Next Article in Journal
Genomic Analysis of a Novel Torradovirus “Rehmannia Torradovirus Virus”: Two Distinct Variants Infecting Rehmannia glutinosa
Previous Article in Journal
Microbiota and Recurrent Pregnancy Loss (RPL); more than a Simple Connection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Postbiotics in Asthma Treatment

by
Konstancja Węgrzyn
1,*,
Agnieszka Jasińska
2,
Kamil Janeczek
3 and
Wojciech Feleszko
2,*
1
Central Clinical Hospital, Medical University of Warsaw, 02-097 Warsaw, Poland
2
Department of Pediatric Pneumonology and Allergy, University Clinical Centre, Medical University of Warsaw, 02-097 Warsaw, Poland
3
Department of Paediatric Pulmonology and Rheumatology, Medical University of Lublin, 20-059 Lublin, Poland
*
Authors to whom correspondence should be addressed.
Microorganisms 2024, 12(8), 1642; https://doi.org/10.3390/microorganisms12081642 (registering DOI)
Submission received: 26 June 2024 / Revised: 1 August 2024 / Accepted: 8 August 2024 / Published: 11 August 2024
(This article belongs to the Section Medical Microbiology)

Abstract

:
In recent years, there has been abundant research concerning human microbiome and its impact on the host’s health. Studies have shown that not only the commensal bacteria itself, but also postbiotics, understood as inanimate microorganisms, possibly with the presence of their components, may themselves have an effect on various elements of human physiology. In this review, we take a closer look at the specific ways in which postbiotics can alter immune response in allergic asthma, which is one of the most prevalent allergic diseases in today’s world and a serious subject of concern. Through altering patients’ immune response, not only to allergens but also to pathogens, postbiotics could have a significant role in lowering the number of asthma exacerbations. We suggest that more profound research should be undertaken in order to launch postbiotics into clinical standards of asthma treatment, given the greatly promising findings in terms of their immunomodulating potential.

1. Introduction

In the 21st century, research on microbial compounds and their favorable impact on immune system has gathered pace, as it is now presumed to be one of the crucial factors that stimulate immune system development [1]. This is the reason why the use of probiotics and prebiotics has attracted recent interest. Probiotics are live bacteria strains that present health benefits for the host, compete with pathogens, promote microbial antagonism, and inhibit bacterial toxin production [2]. Prebiotics as specific dietary ingredients affect gut microbiota composition and support probiotics [3]. Strikingly, in many cases, the beneficial role of probiotics results from the effects of postbiotics (which are inanimate microorganisms) and/or their components that confer a health benefit on the host [4]. Postbiotics’ advantages over probiotics include aspects such as the absence of bacterial translocation and a lesser risk of infection deterioration. Moreover, they promote immune system development, inhibit inflammation, prevent infections, regulate lipid metabolism, and stabilize gut microbiota composition [5]. Among them, the most common are bacterial lysates (BLs), short-chained fatty acids (SCFAs), exopolysaccharides (EPSs), and heat-killed Lactobacilli [6]. Although SCFAs and EPSs cannot be classified as postbiotics in and of themselves, they are still to be found in the presence of bacterial biomass and therefore present beneficial outcomes for the host [4]. For this reason, we decided to discuss their great immunomodulatory potential in atopic asthma within this article (Figure 1).
Asthma, one of the most common of allergic diseases, affecting approximately 14% of children and young adults worldwide, is a disease with strikingly prominent diversity of ontogenetic and environmental factors modulating its course [7,8]. At its core lie the innate immune system’s acts of recognizing and processing internal and external stimuli, leading to various different ways in which the adaptive response is triggered [9]. Previously, asthma was considered to be a single diagnosis with standardized treatment for all patients. However, it is now known as a heterogenous, multifactorial disorder, and a new approach acknowledging the modulation of the immune response with various specific molecules allows researchers to broaden the possibilities in asthma treatment, focusing on the immunomodulatory properties of postbiotics. This publication aims to summarize and explain the immunomodulatory effects of postbiotics, SCFAs, and EPS in the prevention of exacerbations and the treatment of allergic asthma.

2. Methods of Acquiring Data

The authors searched biomedical databases (PubMed, Scopus, and Web of Science) for articles concerning the use of postbiotics in the asthma treatment. In all databases, the following keywords were searched: “asthma”, “postbiotics”, “bacterial lysates”, “short-chained fatty acids”, “exopolysaccharides”, and “heat-killed Lactobacillus”. The research was performed by two independent persons. All the relevant studies were identified by title and abstract reading. Duplicated articles were initially excluded. A careful analysis of full texts was carried out (Figure 2).
In this narrative review, the section focusing on the use of BLs took the form of a systematic review. The authors decided on this approach because BLs represent a well-defined and extensively studied category of postbiotics, warranting a more rigorous and structured analysis. A systematic review methodology ensures a comprehensive and unbiased synthesis of existing evidence, allowing for a more precise evaluation of the efficacy and safety of BLs in asthma treatment. In contrast, the authors chose a narrative approach for other postbiotics due to the lack of available studies on humans. The absence of human studies makes it difficult to apply the same systematic review approach, necessitating a broader, narrative discussion of their potential benefits and theoretical underpinnings.
Studies to be included in the section regarding BLs had to meet all of the following inclusion criteria: clinical trial (double-blind RCT or open-label RCT or sequential trial or cohort study); study on humans (children or/and adults) with asthma; BLs (PMBL or PCBL) as an intervention (alone or combined with standard care); control group receiving only standard asthma treatment or placebo or both. All studies had to be written in English. Study size was not included as a criterion. Studies were excluded if the study design was ineligible, if they included tests performed on animals, or if they evaluated the use of BLs for asthma prevention.
Two authors independently extracted the following data from the eligible studies: study design, sample size, participants’ characteristics, interventions, comparators, and clinical and other outcomes.

3. Bacterial Lysates

BLs are immunomodulatory preparations consisting of antigens derived from the most common respiratory tract bacteria species [10]. They can be obtained with chemical or mechanical lysis—polyvalent mechanical bacterial lysate (PMBL) or polyvalent chemical bacterial lysate (PCBL), which indicates their different biological effects [11]. BLs can be administered orally, sublingually, or intranasally in various forms [12]. Each route of administration provides slightly different effects; thus, it should be chosen considering the type of allergic disease and patients’ preferences [13]. Patients with asthma usually receive the oral form, which mainly affects mucosal immune cells in the intestine [14].
BLs’ fundamental mechanism of action is based on the natural immune response provoked by pathogens. Bacterial antigens continuously stimulate the lymphoid tissue in the mucosa to produce cytokines that modulate the immune response, both locally and generally [15]. These immunological changes provide a beneficial switch in the cytokines’ classes and affect the clinical course of the disease [16].
Toll-like receptors (TLRs) expressed on dendritic cells (DCs) and monocytes are the first-line proteins that recognize and respond to antigens. They stimulate DCs to maturate and influence cytokine release [17]. Studies suggest that BLs’ activity mainly depends on TLR2 and TLR4 signaling which suppresses airway hyperreactivity, mucus production, and Th2-type immune response in the lungs and lowers the risk of asthma development [18,19,20]. BLs promote the production of IL-10, IL-12, and IFN-gamma characteristic of Th1-type immune response [21,22] and suppress the secretion of Th2-type cytokines IL-4, IL-5, IL-13, and IL-17, which maintains the disturbed Th1/Th2 balance [23,24,25]. Sublingually administered BLs increase the level of NK cells in asthmatic children [24,26]. They are also known to activate peripheral blood macrophages and increase serum levels of IgA, IgG, and human beta-defensin 1 (hBD-1) [27,28]. Furthermore, they seem effective in lowering eosinophil counts in bronchoalveolar fluid and blood [29]. BLs’ mechanism of action was described in depth in our previous publication [30].
Nine human studies evaluated the efficacy of BLs in the prevention of exacerbations and the treatment of asthma. The very first clinical trial was conducted in 1987 by Weiss et al., who investigated the impact of Broncho-Vaxom (PCBL) on patients with asthma or chronic obstructive pulmonary disease (COPD). The research showed that PCBL reduces IgE and increases IgG levels in atopic patients. Although the results did not attain statistical significance, they pointed out the possible beneficial effects of BLs and encouraged scientists to conduct further research [31]. The effectiveness of BLs in reducing asthma exacerbations and symptom severity was confirmed in five clinical trials [23,32,33,34,35]. Both PCBLs and PMBLs were more effective in facilitating the clinical course of the disease than standardized care (SC) [23,33,34]. Only the study by de Boer et al. did not show any difference in the number of exacerbations in the PCBL and SC groups. However, FEV1 was increased in the PCBL group; thus, some improvements were observed [21]. On the other hand, Roßberg et al., who evaluated the effect of BLs administered in infancy on the risk of developing atopic dermatitis (AD), allergic rhinitis (AR), and asthma, concluded that they do not reduce the risk of allergic diseases. Nevertheless, patients received a mixture of heat-killed Escherichia coli and Enterococcus faecalis; thus, the effects probably resulted from the specific antigen properties and the results should not be clinching [36]. Therefore, further research on the efficacy of BLs in asthma prevention should be conducted (Table 1).

4. Short-Chained Fatty Acids

Short-chained fatty acids (SCFAs) are found in the human gut as a product of the anaerobic fermentation of non-digestible dietary fiber and amino acids by saprophytic bacteria, as well as being, in marginal amounts, derived directly from the diet [37]. Although the most abundant SCFA in the human gut tends to be acetate, the most beneficial in regard to health are found to be propionate and butyrate, produced mostly by Bacteroidetes and Firmicutes, respectively [38,39].
SCFAs can be either absorbed by the gut lining endothelium cells to serve as an energy source or enter the bloodstream and modulate the immune response [37]. Said immunomodulating effects are enabled via transporters, namely, proton-coupled monocarboxylate transporter isoform 1 (MCT1) and the Na+-coupled monocarboxylate transporter 1 (SMCT1), which can be found on the apical surface of colonocytes as well as on the surface of immune system cells [40,41]. Along with the MCT1 and SMCT2 transporters, SCFA uptake into the immune system cells is also facilitated by the direct activation of butyrate-sensing G protein-coupled receptor (GPCR) class, namely, GPR41/FFAR3 (free fatty acid receptor 3), GPR43/FFAR2, and GPR109A/HCAR2, as well as the activation of peroxisome proliferator-activated receptors (PPARs), which promote the influx of SCFAs into the immune cells, and butyrate-sensitive histone deacetylases (HDACs). The essence of SCFA’s immunomodulation potency lies in the deacetylation of histone complexes’ lysine. As a result, chromatin formation becomes denser and firmer so that gene expression becomes suppressed, inhibiting the immune system cells’ natural functions [37]. In various studies, the knockout of GPR41 and GPR43 has been proven to exacerbate asthma responses in mice, leading to assumptions that SCFAs can have a substantial effect on soothing asthmatic symptoms [42]. In a recent systematic review regarding the effects of SCFAs on allergic diseases in humans, a protective effect of higher SCFA levels was shown against allergic diseases, including asthma [43]. There is, however, still a need for further research in this area.
Th2 cell induction was shown to be suppressed in mice with high SCFA levels [44]. Moreover, DCs were proven to polarize naïve CD4+ T cells away from type 2 maturation and instead lean toward type 1 maturation [45]. Furthermore, in the in vitro model, SCFAs decreased DCs’ migration abilities, resulting in decreased asthmatic symptoms [46]. Another relevant element of the immunological pathway in allergy response are regulatory T lymphocytes (Tregs). Tregs have the ability to suppress the inflammation response, regulate the Th1/Th2 imbalance, and promote the remodeling of the airways via the secretion of IL-10 and transforming growth factor beta (TGF-β). What is more, Tregs can derive from naïve CD4+ T cells in the presence of TGF-β [47]. SCFA levels have been proven to positively correlate with the abundance of Treg cells. They have also been found to play a crucial role in Treg cells’ differentiation not only in mice [48,49] but also in humans [50].
Kim et al. showed in a study on AD that in mice individuals with higher SCFA levels, the eosinophilic percentage and eosinophilic count were decreased [51]. Given the atopic nature of asthma and AD, this correlation of high SCFA level to low eosinophils seems promising in discovering new therapeutic possibilities in asthma. In other studies, butyrate has been revealed to induce eosinophil apoptosis and reduce their adhesion to endothelial cells. The above-mentioned Zn2+-dependent class I, II, and IV HDACs have also been proven to inhibit eosinophilic survival and migration after exposure to propionate and butyrate [52]. In this light, SCFAs might contribute to alleviating symptoms of atopic asthma.
In allergic response, after being stimulated by IL-4 and IL-13, B cells mature into IgE, producing plasma cells. This ability of maturation and class-switching has been reported to be lower in mice with a high-fiber diet through epigenetic alterations [53]. Propionate and butyrate also decreased IL-4 levels, which is essential to differentiate B cells into IgE-producing cells [54]. Moreover, SCFAs were shown to inhibit mast cell degranulation and release airway contractiveness [55].
Another element of allergic diseases, including asthma, is the dysfunction of the airway epithelium barrier. SCFAs have been discovered to enhance the epithelial wall barrier function [56].
The imbalance of gut microbiota is a profound factor contributing to immunological diseases. It has been shown that amongst children suffering from asthma, the levels of SCFAs were significantly lower than in their healthy peers [57].

5. Exopolysaccharides

EPSs are carbohydrate polymers forming the external coating of the bacterial cell wall. They have diverse health effects, such as calcium and magnesium absorption, glycemic control, and anticarcinogenic and antioxidant effects [58]. Notably, EPSs produced by commensal bacteria, like Lactobacillus or Bifidobacterium, present immunomodulatory properties [59]. Schiavi et al. revealed that EPSs derived from Bifidobacterium longum inhibit eosinophilic migration to the airways, which was connected to Th2-associated interleukin IL-4 and IL-13 decrease [60]. A recent study showed that EPSs isolated from Bacillus subtilis divested asthmatic inflammation, linked to the concentration-dependent decrease in IL-4 and IL-5 levels, leading to reduced eosinophilic count [61]. Moreover, EPSs strongly bind the histamine molecules to the surface of bacterial wall and lower their blood count, mitigating the asthmatic response [62]. In a randomized, double-blind, placebo-controlled clinical trial where patients with airborne allergy were administered EPSs derived from Lactobacillus paracasei for 12 weeks, there was a significant alleviation of allergic symptoms reported by patients, which also correlated with the decrease in biochemical signs of allergic inflammation [63].
Knowing the effect of EPSs on various elements of the immune system and promising results on inhalant allergy in humans, those studies shine a new light on the potential use of EPSs, amongst other postbiotics, in controlling atopic asthma symptoms.

6. Heat-Killed Lactobacillus

The genus Lactobacillus is represented by almost 250 species of Gram-positive, anaerobic bacteria colonizing multiple, diverse habitats, which can be used in many industrial and healthcare applications [64]. For years, beneficial effects of Lactobacillus spp. were observed in preventing gut dysbiosis and allergy development [65,66]. Additionally, a recent airway microbiome profiling revealed the presence of Lactobacillus spp. in the nasopharynx and pointed out its remarkable role in local immune changes and the inhibition of respiratory tract pathogens’ growth and virulence [67]. However, there are still insufficient data establishing whether the above-mentioned abilities result from the activity of live bacteria or bacterial products and particles that influence the host’s immunity.
In an allergy model, mice fed with heat-killed L. casei showed significantly lower IgE and IgG1 levels and suppressed T cell production of Th2 type (IL-4, IL-5, IL-10, IL-13) and proinflammatory (IFN-γ and TNF-α) cytokines compared with the placebo group. Moreover, the histological evidence showed the attenuation of lung inflammation and reduced proinflammatory cytokines in bronchoalveolar fluid [68]. Choi et al. observed similar phenomena when examining the impact of heat-killed Lactobacillus spp. on dust-mite-induced AD in mice. Moreover, the above-mentioned supplementation ameliorated symptoms and reduced the number of mast cells and eosinophils in lesions. Among 18 Lactobacillus strains, L. brevis NS1401 induced the greatest IFN-γ and IL-12 secretion and the least IL-4 production [69]. This suggests that the immunomodulatory abilities depend on the Lactobacilli species and are unequal for the whole genus. Hong et al. compared the effects of three heat-killed Lactobacilli on airway hyper-responsiveness in a murine asthma model. The research showed that airway inflammation was suppressed in L. plantarum- and L. curvatus-treated mice, and lower IL-4 and IL-5 levels were observed. On the contrary, in the L. sakei subsp. sakei. group, no differences were found [70]. Similar results were shown by Lee et al., who compared cytokine regulatory effects of three Lactobacilli strains in an in vitro study. Notably, Lactobacilli lysates were more likely to stimulate cytokine production than heat-killed bacteria, cell supernatants, and live strains. Lipoteichoic acid isolated from bacteria promoted TNF-alpha production via TLR2-mediated NF-κB and extracellular-signal-regulated kinase (ERK) activation [71]. Heat-killed Lactobacilli strains stimulate DCs to produce IL-12 p70 and switch T cells to Th1 type immune response [72]. Moreover, they suppress the production of IL-6 and IL-17A, which results in Treg/Th17 balance maintenance [73]. The above-mentioned studies suggest that tyndallization does not reduce the immunomodulatory abilities of Lactobacillus spp. and the results of their use are comparable with those from live bacteria. Furthermore, heat-killed bacteria offer increased safety; thus, their usage in clinical practice should be considered.
Currently, there are insufficient data concerning their use in asthmatic patients. Nevertheless, promising effects of tyndallized Lactobacillus spp. are shown in research on other allergic diseases; thus, maybe in the future, extensive sample studies will be performed [74].

7. Conclusions

As described above, postbiotics seem to be promising therapeutic approaches in asthma treatment. They undoubtedly affect the immune system and promote various changes in cytokine production, T-cell differentiation, immunoglobulin release, and eosinophil infiltration. Lowering the number of asthma exacerbations as the effect of BLs’ immunomodulating qualities could insinuate their appropriate use in asthma treatment. However, their underlying mechanisms of action are not fully elucidated, and it is difficult to determine whether they should be applied in clinical practice. Moreover, postbiotics, excluding BLs, have not been studied in human subjects with asthma, making it challenging to determine their efficacy in treatment. Nevertheless, existing research indicates that their immunomodulatory properties could be promising. Thus, further, large-sample studies should be carried out. Health benefits, side effects, and cost-effectiveness should be examined.

Author Contributions

Conceptualization, K.W. and A.J.; Data curation, K.W. and A.J.; Data analysis, A.J.; Writing—draft preparation, K.W. and A.J.; Writing—review and editing W.F., K.W. and A.J.; Visualization W.F.; Supervision, W.F. and K.J.; Project administration, K.W. and W.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

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 conflicts of interest.

References

  1. Weinberger, M. Can we prevent exacerbations of asthma caused by common cold viruses? J. Allergy Clin. Immunol. 2010, 126, 770–771. [Google Scholar] [CrossRef] [PubMed]
  2. 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]
  3. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  4. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef] [PubMed]
  5. Wegh, C.A.M.; Geerlings, S.Y.; Knol, J.; Roeselers, G.; Belzer, C. Postbiotics and Their Potential Applications in Early Life Nutrition and Beyond. Int. J. Mol. Sci. 2019, 20, 4673. [Google Scholar] [CrossRef] [PubMed]
  6. Zolkiewicz, J.; Marzec, A.; Ruszczynski, M.; Feleszko, W. Postbiotics—A Step beyond Pre- and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef] [PubMed]
  7. Naja, A.S.; Permaul, P.; Phipatanakul, W. Taming Asthma in School-Aged Children: A Comprehensive Review. J. Allergy Clin. Immunol. Pract. 2018, 6, 726–735. [Google Scholar] [CrossRef] [PubMed]
  8. Gans, M.D.; Gavrilova, T. Understanding the immunology of asthma: Pathophysiology, biomarkers, and treatments for asthma endotypes. Paediatr. Respir. Rev. 2020, 36, 118–127. [Google Scholar] [CrossRef] [PubMed]
  9. Nobs, S.P.; Zmora, N.; Elinav, E. Nutrition Regulates Innate Immunity in Health and Disease. Annu. Rev. Nutr. 2020, 40, 189–219. [Google Scholar] [CrossRef]
  10. Bessler, W.G.; Vor dem Esche, U.; Masihi, N. The bacterial extract OM-85 BV protects mice against influenza and Salmonella infection. Int. Immunopharmacol. 2010, 10, 1086–1090. [Google Scholar] [CrossRef]
  11. Suarez, N.; Ferrara, F.; Rial, A.; Dee, V.; Chabalgoity, J.A. Bacterial Lysates as Immunotherapies for Respiratory Infections: Methods of Preparation. Front. Bioeng. Biotechnol. 2020, 8, 545. [Google Scholar] [CrossRef] [PubMed]
  12. Cazzola, M.; Anapurapu, S.; Page, C.P. Polyvalent mechanical bacterial lysate for the prevention of recurrent respiratory infections: A meta-analysis. Pulm. Pharmacol. Ther. 2012, 25, 62–68. [Google Scholar] [CrossRef] [PubMed]
  13. Pivniouk, V.; Gimenes-Junior, J.A.; Ezeh, P.; Michael, A.; Pivniouk, O.; Hahn, S.; VanLinden, S.R.; Malone, S.P.; Abidov, A.; Anderson, D.; et al. Airway administration of OM-85, a bacterial lysate, blocks experimental asthma by targeting dendritic cells and the epithelium/IL-33/ILC2 axis. J. Allergy Clin. Immunol. 2022, 149, 943–956. [Google Scholar] [CrossRef] [PubMed]
  14. Rossi, G.A.; Pohunek, P.; Feleszko, W.; Ballarini, S.; Colin, A.A. Viral infections and wheezing-asthma inception in childhood: Is there a role for immunomodulation by oral bacterial lysates? Clin. Transl. Allergy 2020, 10, 17. [Google Scholar] [CrossRef] [PubMed]
  15. Ver Heul, A.; Planer, J.; Kau, A.L. The Human Microbiota and Asthma. Clin. Rev. Allergy Immunol. 2019, 57, 350–363. [Google Scholar] [CrossRef] [PubMed]
  16. Kearney, S.C.; Dziekiewicz, M.; Feleszko, W. Immunoregulatory and immunostimulatory responses of bacterial lysates in respiratory infections and asthma. Ann. Allergy Asthma Immunol. 2015, 114, 364–369. [Google Scholar] [CrossRef] [PubMed]
  17. Kirtland, M.E.; Tsitoura, D.C.; Durham, S.R.; Shamji, M.H. Toll-Like Receptor Agonists as Adjuvants for Allergen Immunotherapy. Front. Immunol. 2020, 11, 599083. [Google Scholar] [CrossRef] [PubMed]
  18. Haapakoski, R.; Karisola, P.; Fyhrquist, N.; Savinko, T.; Lehtimaki, S.; Wolff, H.; Lauerma, A.; Alenius, H. Toll-like receptor activation during cutaneous allergen sensitization blocks development of asthma through IFN-gamma-dependent mechanisms. J. Investig. Dermatol. 2013, 133, 964–972. [Google Scholar] [CrossRef] [PubMed]
  19. Coviello, S.; Wimmenauer, V.; Polack, F.P.; Irusta, P.M. Bacterial lysates improve the protective antibody response against respiratory viruses through Toll-like receptor 4. Hum. Vaccines Immunother. 2014, 10, 2896–2902. [Google Scholar] [CrossRef]
  20. Li, Y.; Tu, C.; Chen, M.; Tan, C.; Zheng, X.; Wang, Z.; Liang, Y.; Wang, K.; Wu, J.; Li, H.; et al. Establishing a high microbial load maternal-offspring asthma model in adult mice. Int. Immunopharmacol. 2020, 83, 106453. [Google Scholar] [CrossRef]
  21. de Boer, G.M.; Braunstahl, G.J.; van der Ploeg, E.K.; van Zelst, C.M.; van Bruggen, A.; Epping, G.; van Nimwegen, M.; Verhoeven, G.; Birnie, E.; Boxma-de Klerk, B.M.; et al. Bacterial lysate add-on therapy to reduce exacerbations in severe asthma: A double-blind placebo-controlled trial. Clin. Exp. Allergy 2021, 51, 1172–1184. [Google Scholar] [CrossRef]
  22. Han, L.; Zheng, C.P.; Sun, Y.Q.; Xu, G.; Wen, W.; Fu, Q.L. A bacterial extract of OM-85 Broncho-Vaxom prevents allergic rhinitis in mice. Am. J. Rhinol. Allergy 2014, 28, 110–116. [Google Scholar] [CrossRef] [PubMed]
  23. Han, R.F.; Li, H.Y.; Wang, J.W.; Cong, X.J. Study on clinical effect and immunologic mechanism of infants capillary bronchitis secondary bronchial asthma treated with bacterial lysates Broncho-Vaxom. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2151–2155. [Google Scholar] [PubMed]
  24. Lu, Y.; Li, Y.; Xu, L.; Xia, M.; Cao, L. Bacterial lysate increases the percentage of natural killer T cells in peripheral blood and alleviates asthma in children. Pharmacology 2015, 95, 139–144. [Google Scholar] [CrossRef]
  25. Rodrigues, A.; Gualdi, L.P.; de Souza, R.G.; Vargas, M.H.; Nunez, N.K.; da Cunha, A.A.; Jones, M.H.; Pinto, L.A.; Stein, R.T.; Pitrez, P.M. Bacterial extract (OM-85) with human-equivalent doses does not inhibit the development of asthma in a murine model. Allergol. Immunopathol. 2016, 44, 504–511. [Google Scholar] [CrossRef] [PubMed]
  26. Bartkowiak-Emeryk, M.; Emeryk, A.; Rolinski, J.; Wawryk-Gawda, E.; Markut-Miotla, E. Impact of Polyvalent Mechanical Bacterial Lysate on lymphocyte number and activity in asthmatic children: A randomized controlled trial. Allergy Asthma Clin. Immunol. 2021, 17, 10. [Google Scholar] [CrossRef]
  27. Luan, H.; Zhang, Q.; Wang, L.; Wang, C.; Zhang, M.; Xu, X.; Zhou, H.; Li, X.; Xu, Q.; He, F.; et al. OM85-BV induced the productions of IL-1β, IL-6, and TNF-α via TLR4- and TLR2-mediated ERK1/2/NF-κB pathway in RAW264.7 cells. J. Interferon Cytokine Res. 2014, 34, 526–536. [Google Scholar] [CrossRef] [PubMed]
  28. Liao, J.Y.; Zhang, T. Influence of OM-85 BV on hBD-1 and immunoglobulin in children with asthma and recurrent respiratory tract infection. Zhongguo Dang Dai Er Ke Za Zhi 2014, 16, 508–512. [Google Scholar]
  29. Liu, C.; Huang, R.; Yao, R.; Yang, A. The Immunotherapeutic Role of Bacterial Lysates in a Mouse Model of Asthma. Lung 2017, 195, 563–569. [Google Scholar] [CrossRef]
  30. Kaczynska, A.; Klosinska, M.; Janeczek, K.; Zarobkiewicz, M.; Emeryk, A. Promising Immunomodulatory Effects of Bacterial Lysates in Allergic Diseases. Front. Immunol. 2022, 13, 907149. [Google Scholar] [CrossRef]
  31. Weiss, S.; Fux, T. Effect of Broncho-Vaxom on serum IgE and IgG levels in patients with bronchial asthma and chronic obstructive lung disease. A placebo-controlled double-blind study. Schweiz. Med. Wochenschr. 1987, 117, 1514–1518. [Google Scholar]
  32. Emeryk, A.; Bartkowiak-Emeryk, M.; Raus, Z.; Braido, F.; Ferlazzo, G.; Melioli, G. Mechanical bacterial lysate administration prevents exacerbation in allergic asthmatic children—The EOLIA study. Pediatr. Allergy Immunol. 2018, 29, 394–401. [Google Scholar] [CrossRef]
  33. Li, L.; Li, J.; Hu, C.; Di Nardo, M.; Srinivasan, V.; Adamko, D.J.; Sun, J.; Du, Y.; Zeng, X. Effectiveness of polyvalent bacterial lysate for pediatric asthma control: A retrospective propensity score-matched cohort study. Transl. Pediatr. 2022, 11, 1697–1703. [Google Scholar] [CrossRef]
  34. Koatz, A.M.; Coe, N.A.; Ciceran, A.; Alter, A.J. Clinical and Immunological Benefits of OM-85 Bacterial Lysate in Patients with Allergic Rhinitis, Asthma, and COPD and Recurrent Respiratory Infections. Lung 2016, 194, 687–697. [Google Scholar] [CrossRef] [PubMed]
  35. Abdou, M.A.; Hanna, K.M.; El Attar, S.; Abdel Nabi, E.; Hatem, A.; Abdel Ghaffar, M. Influence of a bacterial extract, broncho-vaxom, on clinical and immunological parameters in patients with intrinsic asthma. Int. J. Immunother. 1993, 9, 127–133. [Google Scholar]
  36. Roßberg, S.; Keller, T.; Icke, K.; Siedmann, V.; Lau, I.; Keil, T.; Lau, S. Orally applied bacterial lysate in infants at risk for atopy does not prevent atopic dermatitis, allergic rhinitis, asthma or allergic sensitization at school age: Follow-up of a randomized trial. Allergy 2020, 75, 2020–2025. [Google Scholar] [CrossRef]
  37. Yip, W.; Hughes, M.R.; Li, Y.; Cait, A.; Hirst, M.; Mohn, W.W.; McNagny, K.M. Butyrate Shapes Immune Cell Fate and Function in Allergic Asthma. Front. Immunol. 2021, 12, 628453. [Google Scholar] [CrossRef] [PubMed]
  38. Xiong, R.G.; Zhou, D.D.; Wu, S.X.; Huang, S.Y.; Saimaiti, A.; Yang, Z.J.; Shang, A.; Zhao, C.N.; Gan, R.Y.; Li, H.B. Health Benefits and Side Effects of Short-Chain Fatty Acids. Foods 2022, 11, 2863. [Google Scholar] [CrossRef] [PubMed]
  39. Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef]
  40. Liu, H.; Wang, J.; He, T.; Becker, S.; Zhang, G.; Li, D.; Ma, X. Butyrate: A Double-Edged Sword for Health? Adv. Nutr. 2018, 9, 21–29. [Google Scholar] [CrossRef]
  41. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; Gonzalez, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef]
  42. Ang, Z.; Ding, J.L. GPR41 and GPR43 in Obesity and Inflammation—Protective or Causative? Front. Immunol. 2016, 7, 28. [Google Scholar] [CrossRef] [PubMed]
  43. Sasaki, M.; Suaini, N.H.A.; Afghani, J.; Heye, K.N.; O’Mahony, L.; Venter, C.; Lauener, R.; Frei, R.; Roduit, C. Systematic review of the association between short-chain fatty acids and allergic diseases. Allergy 2024, 79, 1789–1811. [Google Scholar] [CrossRef] [PubMed]
  44. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef] [PubMed]
  45. Kaisar, M.M.M.; Pelgrom, L.R.; van der Ham, A.J.; Yazdanbakhsh, M.; Everts, B. Butyrate Conditions Human Dendritic Cells to Prime Type 1 Regulatory T Cells via both Histone Deacetylase Inhibition and G Protein-Coupled Receptor 109A Signaling. Front. Immunol. 2017, 8, 1429. [Google Scholar] [CrossRef] [PubMed]
  46. Cait, A.; Hughes, M.R.; Antignano, F.; Cait, J.; Dimitriu, P.A.; Maas, K.R.; Reynolds, L.A.; Hacker, L.; Mohr, J.; Finlay, B.B.; et al. Microbiome-driven allergic lung inflammation is ameliorated by short-chain fatty acids. Mucosal Immunol. 2018, 11, 785–795. [Google Scholar] [CrossRef] [PubMed]
  47. Lange, J.; Kozielski, J.; Bartolik, K.; Kabicz, P.; Targowski, T. The incidence of pneumonia in the paediatric population in Poland in light of the maps of health needs. J. Public Health 2023, 31, 457–465. [Google Scholar] [CrossRef]
  48. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  49. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
  50. Hu, M.; Alashkar Alhamwe, B.; Santner-Nanan, B.; Miethe, S.; Harb, H.; Renz, H.; Potaczek, D.P.; Nanan, R.K. Short-Chain Fatty Acids Augment Differentiation and Function of Human Induced Regulatory T Cells. Int. J. Mol. Sci. 2022, 23, 5740. [Google Scholar] [CrossRef]
  51. Kim, J.H.; Kim, K.; Kim, W. Gut microbiota restoration through fecal microbiota transplantation: A new atopic dermatitis therapy. Exp. Mol. Med. 2021, 53, 907–916. [Google Scholar] [CrossRef]
  52. Theiler, A.; Barnthaler, T.; Platzer, W.; Richtig, G.; Peinhaupt, M.; Rittchen, S.; Kargl, J.; Ulven, T.; Marsh, L.M.; Marsche, G.; et al. Butyrate ameliorates allergic airway inflammation by limiting eosinophil trafficking and survival. J. Allergy Clin. Immunol. 2019, 144, 764–776. [Google Scholar] [CrossRef] [PubMed]
  53. Sanchez, H.N.; Moroney, J.B.; Gan, H.; Shen, T.; Im, J.L.; Li, T.; Taylor, J.R.; Zan, H.; Casali, P. B cell-intrinsic epigenetic modulation of antibody responses by dietary fiber-derived short-chain fatty acids. Nat. Commun. 2020, 11, 60. [Google Scholar] [CrossRef] [PubMed]
  54. Shi, Y.; Xu, M.; Pan, S.; Gao, S.; Ren, J.; Bai, R.; Li, H.; He, C.; Zhao, S.; Shi, Z.; et al. Induction of the apoptosis, degranulation and IL-13 production of human basophils by butyrate and propionate via suppression of histone deacetylation. Immunology 2021, 164, 292–304. [Google Scholar] [CrossRef] [PubMed]
  55. Folkerts, J.; Redegeld, F.; Folkerts, G.; Blokhuis, B.; van den Berg, M.P.M.; de Bruijn, M.J.W.; van IJcken, W.F.J.; Junt, T.; Tam, S.Y.; Galli, S.J.; et al. Butyrate inhibits human mast cell activation via epigenetic regulation of FcεRI-mediated signaling. Allergy 2020, 75, 1966–1978. [Google Scholar] [CrossRef] [PubMed]
  56. Richards, L.B.; Li, M.; Folkerts, G.; Henricks, P.A.J.; Garssen, J.; van Esch, B. Butyrate and Propionate Restore the Cytokine and House Dust Mite Compromised Barrier Function of Human Bronchial Airway Epithelial Cells. Int. J. Mol. Sci. 2020, 22, 65. [Google Scholar] [CrossRef] [PubMed]
  57. Bottcher, M.F.; Nordin, E.K.; Sandin, A.; Midtvedt, T.; Bjorksten, B. Microflora-associated characteristics in faeces from allergic and nonallergic infants. Clin. Exp. Allergy 2000, 30, 1590–1596. [Google Scholar] [CrossRef] [PubMed]
  58. Juraskova, D.; Ribeiro, S.C.; Silva, C.C.G. Exopolysaccharides Produced by Lactic Acid Bacteria: From Biosynthesis to Health-Promoting Properties. Foods 2022, 11, 156. [Google Scholar] [CrossRef] [PubMed]
  59. Kaur, N.; Dey, P. Bacterial exopolysaccharides as emerging bioactive macromolecules: From fundamentals to applications. Res. Microbiol. 2023, 174, 104024. [Google Scholar] [CrossRef]
  60. Schiavi, E.; Plattner, S.; Rodriguez-Perez, N.; Barcik, W.; Frei, R.; Ferstl, R.; Kurnik-Lucka, M.; Groeger, D.; Grant, R.; Roper, J.; et al. Exopolysaccharide from Bifidobacterium longum subsp. longum 35624 modulates murine allergic airway responses. Benef. Microbes 2018, 9, 761–773. [Google Scholar] [CrossRef]
  61. Zhang, L.; Yi, H. An exopolysaccharide from Bacillus subtilis alleviates airway inflammatory responses via the NF-κB and STAT6 pathways in asthmatic mice. Biosci. Rep. 2022, 42, BSR20212461. [Google Scholar] [CrossRef] [PubMed]
  62. Kinoshita, H.; Hariu, M.; Nakashima, Y.; Watanabe, K.; Yasuda, S.; Igoshi, K. Lactic acid bacterial exopolysaccharides strongly bind histamine and can potentially be used to remove histamine contamination in food. Microbiology 2021, 167, mic000936. [Google Scholar] [CrossRef] [PubMed]
  63. Noda, M.; Kanno, K.; Danshiitsoodol, N.; Higashikawa, F.; Sugiyama, M. Plant-Derived Lactobacillus paracasei IJH-SONE68 Improves Chronic Allergy Status: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2021, 13, 4022. [Google Scholar] [CrossRef] [PubMed]
  64. Oberg, T.S.; McMahon, D.J.; Culumber, M.D.; McAuliffe, O.; Oberg, C.J. Invited review: Review of taxonomic changes in dairy-related lactobacilli. J. Dairy Sci. 2022, 105, 2750–2770. [Google Scholar] [CrossRef] [PubMed]
  65. Kalliomaki, M.; Salminen, S.; Arvilommi, H.; Kero, P.; Koskinen, P.; Isolauri, E. Probiotics in primary prevention of atopic disease: A randomised placebo-controlled trial. Lancet 2001, 357, 1076–1079. [Google Scholar] [CrossRef] [PubMed]
  66. Ni, Y.; Zhang, Y.; Zheng, L.; Rong, N.; Yang, Y.; Gong, P.; Yang, Y.; Siwu, X.; Zhang, C.; Zhu, L.; et al. Bifidobacterium and Lactobacillus improve inflammatory bowel disease in zebrafish of different ages by regulating the intestinal mucosal barrier and microbiota. Life Sci. 2023, 324, 121699. [Google Scholar] [CrossRef] [PubMed]
  67. Tonetti, F.R.; Tomokiyo, M.; Fukuyama, K.; Elean, M.; Moyano, R.O.; Yamamuro, H.; Shibata, R.; Quilodran-Vega, S.; Kurata, S.; Villena, J.; et al. Post-immunobiotics increase resistance to primary respiratory syncytial virus infection and secondary pneumococcal pneumonia. Benef. Microbes 2023, 14, 209–221. [Google Scholar] [CrossRef] [PubMed]
  68. Lim, L.H.; Li, H.Y.; Huang, C.H.; Lee, B.W.; Lee, Y.K.; Chua, K.Y. The effects of heat-killed wild-type Lactobacillus casei Shirota on allergic immune responses in an allergy mouse model. Int. Arch. Allergy Immunol. 2009, 148, 297–304. [Google Scholar] [CrossRef] [PubMed]
  69. Choi, C.Y.; Kim, Y.H.; Oh, S.; Lee, H.J.; Kim, J.H.; Park, S.H.; Kim, H.J.; Lee, S.J.; Chun, T. Anti-inflammatory potential of a heat-killed Lactobacillus strain isolated from Kimchi on house dust mite-induced atopic dermatitis in NC/Nga mice. J. Appl. Microbiol. 2017, 123, 535–543. [Google Scholar] [CrossRef]
  70. Hong, H.J.; Kim, E.; Cho, D.; Kim, T.S. Differential suppression of heat-killed lactobacilli isolated from kimchi, a Korean traditional food, on airway hyper-responsiveness in mice. J. Clin. Immunol. 2010, 30, 449–458. [Google Scholar] [CrossRef]
  71. Lee, Y.D.; Hong, Y.F.; Jeon, B.; Jung, B.J.; Chung, D.K.; Kim, H. Differential Cytokine Regulatory Effect of Three Lactobacillus Strains Isolated from Fermented Foods. J. Microbiol. Biotechnol. 2016, 26, 1517–1526. [Google Scholar] [CrossRef] [PubMed]
  72. Chuang, L.; Wu, K.G.; Pai, C.; Hsieh, P.S.; Tsai, J.J.; Yen, J.H.; Lin, M.Y. Heat-killed cells of lactobacilli skew the immune response toward T helper 1 polarization in mouse splenocytes and dendritic cell-treated T cells. J. Agric. Food Chem. 2007, 55, 11080–11086. [Google Scholar] [CrossRef] [PubMed]
  73. Li, A.L.; Meng, X.C.; Duan, C.C.; Huo, G.C.; Zheng, Q.L.; Li, D. Suppressive effects of oral administration of heat-killed Lactobacillus acidophilus on T helper-17 immune responses in a bovine β-lactoglobulin-sensitized mice model. Biol. Pharm. Bull. 2013, 36, 202–207. [Google Scholar] [CrossRef] [PubMed]
  74. Jeong, K.; Kim, M.; Jeon, S.A.; Kim, Y.H.; Lee, S. A randomized trial of Lactobacillus rhamnosus IDCC 3201 tyndallizate (RHT3201) for treating atopic dermatitis. Pediatr. Allergy Immunol. 2020, 31, 783–792. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Immunomodulatory properties of postbiotics. Postbiotics act as immunomodulators and promote antiallergic immune responses. They provide the maintenance of violated Th1/Th2 type immune response balance, stimulate IgA and IgG production, and provoke DC maturation. Moreover, they reduce asthma symptoms by decreasing mast cell degranulation and eosinophil count. Created with biorender.com. HKL—Heat-Killed Lactobacillus.
Figure 1. Immunomodulatory properties of postbiotics. Postbiotics act as immunomodulators and promote antiallergic immune responses. They provide the maintenance of violated Th1/Th2 type immune response balance, stimulate IgA and IgG production, and provoke DC maturation. Moreover, they reduce asthma symptoms by decreasing mast cell degranulation and eosinophil count. Created with biorender.com. HKL—Heat-Killed Lactobacillus.
Microorganisms 12 01642 g001
Figure 2. Identification of studies via databases.
Figure 2. Identification of studies via databases.
Microorganisms 12 01642 g002
Table 1. Characteristics of included studies.
Table 1. Characteristics of included studies.
Author, YearStudy DesignSubject (BLs/Control)Mean AgeTreatment RegimenClinical OutcomesImmunological and Other Outcomes
Emeryk et al.
2018
[32]
RCT150 (74/76)6–16 yearsIsmigen (PMBL) vs. Placebo The number of asthma exacerbations was lower in the PMBL group.
de Boer et al.
2021
[21]
RCT75 (38/37)16–60 yearsOM-85 (PCBL) vs. SC Exacerbations were not different between groups after 18 months.FEV1 increased in the PCBL group.
Lu et al.
2015
[28]
RCT60 (24/36)5–15 yearsOM-85 (PCBL) vs. SC Increased serum IFN-γ/IL-4 ratio was observed.
Li et al.
2022
[33]
Retrospective PS-matched cohort study795 (337/458)6 months–14 yearsQIPIAN (PMBL) vs. SCFewer exacerbations were observed in the PMBL group.
Bartkowiak-Emeryk et al.
2021
[26]
RCT49 (21/28)6–15 yearsIsmigen (PMBL) vs. placebo Increased serum T lymphocyte, CD4+ CD25+ FOXP3+, CD8+, CD3− CD16+ CD56+.
Decreased serum CD69+ and CD25+ subset of CD3+.
Koatz et al.
2016
[34]
open-label, prospective, sequential2816–65 years1st year SC;
2nd year OM-85 (PCBL)
Decreased symptom severity and the number of exacerbations.Increased serum and salivary secretory IgA.
Han et al.
2016
[23]
RCT136 (74/62)7 months–5 yearsOM-85 (PCBL) vs. inhaled corticosteroids/aminophylline/antibioticsDecreased the frequency and duration of capillary bronchitis and asthma.Decreased serum IL-4 and IL-17 levels.
Increased serum IL-10 and IFN-g levels.
Abdou et al.
1993
[35]
RCT50(25/25)Not applicableOM-85 vs. SCReduced the duration and number of asthma attacks.Increased FEV1/FVC% ratio.
Increased serum IgA, IgM, and IgG levels.
Decreased serum IgE level.
Decreased eosinophil count in bronchoalveolar fluid.
Increased IgA/albumin ratio.
RCT—randomized controlled trial; PMBL—polyvalent mechanical bacterial lysate; PCBL—polyvalent chemical bacterial lysate; SC—standard care; FEV1—forced expiratory volume during the first second of expiration.
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

Węgrzyn, K.; Jasińska, A.; Janeczek, K.; Feleszko, W. The Role of Postbiotics in Asthma Treatment. Microorganisms 2024, 12, 1642. https://doi.org/10.3390/microorganisms12081642

AMA Style

Węgrzyn K, Jasińska A, Janeczek K, Feleszko W. The Role of Postbiotics in Asthma Treatment. Microorganisms. 2024; 12(8):1642. https://doi.org/10.3390/microorganisms12081642

Chicago/Turabian Style

Węgrzyn, Konstancja, Agnieszka Jasińska, Kamil Janeczek, and Wojciech Feleszko. 2024. "The Role of Postbiotics in Asthma Treatment" Microorganisms 12, no. 8: 1642. https://doi.org/10.3390/microorganisms12081642

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop