogdan, C. Nitric oxide synthase in innate and adaptive immunity: An update. Trends Immunol. 2015, 36, 16178. [Google Scholar] [CrossRef]
  40. Jones-Carson, J.; Zweifel, A.E.; Tapscott, T.; Austin, C.; Brown, J.M.; Jones, K.L.; Voskuil, M.I.; Vázquez-Torres, A. Nitric oxide from IFNγ-primed macrophages modulates the antimicrobial activity of β-lactams against the intracellular pathogens Burkholderia pseudomallei and Non typhoidal Salmonella. PLoS Negl. Trop. Dis. 2014, 14, e3079. [Google Scholar] [CrossRef]
  41. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses (Table of Contents). Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Billack, B. Macrophage activation: Role of toll-like receptors, nitric oxide, and nuclear factor kappa B. Am. J. Pharm. Educ. 2006, 70, 102. [Google Scholar] [CrossRef] [PubMed]
  43. Hunter, C.A.; Jones, S.A. IL-6 as a keystone cytokine in health and disease. Nat. Immunol. 2015, 16, 448–457. [Google Scholar] [CrossRef] [PubMed]
  44. Miettinen, M.; Pietilä, T.E.; Kekkonen, R.A.; Kankainen, M.; Latvala, S.; Pirhonen, J.; Österlund, P.; Korpela, R.; Julkunen, I. Nonpathogenic Lactobacillus rhamnosus activates the inflammasome and antiviral responses in human macrophages. Gut Microbes. 2012, 3, 510–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Couper, K.N.; Blount, D.G.; Riley, E.M. IL-10: The master regulator of immunity to infection. J. Immunol. 2008, 577, 1–7. [Google Scholar] [CrossRef] [PubMed]
  46. Worthington, J.J.; Fenton, T.M.; Czajkowska, B.I.; Klementowicz, J.E.; Travis, M.A. Regulation of TGFβ in the immune system: An emerging role for integrins and dendritic cells. Immunobiology 2012, 217, 1259–1265. [Google Scholar] [CrossRef] [PubMed]
  47. Jung, J.Y.; Shin, J.S.; Lee, S.G.; Rhee, Y.K.; Cho, C.W.; Hong, H.D.; Lee, K.T. Lactobacillus sakei K040706 evokes immunostimulatory effects on macrophages through TLR2-mediated activation. Int. Immunopharmacol. 2015, 28, 88–96. [Google Scholar] [CrossRef]
  48. Kim, J.Y.; Park, M.S.; Ji, G.E. Probiotic modulation of dendritic cells co-cultured with intestinal epithelial cells. World J. Gastroenterol. 2012, 18, 1308–1318. [Google Scholar] [CrossRef]
  49. Shida, K.; Nanno, M.; Nagata, S. Flexible cytokine production by macrophages and T cells in response to probiotic bacteria: A possible mechanism by which probiotics exert multifunctional immune regulatory activities. Gut Microbes 2011, 2, 109–114. [Google Scholar] [CrossRef] [Green Version]
  50. Thakur, B.K.; Saha, P.; Banik, G.; Saha, D.R.; Grover, S.; Batish, V.K.; Das, S. Live and heat-killed probiotic Lactobacillus casei Lbs2 protects from experimental colitis through Toll-like receptor 2-dependent induction of T-regulatory response. Int. Immunopharmacol. 2016, 36, 39–50. [Google Scholar] [CrossRef]
  51. Sun, K.Y.; Xu, D.H.; Xie, C.; Plummer, S.; Tang, J.; Yang, X.F.; Ji, X.H. Lactobacillus paracasei modulates LPS-induced inflammatory cytokine release by monocyte macrophages via the up-regulation of negative regulators of NF-kappaB signaling in a TLR2-dependent manner. Cytokine 2017, 92, 1–11. [Google Scholar] [CrossRef] [PubMed]
  52. Jaffar, N.; Okinaga, T.; Nishihara, T.; Maeda, T. Enhanced phagocytosis of Aggregatibacter actinomycetemcomitans cells by macrophages activated by a probiotic Lactobacillus strain. J. Dairy Sci. 2018, 101, 5789–5798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yanagihara, S.; Goto, H.; Hirota, T.; Fukuda, S.; Ohno, H.; Yamamoto, N. Lactobacillus acidophilus L-92 Cells Activate Expression of Immunomodulatory Genes in THP-1 Cells. Biosci. Microbiota Food Health 2014, 33, 157–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Song, J.; Lang, F.; Zhao, N.; Guo, Y.; Zhang, H. Vaginal Lactobacilli Induce Differentiation of Monocytic Precursors Toward Langerhans-like Cells: In Vitro Evidence. Front. Immunol. 2018, 9, 2437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Lee, Y.; Lee, T.S. Enhancement in ex vivo phagocytic capacity of peritoneal leukocytes in mice by oral delivery of various lactic-acid-producing bacteria. Curr. Microbiol. 2005, 50, 24–27. [Google Scholar] [CrossRef] [PubMed]
  56. Gill, H.S.; Rutherfurd, K.J.; Prasad, J.; Gopal, P.K. Enhancement of natural and acquired immunity by Lactobacillus rhamnosus (HN001), Lactobacillus acidophilus (HN017) and Bifidobacterium lactis (HN019). Br. J. Nutr. 2000, 83, 167–176. [Google Scholar] [CrossRef] [Green Version]
  57. Hatcher, G.E.; Lambrecht, R.S. Augmentation of macrophage phagocytic activity by cell-free extracts of selected lactic acid-producing bacteria. J. Dairy Sci. 1993, 76, 2485–2492. [Google Scholar] [CrossRef]
  58. Ren, D.; Li, C.; Qin, Y.; Yin, R.; Du, S.; Liu, H.; Zhang, Y.; Wang, C.; Rong, F.; Jin, N. Evaluation of immunomodulatory activity of two potential probiotic Lactobacillus strains by in vivo tests. Anaerobe 2015, 35, 22–27. [Google Scholar] [CrossRef]
  59. Li, A.L.; Sun, Y.Q.; Du, P.; Meng, X.C.; Guo, L.; Li, S.; Zhang, C. The Effect of Lactobacillus actobacillus Peptidoglycan on Bovine β-Lactoglobulin-Sensitized Mice via TLR2/NF-κB Pathway. Iran. J. Allergy Asthma Immunol. 2017, 16, 147–158. [Google Scholar]
  60. Kaji, R.; Kiyoshima-Shibata, J.; Nagaoka, M.; Nanno, M.; Shida, K. Bacterial teichoic acids reverse predominant IL-12 production induced by certain lactobacillus strains into predominant IL-10 production via TLR2-dependent ERK activation in macrophages. J. Immunol. 2010, 184, 3505–3513. [Google Scholar] [CrossRef] [Green Version]
  61. Kaji, R.; Kiyoshima-Shibata, J.; Tsujibe, S.; Nanno, M.; Shida, K. Short communication: Probiotic induction of interleukin-10 and interleukin-12 production by macrophages is modulated by co-stimulation with microbial components. J. Dairy Sci. 2018, 10, 2838–2841. [Google Scholar] [CrossRef] [PubMed]
  62. Ren, C.; Zhang, Q.; de Haan, B.J.; Zhang, H.; Faas, M.M.; de Vos, P. Identification of TLR2/TLR6 signalling lactic acid bacteria for supporting immune regulation. Sci. Rep. 2016, 6, 34561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Purity and morphological identification of monocyte-derived macrophages (MDMs). CD14 and HLA-DR surface expression on MDM cells were evaluated after seven days in culture. (a) MDM cells were stained with isotype control Ab (IgG2) or (b) isotype control antibody (IgG1) and anti-HLA-DR-PE, and (c) morphology was identified by standard Giemsa staining. Results are from four healthy donors analyzed.
Figure 1. Purity and morphological identification of monocyte-derived macrophages (MDMs). CD14 and HLA-DR surface expression on MDM cells were evaluated after seven days in culture. (a) MDM cells were stained with isotype control Ab (IgG2) or (b) isotype control antibody (IgG1) and anti-HLA-DR-PE, and (c) morphology was identified by standard Giemsa staining. Results are from four healthy donors analyzed.
Microorganisms 08 00079 g001
Figure 2. Dose-dependent effect of heat-killed Lactobacillus casei IMAU60214 on nitrite (nitric oxide (NO)) release by MDMs. Cells were cultured in duplicate in 96-well plates in the absence or presence of increasing heat-killed L. casei IMAU60214 at ratios of (bacteria/MDM) 50:1, 100:1, 250:1, and 500:1 for 24 h. After incubation, NO release was measured as described in the Materials and Methods section. Each value represents the mean ± SD of four independent experiments. * p < 0.05 was considered for statistically significant data compared to untreated controls.
Figure 2. Dose-dependent effect of heat-killed Lactobacillus casei IMAU60214 on nitrite (nitric oxide (NO)) release by MDMs. Cells were cultured in duplicate in 96-well plates in the absence or presence of increasing heat-killed L. casei IMAU60214 at ratios of (bacteria/MDM) 50:1, 100:1, 250:1, and 500:1 for 24 h. After incubation, NO release was measured as described in the Materials and Methods section. Each value represents the mean ± SD of four independent experiments. * p < 0.05 was considered for statistically significant data compared to untreated controls.
Microorganisms 08 00079 g002
Figure 3. Heat-killed L. casei IMAU60214 dose-dependently enhances pro- and anti-inflammatory cytokine production by MDMs. The effects of L. casei IMAU60214 exposed at various ratios (50:1, 100:1, 250:1, 500:1) for 24 h on cytokine release were assessed by ELISA. Tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-12p70, IL-10, and transforming growth factor (TGF)-β. Each value represents the mean ± SD of four independent experiments.
Figure 3. Heat-killed L. casei IMAU60214 dose-dependently enhances pro- and anti-inflammatory cytokine production by MDMs. The effects of L. casei IMAU60214 exposed at various ratios (50:1, 100:1, 250:1, 500:1) for 24 h on cytokine release were assessed by ELISA. Tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-12p70, IL-10, and transforming growth factor (TGF)-β. Each value represents the mean ± SD of four independent experiments.
Microorganisms 08 00079 g003
Figure 4. Heat-killed L. casei IMAU60214 increases surface expression of CD80 and CD86 by MDMs. Macrophages were cultured in medium alone, 1 µg/mL of the Toll-like receptor 2 (TLR2) agonist Pam3CSK4 (positive control), or L. casei IMAU60214 at a ratio (500:1) for 24 h. Cells were then fixed and stained with isotype control, FITC-labeled anti-CD80, or PE-labeled anti-CD86. (a) CD80 and (b) CD86.
Figure 4. Heat-killed L. casei IMAU60214 increases surface expression of CD80 and CD86 by MDMs. Macrophages were cultured in medium alone, 1 µg/mL of the Toll-like receptor 2 (TLR2) agonist Pam3CSK4 (positive control), or L. casei IMAU60214 at a ratio (500:1) for 24 h. Cells were then fixed and stained with isotype control, FITC-labeled anti-CD80, or PE-labeled anti-CD86. (a) CD80 and (b) CD86.
Microorganisms 08 00079 g004
Figure 5. Heat-killed L. casei IMAU60214 increases the phagocytosis of Staphylococcus aureus by MDMs. Phagocytosis was evaluated by counting phagocytic macrophages, and the number of engulfed particles in randomly selected fields of untreated and treated MDMs with L. casei IMAU60214 (at a ratio of 500:1 for 24 h) was challenged with FITC S. aureus at a ratio 10:1 (bacteria/MDM). (a) Differential contrast (DIC) micrograph showing FITC-S. aureus fluorescence in MDMs. (b) Percentage of phagocytosis. (c) Mean number of particles phagocytized per cell. * A p < 0.05 was considered for statistical significance compared to untreated controls for percentage phagocytosis and mean number particles per cells, respectively.
Figure 5. Heat-killed L. casei IMAU60214 increases the phagocytosis of Staphylococcus aureus by MDMs. Phagocytosis was evaluated by counting phagocytic macrophages, and the number of engulfed particles in randomly selected fields of untreated and treated MDMs with L. casei IMAU60214 (at a ratio of 500:1 for 24 h) was challenged with FITC S. aureus at a ratio 10:1 (bacteria/MDM). (a) Differential contrast (DIC) micrograph showing FITC-S. aureus fluorescence in MDMs. (b) Percentage of phagocytosis. (c) Mean number of particles phagocytized per cell. * A p < 0.05 was considered for statistical significance compared to untreated controls for percentage phagocytosis and mean number particles per cells, respectively.
Microorganisms 08 00079 g005
Figure 6. Effects of heat-killed L. casei IMAU60214 on MDMs are dependent on the activation of Toll-like receptor 2. MDMs were pre-incubated with anti-TLR2 (10 μg/ml) for 1 h prior to stimulation by L. casei IMAU60214 (at ratio of 500:1) or Pam3CSK4 (1 μg) as a positive control for 24 h. Control MDMs were left untreated as a negative control. (a) NO accumulation. (b) Cytokine production. * Statistically significant p < 0.05 difference between conditions. (c) CD80 and (d) CD86.
Figure 6. Effects of heat-killed L. casei IMAU60214 on MDMs are dependent on the activation of Toll-like receptor 2. MDMs were pre-incubated with anti-TLR2 (10 μg/ml) for 1 h prior to stimulation by L. casei IMAU60214 (at ratio of 500:1) or Pam3CSK4 (1 μg) as a positive control for 24 h. Control MDMs were left untreated as a negative control. (a) NO accumulation. (b) Cytokine production. * Statistically significant p < 0.05 difference between conditions. (c) CD80 and (d) CD86.
Microorganisms 08 00079 g006

Share and Cite

MDPI and ACS Style

Rocha-Ramírez, L.M.; Hernández-Ochoa, B.; Gómez-Manzo, S.; Marcial-Quino, J.; Cárdenas-Rodríguez, N.; Centeno-Leija, S.; García-Garibay, M. Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages In Vitro. Microorganisms 2020, 8, 79. https://doi.org/10.3390/microorganisms8010079

AMA Style

Rocha-Ramírez LM, Hernández-Ochoa B, Gómez-Manzo S, Marcial-Quino J, Cárdenas-Rodríguez N, Centeno-Leija S, García-Garibay M. Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages In Vitro. Microorganisms. 2020; 8(1):79. https://doi.org/10.3390/microorganisms8010079

Chicago/Turabian Style

Rocha-Ramírez, Luz María, Beatriz Hernández-Ochoa, Saúl Gómez-Manzo, Jaime Marcial-Quino, Noemí Cárdenas-Rodríguez, Sara Centeno-Leija, and Mariano García-Garibay. 2020. "Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages In Vitro" Microorganisms 8, no. 1: 79. https://doi.org/10.3390/microorganisms8010079

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